201
|
De Toma I, Dierssen M. Network analysis of Down syndrome and SARS-CoV-2 identifies risk and protective factors for COVID-19. Sci Rep 2021; 11:1930. [PMID: 33479353 PMCID: PMC7820501 DOI: 10.1038/s41598-021-81451-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
SARS-CoV-2 infection has spread uncontrollably worldwide while it remains unknown how vulnerable populations, such as Down syndrome (DS) individuals are affected by the COVID-19 pandemic. Individuals with DS have more risk of infections with respiratory complications and present signs of auto-inflammation. They also present with multiple comorbidities that are associated with poorer COVID-19 prognosis in the general population. All this might place DS individuals at higher risk of SARS-CoV-2 infection or poorer clinical outcomes. In order to get insight into the interplay between DS genes and SARS-cov2 infection and pathogenesis we identified the genes associated with the molecular pathways involved in COVID-19 and the host proteins interacting with viral proteins from SARS-CoV-2. We then analyzed the overlaps of these genes with HSA21 genes, HSA21 interactors and other genes consistently differentially expressed in DS (using public transcriptomic datasets) and created a DS-SARS-CoV-2 network. We detected COVID-19 protective and risk factors among HSA21 genes and interactors and/or DS deregulated genes that might affect the susceptibility of individuals with DS both at the infection stage and in the progression to acute respiratory distress syndrome. Our analysis suggests that at the infection stage DS individuals might be more susceptible to infection due to triplication of TMPRSS2, that primes the viral S protein for entry in the host cells. However, as the anti-viral interferon I signaling is also upregulated in DS, this might increase the initial anti-viral response, inhibiting viral genome release, viral replication and viral assembly. In the second pro-inflammatory immunopathogenic phase of the infection, the prognosis for DS patients might worsen due to upregulation of inflammatory genes that might favor the typical cytokine storm of COVID-19. We also detected strong downregulation of the NLRP3 gene, critical for maintenance of homeostasis against pathogenic infections, possibly leading to bacterial infection complications.
Collapse
Affiliation(s)
- Ilario De Toma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Biomedical Research Networking Center On Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
202
|
Name JJ, Vasconcelos AR, Souza ACR, Fávaro WJ. Vitamin D, zinc and glutamine: Synergistic action with OncoTherad immunomodulator in interferon signaling and COVID‑19 (Review). Int J Mol Med 2021; 47:11. [PMID: 33448317 PMCID: PMC7834962 DOI: 10.3892/ijmm.2021.4844] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was identified in December, 2019 in Wuhan, China. Since then, it has continued to spread rapidly in numerous countries, while the search for effective therapeutic options persists. Coronaviruses, including SARS-CoV-2, are known to suppress and evade the antiviral responses of the host organism mediated by interferon (IFN), a family of cytokines that plays an important role in antiviral defenses associated with innate immunity, and has been used therapeutically for chronic viral diseases and cancer. On the other hand, OncoTherad, a safe and effective immunotherapeutic agent in the treatment of non-muscle invasive bladder cancer (NMIBC), increases IFN signaling and has been shown to be a promising therapeutic approach for COVID-19 in a case report that described the rapid recovery of a 78-year-old patient with NMIBC with comorbidities. The present review discusses the possible synergistic action of OncoTherad with vitamin D, zinc and glutamine, nutrients that have been shown to facilitate immune responses mediated by IFN signaling, as well as the potential of this combination as a therapeutic option for COVID-19.
Collapse
Affiliation(s)
- José João Name
- Kilyos Assessoria, Cursos e Palestras (Kilyos Nutrition), São Paulo, SP 01311‑100, Brazil
| | - Andrea Rodrigues Vasconcelos
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP 05508‑000, Brazil
| | | | - Wagner José Fávaro
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, University of Campinas, Campinas, SP 13083‑970, Brazil
| |
Collapse
|
203
|
Duncan CJA, Randall RE, Hambleton S. Genetic Lesions of Type I Interferon Signalling in Human Antiviral Immunity. Trends Genet 2021; 37:46-58. [PMID: 32977999 PMCID: PMC7508017 DOI: 10.1016/j.tig.2020.08.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/08/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022]
Abstract
The concept that type I interferons (IFN-I) are essential to antiviral immunity derives from studies on animal models and cell lines. Virtually all pathogenic viruses have evolved countermeasures to IFN-I restriction, and genetic loss of viral IFN-I antagonists leads to virus attenuation. But just how important is IFN-I to antiviral defence in humans? The recent discovery of genetic defects of IFN-I signalling illuminates this and other questions of IFN biology, including the role of the mucosa-restricted type III IFNs (IFN-III), informing our understanding of the place of the IFN system within the concerted antiviral response. Here we review monogenic lesions of IFN-I signalling pathways and summarise the organising principles which emerge.
Collapse
Affiliation(s)
- Christopher J A Duncan
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK.
| | - Richard E Randall
- School of Biology, University of St Andrew's, St Andrew's KY16 9ST, UK
| | - Sophie Hambleton
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| |
Collapse
|
204
|
Wang Y, Zhu J, Cao Y, Shen J, Yu L. Insight Into Inflammasome Signaling: Implications for Toxoplasma gondii Infection. Front Immunol 2020; 11:583193. [PMID: 33391259 PMCID: PMC7772217 DOI: 10.3389/fimmu.2020.583193] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022] Open
Abstract
Inflammasomes are multimeric protein complexes regulating the innate immune response to invading pathogens or stress stimuli. Recent studies have reported that nucleotide-binding leucine-rich repeat-containing (NLRs) proteins and DNA sensor absent in melanoma 2 (AIM2) serve as inflammasome sentinels, whose stimulation leads to the proteolytic activation of caspase-1, proinflammatory cytokine secretion, and pyroptotic cell death. Toxoplasma gondii, an obligate intracellular parasite of phylum Apicomplexans, is reportedly involved in NLRP1, NLRP3 and AIM2 inflammasomes activation; however, mechanistic evidence regarding the activation of these complexes is preliminary. This review describes the current understanding of inflammasome signaling in rodent and human models of T. gondii infection.
Collapse
Affiliation(s)
- Yang Wang
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jinjin Zhu
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yuanyuan Cao
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jilong Shen
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Li Yu
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
205
|
Malhotra S, Costa C, Eixarch H, Keller CW, Amman L, Martínez-Banaclocha H, Midaglia L, Sarró E, Machín-Díaz I, Villar LM, Triviño JC, Oliver-Martos B, Parladé LN, Calvo-Barreiro L, Matesanz F, Vandenbroeck K, Urcelay E, Martínez-Ginés ML, Tejeda-Velarde A, Fissolo N, Castilló J, Sanchez A, Robertson AAB, Clemente D, Prinz M, Pelegrin P, Lünemann JD, Espejo C, Montalban X, Comabella M. NLRP3 inflammasome as prognostic factor and therapeutic target in primary progressive multiple sclerosis patients. Brain 2020; 143:1414-1430. [PMID: 32282893 DOI: 10.1093/brain/awaa084] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/13/2019] [Accepted: 02/05/2020] [Indexed: 01/24/2023] Open
Abstract
Primary progressive multiple sclerosis is a poorly understood disease entity with no specific prognostic biomarkers and scarce therapeutic options. We aimed to identify disease activity biomarkers in multiple sclerosis by performing an RNA sequencing approach in peripheral blood mononuclear cells from a discovery cohort of 44 untreated patients with multiple sclerosis belonging to different clinical forms and activity phases of the disease, and 12 healthy control subjects. A validation cohort of 58 patients with multiple sclerosis and 26 healthy control subjects was included in the study to replicate the RNA sequencing findings. The RNA sequencing revealed an interleukin 1 beta (IL1B) signature in patients with primary progressive multiple sclerosis. Subsequent immunophenotyping pointed to blood monocytes as responsible for the IL1B signature observed in this group of patients. Functional experiments at baseline measuring apoptosis-associated speck-like protein containing a CARD (ASC) speck formation showed that the NOD-leucine rich repeat and pyrin containing protein 3 (NLRP3) inflammasome was overactive in monocytes from patients with primary progressive multiple sclerosis, and canonical NLRP3 inflammasome activation with a combination of ATP plus lipopolysaccharide was associated with increased IL1B production in this group of patients. Primary progressive multiple sclerosis patients with high IL1B gene expression levels in peripheral blood mononuclear cells progressed significantly faster compared to patients with low IL1B levels based on the time to reach an EDSS of 6.0 and the Multiple Sclerosis Severity Score. In agreement with peripheral blood findings, both NLRP3 and IL1B expression in brain tissue from patients with primary progressive multiple sclerosis was mainly restricted to cells of myeloid lineage. Treatment of mice with a specific NLRP3 inflammasome inhibitor attenuated established experimental autoimmune encephalomyelitis disease severity and improved CNS histopathology. NLRP3 inflammasome-specific inhibition was also effective in reducing axonal damage in a model of lipopolysaccharide-neuroinflammation using organotypic cerebellar cultures. Altogether, these results point to a role of IL1B and the NLRP3 inflammasome as prognostic biomarker and potential therapeutic target, respectively, in patients with primary progressive multiple sclerosis.
Collapse
Affiliation(s)
- Sunny Malhotra
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carme Costa
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Herena Eixarch
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Christian W Keller
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.,Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Lukas Amman
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Helios Martínez-Banaclocha
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Luciana Midaglia
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eduard Sarró
- Renal Physiopathology Group, Institut de Recerca Vall d'Hebron (VHIR) - CIBBIM Nanomedicine, Barcelona, Spain
| | - Isabel Machín-Díaz
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain
| | - Luisa M Villar
- Departments of Immunology and Neurology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid, Spain
| | | | - Begoña Oliver-Martos
- Neuroimmunology and Neuroinflammation Group, Instituto de Investigación Biomédica de Málaga-IBIMA, UGC Neurociencias, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Laura Navarro Parladé
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Calvo-Barreiro
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Fuencisla Matesanz
- Department of Cell Biology and Immunology, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Granada, Spain
| | - Koen Vandenbroeck
- Universidad del País Vasco (UPV/EHU), Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Elena Urcelay
- Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | | | - Amalia Tejeda-Velarde
- Departments of Immunology and Neurology, Multiple Sclerosis Unit, Hospital Ramon y Cajal, (IRYCIS), Madrid, Spain
| | - Nicolás Fissolo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joaquín Castilló
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Sanchez
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain.,Statistics and Bioinformatics Unit, Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Avril A B Robertson
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Diego Clemente
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center for NeuroModulation (NeuroModul), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pablo Pelegrin
- Biomedical Research Institute of Murcia (IMIB-Arrixaca), University Clinical Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Jan D Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany.,Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Carmen Espejo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center for Multiple Sclerosis, St, Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d'Hebron (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
206
|
Akhade AS, Atif SM, Lakshmi BS, Dikshit N, Hughes KT, Qadri A, Subramanian N. Type 1 interferon-dependent repression of NLRC4 and iPLA2 licenses down-regulation of Salmonella flagellin inside macrophages. Proc Natl Acad Sci U S A 2020; 117:29811-29822. [PMID: 33177235 PMCID: PMC7703570 DOI: 10.1073/pnas.2002747117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Inflammasomes have been implicated in the detection and clearance of a variety of bacterial pathogens, but little is known about whether this innate sensing mechanism has any regulatory effect on the expression of stimulatory ligands by the pathogen. During infection with Salmonella and many other pathogens, flagellin is a major activator of NLRC4 inflammasome-mediated macrophage pyroptosis and pathogen eradication. Salmonella switches to a flagellin-low phenotype as infection progresses to avoid this mechanism of clearance by the host. However, the host cues that Salmonella perceives to undergo this switch remain unclear. Here, we report an unexpected role of the NLRC4 inflammasome in promoting expression of its microbial ligand, flagellin, and identify a role for type 1 IFN signaling in switching of Salmonella to a flagellin-low phenotype. Early in infection, activation of NLRC4 by flagellin initiates pyroptosis and concomitant release of lysophospholipids which in turn enhance expression of flagellin by Salmonella thereby amplifying its ability to elicit cell death. TRIF-dependent production of type 1 IFN, however, later represses NLRC4 and the lysophospholipid biosynthetic enzyme iPLA2, causing a decline in intracellular lysophospholipids that results in down-regulation of flagellin expression by Salmonella These findings reveal a previously unrecognized immune-modulating regulatory cross-talk between endosomal TLR signaling and cytosolic NLR activation with significant implications for the establishment of infection with Salmonella.
Collapse
Affiliation(s)
| | - Shaikh M Atif
- Hybridoma Laboratory, National Institute of Immunology, 110067 New Delhi, India
| | | | - Neha Dikshit
- Hybridoma Laboratory, National Institute of Immunology, 110067 New Delhi, India
| | - Kelly T Hughes
- Department of Biology, University of Utah, Salt Lake City, UT 84112
| | - Ayub Qadri
- Hybridoma Laboratory, National Institute of Immunology, 110067 New Delhi, India;
| | - Naeha Subramanian
- Institute for Systems Biology, Seattle, WA 98109;
- Department of Immunology, University of Washington, Seattle, WA 98109
- Department of Global Health, University of Washington, Seattle, WA 98109
| |
Collapse
|
207
|
|
208
|
Multiple Roles of 25-Hydroxycholesterol in Lipid Metabolism, Antivirus Process, Inflammatory Response, and Cell Survival. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8893305. [PMID: 33274010 PMCID: PMC7695496 DOI: 10.1155/2020/8893305] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
As an essential lipid, cholesterol is of great value in keeping cell homeostasis, being the precursor of bile acid and steroid hormones, and stabilizing membrane lipid rafts. As a kind of cholesterol metabolite produced by enzymatic or radical process, oxysterols have drawn much attention in the last decades. Among which, the role of 25-hydroxycholesterol (25-HC) in cholesterol and bile acid metabolism, antivirus process, and inflammatory response has been largely disclosed. This review is aimed at revealing these functions and underlying mechanisms of 25-HC.
Collapse
|
209
|
Hong SM, Lee J, Jang SG, Lee J, Cho ML, Kwok SK, Park SH. Type I Interferon Increases Inflammasomes Associated Pyroptosis in the Salivary Glands of Patients with Primary Sjögren's Syndrome. Immune Netw 2020; 20:e39. [PMID: 33163247 PMCID: PMC7609163 DOI: 10.4110/in.2020.20.e39] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/11/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
Sjögren's syndrome (SS) is a chronic and systemic autoimmune disease characterized by lymphocytic infiltration in the exocrine glands. In SS, type I IFN has a pathogenic role, and recently, inflammasome activation has been observed in both immune and non-immune cells. However, the relationship between type I IFN and inflammasome-associated pyroptosis in SS has not been studied. We measured IL-18, caspase-1, and IFN-stimulated gene 15 (ISG15) in saliva and serum, and compared whether the expression levels of inflammasome and pyroptosis components, including absent in melanoma 2 (AIM2), NLR family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein (ASC), caspase-1, gasdermin D (GSDMD), and gasdermin E (GSDME), in minor salivary gland (MSG) are related to the expression levels of type I IFN signature genes. Expression of type I IFN signature genes was correlated with mRNA levels of caspase-1 and GSDMD in MSG. In confocal analysis, the expression of caspase-1 and GSDMD was higher in salivary gland epithelial cells (SGECs) from SS patients. In the type I IFN-treated human salivary gland epithelial cell line, the expression of caspase-1 and GSDMD was increased, and pyroptosis was accelerated in a caspase-dependent manner upon inflammasome activation. In conclusion, we demonstrate that type I IFN may contribute to inflammasome-associated pyroptosis of the SGECs of SS patients, suggesting another pathogenic role of type I IFN in SS in terms of target tissue -SGECs destruction.
Collapse
Affiliation(s)
- Seung-Min Hong
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jaeseon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Se Gwang Jang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jennifer Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Ki Kwok
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Hwan Park
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
210
|
Nunn AVW, Guy GW, Brysch W, Botchway SW, Frasch W, Calabrese EJ, Bell JD. SARS-CoV-2 and mitochondrial health: implications of lifestyle and ageing. Immun Ageing 2020; 17:33. [PMID: 33292333 PMCID: PMC7649575 DOI: 10.1186/s12979-020-00204-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022]
Abstract
Infection with SARs-COV-2 displays increasing fatality with age and underlying co-morbidity, in particular, with markers of the metabolic syndrome and diabetes, which seems to be associated with a "cytokine storm" and an altered immune response. This suggests that a key contributory factor could be immunosenescence that is both age-related and lifestyle-induced. As the immune system itself is heavily reliant on mitochondrial function, then maintaining a healthy mitochondrial system may play a key role in resisting the virus, both directly, and indirectly by ensuring a good vaccine response. Furthermore, as viruses in general, and quite possibly this new virus, have also evolved to modulate immunometabolism and thus mitochondrial function to ensure their replication, this could further stress cellular bioenergetics. Unlike most sedentary modern humans, one of the natural hosts for the virus, the bat, has to "exercise" regularly to find food, which continually provides a powerful adaptive stimulus to maintain functional muscle and mitochondria. In effect the bat is exposed to regular hormetic stimuli, which could provide clues on how to resist this virus. In this paper we review the data that might support the idea that mitochondrial health, induced by a healthy lifestyle, could be a key factor in resisting the virus, and for those people who are perhaps not in optimal health, treatments that could support mitochondrial function might be pivotal to their long-term recovery.
Collapse
Affiliation(s)
- Alistair V W Nunn
- Department of Life Sciences, Research Centre for Optimal Health, University of Westminster, London, W1W 6UW, UK.
| | | | | | - Stanley W Botchway
- UKRI, STFC, Central Laser Facility, & Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX110QX, UK
| | - Wayne Frasch
- School of Life Sciences, Arizona State University, Tempe, USA
| | - Edward J Calabrese
- Environmental Health Sciences Division, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Jimmy D Bell
- Department of Life Sciences, Research Centre for Optimal Health, University of Westminster, London, W1W 6UW, UK
| |
Collapse
|
211
|
Alnefaie A, Albogami S. Current approaches used in treating COVID-19 from a molecular mechanisms and immune response perspective. Saudi Pharm J 2020; 28:1333-1352. [PMID: 32905015 PMCID: PMC7462599 DOI: 10.1016/j.jsps.2020.08.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), which is caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was declared by the World Health Organization (WHO) as a global pandemic on March 11, 2020. SARS-CoV-2 targets the respiratory system, resulting in symptoms such as fever, headache, dry cough, dyspnea, and dizziness. These symptoms vary from person to person, ranging from mild to hypoxia with acute respiratory distress syndrome (ARDS) and sometimes death. Although not confirmed, phylogenetic analysis suggests that SARS-CoV-2 may have originated from bats; the intermediary facilitating its transfer from bats to humans is unknown. Owing to the rapid spread of infection and high number of deaths caused by SARS-CoV-2, most countries have enacted strict curfews and the practice of social distancing while awaiting the availability of effective U.S. Food and Drug Administration (FDA)-approved medications and/or vaccines. This review offers an overview of the various types of coronaviruses (CoVs), their targeted hosts and cellular receptors, a timeline of their emergence, and the roles of key elements of the immune system in fighting pathogen attacks, while focusing on SARS-CoV-2 and its genomic structure and pathogenesis. Furthermore, we review drugs targeting COVID-19 that are under investigation and in clinical trials, in addition to progress using mesenchymal stem cells to treat COVID-19. We conclude by reviewing the latest updates on COVID-19 vaccine development. Understanding the molecular mechanisms of how SARS-CoV-2 interacts with host cells and stimulates the immune response is extremely important, especially as scientists look for new strategies to guide their development of specific COVID-19 therapies and vaccines.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- AHFS, American Hospital Formula Service
- ANGII, angiotensin II
- APCs, antigen presenting cells
- ARDS, acute respiratory distress syndrome
- COVID-19, coronavirus disease
- CoVs, coronaviruses
- Coronavirus
- GVHD, graft versus host disease
- HCoVs, human coronoaviruses
- IBV, infectious bronchitis coronavirus
- IFN-γ, interferon-gamma
- ILCs, innate lymphoid cells
- Investigational medications
- MERS-CoV, Middle East respiratory syndrome
- NKs, natural killer cells
- ORFs, open reading frames
- PAMPs, pathogen-associated molecular patterns
- Pandemic
- Pathophysiology
- RdRp, RNA-dependent RNA polymerase
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SLE, systemic lupus erythematosus
- TMPRSS2, transmembrane serine protease 2
- Viral immune response
- WHO, World Health Organization
- nsps, nonstructural proteins
Collapse
Affiliation(s)
- Alaa Alnefaie
- Department of Biotechnology, Faculty of Science, Taif University, Taif, Saudi Arabia
| | - Sarah Albogami
- Department of Biotechnology, Faculty of Science, Taif University, Taif, Saudi Arabia
| |
Collapse
|
212
|
Di Stadio A, Romani L, Bernitsas E. Could Sars-Cov2 affect MS progression? Mult Scler Relat Disord 2020; 46:102540. [PMID: 33032060 PMCID: PMC7524432 DOI: 10.1016/j.msard.2020.102540] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/21/2022]
Abstract
A long-term neurologic sequela arising from COVID-19 infection in multiple sclerosis (MS) patients could be related both to the increase of cytokines and the activation of NLRP3 inflammasome by the Sars-CoV2. These two mechanisms may cause a worsening of MS several months after the resolution of the infection.
Collapse
Affiliation(s)
- Arianna Di Stadio
- Otolaryngology Department, University of Perugia, Perugia, Italy; Neuroinflammation Lab, Queen Square UCL Neurology, London, UK.
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | |
Collapse
|
213
|
Molla MD, Akalu Y, Geto Z, Dagnew B, Ayelign B, Shibabaw T. Role of Caspase-1 in the Pathogenesis of Inflammatory-Associated Chronic Noncommunicable Diseases. J Inflamm Res 2020; 13:749-764. [PMID: 33116753 PMCID: PMC7585796 DOI: 10.2147/jir.s277457] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
Caspase-1 is the first and extensively studied inflammatory caspase that is activated through inflammasome assembly. Inflammasome is a cytosolic formation of multiprotein complex that aimed to start inflammatory response against infections or cellular damages. The process leads to an auto-activation of caspase-1 and consequent maturation of caspase-1 target molecules such as interleukin (IL)-1β and IL-18. Recently, the role of caspase-1 and inflammasome in inflammatory-induced noncommunicable diseases (NCDs) like obesity, diabetes mellitus (DM), cardiovascular diseases (CVDs), cancers and chronic respiratory diseases have widely studied. However, their reports are distinct and even they have reported contrasting role of caspase-1 in the development and progression of NCDs. A few studies have reported that caspase-1/inflammasome assembley has a protective role in the initiation and progression of these diseases through the activation of the noncanonical caspase-1 target substrates like gasdermin-D and regulation of immune cells. Conversely, others have revealed that caspase-1 has a direct/indirect effect in the development and progression of several NCDs. Therefore, in this review, we systematically summarized the role of caspase-1 in the development and progression of NCDs, especially in obesity, DM, CVDs and cancers.
Collapse
Affiliation(s)
- Meseret Derbew Molla
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Yonas Akalu
- Department of Human Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Zeleke Geto
- Department of Biomedical Sciences, School of Medicine, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Baye Dagnew
- Department of Human Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tewodros Shibabaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
214
|
Huerga Encabo H, Traveset L, Argilaguet J, Angulo A, Nistal-Villán E, Jaiswal R, Escalante CR, Gekas C, Meyerhans A, Aramburu J, López-Rodríguez C. The transcription factor NFAT5 limits infection-induced type I interferon responses. J Exp Med 2020; 217:132619. [PMID: 31816635 PMCID: PMC7062515 DOI: 10.1084/jem.20190449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 09/23/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022] Open
Abstract
Huerga Encabo et al. show that NFAT5, previously characterized as a pro-inflammatory transcription factor, limits the IFN-I response to control antiviral defenses and preserve HSC quiescence. NFAT5 represses IFN-I and ISG expression through an evolutionarily conserved DNA element that prevents IRF3 recruitment to the IFNB1 enhanceosome. Type I interferon (IFN-I) provides effective antiviral immunity but can exacerbate harmful inflammatory reactions and cause hematopoietic stem cell (HSC) exhaustion; therefore, IFN-I expression must be tightly controlled. While signaling mechanisms that limit IFN-I induction and function have been extensively studied, less is known about transcriptional repressors acting directly on IFN-I regulatory regions. We show that NFAT5, an activator of macrophage pro-inflammatory responses, represses Toll-like receptor 3 and virus-induced expression of IFN-I in macrophages and dendritic cells. Mice lacking NFAT5 exhibit increased IFN-I production and better control of viral burden upon LCMV infection but show exacerbated HSC activation under systemic poly(I:C)-induced inflammation. We identify IFNβ as a primary target repressed by NFAT5, which opposes the master IFN-I inducer IRF3 by binding to an evolutionarily conserved sequence in the IFNB1 enhanceosome that overlaps a key IRF site. These findings illustrate how IFN-I responses are balanced by simultaneously opposing transcription factors.
Collapse
Affiliation(s)
- Hector Huerga Encabo
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laia Traveset
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Jordi Argilaguet
- Infection Biology Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ana Angulo
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Estanislao Nistal-Villán
- Microbiology Section, Departamento de Ciencias, Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, CEU Universities, Madrid, Spain
| | - Rahul Jaiswal
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Carlos R Escalante
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Christos Gekas
- Program in Cancer Research, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Andreas Meyerhans
- Infection Biology Laboratory, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Jose Aramburu
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Cristina López-Rodríguez
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
215
|
Transcriptional Regulation of Inflammasomes. Int J Mol Sci 2020; 21:ijms21218087. [PMID: 33138274 PMCID: PMC7663688 DOI: 10.3390/ijms21218087] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Inflammasomes are multimolecular complexes with potent inflammatory activity. As such, their activity is tightly regulated at the transcriptional and post-transcriptional levels. In this review, we present the transcriptional regulation of inflammasome genes from sensors (e.g., NLRP3) to substrates (e.g., IL-1β). Lineage-determining transcription factors shape inflammasome responses in different cell types with profound consequences on the responsiveness to inflammasome-activating stimuli. Pro-inflammatory signals (sterile or microbial) have a key transcriptional impact on inflammasome genes, which is largely mediated by NF-κB and that translates into higher antimicrobial immune responses. Furthermore, diverse intrinsic (e.g., circadian clock, metabolites) or extrinsic (e.g., xenobiotics) signals are integrated by signal-dependent transcription factors and chromatin structure changes to modulate transcriptionally inflammasome responses. Finally, anti-inflammatory signals (e.g., IL-10) counterbalance inflammasome genes induction to limit deleterious inflammation. Transcriptional regulations thus appear as the first line of inflammasome regulation to raise the defense level in front of stress and infections but also to limit excessive or chronic inflammation.
Collapse
|
216
|
Souza De Lima D, Bomfim CCB, Leal VNC, Reis EC, Soares JLS, Fernandes FP, Amaral EP, Loures FV, Ogusku MM, Lima MRD, Sadahiro A, Pontillo A. Combining Host Genetics and Functional Analysis to Depict Inflammasome Contribution in Tuberculosis Susceptibility and Outcome in Endemic Areas. Front Immunol 2020; 11:550624. [PMID: 33193317 PMCID: PMC7609898 DOI: 10.3389/fimmu.2020.550624] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 08/25/2020] [Indexed: 12/31/2022] Open
Abstract
The interplay between M. tuberculosis (Mtb) and humans is multifactorial. The susceptibility/resistance profile and the establishment of clinical tuberculosis (TB) still remains elusive. The gain-of-function variant rs10754558 in the NLRP3 gene (found in 30% of the world population) confers protection against the development of TB, indicating a prominent role played by NLRP3 inflammasome against Mtb. Through genotype-guided assays and various Mtb strains (BCG, H37Rv, Beijing-1471, MP287/03), we demonstrate that Mtb strains activate inflammasome according to the NLRP3/IL-1ß or NLRC4/IL18 preferential axis. NLRP3 and NLRC4 genetic variants contribute to the presentation of TB. For the first time, we have shown that loss-of-function variants in NLRC4 significantly contribute to the development of extra-pulmonary TB. The analysis of inflammasome activation in a cohort of TB patients and their “household contacts” (CNT) revealed that plasma IL-1ß/IFN-α ratio lets us distinguish patients from Mtb-exposed-but-healthy individuals from an endemic region. Moreover, NLRP3 inflammasome seemed “exhausted” in TB patients compared to CNT, indicating a more efficient activation of inflammasome in resistant individuals. These findings suggest that inflammasome genetics as well as virulence-dependent level of inflammasome activation contribute to the onset of a susceptible/resistant profile among Mtb-exposed individuals.
Collapse
Affiliation(s)
- Dhêmerson Souza De Lima
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Caio C B Bomfim
- Laboratório de Imunologia das Doenças Infecciosas, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Vinícius N C Leal
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Edione C Reis
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Jaíne L S Soares
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Fernanda P Fernandes
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Eduardo P Amaral
- Laboratório de Imunologia das Doenças Infecciosas, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Flavio V Loures
- Instituto de Ciência e Tecnologia, Universidade Federal de São Paulo, São José dos Campos, Brazil
| | - Mauricio M Ogusku
- Laboratório de Micobacteriologia, Instituto Nacional de Pesquisas da Amazônia, Manaus, Brazil
| | - Maria R D'Imperio Lima
- Laboratório de Imunologia das Doenças Infecciosas, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Aya Sadahiro
- Departamento de Parasitologia, Universidade Federal do Amazonas, Manaus, Brazil
| | - Alessandra Pontillo
- Laboratório de Imunogenética, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
217
|
Sakaguchi N, Sasai M, Bando H, Lee Y, Pradipta A, Ma JS, Yamamoto M. Role of Gate-16 and Gabarap in Prevention of Caspase-11-Dependent Excess Inflammation and Lethal Endotoxic Shock. Front Immunol 2020; 11:561948. [PMID: 33042141 PMCID: PMC7522336 DOI: 10.3389/fimmu.2020.561948] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/18/2020] [Indexed: 12/03/2022] Open
Abstract
Sepsis is a life-threating multi-organ disease induced by host innate immunity to pathogen-derived endotoxins including lipopolysaccharide (LPS). Direct sensing of LPS by caspase-11 activates inflammasomes and causes lethal sepsis in mice. Inhibition of caspase-11 inflammasomes is important for the prevention of LPS-induced septic shock; however, whether a caspase-11 inflammasome-specific suppressive mechanism exists is unclear. Here we show that deficiency of GABARAP autophagy-related proteins results in over-activation of caspase-11 inflammasomes but not of canonical inflammasomes. Gate-16−/−Gabarap−/− macrophages exhibited elevated guanylate binding protein 2 (GBP2)-dependent caspase-11 activation and inflammatory responses. Deficiency of GABARAPs resulted in formation of GBP2-containing aggregates that promote IL-1β production. High mortality after low dose LPS challenge in Gate-16−/−Gabarap−/− mice primed with poly(I:C) or polymicrobial sepsis was ameliorated by compound GBP2 deficiency. These results reveal a critical function of Gate-16 and Gabarap to suppress GBP2-dependent caspase-11-induced inflammation and septic shock.
Collapse
Affiliation(s)
- Naoya Sakaguchi
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hironori Bando
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Youngae Lee
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Ariel Pradipta
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ji Su Ma
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
218
|
Mathias LM, Stohl W. Systemic lupus erythematosus (SLE): emerging therapeutic targets. Expert Opin Ther Targets 2020; 24:1283-1302. [PMID: 33034541 DOI: 10.1080/14728222.2020.1832464] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a heterogeneous clinical presentation whose etiologies are multifactorial. A myriad of genetic, hormonal, immunologic, and environmental factors contribute to its pathogenesis, and its diverse biological basis and phenotypic presentations make development of therapeutics difficult. In the past decade, tens of therapeutic targets with hundreds of individual candidate therapeutics have been investigated. AREAS COVERED We used a PUBMED database search through April 2020 to review the relevant literature. This review discusses therapeutic targets in the adaptive and innate immune systems, specifically: B cell surface antigens, B cell survival factors, Bruton's tyrosine kinase, costimulators, IL-12/IL-23, the calcineurin pathway, the JAK/STAT pathway, and interferons. EXPERT OPINION Our ever-improving understanding of SLE pathophysiology in the past decade has allowed us to identify new therapeutic targets. Multiple new drugs are on the horizon that target different elements of the adaptive and innate immune systems. SLE research remains challenging due to the heterogenous clinical presentation of SLE, confounding from background immunosuppressives being taken by SLE patients, animal models that inadequately recapitulate human disease, and imperfect and complicated outcome measures. Despite these limitations, research is promising and ongoing. The search for new therapies that target specific elements of SLE pathophysiology are discussed as well as key findings, pitfalls, and questions surrounding these targets.
Collapse
Affiliation(s)
- Lauren M Mathias
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine , Los Angeles, CA, USA
| | - William Stohl
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine , Los Angeles, CA, USA
| |
Collapse
|
219
|
Robichon K, Maiwald T, Schilling M, Schneider A, Willemsen J, Salopiata F, Teusel M, Kreutz C, Ehlting C, Huang J, Chakraborty S, Huang X, Damm G, Seehofer D, Lang PA, Bode JG, Binder M, Bartenschlager R, Timmer J, Klingmüller U. Identification of Interleukin1β as an Amplifier of Interferon alpha-induced Antiviral Responses. PLoS Pathog 2020; 16:e1008461. [PMID: 33002089 PMCID: PMC7553310 DOI: 10.1371/journal.ppat.1008461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 10/13/2020] [Accepted: 08/20/2020] [Indexed: 12/24/2022] Open
Abstract
The induction of an interferon-mediated response is the first line of defense against pathogens such as viruses. Yet, the dynamics and extent of interferon alpha (IFNα)-induced antiviral genes vary remarkably and comprise three expression clusters: early, intermediate and late. By mathematical modeling based on time-resolved quantitative data, we identified mRNA stability as well as a negative regulatory loop as key mechanisms endogenously controlling the expression dynamics of IFNα-induced antiviral genes in hepatocytes. Guided by the mathematical model, we uncovered that this regulatory loop is mediated by the transcription factor IRF2 and showed that knock-down of IRF2 results in enhanced expression of early, intermediate and late IFNα-induced antiviral genes. Co-stimulation experiments with different pro-inflammatory cytokines revealed that this amplified expression dynamics of the early, intermediate and late IFNα-induced antiviral genes can also be achieved by co-application of IFNα and interleukin1 beta (IL1β). Consistently, we found that IL1β enhances IFNα-mediated repression of viral replication. Conversely, we observed that in IL1β receptor knock-out mice replication of viruses sensitive to IFNα is increased. Thus, IL1β is capable to potentiate IFNα-induced antiviral responses and could be exploited to improve antiviral therapies. Innate immune responses contribute to the control of viral infections and the induction of interferon alpha (IFNα)-mediated antiviral responses is an important component. However, IFNα induces a multitude of antiviral response genes and the expression dynamics of these genes can be classified as early, intermediate and late. Here we show, based on a mathematical modeling approach, that mRNA stability as well as the negative regulator IRF2 control the expression dynamics of IFNα-induced antiviral genes. Knock-down of IRF2 resulted in the amplified IFNα-mediated induction of the antiviral genes and this amplified expression of antiviral genes could be functionally mimicked by co-stimulation with IFNα and IL1β. We observed that co-stimulation with IFNα and IL1β enhanced the repression of virus replication and that knock-out of the IL1 receptor in mice resulted in increased replication of a virus sensitive to IFNα. In sum, our studies identified IL1β as an important amplifier of IFNα-induced antiviral responses.
Collapse
Affiliation(s)
- Katharina Robichon
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Maiwald
- Institute for Physics, University of Freiburg, Germany.,FDM-Freiburg Center for Data Analysis and Modeling, University of Freiburg, Freiburg, Germany
| | - Marcel Schilling
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Annette Schneider
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Joschka Willemsen
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Florian Salopiata
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melissa Teusel
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clemens Kreutz
- Institute for Physics, University of Freiburg, Germany.,Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Christian Ehlting
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Germany
| | - Jun Huang
- Department of Molecular Medicine II, University Hospital, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Germany
| | - Sajib Chakraborty
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Xiaoyun Huang
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, Leipzig, Germany and Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, Berlin, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University of Leipzig, Leipzig, Germany and Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, Berlin, Germany
| | - Philipp A Lang
- Department of Molecular Medicine II, University Hospital, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Germany
| | - Johannes G Bode
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Medical Faculty, Heinrich-Heine-University of Düsseldorf, Germany
| | - Marco Binder
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Jens Timmer
- Institute for Physics, University of Freiburg, Germany.,FDM-Freiburg Center for Data Analysis and Modeling, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Ursula Klingmüller
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
220
|
Alvarenga L, Cardozo LF, Borges NA, Lindholm B, Stenvinkel P, Shiels PG, Fouque D, Mafra D. Can nutritional interventions modulate the activation of the NLRP3 inflammasome in chronic kidney disease? Food Res Int 2020; 136:109306. [DOI: 10.1016/j.foodres.2020.109306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/28/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
|
221
|
Islam MT, Bardaweel SK, Mubarak MS, Koch W, Gaweł-Beben K, Antosiewicz B, Sharifi-Rad J. Immunomodulatory Effects of Diterpenes and Their Derivatives Through NLRP3 Inflammasome Pathway: A Review. Front Immunol 2020; 11:572136. [PMID: 33101293 PMCID: PMC7546345 DOI: 10.3389/fimmu.2020.572136] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
Nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing protein (NLRP) inflammasomes are involved in the molecular pathogenesis of many diseases and disorders. Among NLRPs, the NLRP3 (in humans encoded by the NLRP3 gene) is expressed predominantly in macrophages as a component of the inflammasome and is associated with many diseases, including gout, type 2 diabetes, multiple sclerosis, atherosclerosis, and neurological diseases and disorders. Diterpenes containing repeated isoprenoid units in their structure are a member of some essential oils that possess diverse biological activities and are becoming a landmark in the field of drug discovery and development. This review sketches a current scenario of diterpenes or their derivatives acting through NLRPs, especially NLRP3-associated pathways with anti-inflammatory effects. For this, a literature survey on the subject has been undertaken using a number of known databases with specific keywords. Findings from the aforementioned databases suggest that diterpenes and their derivatives can exert anti-inflammatory effects via NLRPs-related pathways. Andrographolide, triptolide, kaurenoic acid, carnosic acid, oridonin, teuvincenone F, and some derivatives of tanshinone IIA and phorbol have been found to act through NLRP3 inflammasome pathways. In conclusion, diterpenes and their derivatives could be one of the promising compounds for the treatment of NLRP3-mediated inflammatory diseases and disorders.
Collapse
Affiliation(s)
- Muhammad Torequl Islam
- Laboratory of Theoretical and Computational Biophysics, Ton Duc Thang University, Ho Chi Minh City, Vietnam.,Faculty of Pharmacy, Ho Chi Minh City, Vietnam
| | - Sanaa K Bardaweel
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Jordan, Amman, Jordan
| | | | - Wojciech Koch
- Chair and Department of Food and Nutrition, Medical University of Lublin, Lublin, Poland
| | - Katarzyna Gaweł-Beben
- Department of Cosmetology, University of Information Technology and Management in Rzeszów, Rzeszów, Poland
| | - Beata Antosiewicz
- Department of Cosmetology, University of Information Technology and Management in Rzeszów, Rzeszów, Poland
| | - Javad Sharifi-Rad
- Zabol Medicinal Plants Research Center, Zabol University of Medical Sciences, Zabol, Iran
| |
Collapse
|
222
|
Ming SL, Zeng L, Guo YK, Zhang S, Li GL, Ma YX, Zhai YY, Chang WR, Yang L, Wang J, Yang GY, Chu BB. The Human-Specific STING Agonist G10 Activates Type I Interferon and the NLRP3 Inflammasome in Porcine Cells. Front Immunol 2020; 11:575818. [PMID: 33072119 PMCID: PMC7543045 DOI: 10.3389/fimmu.2020.575818] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/28/2020] [Indexed: 01/18/2023] Open
Abstract
Pigs have anatomical and physiological characteristics comparable to those in humans and, therefore, are a favorable model for immune function research. Interferons (IFNs) and inflammasomes have essential roles in the innate immune system. Here, we report that G10, a human-specific agonist of stimulator of interferon genes (STING), activates both type I IFN and the canonical NLRP3 inflammasome in a STING-dependent manner in porcine cells. Without a priming signal, G10 alone transcriptionally stimulated Sp1-dependent p65 expression, thus triggering activation of the nuclear factor-κB (NF-κB) signaling pathway and thereby priming inflammasome activation. G10 was also found to induce potassium efflux- and NLRP3/ASC/Caspase-1-dependent secretion of IL-1β and IL-18. Pharmacological and genetic inhibition of NLRP3 inflammasomes increased G10-induced type I IFN expression, thereby preventing virus infection, suggesting negative regulation of the NLRP3 inflammasome in the IFN response in the context of STING-mediated innate immune activation. Overall, our findings reveal a new mechanism through which G10 activates the NLRP3 inflammasome in porcine cells and provide new insights into STING-mediated innate immunity in pigs compared with humans.
Collapse
Affiliation(s)
- Sheng-Li Ming
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Lei Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yu-Kun Guo
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Shuang Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Guo-Li Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ying-Xian Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yun-Yun Zhai
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Wen-Ru Chang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Le Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Guo-Yu Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
223
|
Interplays between inflammasomes and viruses, bacteria (pathogenic and probiotic), yeasts and parasites. Immunol Lett 2020; 228:1-14. [PMID: 32971149 PMCID: PMC7505743 DOI: 10.1016/j.imlet.2020.09.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/30/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
In recent years, scientists studying the molecular mechanisms of inflammation have discovered an amazing phenomenon - the inflammasome - a component of the innate immune system that can regulate the functional activity of effector cells during inflammation. At present, it is known that inflammasomes are multimolecular complexes (cytosolic multiprotein oligomers of the innate immune system) that contain many copies of receptors recognizing the molecular structures of cell-damaging factors and pathogenic agents. Inflammasomes are mainly formed in myeloid cells, and their main function is participation in the cleavage of the pro-IL-1β and pro-IL-18 cytokines into their biologically active forms (IL-1β, IL-18). Each type of microorganism influences particular inflammasome activation, and long-term exposure of the organism to viruses, bacteria, yeasts or parasites, among others, can induce uncontrolled inflammation and autoinflammatory diseases. Therefore, this review aims to present the most current scientific data on the molecular interplay between inflammasomes and particular microorganisms. Knowledge about the mechanisms responsible for the interaction between the host and certain types of microorganisms could contribute to the individuation of innovative strategies for the treatment of uncontrolled inflammation targeting a specific type of inflammasome activated by a specific type of pathogen.
Collapse
|
224
|
Murthy AMV, Robinson N, Kumar S. Crosstalk between cGAS-STING signaling and cell death. Cell Death Differ 2020; 27:2989-3003. [PMID: 32948836 DOI: 10.1038/s41418-020-00624-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023] Open
Abstract
Cytosolic nucleic acid sensors have a critical role in detecting endogenous nucleic acids to initiate innate immune responses during microbial infections and/or cell death. Several seminal studies over the past decade have delineated the conserved mechanism of cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) and the downstream signaling adapter stimulator of interferon genes (STING) in mediating innate immune signaling pathways as a host defense mechanism. Besides the predominant role in microbial infections and inflammatory diseases, there is an increased attention on alternative functional responses of cGAS-STING-mediated signaling. Here we review the complexity of interactions between the cGAS-STING signaling and cell death pathways. A better understanding of molecular mechanisms of this interplay is important with regard to the development of new therapeutics targeting cGAS-STING signaling in cancer, infectious, and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ambika M V Murthy
- Centre for Cancer Biology, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia.
| | - Nirmal Robinson
- Centre for Cancer Biology, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia.
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia.
| |
Collapse
|
225
|
A Protective Vaccine against Johne's Disease in Cattle. Microorganisms 2020; 8:microorganisms8091427. [PMID: 32957508 PMCID: PMC7564561 DOI: 10.3390/microorganisms8091427] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/12/2020] [Accepted: 09/15/2020] [Indexed: 01/16/2023] Open
Abstract
Johne’s disease (JD) caused by Mycobacterium avium subsp. paratuberculosis (M. paratuberculosis) is a chronic infection characterized by the development of granulomatous enteritis in wild and domesticated ruminants. It is one of the most significant livestock diseases not only in the USA but also globally, accounting for USD 200–500 million losses annually for the USA alone with potential link to cases of Crohn’s disease in humans. Developing safe and protective vaccines is of a paramount importance for JD control in dairy cows. The current study evaluated the safety, immunity and protective efficacy of a novel live attenuated vaccine (LAV) candidate with and without an adjuvant in comparison to an inactivated vaccine. Results indicated that the LAV, irrespective of the adjuvant presence, induced robust T cell immune responses indicated by proinflammatory cytokine production such as IFN-γ, IFN-α, TNF-α and IL-17 as well as strong response to intradermal skin test against M. paratuberculosis antigens. Furthermore, the LAV was safe with minimal tissue pathology. Finally, calves vaccinated with adjuvanted LAV did not shed M. paratuberculosis post-challenge, a much-desired characteristic of an effective vaccine against JD. Together, this data suggests a strong potential of testing LAV in field trials to curb JD in dairy herds.
Collapse
|
226
|
TYK2 licenses non-canonical inflammasome activation during endotoxemia. Cell Death Differ 2020; 28:748-763. [PMID: 32929218 DOI: 10.1038/s41418-020-00621-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
The non-canonical inflammasome is an emerging crucial player in the development of inflammatory and neurodegenerative diseases. It is activated by direct sensing of cytosolic lipopolysaccharide (LPS) by caspase-11 (CASP11), which then induces pyroptosis, an inflammatory form of regulated cell death. Here, we report that tyrosine kinase 2 (TYK2), a cytokine receptor-associated kinase, is a critical upstream regulator of CASP11. Absence of TYK2 or its kinase activity impairs the transcriptional induction of CASP11 in vitro and in vivo and protects mice from LPS-induced lethality. Lack of TYK2 or its enzymatic activity inhibits macrophage pyroptosis and impairs release of mature IL-1β and IL-18 specifically in response to intracellular LPS. Deletion of TYK2 in myeloid cells reduces LPS-induced IL-1β and IL-18 production in vivo, highlighting the importance of these cells in the inflammatory response to LPS. In support of our data generated with genetically engineered mice, pharmacological inhibition of TYK2 reduced LPS-induced upregulation of CASP11 in bone marrow-derived macrophages (BMDMs) and of its homolog CASP5 in human macrophages. Our study provides insights into the regulation of CASP11 in vivo and uncovered a novel link between TYK2 activity and CASP11-dependent inflammation.
Collapse
|
227
|
Saito LB, Fernandes JP, Smith MJ, Doan MAL, Branton WG, Schmitt LM, Wuest M, Monaco MC, Major EO, Wuest F, Power C. Intranasal anti-caspase-1 therapy preserves myelin and glucose metabolism in a model of progressive multiple sclerosis. Glia 2020; 69:216-229. [PMID: 32882086 DOI: 10.1002/glia.23896] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
Inflammatory demyelination and axonal injury in the central nervous system (CNS) are cardinal features of progressive multiple sclerosis (MS), and linked to activated brain macrophage-like cells (BMCs) including resident microglia and trafficking macrophages. Caspase-1 is a pivotal mediator of inflammation and cell death in the CNS. We investigated the effects of caspase-1 activation and its regulation in models of MS. Brains from progressive MS and non-MS patients, as well as cultured human oligodendrocytes were examined by transcriptomic and morphological methods. Next generation transcriptional sequencing of progressive MS compared to non-MS patients' normal appearing white matter (NAWM) showed induction of caspase-1 as well as other inflammasome-associated genes with concurrent suppression of neuron-specific genes. Oligodendrocytes exposed to TNFα exhibited upregulation of caspase-1 with myelin gene suppression in a cell differentiation state-dependent manner. Brains from cuprizone-exposed mice treated by intranasal delivery of the caspase-1 inhibitor, VX-765 or its vehicle, were investigated in morphological and molecular studies, as well as by fluorodeoxyglucose-positron emission tomography (FDG-PET) imaging. Cuprizone exposure resulted in BMC and caspase-1 activation accompanied by demyelination and axonal injury, which was abrogated by intranasal VX-765 treatment. FDG-PET imaging revealed suppressed glucose metabolism in the thalamus, hippocampus and cortex of cuprizone-exposed mice that was restored with VX-765 treatment. These studies highlight the caspase-1 dependent interactions between inflammation, demyelination, and glucose metabolism in progressive MS and associated models. Intranasal delivery of an anti-caspase-1 therapy represents a promising therapeutic approach for progressive MS and other neuro-inflammatory diseases.
Collapse
Affiliation(s)
- Leina B Saito
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Jason P Fernandes
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Mackenzie J Smith
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Matthew A L Doan
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - William G Branton
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Laura M Schmitt
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Melinda Wuest
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - Frank Wuest
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Christopher Power
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
228
|
Govindarajan V, de Rivero Vaccari JP, Keane RW. Role of inflammasomes in multiple sclerosis and their potential as therapeutic targets. J Neuroinflammation 2020; 17:260. [PMID: 32878648 PMCID: PMC7469327 DOI: 10.1186/s12974-020-01944-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system (CNS), and it remains the most common immune-mediated disorder affecting the CNS. While the cause of MS is unclear, the underlying pathomechanisms are thought to be either destruction by autoimmune T cells or dysfunction of myelin-producing cells. Recent advances have indicated that inflammasomes contribute the etiology of MS. Inflammasomes are multiprotein complexes of the innate immune response involved in the processing of caspase-1, the activation of pro-inflammatory cytokines interleukin (IL)-1β and IL-18 as well as the cell death-mediated mechanism of pyroptosis and the activation of the adaptive immune response. Here we review the literature to date on the role of different inflammasome signaling pathways in the pathogenesis of MS and how these pathways may be targeted to reduce deleterious inflammatory processes and improve outcomes in this patient population.
Collapse
Affiliation(s)
- Vaidya Govindarajan
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, 1600 NW 10th Ave RMSB 5058, Miami, FL, 33136, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Robert W Keane
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, 1600 NW 10th Ave RMSB 5058, Miami, FL, 33136, USA. .,Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
229
|
Nehar-Belaid D, Hong S, Marches R, Chen G, Bolisetty M, Baisch J, Walters L, Punaro M, Rossi RJ, Chung CH, Huynh RP, Singh P, Flynn WF, Tabanor-Gayle JA, Kuchipudi N, Mejias A, Collet MA, Lucido AL, Palucka K, Robson P, Lakshminarayanan S, Ramilo O, Wright T, Pascual V, Banchereau JF. Mapping systemic lupus erythematosus heterogeneity at the single-cell level. Nat Immunol 2020; 21:1094-1106. [PMID: 32747814 PMCID: PMC7442743 DOI: 10.1038/s41590-020-0743-0] [Citation(s) in RCA: 258] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/20/2020] [Indexed: 12/12/2022]
Abstract
Patients with systemic lupus erythematosus (SLE) display a complex blood transcriptome whose cellular origin is poorly resolved. Using single-cell RNA sequencing, we profiled ~276,000 peripheral blood mononuclear cells from 33 children with SLE with different degrees of disease activity and 11 matched controls. Increased expression of interferon-stimulated genes (ISGs) distinguished cells from children with SLE from healthy control cells. The high ISG expression signature (ISGhi) derived from a small number of transcriptionally defined subpopulations within major cell types, including monocytes, CD4+ and CD8+ T cells, natural killer cells, conventional and plasmacytoid dendritic cells, B cells and especially plasma cells. Expansion of unique subpopulations enriched in ISGs and/or in monogenic lupus-associated genes classified patients with the highest disease activity. Profiling of ~82,000 single peripheral blood mononuclear cells from adults with SLE confirmed the expansion of similar subpopulations in patients with the highest disease activity. This study lays the groundwork for resolving the origin of the SLE transcriptional signatures and the disease heterogeneity towards precision medicine applications.
Collapse
Affiliation(s)
| | - Seunghee Hong
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Radu Marches
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Guo Chen
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mohan Bolisetty
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jeanine Baisch
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | | | - Marilynn Punaro
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
- UT Southwestern Medical Center, Dallas, TX, USA
| | - Robert J Rossi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Cheng-Han Chung
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Richie P Huynh
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Prashant Singh
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - William F Flynn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Joy-Ann Tabanor-Gayle
- Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Navya Kuchipudi
- Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Asuncion Mejias
- Division of Pediatric Infectious Diseases, Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH, USA
| | - Magalie A Collet
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Anna Lisa Lucido
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Institute for Systems Genomics and Department of Genetics & Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
| | | | - Octavio Ramilo
- Division of Pediatric Infectious Diseases, Nationwide Children's Hospital and the Ohio State University School of Medicine, Columbus, OH, USA
| | - Tracey Wright
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
- UT Southwestern Medical Center, Dallas, TX, USA
| | - Virginia Pascual
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| | | |
Collapse
|
230
|
Jiang S, Xiao H, Wu Z, Yang Z, Ding B, Jin Z, Yang Y. NLRP3 sparks the Greek fire in the war against lipid-related diseases. Obes Rev 2020; 21:e13045. [PMID: 32390276 DOI: 10.1111/obr.13045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022]
Abstract
In recent years, the obesity rate worldwide has reached epidemic proportions and contributed to the growing prevalence of lipid-related diseases. A strong link between inflammation and metabolism is becoming increasingly evident. Compelling evidence has indicated the activation of the nucleotide-binding and oligomerization domain-like receptor, leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome, a cytoplasmic complex containing multiple proteins, in a variety of lipid-related diseases including obesity, atherosclerosis, liver diseases, and type 2 diabetes. Recent studies have further clarified the regulatory mechanisms and the optional therapeutic agents that target NLRP3 inflammasomes. In this study, we review the recent progress in the research on NLRP3 inflammasomes and discuss their implications for a better understanding of inflammation in lipid-related disease and the prospects of targeting the NLRP3 inflammasome for therapeutic intervention.
Collapse
Affiliation(s)
- Shuai Jiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Life of Sciences, Northwest University, Xi'an, China
| | - Haoxiang Xiao
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhen Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Life of Sciences, Northwest University, Xi'an, China
| | - Zhi Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Life of Sciences, Northwest University, Xi'an, China
| | - Baoping Ding
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Life of Sciences, Northwest University, Xi'an, China
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Life of Sciences, Northwest University, Xi'an, China
| |
Collapse
|
231
|
Sun X, Pang H, Li J, Luo S, Huang G, Li X, Xie Z, Zhou Z. The NLRP3 Inflammasome and Its Role in T1DM. Front Immunol 2020; 11:1595. [PMID: 32973739 PMCID: PMC7481449 DOI: 10.3389/fimmu.2020.01595] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/16/2020] [Indexed: 12/15/2022] Open
Abstract
The NLRP3 (nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3) inflammasome is a protein complex expressed in cells. It detects danger signals and induces the production of active caspase-1 and the maturation and release of IL (interleukin)-33, IL-18, IL-1β and other cytokines. T1DM (type 1 diabetes mellitus) is defined as a chronic autoimmune disorder characterized by the autoreactive T cell-mediated elimination of insulin-positive pancreatic beta-cells. Although the exact underlying mechanisms are obscure, researchers have proposed that both environmental and genetic factors are involved in the pathogenesis of T1DM. Furthermore, immune responses, including innate and adaptive immunity, play an important role in this process. Recently, the NLRP3 inflammasome, a critical component of innate immunity, was reported to be associated with T1DM. Here, we review the assembly and function of the NLRP3 inflammasome. In addition, the activation and regulatory mechanisms that enhance or attenuate NLRP3 inflammasome activation are discussed. Finally, we focus on the relationship between the NLRP3 inflammasome and T1DM, as well as its potential value for clinical use.
Collapse
Affiliation(s)
- Xiaoxiao Sun
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Haipeng Pang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Jiaqi Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Shuoming Luo
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Gan Huang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Xia Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Zhiguo Xie
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| |
Collapse
|
232
|
Neuroinflammation Mediated by NLRP3 Inflammasome After Intracerebral Hemorrhage and Potential Therapeutic Targets. Mol Neurobiol 2020; 57:5130-5149. [PMID: 32856203 DOI: 10.1007/s12035-020-02082-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
Intracerebral hemorrhage (ICH) is the most fatal subtype of stroke; there is still a lack of effective treatment. Microglia are a major component of the innate immune system, and they respond to acute brain injury by activating and forming classic M1-like (pro-inflammatory) or alternative M2-like (anti-inflammatory) phenotype. The existence of the polarization indicates that the role of microglia in disease's progression and recovery after ICH is still unclear, perhaps involving microglial secretion of anti-inflammatory or pro-inflammatory cytokines and chemokines. The NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome is considered to be the main participant in neuroinflammation. Recent evidence has shown that NLRP3 inflammasome can be activated after ICH, resulting in inflammatory cascade reactions and aggravating brain injury. Furthermore, previous studies have reported that NLRP3 inflammasome is mainly present in microglia, so we speculate that its activation may be strongly associated with microglial polarization. Many scholars have investigated the role of brain injury caused by NLRP3 inflammasome after ICH, but the precise operating mechanisms remain uncertain. This review summarized the activation mechanism of NLRP3 inflammasome after ICH and the possible mechanism of NLRP3 inflammasome promoting neuroinflammation and aggravating nerve injury and discussed the relevant potential therapeutic targets.
Collapse
|
233
|
Nguyen H, Gazy N, Venketaraman V. A Role of Intracellular Toll-Like Receptors (3, 7, and 9) in Response to Mycobacterium tuberculosis and Co-Infection with HIV. Int J Mol Sci 2020; 21:E6148. [PMID: 32858917 PMCID: PMC7503332 DOI: 10.3390/ijms21176148] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is a highly infectious acid-fast bacillus and is known to cause tuberculosis (TB) in humans. It is a leading cause of death from a sole infectious agent, with an estimated 1.5 million deaths yearly worldwide, and up to one third of the world's population has been infected with TB. The virulence and susceptibility of Mtb are further amplified in the presence of Human Immunodeficiency Virus (HIV). Coinfection with Mtb and HIV forms a lethal combination. Previous studies had demonstrated the synergistic effects of Mtb and HIV, with one disease accelerating the disease progression of the other through multiple mechanisms, including the modulation of the immune response to these two pathogens. The response of the endosomal pattern recognition receptors to these two pathogens, specifically toll-like receptors (TLR)-3, -7, and -9, has not been elucidated, with some studies producing mixed results. This article seeks to review the roles of TLR-3, -7, and -9 in response to Mtb infection, as well as Mtb-HIV-coinfection via Toll-interleukin 1 receptor (TIR) domain-containing adaptor inducing INF-β (TRIF)-dependent and myeloid differentiation factor 88 (MyD88)-dependent pathways.
Collapse
Affiliation(s)
- Huy Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| | - Nicky Gazy
- Beaumont Health System, 5450 Fort St, Trenton, MI 48183, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA
| |
Collapse
|
234
|
Pereira NZ, Branco ACCC, Manfrere KCG, de Lima JF, Yoshikawa FSY, Milanez HMBPM, Pereira NV, Sotto MN, Duarte AJDS, Sato MN. Increased Expression on Innate Immune Factors in Placentas From HIV-Infected Mothers Concurs With Dampened Systemic Immune Activation. Front Immunol 2020; 11:1822. [PMID: 32983090 PMCID: PMC7477039 DOI: 10.3389/fimmu.2020.01822] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 07/07/2020] [Indexed: 01/15/2023] Open
Abstract
Innate immunity is one of the main protection mechanisms against viral infections, but how this system works at the maternal-fetal interface, especially during HIV infection, is still poorly known. In this study, we investigated the relationship between pregnancy and innate mechanisms associated with HIV immunity by evaluating the expression of DAMPs, inflammasome components and type I/III IFNs in placenta and serum samples from HIV-infected mothers and exposed newborns. Our results showed that most of these factors, including HMGB1, IL-1, and IFN, were increased in placental villi from HIV-infected mothers. Curiously, however, these factors were simultaneously repressed in serum from HIV-infected mothers and their exposed newborns, suggesting that pregnancy could restrict HIV immune activation systemically but preserve the immune response at the placental level. An effective local antiviral status associated with a suppressed inflammatory environment can balance the maternal immune response, promoting homeostasis for fetal development and protection against HIV infection in neonates.
Collapse
Affiliation(s)
- Nátalli Zanete Pereira
- Laboratório de Investigação em Dermatologia e Imunodeficiências LIM56, Instituto de Medicina Tropical, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Anna Cláudia Calvielli Castelo Branco
- Laboratório de Investigação em Dermatologia e Imunodeficiências LIM56, Instituto de Medicina Tropical, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Instituto de Ciências Biomédicas - Universidade de São Paulo, São Paulo, Brazil
| | - Kelly Cristina Gomes Manfrere
- Laboratório de Investigação em Dermatologia e Imunodeficiências LIM56, Instituto de Medicina Tropical, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Josenilson Feitosa de Lima
- Laboratório de Investigação em Dermatologia e Imunodeficiências LIM56, Instituto de Medicina Tropical, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Fabio Seiti Yamada Yoshikawa
- Laboratório de Investigação em Dermatologia e Imunodeficiências LIM56, Instituto de Medicina Tropical, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil.,Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
| | | | - Naiura Vieira Pereira
- Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Miriam Nacagami Sotto
- Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Alberto José da Silva Duarte
- Laboratório de Investigação em Dermatologia e Imunodeficiências LIM56, Instituto de Medicina Tropical, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Maria Notomi Sato
- Laboratório de Investigação em Dermatologia e Imunodeficiências LIM56, Instituto de Medicina Tropical, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
235
|
Starobova H, Nadar EI, Vetter I. The NLRP3 Inflammasome: Role and Therapeutic Potential in Pain Treatment. Front Physiol 2020; 11:1016. [PMID: 32973552 PMCID: PMC7468416 DOI: 10.3389/fphys.2020.01016] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Pain is a fundamental feature of inflammation. The immune system plays a critical role in the activation of sensory neurons and there is increasing evidence of neuro-inflammatory mechanisms contributing to painful pathologies. The inflammasomes are signaling multiprotein complexes that are key components of the innate immune system. They are intimately involved in inflammatory responses and their activation leads to production of inflammatory cytokines that in turn can affect sensory neuron function. Accordingly, the contribution of inflammasome activation to pain signaling has attracted considerable attention in recent years. NLRP3 is the best characterized inflammasome and there is emerging evidence of its role in a variety of inflammatory pain conditions. In vitro and in vivo studies have reported the activation and upregulation of NLRP3 in painful conditions including gout and rheumatoid arthritis, while inhibition of NLRP3 function or expression can mediate analgesia. In this review, we discuss painful conditions in which NLRP3 inflammasome signaling has been pathophysiologically implicated, as well as NLRP3 inflammasome-mediated mechanisms and signaling pathways that may lead to the activation of sensory neurons.
Collapse
Affiliation(s)
- Hana Starobova
- Centre for Pain Research, Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
| | - Evelyn Israel Nadar
- Centre for Pain Research, Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
| | - Irina Vetter
- Centre for Pain Research, Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia.,School of Pharmacy, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
236
|
Mollaei M, Abbasi A, Hassan ZM, Pakravan N. The intrinsic and extrinsic elements regulating inflammation. Life Sci 2020; 260:118258. [PMID: 32818542 DOI: 10.1016/j.lfs.2020.118258] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/14/2022]
Abstract
Inflammation is a sophisticated biological tissue response to both extrinsic and intrinsic stimuli. Although the pathological aspects of inflammation are well appreciated, there are still rooms for understanding the physiological functions of the inflammation. Recent studies have focused on mechanisms, context and the role of physiological inflammation. Besides, there have been progress in the comprehension of commensal microbiota, immunometabolism, cancer and intracellular signaling events' roles that impact on the regulation of inflammation. Despite the fact that inflammatory responses are vital through tissue damage, understanding the mechanisms to turn off the finished or unnecessary inflammation is crucial for restoring homeostasis. Inflammation seems to be a smart process that acts like two edges of a sword, meaning that it has both protective and deleterious consequences. Knowing both edges and the regulation processes will help the future understanding and therapy for various diseases.
Collapse
Affiliation(s)
- M Mollaei
- Department of Immunology, School of Medicine, Tarbiat Modares University, Iran.
| | - A Abbasi
- Department of Immunology, School of Medicine, Tarbiat Modares University, Iran
| | - Z M Hassan
- Department of Immunology, School of Medicine, Tarbiat Modares University, Iran
| | - N Pakravan
- Department of Immunology, School of Medicine, Alborz University of Medical Science, Iran
| |
Collapse
|
237
|
Zheng S, Ma M, Li Z, Hao Y, Li H, Fu P, Jin S. Posttreatment of Maresin1 Inhibits NLRP3 inflammasome activation via promotion of NLRP3 ubiquitination. FASEB J 2020; 34:11944-11956. [PMID: 32667092 DOI: 10.1096/fj.202000665rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/16/2022]
Abstract
Maresin1 is a potent lipid mediator exhibiting potential anti-inflammatory activity in a variety of inflammatory diseases, however, the underlying mechanisms remain poorly understood. Excessive activation of NLRP3 inflammasome has been established in multiple inflammatory diseases. Here, we show that Maresin1 dose-dependently inhibited the NLRP3 inflammasome activation and subsequent caspase-1 activation and IL-1β secretion. This inhibitory effect could be reversed by KH7 and H89, the inhibitors of the cAMP-PKA signaling pathway. Activation of PKA kinase induced by Maresin1 led to the K63-linked ubiquitination of NLRP3 in macrophages. Maresin1 attenuated serum IL-1β secretion through inhibition of NLRP3 inflammasome in vivo using Nlrp3-deficient mouse models of lipopolysaccharide (LPS)-induced sepsis. Maresin1 also repressed MSU-induced peritonitis. This study suggests that Maresin1 is an inhibitor of NLRP3 inflammasome activation and can be used clinically in the treatment of NLRP3 inflammasome-driven inflammatory diseases.
Collapse
Affiliation(s)
- Sisi Zheng
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, P.R. China
| | - Minqi Ma
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, P.R. China
| | - Zhongwang Li
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, P.R. China
| | - Yu Hao
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, P.R. China
| | - Hui Li
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, P.R. China
| | - Panhan Fu
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, P.R. China
| | - Shengwei Jin
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, P.R. China
| |
Collapse
|
238
|
Yap JKY, Moriyama M, Iwasaki A. Inflammasomes and Pyroptosis as Therapeutic Targets for COVID-19. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:307-312. [PMID: 32493814 PMCID: PMC7343621 DOI: 10.4049/jimmunol.2000513] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/15/2020] [Indexed: 12/15/2022]
Abstract
The inflammatory response to severe acute respiratory syndrome-related coronavirus 2 infection has a direct impact on the clinical outcomes of coronavirus disease 2019 patients. Of the many innate immune pathways that are engaged by severe acute respiratory syndrome-related coronavirus 2, we highlight the importance of the inflammasome pathway. We discuss available pharmaceutical agents that target a critical component of inflammasome activation, signaling leading to cellular pyroptosis, and the downstream cytokines as a promising target for the treatment of severe coronavirus disease 2019-associated diseases.
Collapse
Affiliation(s)
- Jeremy K Y Yap
- School of Postgraduate Studies, International Medical University, 57000 Kuala Lumpur, Malaysia
| | - Miyu Moriyama
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520;
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06512; and
- Howard Hughes Medical Institute, Chevy Chase, MD 20815
| |
Collapse
|
239
|
Chen HJ, Tas SW, de Winther MPJ. Type-I interferons in atherosclerosis. J Exp Med 2020; 217:132613. [PMID: 31821440 PMCID: PMC7037237 DOI: 10.1084/jem.20190459] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/05/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Chen et al. review the effects of type-I IFNs and the potential of anti–type-I IFN therapies in atherosclerosis. The contribution of dyslipidemia and inflammation in atherosclerosis is well established. Along with effective lipid-lowering treatments, the recent success of clinical trials with anti-inflammatory therapies and the accelerated atherosclerosis in many autoimmune diseases suggest that targeting inflammation may open new avenues for the prevention and the treatment for cardiovascular diseases (CVDs). In the past decades, studies have widened the role of type-I interferons (IFNs) in disease, from antivirus defense to autoimmune responses and immuno-metabolic syndromes. While elevated type-I IFN level in serum is associated with CVD incidence in patients with interferonopathies, experimental data have attested that type-I IFNs affect plaque-residing macrophages, potentiate foam cell and extracellular trap formation, induce endothelial dysfunction, alter the phenotypes of dendritic cells and T and B lymphocytes, and lead to exacerbated atherosclerosis outcomes. In this review, we discuss the production and the effects of type-I IFNs in different atherosclerosis-associated cell types from molecular biology studies, animal models, and clinical observations, and the potential of new therapies against type-I IFN signaling for atherosclerosis.
Collapse
Affiliation(s)
- Hung-Jen Chen
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands
| | - Menno P J de Winther
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
240
|
Multifaceted Functions of CH25H and 25HC to Modulate the Lipid Metabolism, Immune Responses, and Broadly Antiviral Activities. Viruses 2020; 12:v12070727. [PMID: 32640529 PMCID: PMC7411728 DOI: 10.3390/v12070727] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
With the frequent outbreaks of emerging infectious diseases in recent years, an effective broad-spectrum antiviral drug is becoming an urgent need for global public health. Cholesterol-25-hydroxylase (CH25H) and its enzymatic products 25-hydroxycholesterol (25HC), a well-known oxysterol that regulates lipid metabolism, have been reported to play multiple functions in modulating cholesterol homeostasis, inflammation, and immune responses. CH25H and 25HC were recently identified as exerting broadly antiviral activities, including upon a variety of highly pathogenic viruses such as human immunodeficiency virus (HIV), Ebola virus (EBOV), Nipah virus (NiV), Rift Valley fever virus (RVFV), and Zika virus (ZIKV). The underlying mechanisms for its antiviral activities are being extensively investigated but have not yet been fully clarified. In this study, we summarized the current findings on how CH25H and 25HC play multiple roles to modulate cholesterol metabolism, inflammation, immunity, and antiviral infections. Overall, 25HC should be further studied as a potential therapeutic agent to control emerging infectious diseases in the future.
Collapse
|
241
|
Yavarpour-Bali H, Ghasemi-Kasman M. The role of inflammasomes in multiple sclerosis. Mult Scler 2020; 27:1323-1331. [PMID: 32539629 DOI: 10.1177/1352458520932776] [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: 11/17/2022]
Abstract
Multiple sclerosis (MS) is considered as an inflammatory autoimmune disease of the central nervous system (CNS), with a complex and heterogenic etiology. However, the involvement of inflammation in its pathophysiology is well documented and current therapies for MS are mainly immunosuppressive drugs. Although the available drugs reduce new lesions and relapses, their long-term outcome is not completely satisfactory. Inflammasomes are multimeric protein complexes that play a critical role in the inflammatory process. Several lines of evidence suggest an association between inflammasome activation and MS. In this paper, we have reviewed current studies that demonstrate the involvement of inflammasomes in MS development, in both animal model and MS patients. Furthermore, prior studies about the effect of inflammasome inhibitor drugs on development and progression of MS are discussed.
Collapse
Affiliation(s)
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran/Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
242
|
Feng YS, Tan ZX, Wang MM, Xing Y, Dong F, Zhang F. Inhibition of NLRP3 Inflammasome: A Prospective Target for the Treatment of Ischemic Stroke. Front Cell Neurosci 2020; 14:155. [PMID: 32581721 PMCID: PMC7283578 DOI: 10.3389/fncel.2020.00155] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
Stroke is one of the major devastating diseases with no effective medical therapeutics. Because of the high rate of disability and mortality among stroke patients, new treatments are urgently required to decrease brain damage following a stroke. In recent years, the inflammasome is a novel breakthrough point that plays an important role in the stroke, and the inhibition of inflammasome may be an effective method for stroke treatment. Briefly, inflammasome is a multi-protein complex that causes activation of caspase-1 and subsequent production of pro-inflammatory factors including interleukin (IL)-18 and IL-1β. Among them, the NLRP3 inflammasome is the most typical inflammasome, which can detect cell damage and mediate inflammatory response to tissue damage in ischemic stroke. The NLRP3 inflammasome has become a key mediator of post-ischemic inflammation, leading to a cascade of inflammatory reactions and cell death eventually. Thus, NLRP3 inflammasome is an ideal therapeutic target due to its important role in the inflammatory response after ischemic stroke. In this mini review article, we will summarize the structure, assembly, and regulation of NLRP3 inflammasome, the role of NLRP3 inflammasome in ischemic stroke, and several treatments targeting NLRP3 inflammasome in ischemic stroke. The further understanding of the mechanism of NLRP3 inflammasome in patients with ischemic stroke will provide novel targets for the treatment of cerebral ischemic stroke patients.
Collapse
Affiliation(s)
- Ya-Shuo Feng
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Man-Man Wang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Xing
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Provincial Orthopedic Biomechanics Key Laboratory, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
243
|
Riedelberger M, Penninger P, Tscherner M, Hadriga B, Brunnhofer C, Jenull S, Stoiber A, Bourgeois C, Petryshyn A, Glaser W, Limbeck A, Lynes MA, Schabbauer G, Weiss G, Kuchler K. Type I Interferons Ameliorate Zinc Intoxication of Candida glabrata by Macrophages and Promote Fungal Immune Evasion. iScience 2020; 23:101121. [PMID: 32428860 PMCID: PMC7232100 DOI: 10.1016/j.isci.2020.101121] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/09/2020] [Accepted: 04/29/2020] [Indexed: 12/16/2022] Open
Abstract
Host and fungal pathogens compete for metal ion acquisition during infectious processes, but molecular mechanisms remain largely unknown. Here, we show that type I interferons (IFNs-I) dysregulate zinc homeostasis in macrophages, which employ metallothionein-mediated zinc intoxication of pathogens as fungicidal response. However, Candida glabrata can escape immune surveillance by sequestering zinc into vacuoles. Interestingly, zinc-loading is inhibited by IFNs-I, because a Janus kinase 1 (JAK1)-dependent suppression of zinc homeostasis affects zinc distribution in macrophages as well as generation of reactive oxygen species (ROS). In addition, systemic fungal infections elicit IFN-I responses that suppress splenic zinc homeostasis, thereby altering macrophage zinc pools that otherwise exert fungicidal actions. Thus, IFN-I signaling inadvertently increases fungal fitness both in vitro and in vivo during fungal infections. Our data reveal an as yet unrecognized role for zinc intoxication in antifungal immunity and suggest that interfering with host zinc homeostasis may offer therapeutic options to treat invasive fungal infections.
Collapse
Affiliation(s)
- Michael Riedelberger
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Philipp Penninger
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Michael Tscherner
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Bernhard Hadriga
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Carina Brunnhofer
- Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria
| | - Sabrina Jenull
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Anton Stoiber
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Christelle Bourgeois
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Andriy Petryshyn
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Walter Glaser
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Andreas Limbeck
- Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria
| | - Michael A Lynes
- Department of Molecular and Cell Biology, University of Connecticut, CT, USA
| | - Gernot Schabbauer
- Institute for Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Guenter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, and Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Karl Kuchler
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, Vienna, Austria.
| |
Collapse
|
244
|
Rocca C, Pasqua T, Cerra MC, Angelone T. Cardiac Damage in Anthracyclines Therapy: Focus on Oxidative Stress and Inflammation. Antioxid Redox Signal 2020; 32:1081-1097. [PMID: 31928066 DOI: 10.1089/ars.2020.8016] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Despite their serious side effects, anthracyclines (ANTs) are the most prescribed chemotherapeutic drugs because of their strong efficacy in both solid and hematological tumors. A major limitation to ANTs clinical application is the severe cardiotoxicity observed both acutely and chronically. The mechanism underlying cardiac dysfunction under chemotherapy is mainly dependent on the generation of oxidative stress and systemic inflammation, both of which lead to progressive cardiomyopathy and heart failure. Recent Advances: Over the years, the iatrogenic ANTs-induced cardiotoxicity was believed to be simply given by iron metabolism and reactive oxygen species production; however, several experimental data indicate that ANTs may use alternative damaging mechanisms, such as topoisomerase 2β inhibition, inflammation, pyroptosis, immunometabolism, and autophagy. Critical Issues: In this review, we aimed at discussing ANTs-induced cardiac injury from different points of view, updating and focusing on oxidative stress and inflammation, since these pathways are not exclusive or independent from each other but they together importantly contribute to the complexity of ANTs-induced multifactorial cardiotoxicity. Future Directions: A deeper understanding of the mechanistic signaling leading to ANTs side effects could reveal crucial targeting molecules, thus representing strategic knowledge to promote better therapeutic efficacy and lower cardiotoxicity during clinical application.
Collapse
Affiliation(s)
- Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Maria Carmela Cerra
- Laboratory of Organ and System Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| |
Collapse
|
245
|
Campbell LK, Magor KE. Pattern Recognition Receptor Signaling and Innate Responses to Influenza A Viruses in the Mallard Duck, Compared to Humans and Chickens. Front Cell Infect Microbiol 2020; 10:209. [PMID: 32477965 PMCID: PMC7236763 DOI: 10.3389/fcimb.2020.00209] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/16/2020] [Indexed: 12/25/2022] Open
Abstract
Mallard ducks are a natural host and reservoir of avian Influenza A viruses. While most influenza strains can replicate in mallards, the virus typically does not cause substantial disease in this host. Mallards are often resistant to disease caused by highly pathogenic avian influenza viruses, while the same strains can cause severe infection in humans, chickens, and even other species of ducks, resulting in systemic spread of the virus and even death. The differences in influenza detection and antiviral effectors responsible for limiting damage in the mallards are largely unknown. Domestic mallards have an early and robust innate response to infection that seems to limit replication and clear highly pathogenic strains. The regulation and timing of the response to influenza also seems to circumvent damage done by a prolonged or dysregulated immune response. Rapid initiation of innate immune responses depends on viral recognition by pattern recognition receptors (PRRs) expressed in tissues where the virus replicates. RIG-like receptors (RLRs), Toll-like receptors (TLRs), and Nod-like receptors (NLRs) are all important influenza sensors in mammals during infection. Ducks utilize many of the same PRRs to detect influenza, namely RIG-I, TLR7, and TLR3 and their downstream adaptors. Ducks also express many of the same signal transduction proteins including TBK1, TRIF, and TRAF3. Some antiviral effectors expressed downstream of these signaling pathways inhibit influenza replication in ducks. In this review, we summarize the recent advances in our understanding of influenza recognition and response through duck PRRs and their adaptors. We compare basal tissue expression and regulation of these signaling components in birds, to better understand what contributes to influenza resistance in the duck.
Collapse
Affiliation(s)
- Lee K Campbell
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Katharine E Magor
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
246
|
Zhang YH, Aldo P, You Y, Ding J, Kaislasuo J, Petersen JF, Lokkegaard E, Peng G, Paidas MJ, Simpson S, Pal L, Guller S, Liu H, Liao AH, Mor G. Trophoblast-secreted soluble-PD-L1 modulates macrophage polarization and function. J Leukoc Biol 2020; 108:983-998. [PMID: 32386458 DOI: 10.1002/jlb.1a0420-012rr] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/22/2022] Open
Abstract
Decidual macrophages are in close contact with trophoblast cells during placenta development, and an appropriate crosstalk between these cellular compartments is crucial for the establishment and maintenance of a healthy pregnancy. During different phases of gestation, macrophages undergo dynamic changes to adjust to the different stages of fetal development. Trophoblast-secreted factors are considered the main modulators responsible for macrophage differentiation and function. However, the phenotype of these macrophages induced by trophoblast-secreted factors and the factors responsible for their polarization has not been elucidated. In this study, we characterized the phenotype and function of human trophoblast-induced macrophages. Using in vitro models, we found that human trophoblast-educated macrophages were CD14+ CD206+ CD86- and presented an unusual transcriptional profile in response to TLR4/LPS activation characterized by the expression of type I IFN-β expression. IFN-β further enhances the constitutive production of soluble programmed cell death ligand 1 (PD-L1) from trophoblast cells. PD-1 blockage inhibited trophoblast-induced macrophage differentiation. Soluble PD-L1 (sPD-L1) was detected in the blood of pregnant women and increased throughout the gestation. Collectively, our data suggest the existence of a regulatory circuit at the maternal fetal interface wherein IFN-β promotes sPD-L1 expression/secretion by trophoblast cells, which can then initiate a PD-L1/PD-1-mediated macrophage polarization toward an M2 phenotype, consequently decreasing inflammation. Macrophages then maintain the expression of sPD-L1 by the trophoblasts through IFN-β production induced through TLR4 ligation.
Collapse
Affiliation(s)
- Yong-Hong Zhang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA.,Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Paulomi Aldo
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Yuan You
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Jiahui Ding
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Janina Kaislasuo
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Obstetrics and Gynecology, University of Helsinki and the Helsinki University Hospital, Helsinki, Finland
| | - Jesper F Petersen
- Department of Obstetrics and Gynecology, North Zealand Hospital, Hilleroed, Denmark
| | - Ellen Lokkegaard
- Department of Obstetrics and Gynecology, North Zealand Hospital, Hilleroed, Denmark
| | - Gang Peng
- Department of Biostatistics, School of Public Health, Yale University, New Haven, Connecticut, USA
| | - Michael J Paidas
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Samantha Simpson
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lubna Pal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Seth Guller
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hong Liu
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Ai Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Gil Mor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA.,C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
247
|
Han B, García‐Mendoza D, van den Berg H, van den Brink NW. Modulatory Effects of Pb 2+ on Virally Challenged Chicken Macrophage (HD-11) and B-Lymphocyte (DT40) Cell Lines In Vitro. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2020; 39:1060-1070. [PMID: 32124477 PMCID: PMC7277059 DOI: 10.1002/etc.4702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/24/2020] [Accepted: 02/25/2020] [Indexed: 05/08/2023]
Abstract
Elevated levels of lead have been found in waterfowl, due to human activities. Lead may cause immunomodulatory effects, but the mechanisms are largely unknown, especially after viral challenges. To characterize avian immunomodulatory hazards of lead (Pb)2+ , we used chicken macrophage (HD-11) and B-lymphocyte (DT40) cell lines, as in vitro models for the innate and adaptive immune systems, respectively. The cells were activated via toll-like receptor-3 by polyinosinic-polycytidylic acid sodium salt (poly I:C), mimicking viral infections. Our results indicate that Pb2+ is cytotoxic to both cell lines, macrophages being more sensitive. De novo synthesis of glutathione plays an important role in protecting macrophages from Pb2+ intoxication, which might also be closely involved in the induction of nitric oxide after Pb2+ exposure. Stimulatory effects on cell proliferation were noticed at noncytotoxic Pb2+ concentrations as well. Exposure to Pb2+ could also affect the inflammatory status by inhibiting the pro-inflammatory interferon (IFN)-γ while promoting the production of anti-inflammatory type I IFNs in both macrophages and B-cells, and increasing intracellular IgM levels in B-cells. These results suggest that the immunomodulatory effects of Pb2+ in birds are probably closely associated with disruption of immune cell proliferation and cytokine production, potentially causing disorders of the avian immune system. Environ Toxicol Chem 2020;39:1060-1070. © 2020 SETAC.
Collapse
Affiliation(s)
- Biyao Han
- Division of ToxicologyWageningen University and ResearchWageningenThe Netherlands
| | - Diego García‐Mendoza
- Division of ToxicologyWageningen University and ResearchWageningenThe Netherlands
| | - Hans van den Berg
- Division of ToxicologyWageningen University and ResearchWageningenThe Netherlands
| | | |
Collapse
|
248
|
Cerbán FM, Stempin CC, Volpini X, Carrera Silva EA, Gea S, Motran CC. Signaling pathways that regulate Trypanosoma cruzi infection and immune response. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165707. [DOI: 10.1016/j.bbadis.2020.165707] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023]
|
249
|
Zhou L, Zhang Y, Wang Y, Zhang M, Sun W, Dai T, Wang A, Wu X, Zhang S, Wang S, Zhou F. A Dual Role of Type I Interferons in Antitumor Immunity. ACTA ACUST UNITED AC 2020; 4:e1900237. [PMID: 33245214 DOI: 10.1002/adbi.201900237] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022]
Abstract
Type I interferons (IFN-Is) are a family of cytokines that exert direct antiviral effects and regulate innate and adaptive immune responses through direct and indirect mechanisms. It is generally believed that IFN-Is repress tumor development via restricting tumor proliferation and inducing antitumor immune responses. However, recent emerging evidence suggests that IFN-Is play a dual role in antitumor immunity. That is, in the early stage of tumorigenesis, IFN-Is promote the antitumor immune response by enhancing antigen presentation in antigen-presenting cells and activating CD8+ T cells. However, in the late stage of tumor progression, persistent expression of IFN-Is induces the expression of immunosuppressive factors (PD-L1, IDO, and IL-10) on the surface of dendritic cells and other bone marrow cells and inhibits their antitumor immunity. This review outlines these dual functions of IFN-Is in antitumor immunity and elucidates the involved mechanisms, as well as their applications in tumor therapy.
Collapse
Affiliation(s)
- Lili Zhou
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Yuqi Zhang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Yongqiang Wang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Meirong Zhang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Wenhuan Sun
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Tong Dai
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Aijun Wang
- Department of Surgery, School of Medicine, UC Davis, Davis, CA, 95817, USA
| | - Xiaojin Wu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Suping Zhang
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, Base for international Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518055, China
| | - Shuai Wang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Fangfang Zhou
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
250
|
Murray SM, Zhang Y, Douek DC, Sekaly RP. Myeloid Cells Enriched for a Dendritic Cell Population From People Living With HIV Have Altered Gene Expression Not Restored by Antiretroviral Therapy. Front Immunol 2020; 11:261. [PMID: 32194550 PMCID: PMC7064632 DOI: 10.3389/fimmu.2020.00261] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 01/31/2020] [Indexed: 12/22/2022] Open
Abstract
Antiretroviral therapy (ART) for human immunodeficiency virus (HIV) infections has been designed to optimize CD4 T-cell survival and limit HIV replication. Cell types other than CD4 T cells such as monocytes/macrophage, dendritic cells, and granulocytes (collectively known as myeloid cells), are generally not considered in the development of ART protocols. Myeloid dendritic cells (mDCs) are the most potent inducers of CD4 T-cell activation and central to the regulation of immune responses. mDCs in the blood are decreased in number, altered in function, and implicated in promoting HIV latency in people living with HIV (PLWH). We found that cells enriched for mDC in PLWH had transcriptional changes compared to mDC from HIV uninfected individuals, some of which were not completely restored by ART. In contrast, other mDC functions such as interleukin-1 signaling and type I interferon pathways were restored by ART. Some of the transcriptional changes in mDC not completely reversed by ART were enriched in genes that are classically associated with cells of the monocyte/macrophage lineage, but new single-cell RNA sequencing studies show that they are also expressed by a subset of mDC. A cellular enzyme, acyloxyacyl hydrolase (AOAH), important for lipopolysaccharide (LPS) detoxification, had increased transcription in mDC of PLWH, not restored by ART. It is possible that one reason ART is not completely successful in PLWH is the failure to phenotypically change the mDCs. Thus, inability of ART to be completely effective might involve myeloid cells and the failure to restore mDC function as measured by gene transcription. We suggest that mDC and myeloid cells should be considered in future combination ART development.
Collapse
Affiliation(s)
- Shannon M Murray
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Yuwei Zhang
- Vaccine and Gene Therapy Institute Florida, Port St. Lucie, FL, United States
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Rafick P Sekaly
- Vaccine and Gene Therapy Institute Florida, Port St. Lucie, FL, United States
| |
Collapse
|