351
|
Harvey R, McCaughan C, Wise LM, Mercer AA, Fleming SB. Orf virus inhibits interferon stimulated gene expression and modulates the JAK/STAT signalling pathway. Virus Res 2015; 208:180-8. [PMID: 26113305 DOI: 10.1016/j.virusres.2015.06.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 02/07/2023]
Abstract
Interferons (IFNs) play a critical role as a first line of defence against viral infection. Activation of the Janus kinase/signal transducer and activation of transcription (JAK/STAT) pathway by IFNs leads to the production of IFN stimulated genes (ISGs) that block viral replication. The Parapoxvirus, Orf virus (ORFV) induces acute pustular skin lesions of sheep and goats and is transmissible to man. The virus replicates in keratinocytes that are the immune sentinels of skin. We investigated whether or not ORFV could block the expression of ISGs. The human gene GBP1 is stimulated exclusively by type II IFN while MxA is stimulated exclusively in response to type I IFNs. We found that GBP1 and MxA were strongly inhibited in ORFV infected HeLa cells stimulated with IFN-γ or IFN-α respectively. Furthermore we showed that ORFV inhibition of ISG expression was not affected by cells pretreated with adenosine N1-oxide (ANO), a molecule that inhibits poxvirus mRNA translation. This suggested that new viral gene synthesis was not required and that a virion structural protein was involved. We next investigated whether ORFV infection affected STAT1 phosphorylation in IFN-γ or IFN-α treated HeLa cells. We found that ORFV reduced the levels of phosphorylated STAT1 in a dose-dependent manner and was specific for Tyr701 but not Ser727. Treatment of cells with sodium vanadate suggested that a tyrosine phosphatase was responsible for dephosphorylating STAT1-p. ORFV encodes a factor, ORFV057, with homology to the vaccinia virus structural protein VH1 that impairs the JAK/STAT pathway by dephosphorylating STAT1. Our findings show that ORFV has the capability to block ISG expression and modulate the JAK/STAT signalling pathway.
Collapse
Affiliation(s)
- Ryan Harvey
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, New Zealand.
| | - Catherine McCaughan
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, New Zealand.
| | - Lyn M Wise
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, New Zealand.
| | - Andrew A Mercer
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, New Zealand.
| | - Stephen B Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, New Zealand.
| |
Collapse
|
352
|
The immunology and inflammatory responses of human melanocytes in infectious diseases. J Infect 2015; 71:413-21. [PMID: 26092350 DOI: 10.1016/j.jinf.2015.06.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 06/06/2015] [Accepted: 06/10/2015] [Indexed: 12/24/2022]
Abstract
Melanin is a canonical and major defense molecule in invertebrates but its role in mammalian immunity remains unexplored. In contrast, several recent studies have highlighted the emerging innate immune activities of human melanin-producing cells which can sense and respond to bacterial and viral infections. Indeed, the skin is a major portal of entry for pathogens such as arboviruses (Chikungunya, Dengue) and bacteria (mycobacterium leprae, Leptospira spirochetes). Melanocytes of the epidermis could contribute to the phagocytosis of these invading pathogens and to present antigens to competent immune cells. Melanocytes are known to produce key cytokines such as IL-1β, IL6 and TNF-α as well as chemokines. These molecules will subsequently alert macrophages, neutrophils, fibroblasts and keratinocytes through unique crosstalk mechanisms. The infection and the inflammatory responses will control melanocyte's immune and metabolic functions and could contribute to skin manifestations (rash, hyper or de-pigmentation, epidermolysis and psoriasis-like lesions). This review will address the potential role of melanocytes in immunity, inflammation and infection of the skin in health and diseases.
Collapse
|
353
|
Abdul-Sater AA, Majoros A, Plumlee CR, Perry S, Gu AD, Lee C, Shresta S, Decker T, Schindler C. Different STAT Transcription Complexes Drive Early and Delayed Responses to Type I IFNs. THE JOURNAL OF IMMUNOLOGY 2015; 195:210-216. [PMID: 26019270 DOI: 10.4049/jimmunol.1401139] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 04/24/2015] [Indexed: 01/12/2023]
Abstract
IFNs, which transduce pivotal signals through Stat1 and Stat2, effectively suppress the replication of Legionella pneumophila in primary murine macrophages. Although the ability of IFN-γ to impede L. pneumophila growth is fully dependent on Stat1, IFN-αβ unexpectedly suppresses L. pneumophila growth in both Stat1- and Stat2-deficient macrophages. New studies demonstrating that the robust response to IFN-αβ is lost in Stat1-Stat2 double-knockout macrophages suggest that Stat1 and Stat2 are functionally redundant in their ability to direct an innate response toward L. pneumophila. Because the ability of IFN-αβ to signal through Stat1-dependent complexes (i.e., Stat1-Stat1 and Stat1-Stat2 dimers) has been well characterized, the current studies focus on how Stat2 is able to direct a potent response to IFN-αβ in the absence of Stat1. These studies reveal that IFN-αβ is able to drive the formation of a Stat2 and IFN regulatory factor 9 complex that drives the expression of a subset of IFN-stimulated genes, but with substantially delayed kinetics. These observations raise the possibility that this pathway evolved in response to microbes that have devised strategies to subvert Stat1-dependent responses.
Collapse
Affiliation(s)
- Ali A Abdul-Sater
- Department of Microbiology & Immunology, Columbia University, New York, NY 10032 USA
| | - Andrea Majoros
- Department of Microbiology, Immunology & Genetics, University of Vienna, Vienna A-1030 Austria
| | - Courtney R Plumlee
- Department of Microbiology & Immunology, Columbia University, New York, NY 10032 USA
| | - Stuart Perry
- Division of Vaccine Discovery, La Jolla Inst. for Allergy and Immunology, La Jolla, CA 92037 USA
| | - Ai Di Gu
- Department of Microbiology & Immunology, Columbia University, New York, NY 10032 USA
| | - Carolyn Lee
- Department of Microbiology & Immunology, Columbia University, New York, NY 10032 USA
| | - Sujan Shresta
- Division of Vaccine Discovery, La Jolla Inst. for Allergy and Immunology, La Jolla, CA 92037 USA
| | - Thomas Decker
- Department of Microbiology, Immunology & Genetics, University of Vienna, Vienna A-1030 Austria
| | - Christian Schindler
- Department of Microbiology & Immunology, Columbia University, New York, NY 10032 USA.,Department of Medicine, Columbia University, New York, NY 10032 USA
| |
Collapse
|
354
|
Doyle T, Goujon C, Malim MH. HIV-1 and interferons: who's interfering with whom? Nat Rev Microbiol 2015; 13:403-13. [PMID: 25915633 DOI: 10.1038/nrmicro3449] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The ability of interferons (IFNs) to inhibit HIV-1 replication in cell culture models has long been recognized, and the therapeutic administration of IFNα to HIV-1-infected patients who are not receiving antiretroviral therapy produces a clear but transient decrease in plasma viral load. Conversely, studies of chronic HIV-1 infection in humans and SIV-infected animal models of AIDS show positive correlations between elevated plasma levels of IFNs, increased expression of IFN-stimulated genes (ISGs), biomarkers of inflammation and disease progression. In this Review, we discuss the evidence that IFNs can control HIV-1 replication in vivo and debate the controversial role of IFNs in promoting the pathological sequelae of chronic HIV-1 infection.
Collapse
Affiliation(s)
- Tomas Doyle
- Department of Infectious Diseases, King's College London, 2nd Floor, Borough Wing, Guy's Hospital, London Bridge, London SE1 9RT, UK
| | - Caroline Goujon
- Department of Infectious Diseases, King's College London, 2nd Floor, Borough Wing, Guy's Hospital, London Bridge, London SE1 9RT, UK
| | - Michael H Malim
- Department of Infectious Diseases, King's College London, 2nd Floor, Borough Wing, Guy's Hospital, London Bridge, London SE1 9RT, UK
| |
Collapse
|
355
|
Konradi J, Mollenhauer M, Baldus S, Klinke A. Redox-sensitive mechanisms underlying vascular dysfunction in heart failure. Free Radic Res 2015; 49:721-42. [DOI: 10.3109/10715762.2015.1027200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
356
|
Hodgson K, Morris J, Bridson T, Govan B, Rush C, Ketheesan N. Immunological mechanisms contributing to the double burden of diabetes and intracellular bacterial infections. Immunology 2015; 144:171-85. [PMID: 25262977 DOI: 10.1111/imm.12394] [Citation(s) in RCA: 249] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 08/12/2014] [Accepted: 08/19/2014] [Indexed: 12/11/2022] Open
Abstract
Diabetes has been recognized as an important risk factor for a variety of intracellular bacterial infections, but research into the dysregulated immune mechanisms contributing to the impaired host-pathogen interactions is in its infancy. Diabetes is characterized by a chronic state of low-grade inflammation due to activation of pro-inflammatory mediators and increased formation of advanced glycation end products. Increased oxidative stress also exacerbates the chronic inflammatory processes observed in diabetes. The reduced phagocytic and antibacterial activity of neutrophils and macrophages provides an intracellular niche for the pathogen to replicate. Phagocytic and antibacterial dysfunction may be mediated directly through altered glucose metabolism and oxidative stress. Furthermore, impaired activation of natural killer cells contributes to decreased levels of interferon-γ, required for promoting macrophage antibacterial mechanisms. Together with impaired dendritic cell function, this impedes timely activation of adaptive immune responses. Increased intracellular oxidation of antigen-presenting cells in individuals with diabetes alters the cytokine profile generated and the subsequent balance of T-cell immunity. The establishment of acute intracellular bacterial infections in the diabetic host is associated with impaired T-cell-mediated immune responses. Concomitant to the greater intracellular bacterial burden and potential cumulative effect of chronic inflammatory processes, late hyper-inflammatory cytokine responses are often observed in individuals with diabetes, contributing to systemic pathology. The convergence of intracellular bacterial infections and diabetes poses new challenges for immunologists, providing the impetus for multidisciplinary research.
Collapse
Affiliation(s)
- Kelly Hodgson
- Infectious Diseases and Immunopathogenesis Research Group, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Qld, Australia
| | | | | | | | | | | |
Collapse
|
357
|
McNab F, Mayer-Barber K, Sher A, Wack A, O'Garra A. Type I interferons in infectious disease. Nat Rev Immunol 2015; 15:87-103. [PMID: 25614319 DOI: 10.1038/nri3787] [Citation(s) in RCA: 1829] [Impact Index Per Article: 182.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type I interferons (IFNs) have diverse effects on innate and adaptive immune cells during infection with viruses, bacteria, parasites and fungi, directly and/or indirectly through the induction of other mediators. Type I IFNs are important for host defence against viruses. However, recently, they have been shown to cause immunopathology in some acute viral infections, such as influenza virus infection. Conversely, they can lead to immunosuppression during chronic viral infections, such as lymphocytic choriomeningitis virus infection. During bacterial infections, low levels of type I IFNs may be required at an early stage, to initiate cell-mediated immune responses. High concentrations of type I IFNs may block B cell responses or lead to the production of immunosuppressive molecules, and such concentrations also reduce the responsiveness of macrophages to activation by IFNγ, as has been shown for infections with Listeria monocytogenes and Mycobacterium tuberculosis. Recent studies in experimental models of tuberculosis have demonstrated that prostaglandin E2 and interleukin-1 inhibit type I IFN expression and its downstream effects, demonstrating that a cross-regulatory network of cytokines operates during infectious diseases to provide protection with minimum damage to the host.
Collapse
Affiliation(s)
- Finlay McNab
- 1] Allergic Inflammation Discovery Performance Unit, Respiratory Disease Respiratory Research and Development, GlaxoSmithKline, Stevenage, Hertfordshire SG1 2NY, UK. [2] Division of Immunoregulation, Medical Research Council (MRC) National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Katrin Mayer-Barber
- Immunobiology Section, Laboratory of Parasitic Diseases (LPD), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases (LPD), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Andreas Wack
- Division of Immunoregulation, Medical Research Council (MRC) National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Anne O'Garra
- 1] Division of Immunoregulation, Medical Research Council (MRC) National Institute for Medical Research, Mill Hill, London NW7 1AA, UK. [2] National Heart and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
358
|
Meunier E, Broz P. Interferon-induced guanylate-binding proteins promote cytosolic lipopolysaccharide detection by caspase-11. DNA Cell Biol 2015; 34:1-5. [PMID: 25347553 DOI: 10.1089/dna.2014.2701] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lipopolysaccharide (LPS) from gram-negative bacteria is a classical pathogen-associated molecular pattern and a strong inducer of immune responses. While the detection of LPS on the cell surface and in the endosome by Toll-like receptor 4 (TLR4) has been studied for some time, it has only recently been discovered that LPS can also be sensed in the cytosol of cells by a noncanonical inflammasome pathway, resulting in the activation of the cysteine protease caspase-11. Intriguingly, activation of this pathway requires the production of interferons (IFNs) and the induction of a class of IFN-induced GTPases called guanylate-binding proteins (GBPs), which have previously been linked to cell-autonomous killing of intracellular microbes. In this study, we review the recent advances in our understanding of cytosolic LPS sensing and the function of mammalian GBPs.
Collapse
Affiliation(s)
- Etienne Meunier
- Focal Area Infection Biology, Biozentrum, University of Basel , Basel, Switzerland
| | | |
Collapse
|
359
|
Gupta M, Shin DM, Ramakrishna L, Goussetis DJ, Platanias LC, Xiong H, Morse HC, Ozato K. IRF8 directs stress-induced autophagy in macrophages and promotes clearance of Listeria monocytogenes. Nat Commun 2015; 6:6379. [PMID: 25775030 PMCID: PMC4363081 DOI: 10.1038/ncomms7379] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 01/23/2015] [Indexed: 12/11/2022] Open
Abstract
Autophagy, activated by many stresses, plays a critical role in innate immune responses. Here we show that Interferon Regulatory Factor 8 (IRF8) is required for expression of autophagy-related genes in dendritic cells. Furthermore in macrophages, IRF8 is induced by multiple autophagy-inducing stresses, including IFNγ and toll like receptor stimulation, bacterial infection, starvation and by macrophage colony-stimulating factor. IRF8 directly activates many genes involved in various steps of autophagy, promoting autophagosome formation and lysosomal fusion. Consequently, Irf8-/- macrophages are deficient in autophagic activity, and excessively accumulate SQSTM1 and ubiquitin-bound proteins. We show that clearance of Listeria monocytogenes in macrophages requires IRF8-dependent activation of autophagy genes and subsequent autophagic capturing and degradation of Listeria antigens. These processes are defective in Irf8-/- macrophages where uninhibited bacterial growth ensues. Together, these data suggest that IRF8 is a major autophagy regulator in macrophages, essential for macrophage maturation, survival and innate immune responses.
Collapse
Affiliation(s)
- Monica Gupta
- Program in Genomics of Differentiation, NICHD, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Dong-Mi Shin
- 1] Laboratory of Immunopathology, NIAID, National Institutes of Health, 5640 Fishers Lane, Room 1421, Rockville, Maryland 20852, USA [2] Department of Food and Nutrition, Seoul National University, Seoul 151-742, Korea
| | - Lakshmi Ramakrishna
- Program in Genomics of Differentiation, NICHD, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Dennis J Goussetis
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois 60611, USA
| | - Leonidas C Platanias
- 1] Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois 60611, USA [2] Division of Hematology-Oncology, Jesse Brown VA Medical Center, Chicago, Illinois 60612, USA
| | - Huabao Xiong
- Immunology Institute, Mount Sinai School of Medicine, New York, New York 10029, USA
| | - Herbert C Morse
- Laboratory of Immunopathology, NIAID, National Institutes of Health, 5640 Fishers Lane, Room 1421, Rockville, Maryland 20852, USA
| | - Keiko Ozato
- Program in Genomics of Differentiation, NICHD, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
360
|
Davidson S, Maini MK, Wack A. Disease-promoting effects of type I interferons in viral, bacterial, and coinfections. J Interferon Cytokine Res 2015; 35:252-64. [PMID: 25714109 PMCID: PMC4389918 DOI: 10.1089/jir.2014.0227] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
While type I interferons (IFNs) are universally acknowledged for their antiviral and immunostimulatory functions, there is increasing appreciation of the detrimental effects of inappropriate, excessive, or mistimed type I IFN responses in viral and bacterial infections. The underlying mechanisms by which type I IFNs promote susceptibility or severity include direct tissue damage by apoptosis induction or suppression of proliferation in tissue cells, immunopathology due to excessive inflammation, and cell death induced by TRAIL- and Fas-expressing immune cells, as well as immunosuppression through IL-10, IL-27, PD-L1, IL-1Ra, and other regulatory molecules that antagonize the induction or action of IL-1, IL-12, IL-17, IFN-γ, KC, and other effectors of the immune response. Bacterial superinfections following influenza infection are a prominent example of a situation where type I IFNs can misdirect the immune response. This review discusses current understanding of the parameters of signal strength, duration, timing, location, and cellular recipients that determine whether type I IFNs have beneficial or detrimental effects in infection.
Collapse
Affiliation(s)
- Sophia Davidson
- 1 Division of Immunoregulation, MRC National Institute for Medical Research , Mill Hill, London, United Kingdom
| | | | | |
Collapse
|
361
|
Xu C, Evensen Ø, Munang'andu HM. De novo assembly and transcriptome analysis of Atlantic salmon macrophage/dendritic-like TO cells following type I IFN treatment and Salmonid alphavirus subtype-3 infection. BMC Genomics 2015; 16:96. [PMID: 25765343 PMCID: PMC4337061 DOI: 10.1186/s12864-015-1302-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/29/2015] [Indexed: 12/23/2022] Open
Abstract
Background Interferons (IFN) are cytokines secreted by vertebrate cells involved in activation of signaling pathways that direct the synthesis of antiviral genes. To gain a global understanding of antiviral genes induced by type I IFNs in salmonids, we used RNA-seq to characterize the transcriptomic changes induced by type I IFN treatment and salmon alphavirus subtype 3 (SAV-3) infection in TO-cells, a macrophage/dendritic like cell-line derived from Atlantic salmon (Salmo salar L) head kidney leukocytes. Results More than 23 million reads generated by RNA-seq were de novo assembled into 58098 unigenes used to generate a total of 3149 and 23289 differentially expressed genes (DEGs) from TO-cells exposed to type I IFN treatment and SAV-3 infection, respectively. Although the DEGs were classified into genes associated with biological processes, cellular components and molecular function based on gene ontology classification, transcriptomic changes reported here show upregulation of genes belonging to the canonical type I IFN signaling pathways together with a broad spectrum of antiviral genes that block virus replication in host cells. In addition, the transcriptome shows a profile of genes associated with apoptosis as well as genes that activate adaptive immunity. Further, our findings show that the profile of genes expressed by TO-cells is comparable to orthologous genes expressed by mammalian macrophages and dendritic cells in response to type I IFNs. Twenty DEGs randomly selected for qRT-PCR confirmed the validity of the transcriptomic changes detected by RNA-seq by showing that the genes upregulated by RNA-seq were also upregulated by qRT-PCR and that genes downregulated by RNA-seq were also downregulated by qRT-PCR. Conclusions The de novo assembled transcriptome presented here provides a global description of genes induced by type I IFNs in TO-cells that could serve as a repository for future studies in fish cells. Transcriptome analysis shows that a large proportion of IFN genes expressed in this study are comparable to IFNs genes expressed in mammalia. In addition, the study shows that SAV-3 is a potent inducer of type I IFNs and that the responses it induces in TO-cells could serve as a model for studying IFN responses in salmonids.
Collapse
Affiliation(s)
- Cheng Xu
- Faculty of Veterinary Medicine and Biosciences, Department of Basic Sciences and Aquatic Medicine, Section of Aquatic Medicine and Nutrition, Norwegian University of Life Sciences, P.O. Box 8146, Dep. NO-0033, Oslo, Norway.
| | - Øystein Evensen
- Faculty of Veterinary Medicine and Biosciences, Department of Basic Sciences and Aquatic Medicine, Section of Aquatic Medicine and Nutrition, Norwegian University of Life Sciences, P.O. Box 8146, Dep. NO-0033, Oslo, Norway.
| | - Hetron Mweemba Munang'andu
- Faculty of Veterinary Medicine and Biosciences, Department of Basic Sciences and Aquatic Medicine, Section of Aquatic Medicine and Nutrition, Norwegian University of Life Sciences, P.O. Box 8146, Dep. NO-0033, Oslo, Norway.
| |
Collapse
|
362
|
Vidalain PO, Lucas-Hourani M, Helynck O, Tangy F, Munier-Lehmann H. [Stimulation of the antiviral innate immune response by pyrimidine biosynthesis inhibitors: a surprise of phenotypic screening]. Med Sci (Paris) 2015; 31:98-104. [PMID: 25658737 DOI: 10.1051/medsci/20153101019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RNA viruses are responsible for major human diseases such as flu, bronchitis, dengue, hepatitis C or measles. They also represent an emerging threat because of increased worldwide exchanges and human populations penetrating more and more natural ecosystems. Recent progresses in our understanding of cellular pathways controlling viral replication suggest that compounds targeting host cell functions, rather than the virus itself, could inhibit a large panel of RNA viruses. In particular, several academic laboratories and private companies are now seeking molecules that stimulate the host innate antiviral response. One appealing strategy is to identify molecules that induce the large cluster of antiviral genes known as Interferon-Stimulated Genes (ISGs). To reach this goal, we have developed a phenotypic assay based on human cells transfected with a luciferase reporter gene under control of an interferon-stimulated response element (ISRE). This system was used in a high-throughput screening of chemical libraries comprising around 54,000 compounds. Among validated hits, compound DD264 was shown to boost the innate immune response in cell cultures, and displayed a broad-spectrum antiviral activity. While deciphering its mode of action, DD264 was found to target the fourth enzyme of de novo pyrimidine biosynthesis, namely the dihydroorotate dehydrogenase (DHODH). Thus, our data unraveled a yet unsuspected link between pyrimidine biosynthesis and the innate antiviral response.
Collapse
Affiliation(s)
- Pierre-Olivier Vidalain
- Unité de génomique virale et vaccination, département de virologie, Institut Pasteur, CNRS UMR 3569, 28, rue du Docteur Roux, 75724 Paris Cedex 15, France
| | - Marianne Lucas-Hourani
- Unité de génomique virale et vaccination, département de virologie, Institut Pasteur, CNRS UMR 3569, 28, rue du Docteur Roux, 75724 Paris Cedex 15, France
| | - Olivier Helynck
- Unité de chimie et biocatalyse, département de biologie structurale et chimie Institut Pasteur, CNRS UMR3523, 28, rue du Docteur Roux, 75724 Paris Cedex 15, France
| | - Frédéric Tangy
- Unité de génomique virale et vaccination, département de virologie, Institut Pasteur, CNRS UMR 3569, 28, rue du Docteur Roux, 75724 Paris Cedex 15, France
| | - Hélène Munier-Lehmann
- Unité de chimie et biocatalyse, département de biologie structurale et chimie Institut Pasteur, CNRS UMR3523, 28, rue du Docteur Roux, 75724 Paris Cedex 15, France
| |
Collapse
|
363
|
IRGM3 contributes to immunopathology and is required for differentiation of antigen-specific effector CD8+ T cells in experimental cerebral malaria. Infect Immun 2015; 83:1406-17. [PMID: 25644000 DOI: 10.1128/iai.02701-14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gamma interferon (IFN-γ) drives antiparasite responses and immunopathology during infection with Plasmodium species. Immunity-related GTPases (IRGs) are a class of IFN-γ-dependent proteins that are essential for cell autonomous immunity to numerous intracellular pathogens. However, it is currently unknown whether IRGs modulate responses during malaria. We have used the Plasmodium berghei ANKA (PbA) model in which mice develop experimental cerebral malaria (ECM) to study the roles of IRGM1 and IRGM3 in immunopathology. Induction of mRNA for Irgm1 and Irgm3 was found in the brains and spleens of infected mice at times of peak IFN-γ production. Irgm3-/- but not Irgm1-/- mice were completely protected from the development of ECM, and this protection was associated with the decreased induction of inflammatory cytokines, as well as decreased recruitment and activation of CD8+ T cells within the brain. Although antigen-specific proliferation of transferred CD8+ T cells was not diminished compared to that of wild-type recipients following PbA infection, T cells transferred into Irgm3-/- recipients showed a striking impairment of effector differentiation. Decreased induction of several inflammatory cytokines and chemokines (interleukin-6, CCL2, CCL3, and CCL4), as well as enhanced mRNA expression of type-I IFNs, was found in the spleens of Irgm3-/- mice at day 4 postinfection. Together, these data suggest that protection from ECM pathology in Irgm3-/- mice occurs due to impaired generation of CD8+ effector function. This defect is nonintrinsic to CD8+ T cells. Instead, diminished T cell responses most likely result from defective initiation of inflammatory responses in myeloid cells.
Collapse
|
364
|
Giugliano S, Kriss M, Golden-Mason L, Dobrinskikh E, Stone AEL, Soto-Gutierrez A, Mitchell A, Khetani SR, Yamane D, Stoddard M, Li H, Shaw GM, Edwards MG, Lemon SM, Gale M, Shah VH, Rosen HR. Hepatitis C virus infection induces autocrine interferon signaling by human liver endothelial cells and release of exosomes, which inhibits viral replication. Gastroenterology 2015; 148:392-402.e13. [PMID: 25447848 PMCID: PMC4765499 DOI: 10.1053/j.gastro.2014.10.040] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 10/21/2014] [Accepted: 10/28/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Liver sinusoidal endothelial cells (LSECs) make up a large proportion of the nonparenchymal cells in the liver. LSECs are involved in induction of immune tolerance, but little is known about their functions during hepatitis C virus (HCV) infection. METHODS Primary human LSECs (HLSECs) and immortalized liver endothelial cells (TMNK-1) were exposed to various forms of HCV, including full-length transmitted/founder virus, sucrose-purified Japanese fulminant hepatitis-1 (JFH-1), a virus encoding a luciferase reporter, and the HCV-specific pathogen-associated molecular pattern molecules. Cells were analyzed by confocal immunofluorescence, immunohistochemical, and polymerase chain reaction assays. RESULTS HLSECs internalized HCV, independent of cell-cell contacts; HCV RNA was translated but not replicated. Through pattern recognition receptors (Toll-like receptor 7 and retinoic acid-inducible gene 1), HCV RNA induced consistent and broad transcription of multiple interferons (IFNs); supernatants from primary HLSECs transfected with HCV-specific pathogen-associated molecular pattern molecules increased induction of IFNs and IFN-stimulated genes in HLSECs. Recombinant type I and type III IFNs strongly up-regulated HLSEC transcription of IFN λ3 (IFNL3) and viperin (RSAD2), which inhibit replication of HCV. Compared with CD8(+) T cells, HLSECs suppressed HCV replication within Huh7.5.1 cells, also inducing IFN-stimulated genes in co-culture. Conditioned media from IFN-stimulated HLSECs induced expression of antiviral genes by uninfected primary human hepatocytes. Exosomes, derived from HLSECs after stimulation with either type I or type III IFNs, controlled HCV replication in a dose-dependent manner. CONCLUSIONS Cultured HLSECs produce factors that mediate immunity against HCV. HLSECs induce self-amplifying IFN-mediated responses and release of exosomes with antiviral activity.
Collapse
Affiliation(s)
- Silvia Giugliano
- Division of Gastroenterology and Hepatology, Hepatitis C Center, Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Michael Kriss
- Division of Gastroenterology and Hepatology, Hepatitis C Center, Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Lucy Golden-Mason
- Division of Gastroenterology and Hepatology, Hepatitis C Center, Department of Medicine, University of Colorado, Denver, Aurora, Colorado; Integrated Department in Immunology: University of Colorado Denver and National Jewish Health, Denver, Colorado
| | - Evgenia Dobrinskikh
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Amy E L Stone
- Department of Immunology, University of Washington, School of Medicine, Seattle, Washington
| | - Alejandro Soto-Gutierrez
- Department of Pathology, Center for Innovative Regenerative Therapies, Department of Surgery, Transplantation Section, Children's Hospital of Pittsburgh, McGowan Institute for Regenerative Medicine and the Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Angela Mitchell
- Division of Gastroenterology and Hepatology, Hepatitis C Center, Department of Medicine, University of Colorado, Denver, Aurora, Colorado; Integrated Department in Immunology: University of Colorado Denver and National Jewish Health, Denver, Colorado
| | - Salman R Khetani
- Mechanical and Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Daisuke Yamane
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Mark Stoddard
- Department of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Hui Li
- Department of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - George M Shaw
- Department of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Michael G Edwards
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Stanley M Lemon
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Michael Gale
- Department of Immunology, University of Washington, School of Medicine, Seattle, Washington
| | - Vijay H Shah
- Mayo Clinic, Division of Gastroenterology and Hepatology, Rochester, Minnesota
| | - Hugo R Rosen
- Division of Gastroenterology and Hepatology, Hepatitis C Center, Department of Medicine, University of Colorado, Denver, Aurora, Colorado; Integrated Department in Immunology: University of Colorado Denver and National Jewish Health, Denver, Colorado; Eastern Colorado Veteran's Affairs Medical Center, Denver, Colorado.
| |
Collapse
|
365
|
Cho H, Kelsall BL. The role of type I interferons in intestinal infection, homeostasis, and inflammation. Immunol Rev 2015; 260:145-67. [PMID: 24942688 DOI: 10.1111/imr.12195] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Type I interferons are a widely expressed family of effector cytokines that promote innate antiviral and antibacterial immunity. Paradoxically, they can also suppress immune responses by driving production of anti-inflammatory cytokines, and dysregulation of these cytokines can contribute to host-mediated immunopathology and disease progression. Recent studies describe their anti-inflammatory role in intestinal inflammation and the locus containing IFNAR, a heterodimeric receptor for the type I interferons has been identified as a susceptibility region for human inflammatory bowel disease. This review focuses on the role of type I IFNs in the intestine in health and disease and their emerging role as immune modulators. Clear understanding of type I IFN-mediated immune responses may provide avenues for fine-tuning existing IFN treatment for infection and intestinal inflammation.
Collapse
Affiliation(s)
- Hyeseon Cho
- Mucosal Immunobiology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
366
|
Etheridge RD, Alaganan A, Tang K, Lou HJ, Turk BE, Sibley LD. The Toxoplasma pseudokinase ROP5 forms complexes with ROP18 and ROP17 kinases that synergize to control acute virulence in mice. Cell Host Microbe 2015; 15:537-50. [PMID: 24832449 DOI: 10.1016/j.chom.2014.04.002] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 02/24/2014] [Accepted: 03/20/2014] [Indexed: 11/28/2022]
Abstract
Polymorphic rhoptry-secreted kinases (ROPs) are essential virulence factors of Toxoplasma gondii. In particular, the pseudokinase ROP5 is the major determinant of acute virulence in mice, but the underlying mechanisms are unclear. We developed a tandem affinity protein tagging and purification approach in T. gondii and used it to show that ROP5 complexes with the active kinases ROP18 and ROP17. Biochemical analyses indicate that ROP18 and ROP17 have evolved to target adjacent and essential threonine residues in switch region I of immunity-related guanosine triphosphatases (GTPases) (IRGs), a family of host defense molecules that function to control intracellular pathogens. The combined activities of ROP17 and ROP18 contribute to avoidance of IRG recruitment to the intracellular T. gondii-containing vacuole, thus protecting the parasite from clearance in interferon-activated macrophages. These studies reveal an intricate, multilayered parasite survival strategy involving pseudokinases that regulate multiple active kinase complexes to synergistically thwart innate immunity.
Collapse
Affiliation(s)
- Ronald D Etheridge
- Department of Molecular Microbiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Aditi Alaganan
- Department of Molecular Microbiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Keliang Tang
- Department of Molecular Microbiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Hua Jane Lou
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Benjamin E Turk
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
367
|
Harms Pritchard G, Hall AO, Christian DA, Wagage S, Fang Q, Muallem G, John B, Glatman Zaretsky A, Dunn WG, Perrigoue J, Reiner SL, Hunter CA. Diverse roles for T-bet in the effector responses required for resistance to infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:1131-40. [PMID: 25556247 DOI: 10.4049/jimmunol.1401617] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The transcription factor T-bet has been most prominently linked to NK and T cell production of IFN-γ, a cytokine required for the control of a diverse array of intracellular pathogens. Indeed, in mice challenged with the parasite Toxoplasma gondii, NK and T cell responses are characterized by marked increases of T-bet expression. Unexpectedly, T-bet(-/-) mice infected with T. gondii develop a strong NK cell IFN-γ response that controls parasite replication at the challenge site, but display high parasite burdens at secondary sites colonized by T. gondii and succumb to infection. The loss of T-bet had a modest effect on T cell production of IFN-γ but did not impact on the generation of parasite-specific T cells. However, the absence of T-bet resulted in lower T cell expression of CD11a, Ly6C, KLRG-1, and CXCR3 and fewer parasite-specific T cells at secondary sites of infection, associated with a defect in parasite control at these sites. Together, these data highlight T-bet-independent pathways to IFN-γ production and reveal a novel role for this transcription factor in coordinating the T cell responses necessary to control this infection in peripheral tissues.
Collapse
Affiliation(s)
- Gretchen Harms Pritchard
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Aisling O'Hara Hall
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Sagie Wagage
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Qun Fang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Gaia Muallem
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Beena John
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Arielle Glatman Zaretsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - William G Dunn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jacqueline Perrigoue
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Steven L Reiner
- Department of Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, NY 10032; and Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, NY 10032
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
368
|
Affiliation(s)
- J. E. Gern
- Pediatrics and Medicine; University of Wisconsin-Madison; Madison WI USA
| |
Collapse
|
369
|
Renner I, Funk N, Geissler R, Friedrich S, Penzel A, Behrens SE. Antiviral interferon-beta signaling induced by designed transcription activator-like effectors (TALE). PLoS One 2014; 9:e114288. [PMID: 25470486 PMCID: PMC4255017 DOI: 10.1371/journal.pone.0114288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 11/07/2014] [Indexed: 12/24/2022] Open
Abstract
Here we show that designed transcription activator-like effectors (TALEs) that bind to defined areas of the interferon beta promoter are capable to induce IFN-beta expression and signaling in human cells. Importantly, TALE-mediated IFN-beta signaling occurs independently of pathogen pattern recognition but effectively prohibits viral RNA replication as demonstrated with a hepatitis C virus replicon. TALEs were thus indicated to be valuable tools in various applications addressing, for example, virus-host interactions.
Collapse
Affiliation(s)
- Ivonne Renner
- Institute of Biochemistry and Biotechnology, Section Microbial Biotechnology, Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Kurt-Mothes-Str. 3, D-06120, Halle/Saale, Germany
| | - Nancy Funk
- Institute of Biochemistry and Biotechnology, Section Microbial Biotechnology, Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Kurt-Mothes-Str. 3, D-06120, Halle/Saale, Germany
| | - Rene Geissler
- Institute of Biochemistry and Biotechnology, Section Microbial Biotechnology, Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Kurt-Mothes-Str. 3, D-06120, Halle/Saale, Germany
| | - Susann Friedrich
- Institute of Biochemistry and Biotechnology, Section Microbial Biotechnology, Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Kurt-Mothes-Str. 3, D-06120, Halle/Saale, Germany
| | - Anika Penzel
- Institute of Biochemistry and Biotechnology, Section Microbial Biotechnology, Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Kurt-Mothes-Str. 3, D-06120, Halle/Saale, Germany
| | - Sven-Erik Behrens
- Institute of Biochemistry and Biotechnology, Section Microbial Biotechnology, Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Kurt-Mothes-Str. 3, D-06120, Halle/Saale, Germany
| |
Collapse
|
370
|
Dong W, Wei X, Zhang F, Hao J, Huang F, Zhang C, Liang W. A dual character of flavonoids in influenza A virus replication and spread through modulating cell-autonomous immunity by MAPK signaling pathways. Sci Rep 2014; 4:7237. [PMID: 25429875 PMCID: PMC4246350 DOI: 10.1038/srep07237] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 11/06/2014] [Indexed: 12/24/2022] Open
Abstract
Flavonoids are well known as a large class of polyphenolic compounds, which have a variety of physiological activities, including anti-influenza virus activity. The influenza A/WSN/33 infected A549 cells have been used to screen anti-influenza virus drugs from natural flavonoid compounds library. Unexpectedly, some flavonoid compounds significantly inhibited virus replication, while the others dramatically promoted virus replication. In this study, we attempted to understand these differences between flavonoid compounds in their antivirus mechanisms. Hesperidin and kaempferol were chosen as representatives of both sides, each of which exhibited the opposite effects on influenza virus replication. Our investigation revealed that the opposite effects produced by hesperidin and kaempferol on influenza virus were due to inducing the opposite cell-autonomous immune responses by selectively modulating MAP kinase pathways: hesperidin up-regulated P38 and JNK expression and activation, thus resulting in the enhanced cell-autonomous immunity; while kaempferol dramatically down-regulated p38 and JNK expression and activation, thereby suppressing cell-autonomous immunity. In addition, hesperidin restricted RNPs export from nucleus by down-regulating ERK activation, but kaempferol promoted RNPs export by up-regulating ERK activation. Our findings demonstrate that a new generation of anti-influenza virus drugs could be developed based on selective modulation of MAP kinase pathways to stimulate cell-autonomous immunity.
Collapse
Affiliation(s)
- Wenjuan Dong
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Xiuli Wei
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Fayun Zhang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Junfeng Hao
- Facility of pathology, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Feng Huang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Chunling Zhang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Wei Liang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| |
Collapse
|
371
|
da Fonseca Ferreira-da-Silva M, Springer-Frauenhoff HM, Bohne W, Howard JC. Identification of the microsporidian Encephalitozoon cuniculi as a new target of the IFNγ-inducible IRG resistance system. PLoS Pathog 2014; 10:e1004449. [PMID: 25356593 PMCID: PMC4214799 DOI: 10.1371/journal.ppat.1004449] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/04/2014] [Indexed: 11/19/2022] Open
Abstract
The IRG system of IFNγ-inducible GTPases constitutes a powerful resistance mechanism in mice against Toxoplasma gondii and two Chlamydia strains but not against many other bacteria and protozoa. Why only T. gondii and Chlamydia? We hypothesized that unusual features of the entry mechanisms and intracellular replicative niches of these two organisms, neither of which resembles a phagosome, might hint at a common principle. We examined another unicellular parasitic organism of mammals, member of an early-diverging group of Fungi, that bypasses the phagocytic mechanism when it enters the host cell: the microsporidian Encephalitozoon cuniculi. Consistent with the known susceptibility of IFNγ-deficient mice to E. cuniculi infection, we found that IFNγ treatment suppresses meront development and spore formation in mouse fibroblasts in vitro, and that this effect is mediated by IRG proteins. The process resembles that previously described in T. gondii and Chlamydia resistance. Effector (GKS subfamily) IRG proteins accumulate at the parasitophorous vacuole of E. cuniculi and the meronts are eliminated. The suppression of E. cuniculi growth by IFNγ is completely reversed in cells lacking regulatory (GMS subfamily) IRG proteins, cells that effectively lack all IRG function. In addition IFNγ-induced cells infected with E. cuniculi die by necrosis as previously shown for IFNγ-induced cells resisting T. gondii infection. Thus the IRG resistance system provides cell-autonomous immunity to specific parasites from three kingdoms of life: protozoa, bacteria and fungi. The phylogenetic divergence of the three organisms whose vacuoles are now known to be involved in IRG-mediated immunity and the non-phagosomal character of the vacuoles themselves strongly suggests that the IRG system is triggered not by the presence of specific parasite components but rather by absence of specific host components on the vacuolar membrane. For some time we have studied an intracellular resistance system essential for mice to survive infection with the intracellular protozoan, Toxoplasma gondii, that is based on a family of proteins, immunity-related GTPases or IRG proteins. Immediately after the parasite enters a cell, IRG proteins accumulate on the membrane of the vacuole in which the organism resides. Within a few hours the vacuole membrane breaks down and the parasite dies. A puzzle is why this mechanism works on Toxoplasma, but only on one other organism among the many tested, namely the bacterial species, Chlamydia. What do these widely different parasites have in common that so many other bacteria and protozoa lack? Neither Toxoplasma nor Chlamydia is taken up by conventional phagocytosis. In this paper we suggest that this is an important clue by showing that a microsporidian, Encephalitozoon cuniculi, a highly-divergent fungal parasite, which also invades cells bypassing phagocytosis, is resisted by the IRG system. Therefore, we propose here the “missing self” principle: IRG proteins bind to vacuolar membranes only in the absence of a host derived inhibitor that is present on phagosomal membranes but excluded from the plasma membrane invaginated by IRG target organisms during non-phagosomal entry.
Collapse
Affiliation(s)
| | | | - Wolfgang Bohne
- Institute of Medical Microbiology and Hygiene, University of Göttingen, Göttingen, Germany
| | - Jonathan C. Howard
- Institute for Genetics, University of Cologne, Cologne, Germany
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Max-Planck Institute for Plant Breeding Research, Cologne, Germany
- * E-mail:
| |
Collapse
|
372
|
Sistigu A, Yamazaki T, Vacchelli E, Chaba K, Enot DP, Adam J, Vitale I, Goubar A, Baracco EE, Remédios C, Fend L, Hannani D, Aymeric L, Ma Y, Niso-Santano M, Kepp O, Schultze JL, Tüting T, Belardelli F, Bracci L, La Sorsa V, Ziccheddu G, Sestili P, Urbani F, Delorenzi M, Lacroix-Triki M, Quidville V, Conforti R, Spano JP, Pusztai L, Poirier-Colame V, Delaloge S, Penault-Llorca F, Ladoire S, Arnould L, Cyrta J, Dessoliers MC, Eggermont A, Bianchi ME, Pittet M, Engblom C, Pfirschke C, Préville X, Uzè G, Schreiber RD, Chow MT, Smyth MJ, Proietti E, André F, Kroemer G, Zitvogel L. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med 2014; 20:1301-9. [PMID: 25344738 DOI: 10.1038/nm.3708] [Citation(s) in RCA: 809] [Impact Index Per Article: 73.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 09/03/2014] [Indexed: 12/13/2022]
Abstract
Some of the anti-neoplastic effects of anthracyclines in mice originate from the induction of innate and T cell-mediated anticancer immune responses. Here we demonstrate that anthracyclines stimulate the rapid production of type I interferons (IFNs) by malignant cells after activation of the endosomal pattern recognition receptor Toll-like receptor 3 (TLR3). By binding to IFN-α and IFN-β receptors (IFNARs) on neoplastic cells, type I IFNs trigger autocrine and paracrine circuitries that result in the release of chemokine (C-X-C motif) ligand 10 (CXCL10). Tumors lacking Tlr3 or Ifnar failed to respond to chemotherapy unless type I IFN or Cxcl10, respectively, was artificially supplied. Moreover, a type I IFN-related signature predicted clinical responses to anthracycline-based chemotherapy in several independent cohorts of patients with breast carcinoma characterized by poor prognosis. Our data suggest that anthracycline-mediated immune responses mimic those induced by viral pathogens. We surmise that such 'viral mimicry' constitutes a hallmark of successful chemotherapy.
Collapse
Affiliation(s)
- Antonella Sistigu
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] INSERM, U1015, Villejuif, France. [3] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France. [4] Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Takahiro Yamazaki
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] INSERM, U1015, Villejuif, France. [3] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France
| | - Erika Vacchelli
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France. [3] INSERM, U848, Villejuif, France
| | - Kariman Chaba
- 1] INSERM, U848, Villejuif, France. [2] Equipe 11 Labellisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France. [3] Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - David P Enot
- 1] INSERM, U848, Villejuif, France. [2] Equipe 11 Labellisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France. [3] Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Julien Adam
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Department of Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif, France. [3] Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute, Rome, Italy
| | - Aicha Goubar
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] INSERM, U981, Villejuif, France
| | - Elisa E Baracco
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France. [3] INSERM, U848, Villejuif, France
| | - Catarina Remédios
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] INSERM, U1015, Villejuif, France. [3] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France
| | - Laetitia Fend
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] INSERM, U1015, Villejuif, France. [3] Transgene S.A., Illkirch-Graffenstaden, France
| | - Dalil Hannani
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] INSERM, U1015, Villejuif, France. [3] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France
| | - Laetitia Aymeric
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] INSERM, U1015, Villejuif, France. [3] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France
| | - Yuting Ma
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France. [3] INSERM, U848, Villejuif, France
| | - Mireia Niso-Santano
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France. [3] INSERM, U848, Villejuif, France
| | - Oliver Kepp
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France. [3] INSERM, U848, Villejuif, France
| | - Joachim L Schultze
- Laboratory for Genomics and Immunoregulation, Life and Medical Sciences (LIMES), University of Bonn, Bonn, Germany
| | - Thomas Tüting
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital Bonn, Bonn, Germany
| | - Filippo Belardelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Bracci
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina La Sorsa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Ziccheddu
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Sestili
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Urbani
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Delorenzi
- 1] SIB-Swiss Institute of Bioinformatics, Lausanne, Switzerland. [2] National Center of Competence in Research (NCCR) Molecular Oncology, Institut Suisse de Recherche Expérimentale sur le Cancer (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland. [3] Departement de Formation et Recherche, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | | | - Virginie Quidville
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Rosa Conforti
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] INSERM, U1015, Villejuif, France. [3] Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France. [4] Department of Medical Oncology, Hôpital Pitie Salpetriere, Paris, France
| | | | - Lajos Pusztai
- Yale School of Medicine, New Haven, Connecticut, USA
| | - Vichnou Poirier-Colame
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] INSERM, U1015, Villejuif, France. [3] Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France
| | - Suzette Delaloge
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Department of Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Frederique Penault-Llorca
- Department of Pathology, Jean Perrin Center, EA 4677 ERTICa, University of Auvergne, Clermont-Ferrand, France
| | - Sylvain Ladoire
- 1] Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France. [2] INSERM, CRI-866 Faculty of Medicine, Dijon, France. [3] University of Burgundy, Dijon, France
| | - Laurent Arnould
- 1] Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France. [2] INSERM, CRI-866 Faculty of Medicine, Dijon, France. [3] University of Burgundy, Dijon, France
| | - Joanna Cyrta
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Department of Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif, France
| | | | | | - Marco E Bianchi
- San Raffaele University and Scientific Institute, Milan, Italy
| | - Mikael Pittet
- 1] Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Camilla Engblom
- 1] Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Christina Pfirschke
- 1] Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Gilles Uzè
- CNRS UMR5235, University Montpellier II, Place Eugène Bataillon, Montpellier, France
| | - Robert D Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Melvyn T Chow
- Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Mark J Smyth
- 1] Queensland Institute of Medical Research, Herston, Queensland, Australia. [2] School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Enrico Proietti
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Fabrice André
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France. [3] Department of Biology and Pathology, Gustave Roussy Cancer Campus, Villejuif, France. [4] Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France. [5] INSERM, U981, Villejuif, France
| | - Guido Kroemer
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] INSERM, U848, Villejuif, France. [3] Université Paris Descartes, Sorbonne Paris Cité, Paris, France. [4] Metabolomics Platform, Gustave Roussy Cancer Campus, Villejuif, France. [5] Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Laurence Zitvogel
- 1] Gustave Roussy Cancer Campus, Villejuif, France. [2] Université Paris Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France. [3] Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France
| |
Collapse
|
373
|
Sani MRM, Moghaddam MM, Aghamollaei H, Hassanpour K, Taheri RA, Farnoosh G. Investigation of caspase-1 activity and interleukin-1β production in murine macrophage cell lines infected with Leishmania major. ASIAN PAC J TROP MED 2014; 7S1:S70-3. [PMID: 25312194 DOI: 10.1016/s1995-7645(14)60205-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 05/21/2014] [Accepted: 06/10/2014] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To investigate the caspase-1 dependent inflammatory pathway activity and interleukin-1β (IL-1β) secretion in murine macrophage cell lines J774G8 infected with Leishmania major (L. major) using caspase-1 activity assay and ELISA. METHODS Novy-MacNeal-Nicolle biphasic medium was applied to produce promastigote form of L. major. Metacyclic promastigotes in the stationary phase were applied to infect macrophage. Caspase-1 activity and IL-1β secretion were assessed by the CPP32/caspase-1 fluorometric protease assay and ELISA IL-1β kits, respectively, with time intervals of 6, 18 and 30 h. RESULTS Our study showed an increase in caspase-1 activity and IL-1β secretion in infected samples compared to non-infected macrophages. The highest increase in IL-1β production was observed after 6 h of infection. CONCLUSIONS These results arise that the activation of inflammasome pathway could be one of the innate immunity pathways against L. major.
Collapse
Affiliation(s)
| | | | - Hossein Aghamollaei
- Young Researchers and Elites Club, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Kazem Hassanpour
- Medical School, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Ramezan Ali Taheri
- Nanobiotechnology Research Centre, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Gholamreza Farnoosh
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
374
|
Tantawy MA, Hatesuer B, Wilk E, Dengler L, Kasnitz N, Weiß S, Schughart K. The interferon-induced gene Ifi27l2a is active in lung macrophages and lymphocytes after influenza A infection but deletion of Ifi27l2a in mice does not increase susceptibility to infection. PLoS One 2014; 9:e106392. [PMID: 25184786 PMCID: PMC4153650 DOI: 10.1371/journal.pone.0106392] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 08/06/2014] [Indexed: 12/20/2022] Open
Abstract
Interferons represent one of the first and essential host defense mechanisms after infection, and the activation of the IFN-pathway results in the transcriptional activation of hundreds of interferon-stimulated genes. The alpha-inducible protein 27 like 2A (Ifi27l2a) gene (human synonym: ISG12) is strongly up-regulated in the lung after influenza A infection in mice and has been shown in gene expression studies to be highly correlated to other activated genes. Therefore, we investigated the role of Ifi27l2a for the host defense to influenza A infections in more detail. RT-PCR analyses in non-infected mice demonstrated that Ifi27l2a was expressed in several tissues, including the lung. Detailed analyses of reporter gene expression in lungs from Ifi27l2a-LacZ mice revealed that Ifi27l2a was expressed in macrophages and lymphocytes but not in alveolar cells or bronchiolar epithelium cells. The number of macrophages and lymphocyte strongly increased in the lung after infection, but no significant increase in expression levels of the LacZ reporter gene was found within individual immune cells. Also, no reporter gene expression was found in bronchiolar epithelial cells, alveolar cells or infiltrating neutrophils after infection. Thus, up-regulation of Ifi27l2a in infected lungs is mainly due to the infiltration of macrophages and lymphocytes. Most surprisingly, deletion of Ifi27l2a in mouse knock-out lines did not result in increased susceptibility to infections with H1N1 or H7N7 influenza A virus compared to wild type C57BL/6N mice, suggesting a less important role of the gene for the host response to influenza infections than for bacterial infections.
Collapse
Affiliation(s)
- Mohamed A. Tantawy
- Department of Infection Genetics, Helmholtz Centre for Infection Research and University of Veterinary Medicine Hannover, Germany
| | - Bastian Hatesuer
- Department of Infection Genetics, Helmholtz Centre for Infection Research and University of Veterinary Medicine Hannover, Germany
| | - Esther Wilk
- Department of Infection Genetics, Helmholtz Centre for Infection Research and University of Veterinary Medicine Hannover, Germany
| | - Leonie Dengler
- Department of Infection Genetics, Helmholtz Centre for Infection Research and University of Veterinary Medicine Hannover, Germany
| | - Nadine Kasnitz
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Hannover, Germany
| | - Siegfried Weiß
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Hannover, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research and University of Veterinary Medicine Hannover, Germany
- University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
375
|
Bsibsi M, Peferoen LAN, Holtman IR, Nacken PJ, Gerritsen WH, Witte ME, van Horssen J, Eggen BJL, van der Valk P, Amor S, van Noort JM. Demyelination during multiple sclerosis is associated with combined activation of microglia/macrophages by IFN-γ and alpha B-crystallin. Acta Neuropathol 2014; 128:215-29. [PMID: 24997049 DOI: 10.1007/s00401-014-1317-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 06/27/2014] [Accepted: 06/27/2014] [Indexed: 01/08/2023]
Abstract
Activated microglia and macrophages play a key role in driving demyelination during multiple sclerosis (MS), but the factors responsible for their activation remain poorly understood. Here, we present evidence for a dual-trigger role of IFN-γ and alpha B-crystallin (HSPB5) in this context. In MS-affected brain tissue, accumulation of the molecular chaperone HSPB5 by stressed oligodendrocytes is a frequent event. We have shown before that this triggers a TLR2-mediated protective response in surrounding microglia, the molecular signature of which is widespread in normal-appearing brain tissue during MS. Here, we show that IFN-γ, which can be released by infiltrated T cells, changes the protective response of microglia and macrophages to HSPB5 into a robust pro-inflammatory classical response. Exposure of cultured microglia and macrophages to IFN-γ abrogated subsequent IL-10 induction by HSPB5, and strongly promoted HSPB5-triggered release of TNF-α, IL-6, IL-12, IL-1β and reactive oxygen and nitrogen species. In addition, high levels of CXCL9, CXCL10, CXL11, several guanylate-binding proteins and the ubiquitin-like protein FAT10 were induced by combined activation with IFN-γ and HSPB5. As immunohistochemical markers for microglia and macrophages exposed to both IFN-γ and HSPB5, these latter factors were found to be selectively expressed in inflammatory infiltrates in areas of demyelination during MS. In contrast, they were absent from activated microglia in normal-appearing brain tissue. Together, our data suggest that inflammatory demyelination during MS is selectively associated with IFN-γ-induced re-programming of an otherwise protective response of microglia and macrophages to the endogenous TLR2 agonist HSPB5.
Collapse
Affiliation(s)
- Malika Bsibsi
- Delta Crystallon, Zernikedreef 9, 2333, CK Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
376
|
MacMicking JD. Cell-autonomous effector mechanisms against mycobacterium tuberculosis. Cold Spring Harb Perspect Med 2014; 4:cshperspect.a018507. [PMID: 25081628 DOI: 10.1101/cshperspect.a018507] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Few pathogens run the gauntlet of sterilizing immunity like Mycobacterium tuberculosis (Mtb). This organism infects mononuclear phagocytes and is also ingested by neutrophils, both of which possess an arsenal of cell-intrinsic effector mechanisms capable of eliminating it. Here Mtb encounters acid, oxidants, nitrosylating agents, and redox congeners, often exuberantly delivered under low oxygen tension. Further pressure is applied by withholding divalent Fe²⁺, Mn²⁺, Cu²⁺, and Zn²⁺, as well as by metabolic privation in the form of carbon needed for anaplerosis and aromatic amino acids for growth. Finally, host E3 ligases ubiquinate, cationic peptides disrupt, and lysosomal enzymes digest Mtb as part of the autophagic response to this particular pathogen. It is a testament to the evolutionary fitness of Mtb that sterilization is rarely complete, although sufficient to ensure most people infected with this airborne bacterium remain disease-free.
Collapse
Affiliation(s)
- John D MacMicking
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06510
| |
Collapse
|
377
|
Use of transgenic parasites and host reporters to dissect events that promote interleukin-12 production during toxoplasmosis. Infect Immun 2014; 82:4056-67. [PMID: 25024368 DOI: 10.1128/iai.01643-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The intracellular parasite Toxoplasma gondii has multiple strategies to alter host cell function, including the injection of rhoptry proteins into the cytosol of host cells as well as bystander populations, but the consequence of these events is unclear. Here, a reporter system using fluorescent parasite strains that inject Cre recombinase with their rhoptry proteins (Toxoplasma-Cre) was combined with Ai6 Cre reporter mice to identify cells that have been productively infected, that have been rhoptry injected but lack the parasite, or that have phagocytosed T. gondii. The ability to distinguish these host-parasite interactions was then utilized to dissect the events that lead to the production of interleukin-12 p40 (IL-12p40), which is required for resistance to T. gondii. In vivo, the use of invasion-competent or invasion-inhibited (phagocytosed) parasites with IL-12p40 (YET40) reporter mice revealed that dendritic cell (DC) and macrophage populations that phagocytose the parasite or are infected can express IL-12p40 but are not the major source, as larger numbers of uninfected cells secrete this cytokine. Similarly, the use of Toxoplasma-Cre parasite strains indicated that dendritic cells and inflammatory monocytes untouched by the parasite and not cells injected by the parasite are the primary source of IL-12p40. These results imply that a soluble host or parasite factor is responsible for the bulk of IL-12p40 production in vivo, rather than cellular interactions with T. gondii that result in infection, infection and clearance, injection of rhoptry proteins, or phagocytosis of the parasite.
Collapse
|
378
|
Fert I, Cagnard N, Glatigny S, Letourneur F, Jacques S, Smith JA, Colbert RA, Taurog JD, Chiocchia G, Araujo LM, Breban M. Reverse interferon signature is characteristic of antigen-presenting cells in human and rat spondyloarthritis. Arthritis Rheumatol 2014; 66:841-51. [PMID: 24757137 DOI: 10.1002/art.38318] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 12/10/2013] [Indexed: 12/13/2022]
Abstract
OBJECTIVE In HLA-B27-transgenic rats, the development of a disorder that mimics spondyloarthritis (SpA) is highly correlated with dendritic cell (DC) dysfunction. The present study was undertaken to analyze the underlying mechanisms of this via transcriptome analysis. METHODS Transcriptome analysis of ex vivo-purified splenic CD103+CD4+ DCs from B27-transgenic rats and control rats was performed. Transcriptional changes in selected genes were confirmed by quantitative reverse transcriptase-polymerase chain reaction. A meta-analysis of our rat data and published data on gene expression in macrophages from ankylosing spondylitis (AS) patients was further performed. RESULTS Interferon (IFN) signaling was the most significantly affected pathway in DCs from B27-transgenic rats; the majority of genes connected to IFN were underexpressed in B27-transgenic rats as compared to controls. This pattern was already present at disease onset, persisted over time, and was conserved in 2 disease-prone B27-transgenic rat lines. In DCs from B27-transgenic rats, we further found an up-regulation of suppressor of cytokine signaling 3 (which may account for reverse IFN signaling) and a down-regulation of interleukin-27 (a cytokine that opposes Th17 differentiation and promotes Treg cells). The meta-analysis of data on conventional DCs from rats and data on monocyte-derived macrophages from humans revealed 7 IFN-regulated genes that were negatively regulated in both human and rat SpA (i.e., IRF1, STAT1, CXCL9, CXCL10, IFIT3, DDX60, and EPSTI1). CONCLUSION Our results suggest that expression of HLA-B27 leads to a defect in IFNγ signaling in antigen-presenting cells in both B27-transgenic rats and SpA patients, which may result in Th17 expansion and Treg cell alteration (as shown in B27-transgenic rats) and contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Ingrid Fert
- Université Paris-Descartes, Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, Institut Cochin, and INSERM U1016, CNRS (UMR 8104), Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
379
|
Ohba K, Ichiyama K, Yajima M, Gemma N, Nikaido M, Wu Q, Chong P, Mori S, Yamamoto R, Wong JEL, Yamamoto N. In vivo and in vitro studies suggest a possible involvement of HPV infection in the early stage of breast carcinogenesis via APOBEC3B induction. PLoS One 2014; 9:e97787. [PMID: 24858917 PMCID: PMC4032256 DOI: 10.1371/journal.pone.0097787] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/23/2014] [Indexed: 12/13/2022] Open
Abstract
High prevalence of infection with high-risk human papilloma virus (HPV) ranging from 25 to 100% (average 31%) was observed in breast cancer (BC) patients in Singapore using novel DNA chip technology. Early stage of BC demonstrated higher HPV positivity, and BC positive for estrogen receptor (ER) showed significantly higher HPV infection rate. This unique association of HPV with BC in vivo prompted us to investigate a possible involvement of HPV in early stages of breast carcinogenesis. Using normal breast epithelial cells stably transfected with HPV-18, we showed apparent upregulation of mRNA for the cytidine deaminase, APOBEC3B (A3B) which is reported to be a source of mutations in BC. HPV-induced A3B overexpression caused significant γH2AX focus formation, and DNA breaks which were cancelled by shRNA to HPV18 E6, E7 and A3B. These results strongly suggest an active involvement of HPV in the early stage of BC carcinogenesis via A3B induction.
Collapse
Affiliation(s)
- Kenji Ohba
- Infectious Disease program, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Koji Ichiyama
- Infectious Disease program, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Misako Yajima
- Infectious Disease program, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nobuhiro Gemma
- TOSHIBA Research & Development Center, TOSHIBA Corporation, Kawasaki, Kanagawa, Japan
| | - Masaru Nikaido
- Materials and Devices Division, TOSHIBA Corporation, Minato-ku, Tokyo, Japan
| | - Qingqing Wu
- Infectious Disease program, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - PeiPei Chong
- Infectious Disease program, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor Darul Ehsan, Malaysia
| | - Seiichiro Mori
- Pathogen Genomics Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Rain Yamamoto
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - John Eu Li Wong
- Department of Hematology-Medical Oncology, National University Cancer Institute, the Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Naoki Yamamoto
- Infectious Disease program, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
380
|
Abstract
Treatment of tuberculosis (TB) remains challenging, with lengthy treatment durations and complex drug regimens that are toxic and difficult to administer. Similar to the vast majority of antibiotics, drugs for Mycobacterium tuberculosis are directed against microbial targets. Although more effective drugs that target the bacterium may lead to faster cure of patients, it is possible that a biological limit will be reached that can be overcome only by adopting a fundamentally new treatment approach. TB regimens might be improved by including agents that target host pathways. Recent work on host-pathogen interactions, host immunity, and host-directed interventions suggests that supplementing anti-TB therapy with host modulators may lead to shorter treatment times, a reduction in lung damage caused by the disease, and a lower risk of relapse or reinfection. We undertook this review to identify molecular pathways of the host that may be amenable to modulation by small molecules for the treatment of TB. Although several approaches to augmenting standard TB treatment have been proposed, only a few have been explored in detail or advanced to preclinical and clinical studies. Our review focuses on molecular targets and inhibitory small molecules that function within the macrophage or other myeloid cells, on host inflammatory pathways, or at the level of TB-induced lung pathology.
Collapse
|
381
|
Dussurget O, Bierne H, Cossart P. The bacterial pathogen Listeria monocytogenes and the interferon family: type I, type II and type III interferons. Front Cell Infect Microbiol 2014; 4:50. [PMID: 24809023 PMCID: PMC4009421 DOI: 10.3389/fcimb.2014.00050] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/04/2014] [Indexed: 12/14/2022] Open
Abstract
Interferons (IFNs) are secreted proteins of the cytokine family that regulate innate and adaptive immune responses to infection. Although the importance of IFNs in the antiviral response has long been appreciated, their role in bacterial infections is more complex and is currently a major focus of investigation. This review summarizes our current knowledge of the role of these cytokines in host defense against the bacterial pathogen Listeria monocytogenes and highlights recent discoveries on the molecular mechanisms evolved by this intracellular bacterium to subvert IFN responses.
Collapse
Affiliation(s)
- Olivier Dussurget
- Unité des Interactions Bactéries-Cellules, Institut PasteurParis, France
- Inserm, U604Paris, France
- INRA, USC2020Paris, France
- University of Paris Diderot, Sorbonne Paris CitéParis, France
| | - Hélène Bierne
- Unité des Interactions Bactéries-Cellules, Institut PasteurParis, France
- Inserm, U604Paris, France
- INRA, USC2020Paris, France
| | - Pascale Cossart
- Unité des Interactions Bactéries-Cellules, Institut PasteurParis, France
- Inserm, U604Paris, France
- INRA, USC2020Paris, France
| |
Collapse
|
382
|
Pilla DM, Hagar JA, Haldar AK, Mason AK, Degrandi D, Pfeffer K, Ernst RK, Yamamoto M, Miao EA, Coers J. Guanylate binding proteins promote caspase-11-dependent pyroptosis in response to cytoplasmic LPS. Proc Natl Acad Sci U S A 2014; 111:6046-51. [PMID: 24715728 PMCID: PMC4000848 DOI: 10.1073/pnas.1321700111] [Citation(s) in RCA: 261] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IFN receptor signaling induces cell-autonomous immunity to infections with intracellular bacterial pathogens. Here, we demonstrate that IFN-inducible guanylate binding protein (Gbp) proteins stimulate caspase-11-dependent, cell-autonomous immunity in response to cytoplasmic LPS. Caspase-11-dependent pyroptosis is triggered in IFN-activated macrophages infected with the Gram-negative bacterial pathogen Legionella pneumophila. The rapid induction of pyroptosis in IFN-activated macrophages required a cluster of IFN-inducible Gbp proteins encoded on mouse chromosome 3 (Gbp(chr3)). Induction of pyroptosis in naive macrophages by infections with the cytosol-invading ΔsdhA L. pneumophila mutant was similarly dependent on Gbp(chr3), suggesting that these Gbp proteins play a role in the detection of bacteria accessing the cytosol. Cytoplasmic LPS derived from Salmonella ssp. or Escherichia coli has recently been shown to trigger caspase-11 activation and pyroptosis, but the cytoplasmic sensor for LPS and components of the caspase-11 inflammasome are not yet defined. We found that the induction of caspase-11-dependent pyroptosis by cytoplasmic L. pneumophila-derived LPS required Gbp(chr3) proteins. Similarly, pyroptosis induced by cytoplasmic LPS isolated from Salmonella was diminished in Gbp(chr3)-deficient macrophages. These data suggest a role for Gbp(chr3) proteins in the detection of cytoplasmic LPS and the activation of the noncanonical inflammasome.
Collapse
Affiliation(s)
- Danielle M. Pilla
- Departments of Molecular Genetics and Microbiology and Immunology, Duke University Medical Center, Durham, NC 27710
| | - Jon A. Hagar
- Department of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Arun K. Haldar
- Departments of Molecular Genetics and Microbiology and Immunology, Duke University Medical Center, Durham, NC 27710
| | - Ashley K. Mason
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21203
| | - Daniel Degrandi
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University Duesseldorf, Duesseldorf 40225, Germany; and
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University Duesseldorf, Duesseldorf 40225, Germany; and
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21203
| | - Masahiro Yamamoto
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Edward A. Miao
- Department of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jörn Coers
- Departments of Molecular Genetics and Microbiology and Immunology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
383
|
Caspase-11 activation requires lysis of pathogen-containing vacuoles by IFN-induced GTPases. Nature 2014; 509:366-70. [PMID: 24739961 DOI: 10.1038/nature13157] [Citation(s) in RCA: 366] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 02/14/2014] [Indexed: 12/28/2022]
Abstract
Lipopolysaccharide from Gram-negative bacteria is sensed in the host cell cytoplasm by a non-canonical inflammasome pathway that ultimately results in caspase-11 activation and cell death. In mouse macrophages, activation of this pathway requires the production of type-I interferons, indicating that interferon-induced genes have a critical role in initiating this pathway. Here we report that a cluster of small interferon-inducible GTPases, the so-called guanylate-binding proteins, is required for the full activity of the non-canonical caspase-11 inflammasome during infections with vacuolar Gram-negative bacteria. We show that guanylate-binding proteins are recruited to intracellular bacterial pathogens and are necessary to induce the lysis of the pathogen-containing vacuole. Lysis of the vacuole releases bacteria into the cytosol, thus allowing the detection of their lipopolysaccharide by a yet unknown lipopolysaccharide sensor. Moreover, recognition of the lysed vacuole by the danger sensor galectin-8 initiates the uptake of bacteria into autophagosomes, which results in a reduction of caspase-11 activation. These results indicate that host-mediated lysis of pathogen-containing vacuoles is an essential immune function and is necessary for efficient recognition of pathogens by inflammasome complexes in the cytosol.
Collapse
|
384
|
Enteropathogenic Escherichia coli inhibits type I interferon- and RNase L-mediated host defense to disrupt intestinal epithelial cell barrier function. Infect Immun 2014; 82:2802-14. [PMID: 24733098 DOI: 10.1128/iai.00105-14] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) primarily infects children in developing countries and causes diarrhea that can be deadly. EPEC pathogenesis occurs through type III secretion system (T3SS)-mediated injection of effectors into intestinal epithelial cells (IECs); these effectors alter actin dynamics, modulate the immune response, and disrupt tight junction (TJ) integrity. The resulting compromised barrier function and increased gastrointestinal (GI) permeability may be responsible for the clinical symptoms of infection. Type I interferon (IFN) mediates anti-inflammatory activities and serves essential functions in intestinal immunity and homeostasis; however, its role in the immune response to enteric pathogens, such as EPEC, and its impact on IEC barrier function have not been examined. Here, we report that IFN-β is induced following EPEC infection and regulates IEC TJ proteins to maintain barrier function. The EPEC T3SS effector NleD counteracts this protective activity by inhibiting IFN-β induction and enhancing tumor necrosis factor alpha to promote barrier disruption. The endoribonuclease RNase L is a key mediator of IFN induction and action that promotes TJ protein expression and IEC barrier integrity. EPEC infection inhibits RNase L in a T3SS-dependent manner, providing a mechanism by which EPEC evades IFN-induced antibacterial activities. This work identifies novel roles for IFN-β and RNase L in IEC barrier functions that are targeted by EPEC effectors to escape host defense mechanisms and promote virulence. The IFN-RNase L axis thus represents a potential therapeutic target for enteric infections and GI diseases involving compromised barrier function.
Collapse
|
385
|
A transcriptomic network identified in uninfected macrophages responding to inflammation controls intracellular pathogen survival. Cell Host Microbe 2014; 14:357-68. [PMID: 24034621 PMCID: PMC4180915 DOI: 10.1016/j.chom.2013.08.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 06/04/2013] [Accepted: 08/06/2013] [Indexed: 12/11/2022]
Abstract
Intracellular pathogens modulate host cell function to promote their survival. However, in vitro infection studies do not account for the impact of host-derived inflammatory signals. Examining the response of liver-resident macrophages (Kupffer cells) in mice infected with the parasite Leishmania donovani, we identified a transcriptomic network operating in uninfected Kupffer cells exposed to inflammation but absent from Kupffer cells from the same animal that contained intracellular Leishmania. To test the hypothesis that regulated expression of genes within this transcriptomic network might impact parasite survival, we pharmacologically perturbed the activity of retinoid X receptor alpha (RXRα), a key hub within this network, and showed that this intervention enhanced the innate resistance of Kupffer cells to Leishmania infection. Our results illustrate a broadly applicable strategy for understanding the host response to infection in vivo and identify Rxra as the hub of a gene network controlling antileishmanial resistance. Leishmania infection rapidly activates infected and uninfected Kupffer cells in mice Transcriptomics of inflamed and infected KC uncover distinct and overlapping networks A network centered on RXRα is uniquely activated in inflammation-exposed uninfected KCs Manipulation of RXRα function leads to a reduction in early parasite burden
Collapse
|
386
|
Abstract
Type I interferons (IFNs) activate intracellular antimicrobial programmes and influence the development of innate and adaptive immune responses. Canonical type I IFN signalling activates the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway, leading to transcription of IFN-stimulated genes (ISGs). Host, pathogen and environmental factors regulate the responses of cells to this signalling pathway and thus calibrate host defences while limiting tissue damage and preventing autoimmunity. Here, we summarize the signalling and epigenetic mechanisms that regulate type I IFN-induced STAT activation and ISG transcription and translation. These regulatory mechanisms determine the biological outcomes of type I IFN responses and whether pathogens are cleared effectively or chronic infection or autoimmune disease ensues.
Collapse
Affiliation(s)
- Lionel B Ivashkiv
- 1] Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York 10021, USA. [2] Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, New York 10065, USA. [3] Department of Medicine, Weill Cornell Medical College, New York, New York 10065,USA
| | - Laura T Donlin
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York 10021, USA
| |
Collapse
|
387
|
Olekhnovitch R, Ryffel B, Müller AJ, Bousso P. Collective nitric oxide production provides tissue-wide immunity during Leishmania infection. J Clin Invest 2014; 124:1711-22. [PMID: 24614106 DOI: 10.1172/jci72058] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/09/2014] [Indexed: 12/24/2022] Open
Abstract
Nitric oxide (NO) production is critical for the host defense against intracellular pathogens; however, it is unclear whether NO-dependent control of intracellular organisms depends on cell-intrinsic or cell-extrinsic activity of NO. For example, NO production by infected phagocytes may enable these cells to individually control their pathogen burden. Alternatively, the ability of NO to diffuse across cell membranes might be critical for infection control. Here, using a murine ear infection model, we found that, during infection with the intracellular parasite Leishmania major, expression of inducible NO synthase does not confer a cell-intrinsic ability to lower parasite content. We demonstrated that the diffusion of NO promotes equally effective parasite killing in NO-producing and bystander cells. Importantly, the collective production of NO by numerous phagocytes was necessary to reach an effective antimicrobial activity. We propose that, in contrast to a cell-autonomous mode of pathogen control, this cooperative mechanism generates an antimicrobial milieu that provides the basis for pathogen containment at the tissue level.
Collapse
|
388
|
Ohshima J, Lee Y, Sasai M, Saitoh T, Su Ma J, Kamiyama N, Matsuura Y, Pann-Ghill S, Hayashi M, Ebisu S, Takeda K, Akira S, Yamamoto M. Role of mouse and human autophagy proteins in IFN-γ-induced cell-autonomous responses against Toxoplasma gondii. THE JOURNAL OF IMMUNOLOGY 2014; 192:3328-35. [PMID: 24563254 DOI: 10.4049/jimmunol.1302822] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IFN-γ mediates cellular innate immunity against an intracellular parasite, Toxoplasma gondii, by inducing immunity-related GTPases such as p47 IFN-γ-regulated GTPases (IRGs) and p65 guanylate-binding proteins (GBPs), which also participate in antibacterial responses via autophagy. An essential autophagy protein, Atg5, was previously shown to play a critical role in anti-T. gondii cell-autonomous immunity. However, the involvement of other autophagy proteins remains unknown. In this study, we show that essential autophagy proteins differentially participate in anti-T. gondii cellular immunity by recruiting IFN-γ-inducible GTPases. IFN-γ-induced suppression of T. gondii proliferation and recruitment of an IRG Irgb6 and GBPs are profoundly impaired in Atg7- or Atg16L1-deficient cells. In contrast, cells lacking other essential autophagy proteins, Atg9a and Atg14, are capable of mediating the anti-T. gondii response and recruiting Irgb6 and GBPs to the parasites. Although IFN-γ also stimulates anti-T. gondii cellular immunity in humans, whether this response requires GBPs and human autophagy proteins remains to be seen. To analyze the role of human ATG16L1 and GBPs in IFN-γ-mediated anti-T. gondii responses, human cells lacking ATG16L1 or GBPs were generated by the Cas9/CRISPR genome-editing technique. Although both ATG16L1 and GBPs are dispensable for IFN-γ-induced inhibition of T. gondii proliferation in the human cells, human ATG16L1 is also required for the recruitment of GBPs. Taken together, human ATG16L1 and mouse autophagy components Atg7 and Atg16L1, but not Atg9a and Atg14, participate in the IFN-γ-induced recruitment of the immunity-related GTPases to the intracellular pathogen.
Collapse
Affiliation(s)
- Jun Ohshima
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
389
|
Kroczynska B, Mehrotra S, Arslan AD, Kaur S, Platanias LC. Regulation of interferon-dependent mRNA translation of target genes. J Interferon Cytokine Res 2014; 34:289-96. [PMID: 24559173 DOI: 10.1089/jir.2013.0148] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Interferons (IFNs) are released by cells on exposure to various stimuli, including viruses, double-stranded RNA, and other cytokines and various polypeptides. These IFNs play important physiological and pathophysiological roles in humans. Many clinical studies have established activity for these cytokines in the treatment of several malignancies, viral syndromes, and autoimmune disorders. In this review, the regulatory effects of type I and II IFN receptors on the translation-initiation process mediated by mechanistic target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK) pathways and the known mechanisms of control of mRNA translation of IFN-stimulated genes are summarized and discussed.
Collapse
Affiliation(s)
- Barbara Kroczynska
- 1 Division of Hematology-Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Medical School , Chicago, Illinois
| | | | | | | | | |
Collapse
|
390
|
Matsuzawa T, Fujiwara E, Washi Y. Autophagy activation by interferon-γ via the p38 mitogen-activated protein kinase signalling pathway is involved in macrophage bactericidal activity. Immunology 2014; 141:61-9. [PMID: 24032631 DOI: 10.1111/imm.12168] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 08/15/2013] [Accepted: 08/30/2013] [Indexed: 12/26/2022] Open
Abstract
Macrophages are involved in many essential immune functions. Their role in cell-autonomous innate immunity is reinforced by interferon-γ (IFN-γ), which is mainly secreted by proliferating type 1 T helper cells and natural killer cells. Previously, we showed that IFN-γ activates autophagy via p38 mitogen-activated protein kinase (p38 MAPK), but the biological importance of this signalling pathway has not been clear. Here, we found that macrophage bactericidal activity increased by 4 hr after IFN-γ stimulation. Inducible nitric oxide synthase (NOS2) is a major downstream effector of the Janus kinase-signal transducer and activator of transcription 1 signalling pathway that contributes to macrophage bactericidal activity via nitric oxide (NO) generation. However, no NO generation was observed after 4 hr of IFN-γ stimulation, and macrophage bactericidal activity at early stages after IFN-γ stimulation was not affected by the NOS inhibitors, NG-methyl-l-arginine acetate salt and diphenyleneiodonium chloride. These results suggest that an NOS2-independent signalling pathway is involved in IFN-γ-mediated bactericidal activity. We also found that this macrophage activity was attenuated by the addition of the p38 MAPK inhibitors, PD 169316, SB 202190, and SB 203580, or by the expression of short hairpin RNA against p38α or the essential factors for autophagy, Atg5 and Atg7. Collectively, our results suggest that the IFN-γ-mediated autophagy via p38 MAPK, without the involvement of NOS2, also contributes to the ability of macrophages to kill intracellular bacteria. These observations provide direct evidence that p38 MAPK-mediated autophagy can support IFN-γ-mediated cell-autonomous innate immunity.
Collapse
Affiliation(s)
- Takeshi Matsuzawa
- Division of Veterinary Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Osaka, Japan
| | | | | |
Collapse
|
391
|
Melo MB, Nguyen QP, Cordeiro C, Hassan MA, Yang N, McKell R, Rosowski EE, Julien L, Butty V, Dardé ML, Ajzenberg D, Fitzgerald K, Young LH, Saeij JPJ. Transcriptional analysis of murine macrophages infected with different Toxoplasma strains identifies novel regulation of host signaling pathways. PLoS Pathog 2013; 9:e1003779. [PMID: 24367253 PMCID: PMC3868521 DOI: 10.1371/journal.ppat.1003779] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 09/18/2013] [Indexed: 11/19/2022] Open
Abstract
Most isolates of Toxoplasma from Europe and North America fall into one of three genetically distinct clonal lineages, the type I, II and III lineages. However, in South America these strains are rarely isolated and instead a great variety of other strains are found. T. gondii strains differ widely in a number of phenotypes in mice, such as virulence, persistence, oral infectivity, migratory capacity, induction of cytokine expression and modulation of host gene expression. The outcome of toxoplasmosis in patients is also variable and we hypothesize that, besides host and environmental factors, the genotype of the parasite strain plays a major role. The molecular basis for these differences in pathogenesis, especially in strains other than the clonal lineages, remains largely unexplored. Macrophages play an essential role in the early immune response against T. gondii and are also the cell type preferentially infected in vivo. To determine if non-canonical Toxoplasma strains have unique interactions with the host cell, we infected murine macrophages with 29 different Toxoplasma strains, representing global diversity, and used RNA-sequencing to determine host and parasite transcriptomes. We identified large differences between strains in the expression level of known parasite effectors and large chromosomal structural variation in some strains. We also identified novel strain-specifically regulated host pathways, including the regulation of the type I interferon response by some atypical strains. IFNβ production by infected cells was associated with parasite killing, independent of interferon gamma activation, and dependent on endosomal Toll-like receptors in macrophages and the cytoplasmic receptor retinoic acid-inducible gene 1 (RIG-I) in fibroblasts.
Collapse
Affiliation(s)
- Mariane B. Melo
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts, United States of America
| | - Quynh P. Nguyen
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts, United States of America
| | - Cynthia Cordeiro
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts, United States of America
- Internal Medicine Department, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Musa A. Hassan
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts, United States of America
| | - Ninghan Yang
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts, United States of America
| | - Renée McKell
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts, United States of America
| | - Emily E. Rosowski
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts, United States of America
| | - Lindsay Julien
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts, United States of America
| | - Vincent Butty
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts, United States of America
| | - Marie-Laure Dardé
- Centre National de Référence Toxoplasmose/Toxoplasma Biological Resource Center, Centre Hospitalier-Universitaire Dupuytren, Limoges, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1094, Neuroépidémiologie Tropicale, Laboratoire de Parasitologie-Mycologie, Faculté de Médecine, Université de Limoges, Limoges, France
| | - Daniel Ajzenberg
- Centre National de Référence Toxoplasmose/Toxoplasma Biological Resource Center, Centre Hospitalier-Universitaire Dupuytren, Limoges, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 1094, Neuroépidémiologie Tropicale, Laboratoire de Parasitologie-Mycologie, Faculté de Médecine, Université de Limoges, Limoges, France
| | - Katherine Fitzgerald
- University of Massachusetts Medical School, Division of Infectious Diseases and Immunology, Worcester, Massachusetts, United States of America
| | - Lucy H. Young
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jeroen P. J. Saeij
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
392
|
Forster F, Paster W, Supper V, Schatzlmaier P, Sunzenauer S, Ostler N, Saliba A, Eckerstorfer P, Britzen-Laurent N, Schütz G, Schmid JA, Zlabinger GJ, Naschberger E, Stürzl M, Stockinger H. Guanylate binding protein 1-mediated interaction of T cell antigen receptor signaling with the cytoskeleton. THE JOURNAL OF IMMUNOLOGY 2013; 192:771-81. [PMID: 24337748 DOI: 10.4049/jimmunol.1300377] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
GTPases act as important switches in many signaling events in cells. Although small and heterotrimeric G proteins are subjects of intensive studies, little is known about the large IFN-inducible GTPases. In this article, we show that the IFN-γ-inducible guanylate binding protein 1 (GBP-1) is a regulator of T cell activation. Silencing of GBP-1 leads to enhanced activation of early T cell Ag receptor/CD3 signaling molecules, including Lck, that is translated to higher IL-2 production. Mass spectrometry analyses showed that regulatory cytoskeletal proteins, like plastin-2 that bundles actin fibers and spectrin β-chain, brain 1 that links the plasma membrane to the actin cytoskeleton, are binding partners of GBP-1. The spectrin cytoskeleton influences cell spreading and surface expression of TCR/CD3 and the leukocyte phosphatase CD45. We found higher cell spreading and enhanced surface expression of TCR/CD3 and CD45 in GBP-1 silenced T cells that explain their enhanced TCR/CD3 signaling. We conclude that GBP-1 is a downstream processor of IFN-γ via which T cells regulate cytoskeleton-dependent cell functions.
Collapse
Affiliation(s)
- Florian Forster
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
393
|
Identification of IFN-γ-producing innate B cells. Cell Res 2013; 24:161-76. [PMID: 24296781 PMCID: PMC3915900 DOI: 10.1038/cr.2013.155] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/10/2013] [Accepted: 09/16/2013] [Indexed: 12/14/2022] Open
Abstract
Although B cells play important roles in the humoral immune response and the regulation of adaptive immunity, B cell subpopulations with unique phenotypes, particularly those with non-classical immune functions, should be further investigated. By challenging mice with Listeria monocytogenes, Escherichia coli, vesicular stomatitis virus and Toll-like receptor ligands, we identified an inducible CD11a(hi)FcγRIII(hi) B cell subpopulation that is significantly expanded and produces high levels of IFN-γ during the early stage of the immune response. This subpopulation of B cells can promote macrophage activation via generating IFN-γ, thereby facilitating the innate immune response against intracellular bacterial infection. As this new subpopulation is of B cell origin and exhibits the phenotypic characteristics of B cells, we designated these cells as IFN-γ-producing innate B cells. Dendritic cells were essential for the inducible generation of these innate B cells from the follicular B cells via CD40L-CD40 ligation. Increased Bruton's tyrosine kinase activation was found to be responsible for the increased activation of non-canonical NF-κB pathway in these innate B cells after CD40 ligation, with the consequent induction of additional IFN-γ production. The identification of this new population of innate B cells may contribute to a better understanding of B cell functions in anti-infection immune responses and immune regulation.
Collapse
|
394
|
Abstract
Vasculitis of the medium and large arteries, most often presenting as giant cell arteritis (GCA), is an infrequent, but potentially fatal, type of immune-mediated vascular disease. The site of the aberrant immune reaction, the mural layers of the artery, is strictly defined by vascular dendritic cells, endothelial cells, vascular smooth muscle cells and fibroblasts, which engage in an interaction with T cells and macrophages to, ultimately, cause luminal stenosis or aneurysmal wall damage of the vessel. A multitude of effector cytokines, all known as critical mediators in host-protective immunity, have been identified in vasculitic lesions. Two dominant cytokine clusters--the IL-6-IL-17 axis and the IL-12-IFN-γ axis--have been linked to disease activity. These two clusters seem to serve different roles in the vasculitic process. The IL-6-IL-17 cluster is highly responsive to standard corticosteroid therapy, whereas the IL-12-IFN-γ cluster is resistant to steroid-mediated immunosuppression. The information exchange between vascular and immune cells and stabilization of the vasculitic process involves members of the Notch receptor and ligand family. Focusing on elements in the tissue context of GCA, instead of broadly suppressing host immunity, might enable a more tailored therapeutic approach that avoids unwanted adverse effects of aggressive immunosuppression.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, CCSR Building Room 2225, Mail Code 5166, 269 Campus Drive West, Stanford, CA 94305-5166, USA
| | | |
Collapse
|
395
|
Malireddi RKS, Kanneganti TD. Role of type I interferons in inflammasome activation, cell death, and disease during microbial infection. Front Cell Infect Microbiol 2013; 3:77. [PMID: 24273750 PMCID: PMC3824101 DOI: 10.3389/fcimb.2013.00077] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 10/24/2013] [Indexed: 12/17/2022] Open
Abstract
Interferons (IFNs) were discovered over a half-century ago as antiviral factors. The role of type I IFNs has been studied in the pathogenesis of both acute and chronic microbial infections. Deregulated type I IFN production results in a damaging cascade of cell death, inflammation, and immunological host responses that can lead to tissue injury and disease progression. Here, we summarize the role of type I IFNs in the regulation of cell death and disease during different microbial infections, ranging from viruses and bacteria to fungal pathogens. Understanding the specific mechanisms driving type I IFN-mediated cell death and disease could aid in the development of targeted therapies.
Collapse
|
396
|
Hsu YL, Shi SF, Wu WL, Ho LJ, Lai JH. Protective roles of interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) in dengue virus infection of human lung epithelial cells. PLoS One 2013; 8:e79518. [PMID: 24223959 PMCID: PMC3817122 DOI: 10.1371/journal.pone.0079518] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 10/01/2013] [Indexed: 11/18/2022] Open
Abstract
Interferons (IFNs) are critical cytokines that regulate immune response against virus infections. Dengue virus (DV) infections are a major public health concern worldwide, and especially in Asia. In the present study, we investigated the effects and mechanisms of action of IFN-induced protein with tetratricopeptide repeats 3 (IFIT3) in human lung epithelial cells. The results demonstrated that DV infection induced expression of several IFITs, including IFIT1, IFIT2, IFIT3, and IFIT5 in A549 cells. Induction of IFIT3 by DV infection was also observed in human dendritic cells. In a knockdown study, we showed that a signal transducer and activator of transcription 2 (STAT2), but not STAT1 or STAT3, regulated DV-induced IFIT3 production. By using several different methods to evaluate cell death, we demonstrated that knockdown of IFIT3 led to cellular apoptosis. Furthermore, knockdown of IFIT3 induced the expression of several apoptotic regulators such as caspase 3, caspase 8, caspase 9, and Bcl-2-associated X protein (BAX). Such apoptotic effects and mechanisms were synergistically enhanced after DV infection. Moreover, under conditions of IFIT3 deficiency, viral production increased, suggesting an anti-viral effect of IFIT3. Interestingly, DV could suppress IFN-α-induced but not IFN-γ-induced IFIT3 expression, a phenomenon similar to the regulation of STATs by DV. In conclusion, this study revealed some mechanisms of IFIT3 induction, and also demonstrated the protective roles of IFIT3 following IFN-α production in DV infection of human lung epithelial cells.
Collapse
Affiliation(s)
- Yu-Lin Hsu
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | | | | | | | | |
Collapse
|
397
|
Zhang NZ, Huang SY, Zhou DH, Chen J, Xu Y, Tian WP, Lu J, Zhu XQ. Protective immunity against Toxoplasma gondii induced by DNA immunization with the gene encoding a novel vaccine candidate: calcium-dependent protein kinase 3. BMC Infect Dis 2013; 13:512. [PMID: 24176018 PMCID: PMC4228491 DOI: 10.1186/1471-2334-13-512] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 10/29/2013] [Indexed: 11/22/2022] Open
Abstract
Background Toxoplasma gondii can infect almost all warm-blood animals including human beings. The plant-like calcium-dependent protein kinases (CDPKs) harbored by T. gondii are involved in gliding motility, cell invasion, egress and some other developmental processes, and so have been implicated as important virulence factors. Methods In the present study, we constructed a DNA vaccine expressing T. gondii CDPK3 (TgCDPK3) and evaluated its protective efficacy against T. gondii infection in Kunming mice. The gene sequence encoding TgCDPK3 was inserted into the eukaryotic expression vector pVAX I, and mice were immunized with pVAX-CDPK3 intramuscularly. Results The results showed that mice immunized with pVAX-CDPK3 developed a high level of specific antibodies and a strong lymphoproliferative response. The significantly increased levels of IFN-γ, IL-2, IL-12 (p70) and IL-23 and high ratio of IgG2a to IgG1 antibody titers indicated that a Th1 type response was elicited after immunization with pVAX-CDPK3. Furthermore, the percentage of CD4+ T cells in mice vaccinated with pVAX-CDPK3 was significantly increased. After lethal challenge with the tachyzoites of the virulent T. gondii RH strain, the mice immunized with pVAX-CDPK3 prolonged the survival time from 10 days to 24 days (13.5 ± 4.89) compared to untreated mice or those received PBS or pVAX I which died within 7 days (P < 0.05). In chronic infection model (10 cysts of the T. gondii PRU strain), the numbers of brain cysts of the mice immunized with pVAX-CDPK3 reduced significantly when compared with those in control groups (P < 0.05), and the rate of reduction could reach to about 50%. Conclusions TgCDPK3 can generate protective immunity against acute and chronic T. gondii infection in Kunming mice and is a promising vaccine candidate for further development of an effective vaccine against T. gondii.
Collapse
Affiliation(s)
| | - Si-Yang Huang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China.
| | | | | | | | | | | | | |
Collapse
|
398
|
Lucas-Hourani M, Dauzonne D, Jorda P, Cousin G, Lupan A, Helynck O, Caignard G, Janvier G, André-Leroux G, Khiar S, Escriou N, Desprès P, Jacob Y, Munier-Lehmann H, Tangy F, Vidalain PO. Inhibition of pyrimidine biosynthesis pathway suppresses viral growth through innate immunity. PLoS Pathog 2013; 9:e1003678. [PMID: 24098125 PMCID: PMC3789760 DOI: 10.1371/journal.ppat.1003678] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 08/16/2013] [Indexed: 12/19/2022] Open
Abstract
Searching for stimulators of the innate antiviral response is an appealing approach to develop novel therapeutics against viral infections. Here, we established a cell-based reporter assay to identify compounds stimulating expression of interferon-inducible antiviral genes. DD264 was selected out of 41,353 compounds for both its immuno-stimulatory and antiviral properties. While searching for its mode of action, we identified DD264 as an inhibitor of pyrimidine biosynthesis pathway. This metabolic pathway was recently identified as a prime target of broad-spectrum antiviral molecules, but our data unraveled a yet unsuspected link with innate immunity. Indeed, we showed that DD264 or brequinar, a well-known inhibitor of pyrimidine biosynthesis pathway, both enhanced the expression of antiviral genes in human cells. Furthermore, antiviral activity of DD264 or brequinar was found strictly dependent on cellular gene transcription, nuclear export machinery, and required IRF1 transcription factor. In conclusion, the antiviral property of pyrimidine biosynthesis inhibitors is not a direct consequence of pyrimidine deprivation on the virus machinery, but rather involves the induction of cellular immune response. Our therapeutic arsenal to treat viral diseases is extremely limited, and there is a critical need for molecules that could be used against multiple viruses. Among possible strategies, there is a growing interest for molecules stimulating cellular defense mechanisms. We recently developed a functional assay to identify stimulators of antiviral genes, and selected compound DD264 from a chemical library using this approach. While searching for its mode of action, we identified this molecule as an inhibitor of pyrimidine biosynthesis, a metabolic pathway that fuels the cell with pyrimidine nucleobases for both DNA and RNA synthesis. Interestingly, it was recently shown that inhibitors of this metabolic pathway prevent the replication of RNA viruses. Here, we established a functional link between pyrimidine biosynthesis pathway and the induction of antiviral genes, and demonstrated that pyrimidine biosynthesis inhibitors like DD264 or brequinar critically rely on cellular immune response to inhibit virus growth. Thus, pyrimidine deprivation is not directly responsible for the antiviral activity of pyrimidine biosynthesis inhibitors, which rather involves the induction of a metabolic stress and subsequent triggering of cellular defense mechanisms.
Collapse
Affiliation(s)
- Marianne Lucas-Hourani
- Institut Pasteur, Unité de Génomique Virale et Vaccination, Paris, France
- CNRS, UMR3569, Paris, France
| | - Daniel Dauzonne
- Institut Curie, Centre de Recherche, Paris, France
- CNRS, UMR176, Paris, France
| | - Pierre Jorda
- Institut Curie, Centre de Recherche, Paris, France
- CNRS, UMR176, Paris, France
| | - Gaëlle Cousin
- Institut Curie, Centre de Recherche, Paris, France
- CNRS, UMR176, Paris, France
| | - Alexandru Lupan
- Institut Pasteur, Unité de Chimie et Biocatalyse, Paris, France
- CNRS, UMR3523, Paris, France
| | - Olivier Helynck
- Institut Pasteur, Unité de Chimie et Biocatalyse, Paris, France
- CNRS, UMR3523, Paris, France
| | - Grégory Caignard
- Institut Pasteur, Unité de Génomique Virale et Vaccination, Paris, France
- CNRS, UMR3569, Paris, France
| | - Geneviève Janvier
- Institut Pasteur, Unité de Génomique Virale et Vaccination, Paris, France
- CNRS, UMR3569, Paris, France
| | - Gwénaëlle André-Leroux
- Institut Pasteur, Unité de Biochimie Structurale, Paris, France
- CNRS, UMR 3528, Paris, France
| | - Samira Khiar
- Institut Pasteur, Unité de Génomique Virale et Vaccination, Paris, France
- CNRS, UMR3569, Paris, France
| | - Nicolas Escriou
- Institut Pasteur, Unité de Génomique Virale et Vaccination, Paris, France
- CNRS, UMR3569, Paris, France
| | - Philippe Desprès
- Institut Pasteur, Unité Interactions moléculaires Flavivirus-Hôtes, Paris, France
| | - Yves Jacob
- CNRS, UMR3569, Paris, France
- Institut Pasteur, Unité de Génétique Moléculaire des Virus à ARN, Paris, France
- Dana-Farber Cancer Institute, Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Boston, Massachusetts, United States of America
| | - Hélène Munier-Lehmann
- Institut Pasteur, Unité de Chimie et Biocatalyse, Paris, France
- CNRS, UMR3523, Paris, France
- * E-mail: (HML); (FT); (POV)
| | - Frédéric Tangy
- Institut Pasteur, Unité de Génomique Virale et Vaccination, Paris, France
- CNRS, UMR3569, Paris, France
- * E-mail: (HML); (FT); (POV)
| | - Pierre-Olivier Vidalain
- Institut Pasteur, Unité de Génomique Virale et Vaccination, Paris, France
- CNRS, UMR3569, Paris, France
- * E-mail: (HML); (FT); (POV)
| |
Collapse
|
399
|
Identification of TRIM22 single nucleotide polymorphisms associated with loss of inhibition of HIV-1 transcription and advanced HIV-1 disease. AIDS 2013; 27:2335-44. [PMID: 23921607 DOI: 10.1097/01.aids.0000432474.76873.5f] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE(S) Tripartite motif-containing 22 (TRIM22) is an interferon-induced protein that inhibits HIV-1 transcription and replication in vitro. Two single nucleotide missense polymorphisms rs7935564A/G (SNP-1) and rs1063303C/G (SNP-2) characterize the coding sequence of human TRIM22 gene. We tested whether these variants affected the inhibitory effect of TRIM22 on HIV-1 replication and transcription and their potential association with HIV-1 disease. DESIGN The allelic discrimination was determined in 182 HIV-1-negative and among HIV-1-positive individuals with advanced disease progression (advanced progressors; n = 57), normal progressors (n = 76), and long-term nonprogressors (LTNPs; n = 95). METHODS Renilla luciferase activity was measured after infection of activated peripheral blood mononuclear cells (PBMCs) from an additional group of 61 blood donors with a recombinant HIV-1. HIV-1-long terminal repeat (LTR)-driven luciferase activity was tested in the presence of plasmid expressing TRIM22 variants in 293T cells. The SNP genotyping was determined by TaqMan assay. RESULTS HIV-1 replication was more efficient in PBMCs from donors with SNP-1G and SNP-2G than from those with SNP-1A and SNP-2C alleles. Consistently, TRIM22-GG enhanced, whereas TRIM22-AC restricted basal HIV-1 LTR-driven transcription. In vivo, SNP-1G homozygotes and A/G heterozygotes were more frequent in advanced progressors than in LTNPs [odds ratio (OR) = 2.072, P = 0.005] or in normal progressors (OR = 1.809, P = 0.022); in contrast, SNP-2 was not associated with any state of HIV-1 disease progression. Although SNP-2 distribution was similar among the groups, TRIM22-GG haplotype was found more frequently in advanced progressors than in LTNPs (P = 0.02). CONCLUSION TRIM22 genetic diversity affects HIV-1 replication in vitro and it is a potentially novel determinant of HIV-1 disease severity.
Collapse
|
400
|
Affiliation(s)
- Jörn Coers
- Departments of Molecular Genetics and Microbiology and Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|