Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Microbiol ; 6(4): 467-478, 2021 04.
Article in English | MEDLINE | ID: mdl-33727702

ABSTRACT

Activation of the RIG-I-like receptors, retinoic-acid inducible gene I (RIG-I) and melanoma differentiation-associated protein 5 (MDA5), establishes an antiviral state by upregulating interferon (IFN)-stimulated genes (ISGs). Among these is ISG15, the mechanistic roles of which in innate immunity still remain enigmatic. In the present study, we report that ISG15 conjugation is essential for antiviral IFN responses mediated by the viral RNA sensor MDA5. ISGylation of the caspase activation and recruitment domains of MDA5 promotes its oligomerization and thereby triggers activation of innate immunity against a range of viruses, including coronaviruses, flaviviruses and picornaviruses. The ISG15-dependent activation of MDA5 is antagonized through direct de-ISGylation mediated by the papain-like protease of SARS-CoV-2, a recently emerged coronavirus that has caused the COVID-19 pandemic. Our work demonstrates a crucial role for ISG15 in the MDA5-mediated antiviral response, and also identifies a key immune evasion mechanism of SARS-CoV-2, which may be targeted for the development of new antivirals and vaccines to combat COVID-19.


Subject(s)
Coronavirus Papain-Like Proteases/metabolism , Cytokines/metabolism , Immunity, Innate , Interferon-Induced Helicase, IFIH1/antagonists & inhibitors , SARS-CoV-2/enzymology , SARS-CoV-2/immunology , Ubiquitins/metabolism , Aedes , Animals , Chlorocebus aethiops , Cricetinae , HEK293 Cells , Humans , Interferon-Induced Helicase, IFIH1/metabolism , Leukocytes, Mononuclear , Mice , Vero Cells
2.
bioRxiv ; 2020 Oct 27.
Article in English | MEDLINE | ID: mdl-33140045

ABSTRACT

Activation of the RIG-I-like receptors, RIG-I and MDA5, establishes an antiviral state by upregulating interferon (IFN)-stimulated genes (ISGs). Among these is ISG15 whose mechanistic roles in innate immunity still remain enigmatic. Here we report that ISGylation is essential for antiviral IFN responses mediated by the viral RNA sensor MDA5. ISG15 conjugation to the caspase activation and recruitment domains of MDA5 promotes the formation of higher-order assemblies of MDA5 and thereby triggers activation of innate immunity against a range of viruses including coronaviruses, flaviviruses and picornaviruses. The ISG15-dependent activation of MDA5 is antagonized through direct de-ISGylation mediated by the papain-like protease (PLpro) of SARS-CoV-2, a recently emerged coronavirus that causes the COVID-19 pandemic. Our work demonstrates a crucial role for ISG15 in the MDA5-mediated antiviral response, and also identifies a novel immune evasion mechanism of SARS-CoV-2, which may be targeted for the development of new antivirals and vaccines to combat COVID-19.

3.
Immunity ; 38(3): 437-49, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23499489

ABSTRACT

RIG-I and MDA5 have emerged as key cytosolic sensors for the detection of RNA viruses and lead to antiviral interferon (IFN) production. Recent studies have highlighted the importance of posttranslational modifications for controlling RIG-I antiviral activity. However, the regulation of MDA5 signal-transducing ability remains unclear. Here, we show that MDA5 signaling activity is regulated by a dynamic balance between phosphorylation and dephosphorylation of its caspase recruitment domains (CARDs). Employing a phosphatome RNAi screen, we identified PP1α and PP1γ as the primary phosphatases that are responsible for MDA5 and RIG-I dephosphorylation and that lead to their activation. Silencing of PP1α and PP1γ enhanced RIG-I and MDA5 CARD phosphorylation and reduced antiviral IFN-ß production. PP1α- and PP1γ-depleted cells were impaired in their ability to induce IFN-stimulated gene expression, which resulted in enhanced RNA virus replication. This work identifies PP1α and PP1γ as regulators of antiviral innate immune responses to various RNA viruses, including influenza virus, paramyxovirus, dengue virus, and picornavirus.


Subject(s)
DEAD-box RNA Helicases/immunology , Immunity, Innate/immunology , Protein Phosphatase 1/immunology , RNA, Viral/immunology , Animals , Cell Line , Cells, Cultured , Chlorocebus aethiops , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate/genetics , Immunoblotting , Interferon-Induced Helicase, IFIH1 , Interferon-beta/immunology , Interferon-beta/metabolism , Mice , Mice, Knockout , Microscopy, Confocal , Molecular Sequence Data , Mutation , Phosphorylation , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , RNA Interference , RNA, Viral/genetics , RNA, Viral/metabolism , Receptors, Immunologic , Signal Transduction/genetics , Signal Transduction/immunology , Vero Cells
4.
Nat Med ; 18(6): 961-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22635007

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma(1), a highly vascularized tumor originating from lymphatic endothelial cells, and of at least two different B cell malignancies(2,3). A dimeric complex formed by the envelope glycoproteins H and L (gH-gL) is required for entry of herpesviruses into host cells(4). We show that the ephrin receptor tyrosine kinase A2 (EphA2) is a cellular receptor for KSHV gH-gL. EphA2 co-precipitated with both gH-gL and KSHV virions. Infection of human epithelial cells with a GFP-expressing recombinant KSHV strain, as measured by FACS analysis, was increased upon overexpression of EphA2. Antibodies against EphA(2) and siRNAs directed against EphA2 inhibited infection of endothelial cells. Pretreatment of KSHV with soluble EphA2 resulted in inhibition of KSHV infection by up to 90%. This marked reduction of KSHV infection was seen with all the different epithelial and endothelial cells used in this study. Similarly, pretreating epithelial or endothelial cells with the soluble EphA2 ligand ephrinA4 impaired KSHV infection. Deletion of the gene encoding EphA2 essentially abolished KSHV infection of mouse endothelial cells. Binding of gH-gL to EphA2 triggered EphA2 phosphorylation and endocytosis, a major pathway of KSHV entry(5,6). Quantitative RT-PCR and in situ histochemistry revealed a close correlation between KSHV infection and EphA2 expression both in cultured cells derived from human Kaposi's sarcoma lesions or unaffected human lymphatic endothelium, and in situ in Kaposi's sarcoma specimens, respectively. Taken together, our results identify EphA2, a tyrosine kinase with known functions in neovascularization and oncogenesis, as an entry receptor for KSHV.


Subject(s)
Herpesvirus 8, Human/physiology , Receptor, EphA2/physiology , Receptors, Virus/physiology , Animals , Cell Line , Endocytosis , Humans , Mice , Phosphorylation , Viral Envelope Proteins/physiology , Viral Proteins/physiology
5.
J Virol ; 86(3): 1358-71, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22114345

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) is a key sensor for viral RNA in the cytosol, and it initiates a signaling cascade that leads to the establishment of an interferon (IFN)-mediated antiviral state. Because of its integral role in immune signaling, RIG-I activity must be precisely controlled. Recent studies have shown that RIG-I CARD-dependent signaling function is regulated by the dynamic balance between phosphorylation and TRIM25-induced K63-linked ubiquitination. While ubiquitination of RIG-I is critical for RIG-I's ability to induce an antiviral IFN response, phosphorylation of RIG-I at S8 or T170 suppresses RIG-I signal-transducing activity under normal conditions. Here, we not only further define the roles of S8 and T170 phosphorylation for controlling RIG-I activity but also identify conventional protein kinase C-α (PKC-α) and PKC-ß as important negative regulators of the RIG-I signaling pathway. Mutational analysis indicated that while the phosphorylation of S8 or T170 potently inhibits RIG-I downstream signaling, the dephosphorylation of RIG-I at both residues is necessary for optimal TRIM25 binding and ubiquitination-mediated RIG-I activation. Furthermore, exogenous expression, gene silencing, and specific inhibitor treatment demonstrated that PKC-α/ß are the primary kinases responsible for RIG-I S8 and T170 phosphorylation. Coimmunoprecipitation showed that PKC-α/ß interact with RIG-I under normal conditions, leading to its phosphorylation, which suppresses TRIM25 binding, RIG-I CARD ubiquitination, and thereby RIG-I-mediated IFN induction. PKC-α/ß double-knockdown cells exhibited markedly decreased S8/T170 phosphorylation levels of RIG-I and resistance to infection by vesicular stomatitis virus. Thus, these findings demonstrate that PKC-α/ß-induced RIG-I phosphorylation is a critical regulatory mechanism for controlling RIG-I antiviral signal transduction under normal conditions.


Subject(s)
DEAD-box RNA Helicases/metabolism , Protein Kinase C-alpha/physiology , Protein Kinase C/physiology , Signal Transduction/physiology , Animals , Cell Line , DEAD Box Protein 58 , Gene Knockdown Techniques , Humans , Interferon Type I/metabolism , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C beta , Protein Kinase C-alpha/genetics , Receptors, Immunologic , Ubiquitination
6.
J Virol ; 85(9): 4530-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21345951

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) carries four genes with homology to human interferon regulatory factors (IRFs). One of these IRFs, the viral interferon regulatory factor 3 (vIRF-3), is expressed in latently infected primary effusion lymphoma (PEL) cells and required for their continuous proliferation. Moreover, vIRF-3 is known to be involved in modulation of the type I interferon (IFN) response. We now show that vIRF-3 also interferes with the type II interferon system and antigen presentation to the adaptive immune system. Starting with an analysis of the transcriptome, we show that vIRF-3 inhibits expression of major histocompatibility complex class II (MHC II) molecules: small interfering RNA (siRNA)-mediated knockdown of vIRF-3 in KSHV-infected PEL cell lines resulted in increased MHC II levels; overexpression of vIRF-3 in KSHV-negative B cells leads to downmodulation of MHC II. This regulation could be traced back to inhibition of class II transactivator (CIITA) transcription by vIRF-3. Reporter assays revealed that the gamma interferon (IFN-γ)-sensitive CIITA promoters PIV and PIII were inhibited by vIRF-3. Consistently, IFN-γ levels increased upon vIRF-3 knockdown in PEL cells. IFN-γ regulation by vIRF-3 was confirmed in reporter assays as well as by upregulation of typical IFN-γ target genes upon knockdown of vIRF-3 in PEL cells. In summary, we conclude that vIRF-3 contributes to the viral immunoevasion by downregulation of IFN-γ and CIITA and thus MHC II expression.


Subject(s)
Herpesvirus 8, Human/immunology , Herpesvirus 8, Human/pathogenicity , Histocompatibility Antigens Class II/biosynthesis , Interferon Regulatory Factors/metabolism , Interferon-gamma/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors , Viral Proteins/metabolism , Antigen Presentation , Gene Expression Profiling , Humans , Immune Evasion
7.
J Biol Chem ; 284(13): 8525-38, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19129183

ABSTRACT

Kaposi's sarcoma-associated herpesvirus encodes four genes with homology to the family of interferon regulatory factors (IRFs). At least one of these viral IRFs, vIRF-3, is expressed in latently Kaposi's sarcoma-associated herpesvirus-infected primary effusion lymphoma (PEL) cells and is essential for the survival of PEL cells. We now report that vIRF-3 interacts with cellular IRF-5, thereby inhibiting binding of IRF-5 to interferon-responsive promoter elements. Consequently, vIRF-3 blocked IRF-5-mediated promoter activation. A central double helix motif present in vIRF-3 was sufficient to abrogate both DNA binding and transcriptional transactivation by IRF-5. Upon DNA damage or activation of the interferon or Toll-like receptor pathways, cytoplasmic IRF-5 has been reported to be translocated to the nucleus, which results in induction of both p53-independent apoptosis and p21-mediated cell cycle arrest. We report here that IRF-5 is present in the nuclei of PEL cells without interferon stimulation. Silencing of vIRF-3 expression in PEL cells was accompanied by increased sensitivity to interferon-mediated apoptosis and up-regulation of IRF-5 target genes. In addition, vIRF-3 antagonized IRF-5-mediated activation of the p21 promoter. The data presented here indicate that vIRF-3 contributes to immune evasion and sustained proliferation of PEL cells by releasing IRF-5 from transcription complexes.


Subject(s)
Cell Nucleus/metabolism , Herpesvirus 8, Human/metabolism , Interferon Regulatory Factors/metabolism , Response Elements , Viral Proteins/metabolism , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/immunology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Apoptosis/genetics , Apoptosis/immunology , Cell Nucleus/genetics , Cell Nucleus/immunology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/immunology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Silencing , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/immunology , Lymphoma, Primary Effusion/metabolism , Protein Binding/genetics , Protein Binding/immunology , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Receptors, Interferon/metabolism , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Transcriptional Activation/genetics , Transcriptional Activation/immunology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism , Up-Regulation/genetics , Up-Regulation/immunology , Viral Proteins/genetics , Viral Proteins/immunology
8.
J Virol ; 83(6): 2563-74, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19129458

ABSTRACT

Human herpesvirus 8 (HHV-8) is the etiologic agent of Kaposi's sarcoma and primary effusion lymphoma. Activation of the cellular transcription factor nuclear factor-kappa B (NF-kappaB) is essential for latent persistence of HHV-8, survival of HHV-8-infected cells, and disease progression. We used reverse-transfected cell microarrays (RTCM) as an unbiased systems biology approach to systematically analyze the effects of HHV-8 genes on the NF-kappaB signaling pathway. All HHV-8 genes individually (n = 86) and, additionally, all K and latent genes in pairwise combinations (n = 231) were investigated. Statistical analyses of more than 14,000 transfections identified ORF75 as a novel and confirmed K13 as a known HHV-8 activator of NF-kappaB. K13 and ORF75 showed cooperative NF-kappaB activation. Small interfering RNA-mediated knockdown of ORF75 expression demonstrated that this gene contributes significantly to NF-kappaB activation in HHV-8-infected cells. Furthermore, our approach confirmed K10.5 as an NF-kappaB inhibitor and newly identified K1 as an inhibitor of both K13- and ORF75-mediated NF-kappaB activation. All results obtained with RTCM were confirmed with classical transfection experiments. Our work describes the first successful application of RTCM for the systematic analysis of pathofunctions of genes of an infectious agent. With this approach, ORF75 and K1 were identified as novel HHV-8 regulatory molecules on the NF-kappaB signal transduction pathway. The genes identified may be involved in fine-tuning of the balance between latency and lytic replication, since this depends critically on the state of NF-kappaB activity.


Subject(s)
Herpesvirus 8, Human/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/biosynthesis , Systems Biology/methods , Viral Proteins/metabolism , Cell Line , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Oligonucleotide Array Sequence Analysis , Viral Proteins/genetics
9.
J Virol ; 83(1): 396-407, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18945775

ABSTRACT

The attachment, entry, and fusion of Kaposi's sarcoma-associated herpesvirus (KSHV) with target cells are mediated by complex machinery containing, among others, viral glycoprotein H (gH) and its alleged chaperone, gL. We observed that KSHV gH, in contrast to its homologues in several other herpesviruses, is transported to the cytoplasm membrane independently from gL, but not vice versa. Mutational analysis revealed that the N terminus of gH is sufficient for gL interaction. However, the entire extracellular part of gH is required for efficient gL secretion. The soluble ectodomain of gH was sufficient to interact with the surfaces of potential target cells in a heparin-dependent manner, and binding was further enhanced by coexpression of gL. Surface plasmon resonance revealed a remarkably high affinity of gH for glycosaminoglycans. Heparan sulfate (HS) proteoglycans of the syndecan family act as cellular receptors for the gH/gL complex. They promoted KSHV infection, and expression of gH/gL on target cells inhibited subsequent KSHV infection. Whereas gH alone was able to bind to HS, we observed that only the gH/gL complex adhered to heparan sulfate-negative cells at lamellipodium-like structures.


Subject(s)
Herpesvirus 8, Human/physiology , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism , Virus Internalization , Cell Line , Cell Membrane/chemistry , DNA Mutational Analysis , Glycosaminoglycans/metabolism , Heparan Sulfate Proteoglycans/metabolism , Humans , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Transport , Surface Plasmon Resonance , Viral Envelope Proteins/genetics
10.
Comb Chem High Throughput Screen ; 11(2): 159-72, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18336209

ABSTRACT

Reversely transfected cell microarrays (RTCM) have been introduced as a method for parallel high throughput analysis of gene functions in mammalian cells. Hundreds to thousands of different recombinant DNA or RNA molecules can be transfected into different cell clusters at the same time on a single glass slide with this method. This allows either the simultaneous overexpression or--by using the recently developed RNA interference (RNAi) techniques--knockdown of a huge number of target genes. A growing number of sophisticated detection systems have been established to determine quantitatively the effects of the transfected molecules on the cell phenotype. Several different cell types have been successfully used for this procedure. This review summarizes the presently available knowledge on this technique and provides a laboratory protocol.


Subject(s)
Drug Evaluation, Preclinical/methods , Genes/genetics , Genes/physiology , Reverse Transcription/genetics , Tissue Array Analysis/methods , Transfection/methods , Animals , Cells, Cultured , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Gene Silencing , Humans
11.
J Virol ; 82(4): 1908-22, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18077714

ABSTRACT

Human herpesvirus 8 (HHV-8) is the etiological agent of Kaposi's sarcoma. We present a localization map of 85 HHV-8-encoded proteins in mammalian cells. Viral open reading frames were cloned with a Myc tag in expression plasmids, confirmed by full-length sequencing, and expressed in HeLa cells. Protein localizations were analyzed by immunofluorescence microscopy. Fifty-one percent of all proteins were localized in the cytoplasm, 22% were in the nucleus, and 27% were found in both compartments. Surprisingly, we detected viral FLIP (v-FLIP) in the nucleus and in the cytoplasm, whereas cellular FLIPs are generally localized exclusively in the cytoplasm. This suggested that v-FLIP may exert additional or alternative functions compared to cellular FLIPs. In addition, it has been shown recently that the K10 protein can bind to at least 15 different HHV-8 proteins. We noticed that K10 and only five of its 15 putative binding factors were localized in the nucleus when the proteins were expressed in HeLa cells individually. Interestingly, in coexpression experiments K10 colocalized with 87% (13 of 15) of its putative binding partners. Colocalization was induced by translocation of either K10 alone or both proteins. These results indicate active intracellular translocation processes in virus-infected cells. Specifically in this framework, the localization map may provide a useful reference to further elucidate the function of HHV-8-encoded genes in human diseases.


Subject(s)
Herpesvirus 8, Human/metabolism , Viral Proteins/analysis , Amino Acid Sequence , Cell Nucleus/chemistry , Chromosome Mapping , Cloning, Molecular , Cytoplasm/chemistry , Endoplasmic Reticulum/chemistry , Golgi Apparatus/chemistry , HeLa Cells , Herpesvirus 8, Human/genetics , Humans , Molecular Sequence Data , Open Reading Frames , Viral Proteins/genetics , Viral Proteins/metabolism
12.
Blood ; 111(1): 320-7, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17890449

ABSTRACT

Human herpesvirus-8 (HHV-8), also known as Kaposi sarcoma-associated herpesvirus (KSHV), is etiologically linked to primary effusion lymphoma (PEL). At least 10 KSHV-encoded proteins with potential roles in KSHV-associated neoplasia have been identified. However, with few exceptions, these putative oncogenes were analyzed in heterologous systems only using overexpression of single genes. Thus, the pathogenetic relevance of most of these putative oncogenes remains essentially unclear. We used RNA interference (RNAi) to knock down the expression of several KSHV genes in cultured PEL cells carrying the KSHV genome. The viral interferon-regulatory factor-3 (vIRF-3) was found to be required for proliferation and survival of cultured PEL cells. Knock-down of vIRF-3 expression by various RNAi approaches unequivocally resulted in reduced proliferation and increased activity of caspase-3 and/or caspase-7. Thus, vIRF-3 can be seen as a bona fide oncogene of KSHV-associated lymphoma. Surprisingly, although the related Epstein-Barr virus (EBV) is usually sufficient to immortalize human B lymphocytes, silencing of vIRF-3 reduced the viability of both EBV(-) and EBV(+) PEL cells. This suggests that KSHV is the driving force in the pathogenesis of PEL.


Subject(s)
Herpesvirus 8, Human/genetics , Interferon Regulatory Factors/genetics , Lymphoma/virology , Pleural Effusion/virology , Sarcoma, Kaposi/virology , Viral Proteins/genetics , Apoptosis , Burkitt Lymphoma , Caspase 3/metabolism , Caspase 7/metabolism , Cell Division , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Humans , Lymphoma/pathology , Multiple Myeloma , Pleural Effusion/pathology , RNA, Small Interfering , Sarcoma, Kaposi/pathology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...