2201
|
Molecular and cellular insights into T cell exhaustion. NATURE REVIEWS. IMMUNOLOGY 2015. [PMID: 26205583 DOI: 10.1038/nri3862.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In chronic infections and cancer, T cells are exposed to persistent antigen and/or inflammatory signals. This scenario is often associated with the deterioration of T cell function: a state called 'exhaustion'. Exhausted T cells lose robust effector functions, express multiple inhibitory receptors and are defined by an altered transcriptional programme. T cell exhaustion is often associated with inefficient control of persisting infections and tumours, but revitalization of exhausted T cells can reinvigorate immunity. Here, we review recent advances that provide a clearer molecular understanding of T cell exhaustion and reveal new therapeutic targets for persisting infections and cancer.
Collapse
|
2202
|
Shi L, Song L, Maurer K, Sharp J, Zhang Z, Sullivan KE. Endotoxin tolerance in monocytes can be mitigated by α2-interferon. J Leukoc Biol 2015. [PMID: 26206900 DOI: 10.1189/jlb.4a0914-450rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Endotoxin tolerance is characterized by diminished expression of inflammatory cytokines after sequential exposure to Toll-like receptor stimuli. Many mechanisms contribute to tolerance; however, chromatin remodeling appears to be the most significant regulator. The type II interferon, IFN-γ, has been recognized as being able to reverse or abrogate the establishment of tolerance. Type I interferons have not been investigated previously, and they bind a distinct receptor. We found that α2-interferon was able to abrogate or diminish tolerance by endotoxin, as defined by measuring mRNA levels at recognized tolerance targets. We also found that α2-interferon treatment during tolerization was associated with increased H3K4me3 and H3K4me2 levels at promoters of tolerance targets in THP1 cells. These marks were normalized after exposure of the cells to α2-interferon. Interferon regulatory factor 1 is a transcription factor activated and induced by types I and II interferons. We found recruitment of this transcription factor paralleled tolerance and inhibition of tolerance at target genes. Therefore, there are at least 2 distinct pathways by which endotoxin tolerance may be mitigated. A type I interferon, in spite of binding to a different receptor, was just as able to inhibit tolerance as the type II interferon and also appeared to act by modifying chromatin at tolerance target genes.
Collapse
Affiliation(s)
- Lihua Shi
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Li Song
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kelly Maurer
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - James Sharp
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Zhe Zhang
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kathleen E Sullivan
- *Division of Allergy Immunology and The Center for Biomedical Informatics, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2203
|
Bossini-Castillo L, López-Isac E, Martín J. Immunogenetics of systemic sclerosis: Defining heritability, functional variants and shared-autoimmunity pathways. J Autoimmun 2015. [PMID: 26212856 DOI: 10.1016/j.jaut.2015.07.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Systemic sclerosis (SSc) is a clinically heterogeneous connective tissue disorder of complex etiology. The development of large-scale genetic studies, such as genome-wide association studies (GWASs) or the Immunochip platform, has achieved remarkable progress in the knowledge of the genetic background of SSc. Herein, we provide an updated picture SSc genetic factors, offering an insight into their role in pathogenic mechanisms that characterize the disease. We review the most recent findings in the HLA region and the well-established non-HLA loci. Up to 18 non-HLA risk factors fulfilled the selected criteria and they were classified according to their role in the innate or adaptive immune response, in apoptosis, autophagy or fibrosis. Additionally, SSc heritability has remained as a controversial question since twin studies provided low SSc heritability estimates. However, we have recalculated the lower bond of narrow sense SSc heritability using GWAS data. Remarkably, our results suggest a greater influence of genetics on SSc than previously reported. Furthermore, we also offer a functional classification of SSc-associated SNPs and their proxies, based on annotated data, to provide clues for the identification of causal variants in these loci. Finally, we explore the genetic overlap between SSc and other autoimmune diseases (ADs). The vast majority of SSc risk loci are shared with at least one additional AD, being the overlap between SSc and systemic lupus erythematous the largest. Nevertheless, we found that an important portion of SSc risk factors are also common to rheumatoid arthritis or primary biliary cirrhosis. Considering all these evidences, we are confident that future research will be successful in understanding the relevant altered pathways in SSc and in identifying new biomarkers and therapeutic targets for the disease.
Collapse
Affiliation(s)
- Lara Bossini-Castillo
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Parque Tecnológico de La Salud (PTS), Granada, Spain.
| | - Elena López-Isac
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Parque Tecnológico de La Salud (PTS), Granada, Spain
| | - Javier Martín
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas (IPBLN-CSIC), Parque Tecnológico de La Salud (PTS), Granada, Spain.
| |
Collapse
|
2204
|
Ramos I, Fernandez-Sesma A. Modulating the Innate Immune Response to Influenza A Virus: Potential Therapeutic Use of Anti-Inflammatory Drugs. Front Immunol 2015; 6:361. [PMID: 26257731 PMCID: PMC4507467 DOI: 10.3389/fimmu.2015.00361] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/04/2015] [Indexed: 12/27/2022] Open
Abstract
Infection by influenza A viruses (IAV) is frequently characterized by robust inflammation that is usually more pronounced in the case of avian influenza. It is becoming clearer that the morbidity and pathogenesis caused by IAV are consequences of this inflammatory response, with several components of the innate immune system acting as the main players. It has been postulated that using a therapeutic approach to limit the innate immune response in combination with antiviral drugs has the potential to diminish symptoms and tissue damage caused by IAV infection. Indeed, some anti-inflammatory agents have been shown to be effective in animal models in reducing IAV pathology as a proof of principle. The main challenge in developing such therapies is to selectively modulate signaling pathways that contribute to lung injury while maintaining the ability of the host cells to mount an antiviral response to control virus replication. However, the dissection of those pathways is very complex given the numerous components regulated by the same factors (i.e., NF kappa B transcription factors) and the large number of players involved in this regulation, some of which may be undescribed or unknown. This article provides a comprehensive review of the current knowledge regarding the innate immune responses associated with tissue damage by IAV infection, the understanding of which is essential for the development of effective immunomodulatory drugs. Furthermore, we summarize the recent advances on the development and evaluation of such drugs as well as the lessons learned from those studies.
Collapse
Affiliation(s)
- Irene Ramos
- Department of Microbiology, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| | - Ana Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| |
Collapse
|
2205
|
Kim TH, Zhou H. Functional Analysis of Chicken IRF7 in Response to dsRNA Analog Poly(I:C) by Integrating Overexpression and Knockdown. PLoS One 2015; 10:e0133450. [PMID: 26186542 PMCID: PMC4505898 DOI: 10.1371/journal.pone.0133450] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/25/2015] [Indexed: 12/13/2022] Open
Abstract
In order to develop novel strategies to protect against increasingly virulent bird-linked pathogens, a better understanding of the avian antiviral response mechanism is essential. Type I interferons (IFNs) are recognized as the first line of defense in a host's antiviral response; and it has been suggested that IRF7, a member of the IFN regulatory factor (IRF) family, plays an important role in modulating the immune response to avian influenza virus infection in chickens. The objective of this study was to identify candidate genes and pathways associated with IRF7 regulation at the transcriptome level as a first step towards elucidating the underlying cellular mechanisms of IRF7 modulation in the chicken antiviral response. IRF7 overexpression and knockdown DF-1 cell lines were established and stimulated by various pathogen-associated molecular patterns. Significant IRF7 and type I IFN expression changes were observed in both the IRF7 overexpression cell line and the IRF7 knockdown cell line upon exposure to the double stranded RNA (dsRNA) analog poly(I:C). Using RNA-seq based transcriptome analysis, we identified potential novel genes that IRF7 may help regulate as part of the host immune response to dsRNA; potential biomarkers and therapeutic targets revealed as a result of this study warrant further investigation. Based on our results, we suggest that IRF7 may have conserved functional activity in the avian antiviral response, and plays a crucial role in type I IFN regulation.
Collapse
Affiliation(s)
- Tae Hyun Kim
- Integrative Genetics and Genomics Graduate Group, Department of Animal Science, University of California, Davis, California, United States of America
| | - Huaijun Zhou
- Integrative Genetics and Genomics Graduate Group, Department of Animal Science, University of California, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
2206
|
The DNA damage response and immune signaling alliance: Is it good or bad? Nature decides when and where. Pharmacol Ther 2015; 154:36-56. [PMID: 26145166 DOI: 10.1016/j.pharmthera.2015.06.011] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 06/10/2015] [Indexed: 12/15/2022]
Abstract
The characteristic feature of healthy living organisms is the preservation of homeostasis. Compelling evidence highlight that the DNA damage response and repair (DDR/R) and immune response (ImmR) signaling networks work together favoring the harmonized function of (multi)cellular organisms. DNA and RNA viruses activate the DDR/R machinery in the host cells both directly and indirectly. Activation of DDR/R in turn favors the immunogenicity of the incipient cell. Hence, stimulation of DDR/R by exogenous or endogenous insults triggers innate and adaptive ImmR. The immunogenic properties of ionizing radiation, a prototypic DDR/R inducer, serve as suitable examples of how DDR/R stimulation alerts host immunity. Thus, critical cellular danger signals stimulate defense at the systemic level and vice versa. Disruption of DDR/R-ImmR cross talk compromises (multi)cellular integrity, leading to cell-cycle-related and immune defects. The emerging DDR/R-ImmR concept opens up a new avenue of therapeutic options, recalling the Hippocrates quote "everything in excess is opposed by nature."
Collapse
|
2207
|
Rivadeneyra L, Pozner RG, Meiss R, Fondevila C, Gómez RM, Schattner M. Poly (I:C) downregulates platelet production and function through type I interferon. Thromb Haemost 2015; 114:982-93. [PMID: 26134179 DOI: 10.1160/th14-11-0951] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 05/24/2015] [Indexed: 12/21/2022]
Abstract
Thrombocytopenia is a frequent complication of viral infections; the underlying mechanisms appear to depend on the identity of the virus involved. Previous research, including reports from our group, indicates that as well as having antiviral activity type I interferons (IFN I) selectively downregulate platelet production. In this study we extended understanding of the role of endogenous IFN I in megakaryo/thrombopoiesis by evaluating platelet and megakaryocyte physiology in mice treated with polyinosinic:polycytidylic acid [poly (I:C)], a synthetic analogue of double-stranded RNA, Toll-like receptor-3 ligand and strong IFNβ inducer. Mice-treated with poly (I:C) showed thrombocytopaenia, an increase in mean platelet volume and abnormal haemostatic and inflammatory platelet-mediated functionality, indicated by decreased fibrinogen binding and platelet adhesion, prolonged tail bleeding times and impaired P-Selectin externalisation, RANTES release and thrombin-induced platelet-neutrophil aggregate formation. These changes were associated with an increase in size and an abnormal distribution of bone marrow megakaryocytes within the vascular niche and were directly correlated with the plasmatic and bone marrow IFNβ levels. All these effects were absent in genetically modified mice lacking the IFN I receptor. Our results suggest that IFN I is the central mediator of poly (I:C)-induced thrombocytopenia and platelet dysfunction and indicate that these abnormalities are due to changes in the last stages of megakaryocyte development. These data provide new evidence for the role of IFN I in megakaryocyte distribution in the bone marrow niches and its influence on thrombopoiesis and haemostasis.
Collapse
Affiliation(s)
| | | | | | | | | | - Mirta Schattner
- Mirta Schattner, Institute of Experimental Medicine, Pacheco de Melo 3081, Buenos Aires 1425, Argentina, Tel.: +5411 48073926, Fax: +5411 48050712,
| |
Collapse
|
2208
|
Schirrmacher V. Signaling through RIG-I and type I interferon receptor: Immune activation by Newcastle disease virus in man versus immune evasion by Ebola virus (Review). Int J Mol Med 2015; 36:3-10. [PMID: 25998621 DOI: 10.3892/ijmm.2015.2213] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/24/2015] [Indexed: 11/06/2022] Open
Abstract
In this review, two types of RNA viruses are compared with regard to the type I interferon (IFN) response in order to obtain a better understanding of the molecular mechanisms of immune activation or evasion. Upon human infection, both viruses exert either beneficial or detrimental effects. The Newcastle disease virus (NDV), is a type strain for avian paramyxoviruses, while the Ebola virus (EBOV), is a virus affecting primates. During evolution, both viruses specifically adapted to their respective hosts, acquiring sophisticated viral escape mechanisms. Two types of receptors play an important role in the life cycle of these two viruses: cytoplasmic retinoic acid‑inducible gene I (RIG‑I) and membrane expressed type I IFN receptor (IFNAR). In mouse and human cells, NDV is a strong inducer of the type I IFN response. The early phase of this is initiated by signaling through RIG‑I and the late response by signaling through IFNAR. EBOV does not induce type I IFN responses in humans as it has viral proteins that specifically and strongly interfere with RIG‑I and IFNAR signaling, as well as immune activation. In this review, we discuss whether the beneficial effects of one virus can be exploited in the fight against the detrimental effects of the other.
Collapse
Affiliation(s)
- Volker Schirrmacher
- Division of Tumorimmunology, The Immunological and Oncological Center of Cologne, D‑50674 Cologne, Germany
| |
Collapse
|
2209
|
Interferon-γ regulates cellular metabolism and mRNA translation to potentiate macrophage activation. Nat Immunol 2015; 16:838-849. [PMID: 26147685 PMCID: PMC4509841 DOI: 10.1038/ni.3205] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 05/19/2015] [Indexed: 12/14/2022]
Abstract
Interferon-γ (IFN-γ) primes macrophages for enhanced inflammatory activation by Toll-like receptors (TLRs) and microbial killing, but little is known about the regulation of cell metabolism or mRNA translation during priming. We found that IFN-γ regulates human macrophage metabolism and translation by targeting the kinases mTORC1 and MNK that both converge on the selective regulator of translation initiation eIF4E. Physiological downregulation of mTORC1 by IFN-γ was associated with autophagy and translational suppression of repressors of inflammation such as HES1. Genome-wide ribosome profiling in TLR2-stimulated macrophages revealed that IFN-γ selectively modulates the macrophage translatome to promote inflammation, further reprogram metabolic pathways, and modulate protein synthesis. These results add IFN-γ-mediated metabolic reprogramming and translational regulation as key components of classical inflammatory macrophage activation.
Collapse
|
2210
|
Tetherin Sensitivity of Influenza A Viruses Is Strain Specific: Role of Hemagglutinin and Neuraminidase. J Virol 2015; 89:9178-88. [PMID: 26109730 DOI: 10.1128/jvi.00615-15] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/15/2015] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED The expression of the antiviral host cell factor tetherin is induced by interferon and can inhibit the release of enveloped viruses from infected cells. The Vpu protein of HIV-1 antagonizes the antiviral activity of tetherin, and tetherin antagonists with Vpu-like activity have been identified in other viruses. In contrast, it is incompletely understood whether tetherin inhibits influenza A virus (FLUAV) release and whether FLUAV encodes tetherin antagonists. Here, we show that release of several laboratory-adapted FLUAV strains and a seasonal FLUAV strain is inhibited by tetherin, while pandemic FLUAV A/Hamburg/4/2009 is resistant. Studies with a virus-like particle system and analysis of reassortant viruses provided evidence that the viral hemagglutinin (HA) is an important determinant of tetherin antagonism but requires the presence of its cognate neuraminidase (NA) to inhibit tetherin. Finally, tetherin antagonism by FLUAV was dependent on the virion context, since retrovirus release from tetherin-positive cells was not rescued, and correlated with an HA- and NA-dependent reduction in tetherin expression. In sum, our study identifies HA and NA proteins of certain pandemic FLUAV as tetherin antagonists, which has important implications for understanding FLUAV pathogenesis. IMPORTANCE Influenza A virus (FLUAV) infection is responsible for substantial global morbidity and mortality, and understanding how the virus evades the immune defenses of the host may uncover novel targets for antiviral intervention. Tetherin is an antiviral effector molecule of the innate immune system which can contribute to control of viral invasion. However, it has been unclear whether FLUAV is inhibited by tetherin and whether these viruses encode tetherin-antagonizing proteins. Our observation that several pandemic FLUAV strains can counteract tetherin via their HA and NA proteins identifies these proteins as novel tetherin antagonists and indicates that HA/NA-dependent inactivation of innate defenses may contribute to the efficient spread of pandemic FLUAV.
Collapse
|
2211
|
Xiao Y, Jin J, Zou Q, Hu H, Cheng X, Sun SC. Peli1 negatively regulates type I interferon induction and antiviral immunity in the CNS. Cell Biosci 2015; 5:34. [PMID: 26131354 PMCID: PMC4486122 DOI: 10.1186/s13578-015-0024-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/12/2015] [Indexed: 12/31/2022] Open
Abstract
Background Type I interferons (IFN-Is) serve as mediators of antiviral innate immunity and also regulate adaptive immune responses. The molecular mechanism that regulates virus-induced IFN-I production, particularly in tissue-resident immune cells, is incompletely understood. Results Here we identified the E3 ubiquitin ligase Peli1 as a negative regulator of IFN-I induction in microglia, innate immune cells of the central nervous system (CNS). Peli1 deficiency profoundly promoted IFN-β expression in microglia in response to in vitro stimulation by toll-like receptor (TLR) ligands or a CNS-tropic virus, the vascular stomatitis virus (VSV). Upon intranasal infection with VSV, the Peli1-deficient mice displayed heightened in vivo IFN-I responses in the CNS, coupled with reduced brain viral titer and increased survival rate. Conclusions These results establish Peli1 as an innate immune regulator in the CNS that modulates the threshold of IFN-I responses against viral infections.
Collapse
Affiliation(s)
- Yichuan Xiao
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX 77030 USA ; Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Science/Shanghai Jiao Tong University School of Medicine, Shanghai, 200031 China
| | - Jin Jin
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX 77030 USA
| | - Qiang Zou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX 77030 USA
| | - Hongbo Hu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX 77030 USA
| | - Xuhong Cheng
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX 77030 USA
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX 77030 USA ; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030 USA
| |
Collapse
|
2212
|
Stifter SA, Feng CG. Interfering with immunity: detrimental role of type I IFNs during infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:2455-65. [PMID: 25747907 DOI: 10.4049/jimmunol.1402794] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type I IFNs are known to inhibit viral replication and mediate protection against viral infection. However, recent studies revealed that these cytokines play a broader and more fundamental role in host responses to infections beyond their well-established antiviral function. Type I IFN induction, often associated with microbial evasion mechanisms unique to virulent microorganisms, is now shown to increase host susceptibility to a diverse range of pathogens, including some viruses. This article presents an overview of the role of type I IFNs in infections with bacterial, fungal, parasitic, and viral pathogens and discusses the key mechanisms mediating the regulatory function of type I IFNs in pathogen clearance and tissue inflammation.
Collapse
Affiliation(s)
- Sebastian A Stifter
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney 2006, New South Wales, Australia; and Mycobacterial Research Program, Centenary Institute, Sydney 2050, New South Wales, Australia
| | - Carl G Feng
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney 2006, New South Wales, Australia; and Mycobacterial Research Program, Centenary Institute, Sydney 2050, New South Wales, Australia
| |
Collapse
|
2213
|
Li G, Ju J, Weyand CM, Goronzy JJ. Age-Associated Failure To Adjust Type I IFN Receptor Signaling Thresholds after T Cell Activation. THE JOURNAL OF IMMUNOLOGY 2015; 195:865-74. [PMID: 26091718 DOI: 10.4049/jimmunol.1402389] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 05/20/2015] [Indexed: 12/23/2022]
Abstract
With increasing age, naive CD4 T cells acquire intrinsic defects that compromise their ability to respond and differentiate. Type I IFNs, pervasive constituents of the environment in which adaptive immune responses occur, are known to regulate T cell differentiation and survival. Activated naive CD4 T cells from older individuals have reduced responses to type I IFN, a defect that develops during activation and that is not observed in quiescent naive CD4 T cells. Naive CD4 T cells from young adults upregulate the expression of STAT1 and STAT5 after activation, lowering their threshold to respond to type I IFN stimulation. The heightened STAT signaling is critical to maintain the expression of CD69 that regulates lymphocyte egress and the ability to produce IL-2 and to survive. Although activation of T cells from older adults also induces transcription of STAT1 and STAT5, failure to exclude SHP-1 from the signaling complex blunts their type I IFN response. In summary, our data show that type I IFN signaling thresholds in naive CD4 T cells after activation are dynamically regulated to respond to environmental cues for clonal expansion and memory cell differentiation. Naive CD4 T cells from older adults have a defect in this threshold calibration. Restoring their ability to respond to type I IFN emerges as a promising target to restore T cell responses and to improve the induction of T cell memory.
Collapse
Affiliation(s)
- Guangjin Li
- Department of Medicine, Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304; and Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jihang Ju
- Department of Medicine, Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304; and Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Cornelia M Weyand
- Department of Medicine, Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304; and Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jörg J Goronzy
- Department of Medicine, Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304; and Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
2214
|
Shen YJ, Ho SSW, Tan NY, Koo CX, Khatoo M, Cheung FSG, Gasser S. Genome-derived cytosolic DNA contributes to type I interferon expression and immunogenicity of B-cell lymphoma cells. Cytokine 2015; 76:581-582. [PMID: 26070935 DOI: 10.1016/j.cyto.2015.05.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 01/21/2023]
Abstract
We recently provided evidence that genome-derived DNA is present in the cytosol of many tumor cells. Genomic loci that give rise to cytosolic DNA can potentially form non-B DNA structures including triple-stranded RNA:DNA structures (R-loops). The RNA:DNA-specific endonuclease RNaseh1 reduced the levels of cytosolic DNA and type I interferon-dependent rejection of B-cell lymphoma suggesting that cytosolic DNA may contribute to immune surveillance of B-cell lymphoma.
Collapse
Affiliation(s)
- Yu J Shen
- Immunology Programme and Department of Microbiology, Centre for Life Science, National University of Singapore, 117456, Singapore; NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, 117456, Singapore
| | - Samantha S W Ho
- Immunology Programme and Department of Microbiology, Centre for Life Science, National University of Singapore, 117456, Singapore
| | - Nikki Y Tan
- Immunology Programme and Department of Microbiology, Centre for Life Science, National University of Singapore, 117456, Singapore
| | - Christine Xing'Er Koo
- Immunology Programme and Department of Microbiology, Centre for Life Science, National University of Singapore, 117456, Singapore; NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, 117456, Singapore
| | - Muznah Khatoo
- Immunology Programme and Department of Microbiology, Centre for Life Science, National University of Singapore, 117456, Singapore
| | - Florence S G Cheung
- Immunology Programme and Department of Microbiology, Centre for Life Science, National University of Singapore, 117456, Singapore
| | - Stephan Gasser
- Immunology Programme and Department of Microbiology, Centre for Life Science, National University of Singapore, 117456, Singapore; NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, 117456, Singapore.
| |
Collapse
|
2215
|
Chan YK, Gack MU. RIG-I-like receptor regulation in virus infection and immunity. Curr Opin Virol 2015; 12:7-14. [PMID: 25644461 PMCID: PMC5076476 DOI: 10.1016/j.coviro.2015.01.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/07/2015] [Indexed: 02/07/2023]
Abstract
Mammalian cells have the intrinsic capacity to detect viral pathogens and to initiate an antiviral response that is characterized by the induction of interferons (IFNs) and proinflammatory cytokines. A delicate regulation of the signaling pathways that lead to cytokine production is needed to ensure effective clearance of the virus, while preventing tissue damage caused by excessive cytokine release. Here, we focus on the mechanisms that modulate the signal transduction triggered by RIG-I-like receptors (RLRs) and their adaptor protein MAVS, key components of the host machinery for sensing foreign RNA. Specifically, we summarize recent advances in understanding how RLR signaling is regulated by posttranslational and posttranscriptional mechanisms, microRNAs (miRNAs) and autophagy. We further discuss how viruses target these regulatory mechanisms for immune evasion.
Collapse
Affiliation(s)
- Ying Kai Chan
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Michaela U Gack
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
2216
|
Yıldırım C, Nieuwenhuis S, Teunissen PF, Horrevoets AJ, van Royen N, van der Pouw Kraan TC. Interferon-Beta, a Decisive Factor in Angiogenesis and Arteriogenesis. J Interferon Cytokine Res 2015; 35:411-20. [DOI: 10.1089/jir.2014.0184] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Cansu Yıldırım
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Sylvia Nieuwenhuis
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul F. Teunissen
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Anton J.G. Horrevoets
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Niels van Royen
- Department of Cardiology, VU University Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
2217
|
Molina PE, Amedee AM, Winsauer P, Nelson S, Bagby G, Simon L. Behavioral, Metabolic, and Immune Consequences of Chronic Alcohol or Cannabinoids on HIV/AIDs: Studies in the Non-Human Primate SIV Model. J Neuroimmune Pharmacol 2015; 10:217-32. [PMID: 25795088 PMCID: PMC4470723 DOI: 10.1007/s11481-015-9599-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 03/05/2015] [Indexed: 02/07/2023]
Abstract
HIV-associated mortality has been significantly reduced with antiretroviral therapy (ART), and HIV infection has become a chronic disease that frequently coexists with many disorders, including substance abuse (Azar et al. Drug Alcohol Depend 112:178-193, 2010; Phillips et al. J Gen Int Med 16:165, 2001). Alcohol and drugs of abuse may modify host-pathogen interactions at various levels including behavioral, metabolic, and immune consequences of HIV infection, as well as the ability of the virus to integrate into the genome and replicate in host cells. Identifying mechanisms responsible for these interactions is complicated by many factors, such as the tissue specific responses to viral infection, multiple cellular mechanisms involved in inflammatory responses, neuroendocrine and localized responses to infection, and kinetics of viral replication. An integrated physiological analysis of the biomedical consequences of chronic alcohol and drug use or abuse on disease progression is possible using rhesus macaques infected with simian immunodeficiency virus (SIV), a relevant model of HIV infection. This review will provide an overview of the data gathered using this model to show that chronic administration of two of the most commonly abused substances, alcohol and cannabinoids (Δ(9)-Tetrahydrocannabinol; THC), affect host-pathogen interactions.
Collapse
Affiliation(s)
- Patricia E Molina
- Department of Physiology, Louisiana State University Health Sciences Center, School of Medicine, 1901 Perdido Street, New Orleans, LA, 70112, USA,
| | | | | | | | | | | |
Collapse
|
2218
|
Falasca L, Agrati C, Petrosillo N, Di Caro A, Capobianchi MR, Ippolito G, Piacentini M. Molecular mechanisms of Ebola virus pathogenesis: focus on cell death. Cell Death Differ 2015; 22:1250-9. [PMID: 26024394 PMCID: PMC4495366 DOI: 10.1038/cdd.2015.67] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/31/2015] [Accepted: 04/20/2015] [Indexed: 12/28/2022] Open
Abstract
Ebola virus (EBOV) belongs to the Filoviridae family and is responsible for a severe disease characterized by the sudden onset of fever and malaise accompanied by other non-specific signs and symptoms; in 30–50% of cases hemorrhagic symptoms are present. Multiorgan dysfunction occurs in severe forms with a mortality up to 90%. The EBOV first attacks macrophages and dendritic immune cells. The innate immune reaction is characterized by a cytokine storm, with secretion of numerous pro-inflammatory cytokines, which induces a huge number of contradictory signals and hurts the immune cells, as well as other tissues. Other highly pathogenic viruses also trigger cytokine storms, but Filoviruses are thought to be particularly lethal because they affect a wide array of tissues. In addition to the immune system, EBOV attacks the spleen and kidneys, where it kills cells that help the body to regulate its fluid and chemical balance and that make proteins that help the blood to clot. In addition, EBOV causes liver, lungs and kidneys to shut down their functions and the blood vessels to leak fluid into surrounding tissues. In this review, we analyze the molecular mechanisms at the basis of Ebola pathogenesis with a particular focus on the cell death pathways induced by the virus. We also discuss how the treatment of the infection can benefit from the recent experience of blocking/modulating cell death in human degenerative diseases.
Collapse
Affiliation(s)
- L Falasca
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - C Agrati
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - N Petrosillo
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - A Di Caro
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - M R Capobianchi
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - G Ippolito
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - M Piacentini
- 1] National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy [2] Department of Biology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
2219
|
Sheehan KCF, Lazear HM, Diamond MS, Schreiber RD. Selective Blockade of Interferon-α and -β Reveals Their Non-Redundant Functions in a Mouse Model of West Nile Virus Infection. PLoS One 2015; 10:e0128636. [PMID: 26010249 PMCID: PMC4444312 DOI: 10.1371/journal.pone.0128636] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 04/30/2015] [Indexed: 01/12/2023] Open
Abstract
Although type I interferons (IFNs) were first described almost 60 years ago, the ability to monitor and modulate the functional activities of the individual IFN subtypes that comprise this family has been hindered by a lack of reagents. The major type I IFNs, IFN-β and the multiple subtypes of IFN-α, are expressed widely and induce their effects on cells by interacting with a shared heterodimeric receptor (IFNAR). In the mouse, the physiologic actions of IFN-α and IFN-β have been defined using polyclonal anti-type I IFN sera, by targeting IFNAR using monoclonal antibodies or knockout mice, or using Ifnb-/- mice. However, the corresponding analysis of IFN-α has been difficult because of its polygenic nature. Herein, we describe two monoclonal antibodies (mAbs) that differentially neutralize murine IFN-β or multiple subtypes of murine IFN-α. Using these mAbs, we distinguish specific contributions of IFN-β versus IFN-α in restricting viral pathogenesis and identify IFN-α as the key mediator of the antiviral response in mice infected with West Nile virus. This study thus suggests the utility of these new reagents in dissecting the antiviral and immunomodulatory roles of IFN-β versus IFN-α in murine models of infection, immunity, and autoimmunity.
Collapse
Affiliation(s)
- Kathleen C. F. Sheehan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Helen M. Lazear
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael S. Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Robert D. Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
2220
|
Tummers B, Burg SHVD. High-risk human papillomavirus targets crossroads in immune signaling. Viruses 2015; 7:2485-506. [PMID: 26008697 PMCID: PMC4452916 DOI: 10.3390/v7052485] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/08/2015] [Indexed: 12/21/2022] Open
Abstract
Persistent infections with a high-risk type human papillomavirus (hrHPV) can progress to cancer. High-risk HPVs infect keratinocytes (KCs) and successfully suppress host immunity for up to two years despite the fact that KCs are well equipped to detect and initiate immune responses to invading pathogens. Viral persistence is achieved by active interference with KCs innate and adaptive immune mechanisms. To this end hrHPV utilizes proteins encoded by its viral genome, as well as exploits cellular proteins to interfere with signaling of innate and adaptive immune pathways. This results in impairment of interferon and pro-inflammatory cytokine production and subsequent immune cell attraction, as well as resistance to incoming signals from the immune system. Furthermore, hrHPV avoids the killing of infected cells by interfering with antigen presentation to antigen-specific cytotoxic T lymphocytes. Thus, hrHPV has evolved multiple mechanisms to avoid detection and clearance by both the innate and adaptive immune system, the molecular mechanisms of which will be dealt with in detail in this review.
Collapse
Affiliation(s)
- Bart Tummers
- Department of Clinical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands.
| | - Sjoerd H Van Der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
2221
|
Zhu M, Fang T, Li S, Meng K, Guo D. Bipartite Nuclear Localization Signal Controls Nuclear Import and DNA-Binding Activity of IFN Regulatory Factor 3. THE JOURNAL OF IMMUNOLOGY 2015; 195:289-97. [DOI: 10.4049/jimmunol.1500232] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/03/2015] [Indexed: 11/19/2022]
|
2222
|
Varicella Viruses Inhibit Interferon-Stimulated JAK-STAT Signaling through Multiple Mechanisms. PLoS Pathog 2015; 11:e1004901. [PMID: 25973608 PMCID: PMC4431795 DOI: 10.1371/journal.ppat.1004901] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 04/21/2015] [Indexed: 12/17/2022] Open
Abstract
Varicella zoster virus (VZV) causes chickenpox in humans and, subsequently, establishes latency in the sensory ganglia from where it reactivates to cause herpes zoster. Infection of rhesus macaques with simian varicella virus (SVV) recapitulates VZV pathogenesis in humans thus representing a suitable animal model for VZV infection. While the type I interferon (IFN) response has been shown to affect VZV replication, the virus employs counter mechanisms to prevent the induction of anti-viral IFN stimulated genes (ISG). Here, we demonstrate that SVV inhibits type I IFN-activated signal transduction via the JAK-STAT pathway. SVV-infected rhesus fibroblasts were refractory to IFN stimulation displaying reduced protein levels of IRF9 and lacking STAT2 phosphorylation. Since previous work implicated involvement of the VZV immediate early gene product ORF63 in preventing ISG-induction we studied the role of SVV ORF63 in generating resistance to IFN treatment. Interestingly, SVV ORF63 did not affect STAT2 phosphorylation but caused IRF9 degradation in a proteasome-dependent manner, suggesting that SVV employs multiple mechanisms to counteract the effect of IFN. Control of SVV ORF63 protein levels via fusion to a dihydrofolate reductase (DHFR)-degradation domain additionally confirmed its requirement for viral replication. Our results also show a prominent reduction of IRF9 and inhibition of STAT2 phosphorylation in VZV-infected cells. In addition, cells expressing VZV ORF63 blocked IFN-stimulation and displayed reduced levels of the IRF9 protein. Taken together, our data suggest that varicella ORF63 prevents ISG-induction both directly via IRF9 degradation and indirectly via transcriptional control of viral proteins that interfere with STAT2 phosphorylation. SVV and VZV thus encode multiple viral gene products that tightly control IFN-induced anti-viral responses. In this manuscript we demonstrate that the immediate early protein ORF63 encoded by varicella zoster virus (VZV) and simian varicella virus (SVV) interferes with interferon type I-mediated activation of JAK-STAT signaling and thereby inhibits the expression of interferon stimulated genes. ORF63 blocks this pathway by degrading IRF9, which plays a central role in JAK-STAT signaling. In addition, both viruses code for immune evasion mechanisms affecting the JAK-STAT pathway upstream of IRF9, which results in the inhibition of STAT2 phosphorylation. By fusing a degradation domain derived from dihydrofolate reductase (DHFR) to ORF63 we further demonstrate that this protein is essential for SVV growth and gene expression, indicating that ORF63 also affects IFN-signaling indirectly by regulating the expression of other immune evasion genes.
Collapse
|
2223
|
A Rhesus Rhadinovirus Viral Interferon (IFN) Regulatory Factor Is Virion Associated and Inhibits the Early IFN Antiviral Response. J Virol 2015; 89:7707-21. [PMID: 25972548 DOI: 10.1128/jvi.01175-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED The interferon (IFN) response is the earliest host immune response dedicated to combating viral infection. As such, viruses have evolved strategies to subvert this potent antiviral response. Two closely related gammaherpesviruses, Kaposi's sarcoma-associated herpesvirus (KSHV) and rhesus macaque rhadinovirus (RRV), are unique in that they express viral homologues to cellular interferon regulatory factors (IRFs), termed viral IRFs (vIRFs). Cellular IRFs are a family of transcription factors that are particularly important for the transcription of type I IFNs. Here, we demonstrate a strategy employed by RRV to ensure rapid inhibition of virus-induced type I IFN induction. We found that RRV vIRF R6, when expressed ectopically, interacts with a transcriptional coactivator, CREB-binding protein (CBP), in the nucleus. As a result, phosphorylated IRF3, an important transcriptional regulator in beta interferon (IFN-β) transcription, fails to effectively bind to the IFN-β promoter, thus inhibiting the activation of IFN-β genes. In addition, we found R6 within RRV virion particles via immunoelectron microscopy and, furthermore, that virion-associated R6 is capable of inhibiting the type I IFN response by preventing efficient binding of IRF3/CBP complexes to the IFN-β promoter in the context of infection. The work shown here is the first example of a vIRF being associated with either the KSHV or RRV virion. The presence of this immunomodulatory protein in the RRV virion provides the virus with an immediate mechanism to evade the host IFN response, thus enabling the virus to effectively establish an infection within the host. IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) and the closely related rhesus macaque rhadinovirus (RRV) are the only viruses known to encode viral homologues to cellular interferon regulatory factors (IRFs), known as vIRFs. In KSHV, these proteins have been shown to play major roles in a variety of cellular processes and are particularly important in the evasion of the host type I interferon (IFN) response. In this study, we delineate the immunomodulatory mechanism of an RRV vIRF and its ability to assist the virus in rapid immune evasion by being prepackaged within the virion, thus providing evidence, for the first time, of a virion-associated vIRF. This work further contributes to our understanding of the mechanisms behind immunomodulation by the RRV vIRFs during infection.
Collapse
|
2224
|
Boshuizen MCS, de Winther MPJ. Interferons as Essential Modulators of Atherosclerosis. Arterioscler Thromb Vasc Biol 2015; 35:1579-88. [PMID: 25953648 DOI: 10.1161/atvbaha.115.305464] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/27/2015] [Indexed: 12/11/2022]
Abstract
Interferons (IFNs) are key regulators of both innate and adaptive immune responses. The family of IFN cytokines can be divided into 3 main subtypes of which type I and type II IFNs are most well-defined. IFNs are known to be important mediators in atherosclerosis. Evidence from both in vitro and in vivo studies shows that the IFNs are generally proatherosclerotic. However, their role in atherosclerosis is complex, with distinct roles for these cytokines throughout different stages of the disease. In this review, we will discuss the current knowledge on the role of type I and type II IFNs in atherosclerosis development, specifically focusing on their role in endothelial activation, cell recruitment, foam cell formation, and regulation of apoptosis. Furthermore, we will discuss whether IFNs could be considered as new molecular targets for therapeutic intervention in atherosclerosis.
Collapse
Affiliation(s)
- Marieke C S Boshuizen
- From the Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Menno P J de Winther
- From the Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
2225
|
Nistal-Villán E, Rodríguez-García E, Di Scala M, Ferrero-Laborda R, Olagüe C, Vales Á, Carte-Abad B, Crespo I, García-Sastre A, Prieto J, Larrea E, González-Aseguinolaza G. A RIG-I 2CARD-MAVS200 Chimeric Protein Reconstitutes IFN-β Induction and Antiviral Response in Models Deficient in Type I IFN Response. J Innate Immun 2015; 7:466-81. [PMID: 25966783 PMCID: PMC4553127 DOI: 10.1159/000375262] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 01/15/2015] [Accepted: 01/15/2015] [Indexed: 01/12/2023] Open
Abstract
RIG-I-like receptors (RLRs) are cellular sensor proteins that detect certain RNA species produced during viral infections. RLRs activate a signaling cascade that results in the production of IFN-β as well as several other cytokines with antiviral and proinflammatory activities. We explored the potential of different constructs based on RLRs to induce the IFN-β pathway and create an antiviral state in type I IFN-unresponsive models. A chimeric construct composed of RIG-I 2CARD and the first 200 amino acids of MAVS (2CARD-MAVS200) showed an enhanced ability to induce IFN-β when compared to other stimulatory constructs. Furthermore, this human chimeric construct showed a superior ability to activate IFN-β expression in cells from various species. This construct was found to overcome the restrictions of blocking IFN-β induction or signaling by a number of viral IFN-antagonist proteins. Additionally, the antiviral activity of this chimera was demonstrated in influenza virus and HBV infection mouse models using adeno-associated virus (AAV) vectors as a delivery vehicle. We propose that AAV vectors expressing 2CARD-MAVS200 chimeric protein can reconstitute IFN-β induction and recover a partial antiviral state in different models that do not respond to recombinant IFN-β treatment.
Collapse
Affiliation(s)
- Estanislao Nistal-Villán
- Centro de Investigación Médica Aplicada, Gene Therapy and Regulation of Gene Expression Program, University of León, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, University of León, Pamplona, Spain
| | - Estefanía Rodríguez-García
- Centro de Investigación Médica Aplicada, Gene Therapy and Regulation of Gene Expression Program, University of León, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, University of León, Pamplona, Spain
| | - Marianna Di Scala
- Centro de Investigación Médica Aplicada, Gene Therapy and Regulation of Gene Expression Program, University of León, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, University of León, Pamplona, Spain
| | - Roberto Ferrero-Laborda
- Centro de Investigación Médica Aplicada, Gene Therapy and Regulation of Gene Expression Program, University of León, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, University of León, Pamplona, Spain
| | - Cristina Olagüe
- Centro de Investigación Médica Aplicada, Gene Therapy and Regulation of Gene Expression Program, University of León, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, University of León, Pamplona, Spain
| | - África Vales
- Centro de Investigación Médica Aplicada, Gene Therapy and Regulation of Gene Expression Program, University of León, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, University of León, Pamplona, Spain
| | - Beatriz Carte-Abad
- Centro de Investigación Médica Aplicada, Gene Therapy and Regulation of Gene Expression Program, University of León, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, University of León, Pamplona, Spain
| | - Irene Crespo
- Institute of Biomedicine (IBIOMED), CIBERehd, University of León, León, Spain
| | - Adolfo García-Sastre
- Department of Microbiology, New York, N.Y., USA
- Global Health and Emerging Pathogens Institute, New York, N.Y., USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, N.Y., USA
| | - Jesús Prieto
- Centro de Investigación Médica Aplicada, Gene Therapy and Regulation of Gene Expression Program, University of León, Pamplona, Spain
- Clínica Universidad de Navarra, CIBERehd, University of León, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, University of León, Pamplona, Spain
| | - Esther Larrea
- Centro de Investigación Médica Aplicada, Gene Therapy and Regulation of Gene Expression Program, University of León, Pamplona, Spain
- Instituto de Salud Tropical, University of Navarra, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, University of León, Pamplona, Spain
| | - Gloria González-Aseguinolaza
- Centro de Investigación Médica Aplicada, Gene Therapy and Regulation of Gene Expression Program, University of León, Pamplona, Spain
- IdiSNA Navarra Institute for Health Research, University of León, Pamplona, Spain
| |
Collapse
|
2226
|
Basler CF. Innate immune evasion by filoviruses. Virology 2015; 479-480:122-30. [DOI: 10.1016/j.virol.2015.03.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 03/17/2015] [Indexed: 01/07/2023]
|
2227
|
Davis ME, Gack MU. Ubiquitination in the antiviral immune response. Virology 2015; 479-480:52-65. [PMID: 25753787 PMCID: PMC4774549 DOI: 10.1016/j.virol.2015.02.033] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/12/2015] [Accepted: 02/17/2015] [Indexed: 01/07/2023]
Abstract
Ubiquitination has long been known to regulate fundamental cellular processes through the induction of proteasomal degradation of target proteins. More recently, 'atypical' non-degradative types of polyubiquitin chains have been appreciated as important regulatory moieties by modulating the activity or subcellular localization of key signaling proteins. Intriguingly, many of these non-degradative types of ubiquitination regulate the innate sensing pathways initiated by pattern recognition receptors (PRRs), ultimately coordinating an effective antiviral immune response. Here we discuss recent advances in understanding the functional roles of degradative and atypical types of ubiquitination in innate immunity to viral infections, with a specific focus on the signaling pathways triggered by RIG-I-like receptors, Toll-like receptors, and the intracellular viral DNA sensor cGAS.
Collapse
Affiliation(s)
- Meredith E Davis
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, United States
| | - Michaela U Gack
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
2228
|
Génin P, Cuvelier F, Lambin S, Côrte-Real Filipe J, Autrusseau E, Laurent C, Laplantine E, Weil R. Optineurin regulates the interferon response in a cell cycle-dependent manner. PLoS Pathog 2015; 11:e1004877. [PMID: 25923723 PMCID: PMC4414543 DOI: 10.1371/journal.ppat.1004877] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 04/13/2015] [Indexed: 01/12/2023] Open
Abstract
Viral invasion into a host is initially recognized by the innate immune system, mainly through activation of the intracellular cytosolic signaling pathway and coordinated activation of interferon regulatory factor 3 (IRF3) and nuclear factor kappa B (NF-κB) transcription factors that promote type I interferon gene induction. The TANK-binding Kinase 1 (TBK1) phosphorylates and activates IRF3. Here, we show that Optineurin (Optn) dampens the antiviral innate immune response by targeting the deubiquitinating enzyme CYLD to TBK1 in order to inhibit its enzymatic activity. Importantly, we found that this regulatory mechanism is abolished at the G2/M phase as a consequence of the nuclear translocation of CYLD and Optn. As a result, we observed, at this cell division stage, an increased activity and phosphorylation of TBK1 that lead to its relocalization to mitochondria and to enhanced interferon production, suggesting that this process, which relies on Optn function, might be of major importance to mount a preventive antiviral response during mitosis. The innate immune system has evolved to detect and neutralize viral invasion. Triggering of this defense mechanism relies on the production and secretion of soluble factors that stimulate an intracellular antiviral defense mechanism. The protein Optineurin was shown to negatively regulate this process. Importantly, we discovered the mechanism by which Optineurin inhibits antiviral activity and showed that this regulation is prevented during a critical step of cell division leading to enhancement of the cellular defense mechanism. This paper shows that the antiviral immune system is controlled during the cell cycle and that Optineurin-mediated induction of this system might serve to protect cells from infection during cell division.
Collapse
Affiliation(s)
- Pierre Génin
- Laboratoire de Signalisation et Pathogenèse, CNRS UMR3691, Institut Pasteur, Paris, France
- * E-mail:
| | - Frédérique Cuvelier
- Laboratoire de Signalisation et Pathogenèse, CNRS UMR3691, Institut Pasteur, Paris, France
| | - Sandrine Lambin
- Laboratoire de Signalisation et Pathogenèse, CNRS UMR3691, Institut Pasteur, Paris, France
| | | | - Elodie Autrusseau
- Laboratoire de Signalisation et Pathogenèse, CNRS UMR3691, Institut Pasteur, Paris, France
| | | | - Emmanuel Laplantine
- Laboratoire de Signalisation et Pathogenèse, CNRS UMR3691, Institut Pasteur, Paris, France
| | - Robert Weil
- Laboratoire de Signalisation et Pathogenèse, CNRS UMR3691, Institut Pasteur, Paris, France
| |
Collapse
|
2229
|
Bezu L, Gomes-de-Silva LC, Dewitte H, Breckpot K, Fucikova J, Spisek R, Galluzzi L, Kepp O, Kroemer G. Combinatorial strategies for the induction of immunogenic cell death. Front Immunol 2015; 6:187. [PMID: 25964783 PMCID: PMC4408862 DOI: 10.3389/fimmu.2015.00187] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/06/2015] [Indexed: 12/12/2022] Open
Abstract
The term "immunogenic cell death" (ICD) is commonly employed to indicate a peculiar instance of regulated cell death (RCD) that engages the adaptive arm of the immune system. The inoculation of cancer cells undergoing ICD into immunocompetent animals elicits a specific immune response associated with the establishment of immunological memory. Only a few agents are intrinsically endowed with the ability to trigger ICD. These include a few chemotherapeutics that are routinely employed in the clinic, like doxorubicin, mitoxantrone, oxaliplatin, and cyclophosphamide, as well as some agents that have not yet been approved for use in humans. Accumulating clinical data indicate that the activation of adaptive immune responses against dying cancer cells is associated with improved disease outcome in patients affected by various neoplasms. Thus, novel therapeutic regimens that trigger ICD are urgently awaited. Here, we discuss current combinatorial approaches to convert otherwise non-immunogenic instances of RCD into bona fide ICD.
Collapse
Affiliation(s)
- Lucillia Bezu
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Campus Cancer , Villejuif , France ; Faculté de Medecine, Université Paris-Sud , Le Kremlin-Bicêtre , France
| | - Ligia C Gomes-de-Silva
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Campus Cancer , Villejuif , France ; Department of Chemistry, University of Coimbra , Coimbra , Portugal
| | - Heleen Dewitte
- Laboratory for General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University , Ghent , Belgium ; Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel , Jette , Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel , Jette , Belgium
| | - Jitka Fucikova
- Sotio a.c. , Prague , Czech Republic ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| | - Radek Spisek
- Sotio a.c. , Prague , Czech Republic ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| | - Lorenzo Galluzzi
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Gustave Roussy Campus Cancer , Villejuif , France ; Université Paris Descartes , Paris , France ; Université Pierre et Marie Curie , Paris , France
| | - Oliver Kepp
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Campus Cancer , Villejuif , France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Campus Cancer , Villejuif , France ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic ; Université Paris Descartes , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP , Paris , France
| |
Collapse
|
2230
|
Fluorescence-Activated Cell Sorting-Based Analysis Reveals an Asymmetric Induction of Interferon-Stimulated Genes in Response to Seasonal Influenza A Virus. J Virol 2015; 89:6982-93. [PMID: 25903337 DOI: 10.1128/jvi.00857-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/18/2015] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Influenza A virus (IAV) infection provokes an antiviral response involving the expression of type I and III interferons (IFN) and IFN-stimulated genes (ISGs) in infected cell cultures. However, the spatiotemporal dynamics of the IFN reaction are incompletely understood, as previous studies investigated mainly the population responses of virus-infected cultures, although substantial cell-to-cell variability has been documented. We devised a fluorescence-activated cell sorting-based assay to simultaneously quantify expression of viral antigens and ISGs, such as ISG15, MxA, and IFIT1, in IAV-infected cell cultures at the single-cell level. This approach revealed that seasonal IAV triggers an unexpected asymmetric response, as the major cell populations expressed either viral antigen or ISG, but rarely both. Further investigations identified a role of the viral NS1 protein in blocking ISG expression in infected cells, which surprisingly did not reduce paracrine IFN signaling to noninfected cells. Interestingly, viral ISG control was impaired in cultures infected with avian-origin IAV, including the H7N9 virus from eastern China. This phenotype was traced back to polymorphic NS1 amino acids known to be important for stable binding of the polyadenylation factor CPSF30 and concomitant suppression of host cell gene expression. Most significantly, mutation of two amino acids within the CPSF30 attachment site of NS1 from seasonal IAV diminished the strict control of ISG expression in infected cells and substantially attenuated virus replication. In conclusion, our approach revealed an asymmetric, NS1-dependent ISG induction in cultures infected with seasonal IAV, which appears to be essential for efficient virus propagation. IMPORTANCE Interferons are expressed by infected cells in response to IAV infection and play important roles in the antiviral immune response by inducing hundreds of interferon-stimulated genes (ISGs). Unlike many previous studies, we investigated the ISG response at the single-cell level, enabling novel insights into this virus-host interaction. Hence, cell cultures infected with seasonal IAV displayed an asymmetric ISG induction that was confined almost exclusively to noninfected cells. In comparison, ISG expression was observed in larger cell populations infected with avian-origin IAV, suggesting a more resolute antiviral response to these strains. Strict control of ISG expression by seasonal IAV was explained by the binding of the viral NS1 protein to the polyadenylation factor CPSF30, which reduces host cell gene expression. Mutational disruption of CPSF30 binding within NS1 concomitantly attenuated ISG control and replication of seasonal IAV, illustrating the importance of maintaining an asymmetric ISG response for efficient virus propagation.
Collapse
|
2231
|
Saleiro D, Mehrotra S, Kroczynska B, Beauchamp EM, Lisowski P, Majchrzak-Kita B, Bhagat TD, Stein BL, McMahon B, Altman JK, Kosciuczuk EM, Baker DP, Jie C, Jafari N, Thompson CB, Levine RL, Fish EN, Verma AK, Platanias LC. Central role of ULK1 in type I interferon signaling. Cell Rep 2015; 11:605-17. [PMID: 25892232 DOI: 10.1016/j.celrep.2015.03.056] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 02/16/2015] [Accepted: 03/25/2015] [Indexed: 11/17/2022] Open
Abstract
We provide evidence that the Unc-51-like kinase 1 (ULK1) is activated during engagement of the type I interferon (IFN) receptor (IFNR). Our studies demonstrate that the function of ULK1 is required for gene transcription mediated via IFN-stimulated response elements (ISRE) and IFNγ activation site (GAS) elements and controls expression of key IFN-stimulated genes (ISGs). We identify ULK1 as an upstream regulator of p38α mitogen-activated protein kinase (MAPK) and establish that the regulatory effects of ULK1 on ISG expression are mediated possibly by engagement of the p38 MAPK pathway. Importantly, we demonstrate that ULK1 is essential for antiproliferative responses and type I IFN-induced antineoplastic effects against malignant erythroid precursors from patients with myeloproliferative neoplasms. Together, these data reveal a role for ULK1 as a key mediator of type I IFNR-generated signals that control gene transcription and induction of antineoplastic responses.
Collapse
Affiliation(s)
- Diana Saleiro
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Swarna Mehrotra
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Barbara Kroczynska
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Elspeth M Beauchamp
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Pawel Lisowski
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, 05-552 Jastrzebiec n/Warsaw, Poland; iPS Cell-Based Disease Modeling Group, Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, 13092 Berlin, Germany
| | - Beata Majchrzak-Kita
- Toronto General Research Institute, University Health Network and Department of Immunology, University of Toronto, Toronto, ON M5G 2M1, Canada
| | - Tushar D Bhagat
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Brady L Stein
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Brandon McMahon
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jessica K Altman
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Ewa M Kosciuczuk
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Darren P Baker
- Biogen Idec Inc., 14 Cambridge Center, Cambridge, MA 02142, USA
| | - Chunfa Jie
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nadereh Jafari
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program, and Leukemia Service, Memorial Sloan Kettering Cancer Center; and Weill Cornell Medical College, New York, NY 10065, USA
| | - Eleanor N Fish
- Toronto General Research Institute, University Health Network and Department of Immunology, University of Toronto, Toronto, ON M5G 2M1, Canada
| | - Amit K Verma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
2232
|
Coccia EM, Battistini A. Early IFN type I response: Learning from microbial evasion strategies. Semin Immunol 2015; 27:85-101. [PMID: 25869307 PMCID: PMC7129383 DOI: 10.1016/j.smim.2015.03.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 03/10/2015] [Indexed: 12/12/2022]
Abstract
Type I interferon (IFN) comprises a class of cytokines first discovered more than 50 years ago and initially characterized for their ability to interfere with viral replication and restrict locally viral propagation. As such, their induction downstream of germ-line encoded pattern recognition receptors (PRRs) upon recognition of pathogen-associated molecular patterns (PAMPs) is a hallmark of the host antiviral response. The acknowledgment that several PAMPs, not just of viral origin, may induce IFN, pinpoints at these molecules as a first line of host defense against a number of invading pathogens. Acting in both autocrine and paracrine manner, IFN interferes with viral replication by inducing hundreds of different IFN-stimulated genes with both direct anti-pathogenic as well as immunomodulatory activities, therefore functioning as a bridge between innate and adaptive immunity. On the other hand an inverse interference to escape the IFN system is largely exploited by pathogens through a number of tactics and tricks aimed at evading, inhibiting or manipulating the IFN pathway, that result in progression of infection or establishment of chronic disease. In this review we discuss the interplay between the IFN system and some selected clinically important and challenging viruses and bacteria, highlighting the wide array of pathogen-triggered molecular mechanisms involved in evasion strategies.
Collapse
Affiliation(s)
- Eliana M Coccia
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy
| | - Angela Battistini
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy.
| |
Collapse
|
2233
|
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: 1890] [Impact Index Per Article: 189.0] [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
|
2234
|
Antonelli G, Scagnolari C, Moschella F, Proietti E. Twenty-five years of type I interferon-based treatment: a critical analysis of its therapeutic use. Cytokine Growth Factor Rev 2015; 26:121-31. [PMID: 25578520 PMCID: PMC7108252 DOI: 10.1016/j.cytogfr.2014.12.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 12/19/2014] [Indexed: 02/06/2023]
Abstract
The clinical exploitation of type I interferon (IFN) as an antiviral and antineoplastic agent is based on the properties originally attributed to this cytokine family, with schedules reflecting only their antiviral and antiproliferative activities. Nevertheless, type I IFN has emerged as a central activator of the innate immunity. As current schedules of treatment for chronic hepatitis C and for hematological and solid tumors, based on the continuous administration of recombinant type I IFN or pegylated formulations, disregard viral resistance, host genetic variants predicting treatment outcome and mechanisms of refractoriness, new administration schedules, the combination of type I IFN with new drugs and the increased monitoring of patients' susceptibility to type I IFN are expected to provide a new life to this valuable cytokine.
Collapse
Affiliation(s)
- Guido Antonelli
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University, Rome, Italy
| | - Carolina Scagnolari
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University, Rome, Italy
| | - Federica Moschella
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Enrico Proietti
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
2235
|
Capobianchi MR, Uleri E, Caglioti C, Dolei A. Type I IFN family members: similarity, differences and interaction. Cytokine Growth Factor Rev 2015; 26:103-11. [PMID: 25466633 PMCID: PMC7108279 DOI: 10.1016/j.cytogfr.2014.10.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/22/2014] [Indexed: 02/07/2023]
Abstract
Interferons (IFN) are key cytokines with multifaceted antiviral and cell-modulatory properties. Three distinct types of IFN are recognized (I-III) based on structural features, receptor usage, cellular source and biological activities. The action of IFNs is mediated by a complex, partially overlapping, transcriptional program initiated by the interaction with specific receptors. Genetic diversity, with polymorphisms and mutations, can modulate the extent of IFN responses and the susceptibility to infections. Almost all viruses developed mechanisms to subvert the IFN response, involving both IFN induction and effector mechanisms. Interactions between IFN types may occur, for both antiviral and cell-modulatory effects, in a complex interplay, involving both synergistic and antagonistic effects. Interferon-associated diseases, not related to virus infections may occur, some of them frequently observed in IFN-treated patients. On the whole, IFNs are pleiotropic biologic response modifiers, that, upon activation of thousands genes, induce a broad spectrum of activities, regulating cell cycle, differentiation, plasma membrane molecules, release of mediators, etc., that can be relevant for cell proliferation, innate and adaptive immunity, hematopoiesis, angiogenesis and other body functions.
Collapse
Affiliation(s)
- Maria Rosaria Capobianchi
- Laboratory of Virology, National Institute for Infectious Diseases "L. Spallanzani", Via Portuense 292, Rome, Italy
| | - Elena Uleri
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Claudia Caglioti
- Laboratory of Virology, National Institute for Infectious Diseases "L. Spallanzani", Via Portuense 292, Rome, Italy
| | - Antonina Dolei
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.
| |
Collapse
|
2236
|
Divergent effects of type-I interferons on regulatory T cells. Cytokine Growth Factor Rev 2015; 26:133-41. [DOI: 10.1016/j.cytogfr.2014.10.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/22/2014] [Indexed: 02/07/2023]
|
2237
|
Abstract
Systemic lupus erythematosus (SLE) is a multisystem autoimmune disorder that has a broad spectrum of effects on the majority of organs, including the kidneys. Approximately 40-70% of patients with SLE will develop lupus nephritis. Renal assault during SLE is initiated by genes that breach immune tolerance and promote autoantibody production. These genes might act in concert with other genetic factors that augment innate immune signalling and IFN-I production, which in turn can generate an influx of effector leucocytes, inflammatory mediators and autoantibodies into end organs, such as the kidneys. The presence of cognate antigens in the glomerular matrix, together with intrinsic molecular abnormalities in resident renal cells, might further accentuate disease progression. This Review discusses the genetic insights and molecular mechanisms for key pathogenic contributors in SLE and lupus nephritis. We have categorized the genes identified in human studies of SLE into one of four pathogenic events that lead to lupus nephritis. We selected these categories on the basis of the cell types in which these genes are expressed, and the emerging paradigms of SLE pathogenesis arising from murine models. Deciphering the molecular basis of SLE and/or lupus nephritis in each patient will help physicians to tailor specific therapies.
Collapse
|
2238
|
Moghaddas F, Masters SL. Monogenic autoinflammatory diseases: Cytokinopathies. Cytokine 2015; 74:237-46. [PMID: 25814341 DOI: 10.1016/j.cyto.2015.02.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 12/17/2022]
Abstract
Rapid advances in genetics are providing unprecedented insight into functions of the innate immune system with identification of the mutations that cause monogenic autoinflammatory disease. Cytokine antagonism is profoundly effective in a subset of these conditions, particularly those associated with increased interleukin-1 (IL-1) activity, the inflammasomopathies. These include syndromes where the production of IL-1 is increased by mutation of innate immune sensors such as NLRP3, upstream signalling molecules such as PSTPIP1 and receptors or downstream signalling molecules, such as IL-1Ra. Another example of this is interferon (IFN) and the interferonopathies, with mutations in the sensors STING and MDA5, the upstream signalling regulator AP1S3, and a downstream inhibitor of IFN signalling, ISG15. We propose that this can be extended to cytokines such as IL-36, with mutations in IL-36Ra, and IL-10, with mutations in IL-10RA and IL-10RB, however mutations in sensors or upstream signalling molecules are yet to be described in these instances. Additionally, autoinflammatory diseases can be caused by multiple cytokines, for example with the activation of NF-κB/Rel, for which we propose the term Relopathies. This nosology is limited in that some cytokine pathways may be degenerate in their generation or execution, however provides insight into likely autoinflammatory disease candidates and the cytokines with which newly identified mutations may be associated, and therefore targeted.
Collapse
Affiliation(s)
- Fiona Moghaddas
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Seth L Masters
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
2239
|
de Jong TD, Vosslamber S, Blits M, Wolbink G, Nurmohamed MT, van der Laken CJ, Jansen G, Voskuyl AE, Verweij CL. Effect of prednisone on type I interferon signature in rheumatoid arthritis: consequences for response prediction to rituximab. Arthritis Res Ther 2015; 17:78. [PMID: 25889713 PMCID: PMC4416246 DOI: 10.1186/s13075-015-0564-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/18/2015] [Indexed: 12/20/2022] Open
Abstract
Introduction Elevated type I interferon (IFN) response gene (IRG) expression has proven clinical relevance in predicting rituximab non-response in rheumatoid arthritis (RA). Interference between glucocorticoids (GCs) and type I IFN signaling has been demonstrated in vitro. Since GC use and dose are highly variable among patients before rituximab treatment, the aim of this study was to determine the effect of GC use on IRG expression in relation to rituximab response prediction in RA. Methods In two independent cohorts of 32 and 182 biologic-free RA patients and a third cohort of 40 rituximab-starting RA patients, peripheral blood expression of selected IRGs was determined by microarray or quantitative real-time polymerase chain reaction (qPCR), and an IFN-score was calculated. The baseline IFN-score was tested for its predictive value towards rituximab response in relation to GC use using receiver operating characteristics (ROC) analysis in the rituximab cohort. Patients with a decrease in disease activity score (∆DAS28) >1.2 after 6 months of rituximab were considered responders. Results We consistently observed suppression of IFN-score in prednisone users (PREDN+) compared to non-users (PREDN−). In the rituximab cohort, analysis on PREDN− patients (n = 13) alone revealed improved prediction of rituximab non-response based on baseline IFN-score, with an area under the curve (AUC) of 0.975 compared to 0.848 in all patients (n = 40). Using a group-specific IFN-score cut-off for all patients and PREDN− patients alone, sensitivity increased from 41% to 88%, respectively, combined with 100% specificity. Conclusions Because of prednisone-related suppression of IFN-score, higher accuracy of rituximab response prediction was achieved in PREDN− patients. These results suggest that the IFN-score-based rituximab response prediction model could be improved upon implementation of prednisone use.
Collapse
Affiliation(s)
- Tamarah D de Jong
- Department of Pathology, VU University Medical Center, P.O. Box 7075, 1007, MB, Amsterdam, The Netherlands.
| | - Saskia Vosslamber
- Department of Pathology, VU University Medical Center, P.O. Box 7075, 1007, MB, Amsterdam, The Netherlands.
| | - Marjolein Blits
- Department of Pathology, VU University Medical Center, P.O. Box 7075, 1007, MB, Amsterdam, The Netherlands.
| | | | | | - Conny J van der Laken
- Department of Rheumatology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Gerrit Jansen
- Department of Rheumatology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Alexandre E Voskuyl
- Department of Rheumatology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Cornelis L Verweij
- Department of Pathology, VU University Medical Center, P.O. Box 7075, 1007, MB, Amsterdam, The Netherlands. .,Department of Rheumatology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
2240
|
Snell LM, Brooks DG. New insights into type I interferon and the immunopathogenesis of persistent viral infections. Curr Opin Immunol 2015; 34:91-8. [PMID: 25771184 DOI: 10.1016/j.coi.2015.03.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/25/2015] [Accepted: 03/02/2015] [Indexed: 02/07/2023]
Abstract
Most viruses generate potent T cell responses that rapidly control infection. However, certain viruses can subvert the immune response to establish persistent infections. The inability to clear virus induces an immunosuppressive program leading to the sustained expression of many immunoregulatory molecules that down-regulate T cell responses. Further, viral persistence is associated with multiple immune dysfunctions including lymphoid disorganization, defective antigen presentation, aberrant B cell responses and hypergammaglobulinemia. Although best known for its antiviral activity, recent data has highlighted the role of type I IFN (IFN-I) signaling as a central mediator of immunosuppression during viral persistence. It is also becoming increasingly apparent that many of the immune dysfunctions during persistent virus infection can be attributed directly or indirectly to the effects of chronic IFN-I signaling. This review explores the increasingly complex role of IFN-I in the regulation of immunity against persistently replicating virus infections and examines current and potential uses of IFN-I and blockade of IFN-I signaling to dampen chronic inflammation and activation in the clinic.
Collapse
Affiliation(s)
- Laura M Snell
- Department of Microbiology, Immunology and Molecular Genetics and UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, United States
| | - David G Brooks
- Department of Microbiology, Immunology and Molecular Genetics and UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, United States; Princess Margaret Cancer Center, University Health Network and the Department of Immunology, University of Toronto, Toronto, Ontario, M5G 2M9 Canada.
| |
Collapse
|
2241
|
Herdy B, Karonitsch T, Vladimer GI, Tan CSH, Stukalov A, Trefzer C, Bigenzahn JW, Theil T, Holinka J, Kiener HP, Colinge J, Bennett KL, Superti-Furga G. The RNA-binding protein HuR/ELAVL1 regulates IFN-β mRNA abundance and the type I IFN response. Eur J Immunol 2015; 45:1500-11. [PMID: 25678110 DOI: 10.1002/eji.201444979] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 12/18/2014] [Accepted: 02/06/2015] [Indexed: 12/26/2022]
Abstract
Secretion of type I interferon (IFN) is the first cellular reaction to invading pathogens. Despite the protective function of these cytokines, an excessive response to their action can contribute to serious pathologies, such as autoimmune diseases. Transcripts of most cytokines contain adenylate-uridylate (A/U)-rich elements (AREs) that make them highly unstable. RNA-binding proteins (RBPs) are mediators of the regulatory mechanisms that determine the fate of mRNAs containing AREs. Here, we applied an affinity proteomic approach and identified lethal, abnormal vision, drosophila-like 1 (ELAVL1)/Hu antigen R (HuR) as the predominant RBP of the IFN-β mRNA ARE. Reduced expression or chemical inhibition of HuR severely hampered the type I IFN response in various cell lines and fibroblast-like synoviocytes isolated from joints of rheumatoid arthritis patients. These results define a role for HuR as a potent modulator of the type I IFN response. Taken together, HuR could be used as therapeutic target for diseases where type I IFN production is exaggerated.
Collapse
Affiliation(s)
- Barbara Herdy
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Karonitsch
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Division of Rheumatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gregory I Vladimer
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Chris S H Tan
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Alexey Stukalov
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Claudia Trefzer
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Johannes W Bigenzahn
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tamara Theil
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Johannes Holinka
- Department of Orthopedics, Medical University of Vienna, Vienna, Austria
| | - Hans P Kiener
- Division of Rheumatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Jacques Colinge
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Keiryn L Bennett
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Giulio Superti-Furga
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| |
Collapse
|
2242
|
Interferon-alpha competing endogenous RNA network antagonizes microRNA-1270. Cell Mol Life Sci 2015; 72:2749-61. [PMID: 25746225 PMCID: PMC4477080 DOI: 10.1007/s00018-015-1875-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 02/24/2015] [Accepted: 02/26/2015] [Indexed: 12/18/2022]
Abstract
A new form of circuitry for gene regulation has been identified in which RNAs can crosstalk by competing for shared microRNAs (miRNAs). Such competing endogenous RNAs (ceRNAs) form a network via shared miRNA response elements (MREs) to antagonize miRNA function. We previously reported natural antisense RNA (AS) as an important modulator of interferon-α1 (IFN-α1) mRNA levels by promoting IFN-α1 mRNA stability. We show that IFN-α1 AS forms a ceRNA network with specific IFN-α AS (IFN-α7/-α8/-α10/-α14) and mRNA (IFN-α8/-α10/-α14/-α17) subtypes from the IFN-α gene (IFNA) family to antagonize miRNA-1270 (miR-1270), thereby modulating IFN-α1 mRNA levels. Bioinformatic analysis demonstrated that IFN-α1 AS harbors multiple miR-1270 MREs (MRE-1270s), whose presence was substantiated by miR-1270 overexpression and transfection of antimiR-1270. The antimiR-1270, complementary to the miR-1270 seed region, revealed that IFN-α1 AS likely shares the MRE-1270 with IFN-α1 mRNA and specific IFN-α AS and mRNA subtypes. Subsequent bioinformatic analysis for MRE-1270s showed that IFN-α1 AS and other RNA subtypes shared the 6-mer MRE-1270 site. Further MRE-mapping demonstrated that the total number of MRE-1270s in IFN-α1 AS accounted for approximately 30 % of the miR-1270 population. AntimiR-1270 transfection also caused specific de-repression of five cellular mRNAs, including that of CAPRIN1. These results suggest that IFN-α1 AS, together with specific IFN-α AS and mRNA subtypes, as well as the five cellular mRNAs, participate as competing molecules in the ceRNA network against miR-1270. This coordinated regulatory architecture suggests a vital function for the innate immune system in maintaining precise physiological type I IFN levels via post-transcriptional regulatory mechanisms.
Collapse
|
2243
|
How do viruses control mitochondria-mediated apoptosis? Virus Res 2015; 209:45-55. [PMID: 25736565 PMCID: PMC7114537 DOI: 10.1016/j.virusres.2015.02.026] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 12/16/2022]
Abstract
There is no doubt that viruses require cells to successfully reproduce and effectively infect the next host. The question is what is the fate of the infected cells? All eukaryotic cells can "sense" viral infections and exhibit defence strategies to oppose viral replication and spread. This often leads to the elimination of the infected cells by programmed cell death or apoptosis. This "sacrifice" of infected cells represents the most primordial response of multicellular organisms to viruses. Subverting host cell apoptosis, at least for some time, is therefore a crucial strategy of viruses to ensure their replication, the production of essential viral proteins, virus assembly and the spreading to new hosts. For that reason many viruses harbor apoptosis inhibitory genes, which once inside infected cells are expressed to circumvent apoptosis induction during the virus reproduction phase. On the other hand, viruses can take advantage of stimulating apoptosis to (i) facilitate shedding and hence dissemination, (ii) to prevent infected cells from presenting viral antigens to the immune system or (iii) to kill non-infected bystander and immune cells which would limit viral propagation. Hence the decision whether an infected host cell undergoes apoptosis or not depends on virus type and pathogenicity, its capacity to oppose antiviral responses of the infected cells and/or to evade any attack from immune cells. Viral genomes have therefore been adapted throughout evolution to satisfy the need of a particular virus to induce or inhibit apoptosis during its life cycle. Here we review the different strategies used by viruses to interfere with the two major apoptosis as well as with the innate immune signaling pathways in mammalian cells. We will focus on the intrinsic mitochondrial pathway and discuss new ideas about how particular viruses could activately engage mitochondria to induce apoptosis of their host.
Collapse
|
2244
|
The molecular basis for functional plasticity in type I interferon signaling. Trends Immunol 2015; 36:139-49. [DOI: 10.1016/j.it.2015.01.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 01/13/2015] [Accepted: 01/13/2015] [Indexed: 01/16/2023]
|
2245
|
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: 144] [Impact Index Per Article: 14.4] [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
|
2246
|
Vogelpoel LTC, Baeten DLP, de Jong EC, den Dunnen J. Control of cytokine production by human fc gamma receptors: implications for pathogen defense and autoimmunity. Front Immunol 2015; 6:79. [PMID: 25759693 PMCID: PMC4338787 DOI: 10.3389/fimmu.2015.00079] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/10/2015] [Indexed: 01/21/2023] Open
Abstract
Control of cytokine production by immune cells is pivotal for counteracting infections via orchestration of local and systemic inflammation. Although their contribution has long been underexposed, it has recently become clear that human Fc gamma receptors (FcγRs), which are receptors for the Fc region of immunoglobulin G (IgG) antibodies, play a critical role in this process by controlling tissue- and pathogen-specific cytokine production. Whereas individual stimulation of FcγRs does not evoke cytokine production, FcγRs cell-type specifically interact with various other receptors for selective amplification or inhibition of particular cytokines, thereby tailoring cytokine responses to the immunological context. The physiological function of FcγR-mediated control of cytokine production is to counteract infections with various classes of pathogens. Upon IgG opsonization, pathogens are simultaneously recognized by FcγRs as well as by various pathogen-sensing receptors, leading to the induction of pathogen class-specific immune responses. However, when erroneously activated, the same mechanism also contributes to the development of autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus. In this review, we discuss control of cytokine production as a novel function of FcγRs in human innate immune cells in the context of homeostasis, infection, and autoimmunity and address the possibilities for future therapeutic exploitation.
Collapse
Affiliation(s)
- Lisa T C Vogelpoel
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam , Amsterdam , Netherlands
| | - Dominique L P Baeten
- Department of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam , Amsterdam , Netherlands
| | - Esther C de Jong
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam , Amsterdam , Netherlands
| | - Jeroen den Dunnen
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam , Amsterdam , Netherlands
| |
Collapse
|
2247
|
Di Scala M, Gil-Fariña I, Vanrell L, Sánchez-Bayona R, Alignani D, Olagüe C, Vales A, Berraondo P, Prieto J, González-Aseguinolaza G. Chronic exposure to IFNα drives medullar lymphopoiesis towards T-cell differentiation in mice. Haematologica 2015; 100:1014-22. [PMID: 25715405 DOI: 10.3324/haematol.2014.115410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 02/12/2015] [Indexed: 12/23/2022] Open
Abstract
Interferon-α is a potent antiviral agent and a vigorous adjuvant in the induction of T-cell responses but its use is limited by hematologic toxicity. Interferon-α alters hematopoietic stem cell dormancy and impairs myelocytic and erythrocytic/megakaryocytic differentiation from hematopoietic progenitors. However, the effect of chronic interferon-α exposure on hematopoietic precursors has still not been well characterized. Here, we transduced the liver of mice with an adenoassociated vector encoding interferon-α to achieve sustained high serum levels of the cytokine. The bone marrow of these animals showed diminished long-term and short-term hematopoietic stem cells, reduction of multipotent progenitor cells, and marked decrease of B cells, but significant increase in the proportion of CD8(+) and CD4(+)CD8(+) T cells. Upon adoptive transfer to RAG(-/-) mice, bone marrow cells from interferon-α-treated animals generated CD4(+) and CD8(+) T cells while CD19(+), CD11b(+) and NK1.1(+) lineages failed to develop. These effects are associated with the transcriptional downregulation of transcription factors involved in B-cell differentiation and modulation of key factors for T-cell development. Thus, sustained interferon-α exposure causes hematopoietic stem cells exhaustion and drives common lymphoid progenitors towards T-cell generation.
Collapse
Affiliation(s)
- Marianna Di Scala
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Irene Gil-Fariña
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Lucia Vanrell
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Rodrigo Sánchez-Bayona
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Diego Alignani
- Department of Instrumental Techniques-Cytometry Unit, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Cristina Olagüe
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Africa Vales
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Pedro Berraondo
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Jesús Prieto
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain University Clinic of Navarra, University of Navarra, Pamplona, Spain CIBERehd, University of Navarra, Pamplona, Spain
| | - Gloria González-Aseguinolaza
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| |
Collapse
|
2248
|
de Jesus AA, Canna SW, Liu Y, Goldbach-Mansky R. Molecular mechanisms in genetically defined autoinflammatory diseases: disorders of amplified danger signaling. Annu Rev Immunol 2015; 33:823-74. [PMID: 25706096 DOI: 10.1146/annurev-immunol-032414-112227] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Patients with autoinflammatory diseases present with noninfectious fever flares and systemic and/or disease-specific organ inflammation. Their excessive proinflammatory cytokine and chemokine responses can be life threatening and lead to organ damage over time. Studying such patients has revealed genetic defects that have helped unravel key innate immune pathways, including excessive IL-1 signaling, constitutive NF-κB activation, and, more recently, chronic type I IFN signaling. Discoveries of monogenic defects that lead to activation of proinflammatory cytokines have inspired the use of anticytokine-directed treatment approaches that have been life changing for many patients and have led to the approval of IL-1-blocking agents for a number of autoinflammatory conditions. In this review, we describe the genetically characterized autoinflammatory diseases, we summarize our understanding of the molecular pathways that drive clinical phenotypes and that continue to inspire the search for novel treatment targets, and we provide a conceptual framework for classification.
Collapse
Affiliation(s)
- Adriana Almeida de Jesus
- Translational Autoinflammatory Diseases Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, Maryland 20892;
| | | | | | | |
Collapse
|
2249
|
Rigby RE, Rehwinkel J. RNA degradation in antiviral immunity and autoimmunity. Trends Immunol 2015; 36:179-88. [PMID: 25709093 PMCID: PMC4358841 DOI: 10.1016/j.it.2015.02.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/02/2015] [Accepted: 02/02/2015] [Indexed: 01/09/2023]
Abstract
The nonsense-mediated decay (NMD) pathway defends cells against RNA virus invasion. NMD targets viral RNAs for degradation, including by the RNA exosome. Genetic deficiencies in NMD and RNA exosome components cause autoimmunity. NMD and the RNA exosome prevent aberrant activation of innate immune responses.
Post-transcriptional control determines the fate of cellular RNA molecules. Nonsense-mediated decay (NMD) provides quality control of mRNA, targeting faulty cellular transcripts for degradation by multiple nucleases including the RNA exosome. Recent findings have revealed a role for NMD in targeting viral RNA molecules, thereby restricting virus infection. Interestingly, NMD is also linked to immune responses at another level: mutations affecting the NMD or RNA exosome machineries cause chronic activation of defence programmes, resulting in autoimmune phenotypes. Here we place these observations in the context of other links between innate antiviral immunity and type I interferon mediated disease and examine two models: one in which expression or function of pathogen sensors is perturbed and one wherein host-derived RNA molecules with a propensity to activate such sensors accumulate.
Collapse
Affiliation(s)
- Rachel E Rigby
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Jan Rehwinkel
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
2250
|
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
|