Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1286617, 2023.
Article in English | MEDLINE | ID: mdl-37876935

ABSTRACT

Pioneer factors are transcription factors sharing the fascinating ability to bind to compact chromatin and thereby alter its transcriptional fate. Most pioneer factors are known for their importance during embryonic development, for instance, in inducing zygotic genome activation or cell fate decision. Some pioneer factors are actively induced or downregulated by viral infection. With this, viruses are capable to modulate different signaling pathways resulting for example in MHC-receptor up/downregulation which contributes to viral immune evasion. In this article, we review the current state of research on how different viruses (Herpesviruses, Papillomaviruses and Hepatitis B virus) use pioneer factors for their viral replication and persistence in the host, as well as for the development of viral cancer.


Subject(s)
Virus Diseases , Virus Replication , Humans , Virus Replication/physiology , Transcription Factors/metabolism , Signal Transduction
2.
Res Sq ; 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38168299

ABSTRACT

DUX4 is a germline transcription factor and a master regulator of zygotic genome activation. During early embryogenesis, DUX4 is crucial for maternal to zygotic transition at the 2-8-cell stage in order to overcome silencing of genes and enable transcription from the zygotic genome. In adult somatic cells, DUX4 expression is silenced and its activation in adult muscle cells causes the genetic disorder Facioscapulohumeral Muscular Dystrophy (FSHD). Here we show that herpesviruses from alpha-, beta- and gamma-herpesvirus subfamilies as well as papillomaviruses actively induce DUX4 expression to promote viral transcription and replication. We demonstrate that HSV-1 immediate early proteins directly induce expression of DUX4 and its target genes including endogenous retroelements, which mimics zygotic genome activation. We further show that DUX4 directly binds to the viral genome and promotes viral transcription. DUX4 is functionally required for herpesvirus infection, since genetic depletion of DUX4 by CRISPR/Cas9 abrogates viral replication. Our results show that herpesviruses induce DUX4 expression and its downstream germline-specific genes and retroelements, thus mimicking an early embryonic-like transcriptional program that prevents epigenetic silencing of the viral genome and facilitates herpesviral gene expression.

3.
Int J Mol Sci ; 23(19)2022 Oct 05.
Article in English | MEDLINE | ID: mdl-36233116

ABSTRACT

The complex host interaction network of human cytomegalovirus (HCMV) involves the regulatory protein kinase pUL97, which represents a viral cyclin-dependent kinase (CDK) ortholog. pUL97 interacts with the three human cyclin types T1, H, and B1, whereby the binding region of cyclin T1 and the pUL97 oligomerization region were both assigned to amino acids 231-280. We further addressed the question of whether HCMVs harboring mutations in ORF-UL97, i.e., short deletions or resistance-conferring point mutations, are affected in the interaction with human cyclins and viral replication. To this end, clinically relevant UL97 drug-resistance-conferring mutants were analyzed by whole-genome sequencing and used for genetic marker transfer experiments. The recombinant HCMVs indicated conservation of pUL97-cyclin interaction, since all viral UL97 point mutants continued to interact with the analyzed cyclin types and exerted wild-type-like replication fitness. In comparison, recombinant HCMVs UL97 Δ231-280 and also the smaller deletion Δ236-275, but not Δ241-270, lost interaction with cyclins T1 and H, showed impaired replication efficiency, and also exhibited reduced kinase activity. Moreover, a cellular knock-out of cyclins B1 or T1 did not alter HCMV replication phenotypes or pUL97 kinase activity, possibly indicating alternative, compensatory pUL97-cyclin interactions. In contrast, however, cyclin H knock-out, similar to virus deletion mutants in the pUL97-cyclin H binding region, exhibited strong defective phenotypes of HCMV replication, as supported by reduced pUL97 kinase activity in a cyclin H-dependent coexpression setting. Thus, cyclin H proved to be a very relevant determinant of pUL97 kinase activity and viral replication efficiency. As a conclusion, the results provide evidence for the functional importance of pUL97-cyclin interaction. High selective pressure on the formation of pUL97-cyclin complexes was identified by the use of clinically relevant mutants.


Subject(s)
Cyclin H , Cytomegalovirus , Viral Proteins , Amino Acids/metabolism , Cyclin H/genetics , Cyclin H/metabolism , Cyclin T/genetics , Cyclin T/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cytomegalovirus/physiology , Genetic Markers , Humans , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Viral Proteins/genetics , Virus Replication/genetics
4.
Elife ; 112022 03 23.
Article in English | MEDLINE | ID: mdl-35319461

ABSTRACT

PML nuclear bodies (PML-NBs) are dynamic interchromosomal macromolecular complexes implicated in epigenetic regulation as well as antiviral defense. During herpesvirus infection, PML-NBs induce epigenetic silencing of viral genomes, however, this defense is antagonized by viral regulatory proteins such as IE1 of human cytomegalovirus (HCMV). Here, we show that PML-NBs undergo a drastic rearrangement into highly enlarged PML cages upon infection with IE1-deficient HCMV. Importantly, our results demonstrate that dual signaling by interferon and DNA damage response is required to elicit giant PML-NBs. DNA labeling revealed that invading HCMV genomes are entrapped inside PML-NBs and remain stably associated with PML cages in a transcriptionally repressed state. Intriguingly, by correlative light and transmission electron microscopy (EM), we observed that PML cages also entrap newly assembled viral capsids demonstrating a second defense layer in cells with incomplete first-line response. Further characterization by 3D EM showed that hundreds of viral capsids are tightly packed into several layers of fibrous PML. Overall, our data indicate that giant PML-NBs arise via combined interferon and DNA damage signaling which triggers entrapment of both nucleic acids and proteinaceous components. This represents a multilayered defense strategy to act in a cytoprotective manner and to combat viral infections.


Subject(s)
Interferons , Nuclear Proteins , Antiviral Agents , DNA Damage , Epigenesis, Genetic , Humans , Interferons/metabolism , Nuclear Bodies , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/genetics , Transcription Factors/metabolism
5.
mBio ; 12(1)2021 01 19.
Article in English | MEDLINE | ID: mdl-33468685

ABSTRACT

The HIV-1 Rev protein is a nuclear export factor for unspliced and incompletely spliced HIV-1 RNAs. Without Rev, these intron-retaining RNAs are trapped in the nucleus. A genome-wide screen identified nine proteins of the spliceosome, which all enhanced expression from the HIV-1 unspliced RNA after CRISPR/Cas knockdown. Depletion of DHX38, WDR70, and four proteins of the Prp19-associated complex (ISY1, BUD31, XAB2, and CRNKL1) resulted in a more than 20-fold enhancement of unspliced HIV-1 RNA levels in the cytoplasm. Targeting of CRNKL1, DHX38, and BUD31 affected nuclear export efficiencies of the HIV-1 unspliced RNA to a much larger extent than splicing. Transcriptomic analyses further revealed that CRNKL1 also suppresses cytoplasmic levels of a subset of cellular mRNAs, including some with selectively retained introns. Thus, CRNKL1-dependent nuclear retention is a novel cellular mechanism for the regulation of cytoplasmic levels of intron-retaining HIV-1 mRNAs, which HIV-1 may have harnessed to direct its complex splicing pattern.IMPORTANCE To regulate its complex splicing pattern, HIV-1 uses the adaptor protein Rev to shuttle unspliced or partially spliced mRNA from the nucleus to the cytoplasm. In the absence of Rev, these RNAs are retained in the nucleus, but it is unclear why. Here we identify cellular proteins whose depletion enhances cytoplasmic levels of the HIV-1 unspliced RNA. Depletion of one of them, CRNKL1, also increases cytoplasmic levels of a subset of intron-retaining cellular mRNA, suggesting that CRNKL1-dependent nuclear retention may be a basic cellular mechanism exploited by HIV-1.


Subject(s)
HIV-1/genetics , Nuclear Proteins/genetics , RNA Splicing , RNA, Messenger/genetics , RNA, Viral/genetics , rev Gene Products, Human Immunodeficiency Virus/genetics , Cell Nucleus/genetics , Cell Nucleus/virology , Cytosol/metabolism , Cytosol/virology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Exons , Gene Regulatory Networks , HIV-1/metabolism , Host-Pathogen Interactions/genetics , Humans , Introns , Jurkat Cells , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Interaction Mapping , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Viral/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , Transcriptome , rev Gene Products, Human Immunodeficiency Virus/metabolism
6.
J Clin Med ; 8(9)2019 Sep 07.
Article in English | MEDLINE | ID: mdl-31500286

ABSTRACT

Herpesviruses are important pathogens that can cause significant morbidity and mortality in the human population. Herpesviruses have a double-stranded DNA genome, and viral genome replication takes place inside the nucleus. Upon entering the nucleus, herpesviruses have to overcome the obstacle of cellular proteins in order to enable viral gene expression and genome replication. In this review, we want to highlight cellular proteins that sense incoming viral genomes of the DNA-damage repair (DDR) pathway and of PML-nuclear bodies (PML-NBs) that all can act as antiviral restriction factors within the first hours after the viral genome is released into the nucleus. We show the function and significance of both nuclear DNA sensors, the DDR and PML-NBs, and demonstrate for three human herpesviruses of the alpha-, beta- and gamma-subfamilies, HSV-1, HCMV and KSHV respectively, how viral tegument proteins antagonize these pathways.

7.
Nat Microbiol ; 4(1): 164-176, 2019 01.
Article in English | MEDLINE | ID: mdl-30420784

ABSTRACT

Tripartite motif (TRIM) proteins mediate antiviral host defences by either directly targeting viral components or modulating innate immune responses. Here we identify a mechanism of antiviral restriction in which a TRIM E3 ligase controls viral replication by regulating the structure of host cell centrosomes and thereby nuclear lamina integrity. Through RNAi screening we identified several TRIM proteins, including TRIM43, that control the reactivation of Kaposi's sarcoma-associated herpesvirus. TRIM43 was distinguished by its ability to restrict a broad range of herpesviruses and its profound upregulation during herpesvirus infection as part of a germline-specific transcriptional program mediated by the transcription factor DUX4. TRIM43 ubiquitinates the centrosomal protein pericentrin, thereby targeting it for proteasomal degradation, which subsequently leads to alterations of the nuclear lamina that repress active viral chromatin states. Our study identifies a role of the TRIM43-pericentrin-lamin axis in intrinsic immunity, which may be targeted for therapeutic intervention against herpesviral infections.


Subject(s)
Antigens/metabolism , Centrosome/metabolism , Herpesviridae Infections/immunology , Herpesvirus 8, Human/growth & development , Tripartite Motif Proteins/metabolism , Virus Replication/physiology , A549 Cells , Animals , Cell Line, Tumor , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Herpesviridae Infections/prevention & control , Herpesviridae Infections/virology , Homeodomain Proteins/metabolism , Humans , Nuclear Lamina/physiology , RNA Interference , RNA, Small Interfering/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/physiology , Ubiquitination , Vero Cells , Virus Replication/genetics
8.
J Exp Med ; 215(9): 2289-2310, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30068544

ABSTRACT

Patients with epidermodysplasia verruciformis (EV) and biallelic null mutations of TMC6 (encoding EVER1) or TMC8 (EVER2) are selectively prone to disseminated skin lesions due to keratinocyte-tropic human ß-papillomaviruses (ß-HPVs), which lack E5 and E8. We describe EV patients homozygous for null mutations of the CIB1 gene encoding calcium- and integrin-binding protein-1 (CIB1). CIB1 is strongly expressed in the skin and cultured keratinocytes of controls but not in those of patients. CIB1 forms a complex with EVER1 and EVER2, and CIB1 proteins are not expressed in EVER1- or EVER2-deficient cells. The known functions of EVER1 and EVER2 in human keratinocytes are not dependent on CIB1, and CIB1 deficiency does not impair keratinocyte adhesion or migration. In keratinocytes, the CIB1 protein interacts with the HPV E5 and E8 proteins encoded by α-HPV16 and γ-HPV4, respectively, suggesting that this protein acts as a restriction factor against HPVs. Collectively, these findings suggest that the disruption of CIB1-EVER1-EVER2-dependent keratinocyte-intrinsic immunity underlies the selective susceptibility to ß-HPVs of EV patients.


Subject(s)
Betapapillomavirus/immunology , Calcium-Binding Proteins/immunology , Epidermodysplasia Verruciformis/immunology , Immunity, Innate , Keratinocytes/immunology , Membrane Proteins/immunology , Multiprotein Complexes/immunology , Adult , Aged , Aged, 80 and over , Cell Adhesion/immunology , Cell Movement/immunology , Epidermodysplasia Verruciformis/pathology , Female , Human papillomavirus 16/immunology , Humans , Keratinocytes/pathology , Male , Middle Aged , Oncogene Proteins, Viral/immunology
9.
Viruses ; 9(10)2017 10 21.
Article in English | MEDLINE | ID: mdl-29065450

ABSTRACT

Gammaherpesviruses like Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) subvert the ubiquitin proteasome system for their own benefit in order to facilitate viral gene expression and replication. In particular, viral tegument proteins that share sequence homology to the formylglycineamide ribonucleotide amidotransferase (FGARAT, or PFAS), an enzyme in the cellular purine biosynthesis, are important for disrupting the intrinsic antiviral response associated with Promyelocytic Leukemia (PML) protein-associated nuclear bodies (PML-NBs) by proteasome-dependent and independent mechanisms. In addition, all herpesviruses encode for a potent ubiquitin protease that can efficiently remove ubiquitin chains from proteins and thereby interfere with several different cellular pathways. In this review, we discuss mechanisms and functional consequences of virus-induced ubiquitination and deubiquitination for early events in gammaherpesviral infection.


Subject(s)
Herpesvirus 8, Human/chemistry , Host-Pathogen Interactions , Promyelocytic Leukemia Protein/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/genetics , DNA Replication/genetics , Herpesviridae Infections/virology , Herpesvirus 8, Human/enzymology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Humans , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/genetics , Ubiquitination , Virus Replication
10.
Nat Microbiol ; 2(11): 1543-1557, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28871090

ABSTRACT

Autophagy and interferon (IFN)-mediated innate immunity are critical antiviral defence mechanisms, and recent evidence indicated that tripartite motif (TRIM) proteins are important regulators of both processes. Although the role of TRIM proteins in modulating antiviral cytokine responses has been well established, much less is known about their involvement in autophagy in response to different viral pathogens. Through a targeted RNAi screen examining the relevance of selected TRIM proteins in autophagy induced by herpes simplex virus 1 (HSV-1), encephalomyocarditis virus (EMCV) and influenza A virus (IAV), we identified several TRIM proteins that regulate autophagy in a virus-species-specific manner, as well as a few TRIM proteins that were essential for autophagy triggered by all three viruses and rapamycin, among them TRIM23. TRIM23 was critical for autophagy-mediated restriction of multiple viruses, and this activity was dependent on both its RING E3 ligase and ADP-ribosylation factor (ARF) GTPase activity. Mechanistic studies revealed that unconventional K27-linked auto-ubiquitination of the ARF domain is essential for the GTP hydrolysis activity of TRIM23 and activation of TANK-binding kinase 1 (TBK1) by facilitating its dimerization and ability to phosphorylate the selective autophagy receptor p62. Our work identifies the TRIM23-TBK1-p62 axis as a key component of selective autophagy and further reveals a role for K27-linked ubiquitination in GTPase-dependent TBK1 activation.


Subject(s)
Autophagy , GTP-Binding Proteins/metabolism , Host-Pathogen Interactions , Protein Serine-Threonine Kinases/metabolism , Virus Physiological Phenomena , Encephalomyocarditis virus/physiology , Enzyme Activation , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Guanosine Triphosphate/metabolism , Herpesvirus 1, Human/physiology , Humans , Hydrolysis , Immunity, Innate , Influenza A virus/physiology , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , RNA Interference , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
11.
Front Microbiol ; 7: 844, 2016.
Article in English | MEDLINE | ID: mdl-27375569

ABSTRACT

In order to explore the potential of HLA-independent T cell therapy for human cytomegalovirus (HCMV) infections, we developed a chimeric antigen receptor (CAR) directed against the HCMV encoded glycoprotein B (gB), which is expressed at high levels on the surface of infected cells. T cells engineered with this anti-gB CAR recognized HCMV-infected cells and released cytokines and cytotoxic granules. Unexpectedly, and in contrast to analogous approaches for HIV, Hepatitis B or Hepatitis C virus, we found that HCMV-infected cells were resistant to killing by the CAR-modified T cells. In order to elucidate whether this phenomenon was restricted to the use of CARs, we extended our experiments to T cell receptor (TCR)-mediated recognition of infected cells. To this end we infected fibroblasts with HCMV-strains deficient in viral inhibitors of antigenic peptide presentation and targeted these HLA-class I expressing peptide-loaded infected cells with peptide-specific cytotoxic T cells (CTLs). Despite strong degranulation and cytokine production by the T cells, we again found significant inhibition of lysis of HCMV-infected cells. Impairment of cell lysis became detectable 1 day after HCMV infection and gradually increased during the following 3 days. We thus postulate that viral anti-apoptotic factors, known to inhibit suicide of infected host cells, have evolved additional functions to directly abrogate T cell cytotoxicity. In line with this hypothesis, CAR-T cell cytotoxicity was strongly inhibited in non-infected fibroblasts by expression of the HCMV-protein UL37x1, and even more so by additional expression of UL36. Our data extend the current knowledge on Betaherpesviral evasion from T cell immunity and show for the first time that, beyond impaired antigen presentation, infected cells are efficiently protected by direct blockade of cytotoxic effector functions through viral proteins.

13.
Cell Host Microbe ; 16(1): 19-30, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-25011105

ABSTRACT

The cytosolic sensor MDA5 is crucial for antiviral innate immune defense against various RNA viruses including measles virus; as such, many viruses have evolved strategies to antagonize the antiviral activity of MDA5. Here, we show that measles virus escapes MDA5 detection by targeting the phosphatases PP1α and PP1γ, which regulate MDA5 activity by removing an inhibitory phosphorylation mark. The V proteins of measles virus and the related paramyxovirus Nipah virus interact with PP1α/γ, preventing PP1-mediated dephosphorylation of MDA5 and thereby its activation. The PP1 interaction with the measles V protein is mediated by a conserved PP1-binding motif in the C-terminal region of the V protein. A recombinant measles virus expressing a mutant V protein deficient in PP1 binding is unable to antagonize MDA5 and is growth impaired due to its inability to suppress interferon induction. This identifies PP1 antagonism as a mechanism employed by paramyxoviruses for evading innate immune recognition.


Subject(s)
DEAD-box RNA Helicases/metabolism , Host-Pathogen Interactions , Immune Evasion , Measles virus/immunology , Measles virus/physiology , Phosphoproteins/metabolism , Protein Phosphatase 1/antagonists & inhibitors , Viral Proteins/metabolism , Cell Line , Humans , Interferon-Induced Helicase, IFIH1 , Nipah Virus/immunology , Nipah Virus/physiology , Viral Structural Proteins/metabolism
14.
Immunity ; 40(4): 453-4, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24745327

ABSTRACT

Prostaglandins are lipid mediators that are involved in a plethora of biological processes. In this issue of Immunity, Coulombe et al. (2014) report that prostaglandin E2 suppresses innate and adaptive immune responses to influenza virus.


Subject(s)
Dinoprostone/metabolism , Influenza A virus/physiology , Interferon Type I/metabolism , Intramolecular Oxidoreductases/antagonists & inhibitors , Macrophages/drug effects , Orthomyxoviridae Infections/drug therapy , Animals , Prostaglandin-E Synthases
15.
PLoS Pathog ; 10(1): e1003863, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24453968

ABSTRACT

Nuclear domain 10 (ND10) components are restriction factors that inhibit herpesviral replication. Effector proteins of different herpesviruses can antagonize this restriction by a variety of strategies, including degradation or relocalization of ND10 proteins. We investigated the interplay of Kaposi's Sarcoma-Associated Herpesvirus (KSHV) infection and cellular defense by nuclear domain 10 (ND10) components. Knock-down experiments in primary human cells show that KSHV-infection is restricted by the ND10 components PML and Sp100, but not by ATRX. After KSHV infection, ATRX is efficiently depleted and Daxx is dispersed from ND10, indicating that these two ND10 components can be antagonized by KSHV. We then identified the ORF75 tegument protein of KSHV as the viral factor that induces the disappearance of ATRX and relocalization of Daxx. ORF75 belongs to a viral protein family (viral FGARATs) that has homologous proteins in all gamma-herpesviruses. Isolated expression of ORF75 in primary cells induces a relocalization of PML and dispersal of Sp100, indicating that this viral effector protein is able to influence multiple ND10 components. Moreover, by constructing a KSHV mutant harboring a stop codon at the beginning of ORF75, we could demonstrate that ORF75 is absolutely essential for viral replication and the initiation of viral immediate-early gene expression. Using recombinant viruses either carrying Flag- or YFP-tagged variants of ORF75, we could further corroborate the role of ORF75 in the antagonization of ND10-mediated intrinsic immunity, and show that it is independent of the PML antagonist vIRF3. Members of the viral FGARAT family target different ND10 components, suggesting that the ND10 targets of viral FGARAT proteins have diversified during evolution. We assume that overcoming ND10 intrinsic defense constitutes a critical event in the replication of all herpesviruses; on the other hand, restriction of herpesviral replication by ND10 components may also promote latency as the default outcome of infection.


Subject(s)
Herpesviridae Infections/immunology , Herpesvirus 8, Human/physiology , Immunity, Innate , Nuclear Proteins/immunology , Viral Structural Proteins/immunology , Virus Replication/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Cells, Cultured , Co-Repressor Proteins , Codon, Terminator/genetics , Codon, Terminator/immunology , DNA Helicases/genetics , DNA Helicases/immunology , Gene Knockdown Techniques , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Humans , Male , Molecular Chaperones , Mutation , Nuclear Proteins/genetics , Viral Structural Proteins/genetics , X-linked Nuclear Protein
16.
PLoS One ; 7(2): e31210, 2012.
Article in English | MEDLINE | ID: mdl-22355347

ABSTRACT

We explored the possibility to target Ewing's sarcoma family of tumors (ESFT) by redirecting T cells. To this aim, we considered NKG2D-ligands (NKG2D-Ls) as possible target antigens. Detailed analysis of the expression of MICA, MICB, ULBP-1, -2, and -3 in fourteen ESFT cell lines revealed consistent expression of at least one NKG2D-L. Thus, for redirecting T cells, we fused a CD3ζ/CD28-derived signaling domain to the ectodomain of NKG2D, however, opposite transmembrane orientation of this signaling domain and NKG2D required inverse orientation fusion of either of them. We hypothesized that the particularly located C-terminus of the NKG2D ectodomain should allow reengineering of the membrane anchoring from a native N-terminal to an artificial C-terminal linkage. Indeed, the resulting chimeric NKG2D receptor (chNKG2D) was functional and efficiently mediated ESFT cell death triggered by activated T cells. Notably, ESFT cells with even low NKG2D-L expression were killed by CD8(pos) and also CD4(pos) cells. Both, mRNA transfection and lentiviral transduction resulted in high level surface expression of chNKG2D. However, upon target-cell recognition receptor surface levels were maintained by tranfected RNA only during the first couple of hours after transfection. Later, target-cell contact resulted in strong and irreversible receptor down-modulation, whereas lentivirally mediated expression of chNKG2D remained constant under these conditions. Together, our study defines NKG2D-Ls as targets for a CAR-mediated T cell based immunotherapy of ESFT. A comparison of two different methods of gene transfer reveals strong differences in the susceptibility to ligand-induced receptor down-modulation with possible implications for the applicability of RNA transfection.


Subject(s)
Immunotherapy , NK Cell Lectin-Like Receptor Subfamily K/metabolism , RNA, Messenger/administration & dosage , Recombinant Fusion Proteins/metabolism , Sarcoma, Ewing/immunology , Sarcoma, Ewing/therapy , T-Lymphocytes/immunology , Apoptosis , Blotting, Western , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K/genetics , Recombinant Fusion Proteins/genetics , T-Lymphocytes/transplantation , Tumor Cells, Cultured
17.
J Virol ; 86(7): 3541-53, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22278248

ABSTRACT

In recent studies, the nuclear domain 10 (ND10) components PML, Sp100, human Daxx (hDaxx), and ATRX were identified to be cellular restriction factors that are able to inhibit the replication of several herpesviruses. The antiviral function of ND10, however, is antagonized by viral effector proteins by a variety of strategies, including degradation of PML or relocalization of ND10 proteins. In this study, we analyzed the interplay between infection with herpesvirus saimiri (HVS), the prototypic rhadinovirus, and cellular defense by ND10. In contrast to other herpesviruses, we found that HVS specifically degraded the cellular ND10 component Sp100, whereas other factors like PML or hDaxx remained intact. We could further identify the ORF3 tegument protein of HVS, which shares homology with the cellular formylglycinamide ribotide amidotransferase (FGARAT) enzyme, to be the viral factor that induces the proteasomal degradation of Sp100. Interestingly, recent studies showed that the ORF3-homologous proteins ORF75c of murine gammaherpesvirus 68 and BNRF-1 of Epstein-Barr virus modulate the ND10 proteins PML and ATRX, respectively, suggesting that the ND10 targets of viral FGARAT-homologous proteins diversified during evolution. Furthermore, a virus with the ORF3 deletion was efficiently complemented in Sp100-depleted cells, indicating that Sp100 is able to inhibit HVS in the absence of antagonistic mechanisms. In contrast, we observed that PML, which was neither degraded nor redistributed after HVS infection, strongly restricted both wild-type HVS and virus with the ORF3 deletion. Thus, HVS may lack a factor that efficiently counteracts the repressive function of PML, which may foster latency as the outcome of infection.


Subject(s)
Antigens, Nuclear/metabolism , Autoantigens/metabolism , Herpesviridae Infections/metabolism , Herpesvirus 2, Saimiriine/metabolism , Nuclear Proteins/immunology , Open Reading Frames , Viral Proteins/metabolism , Antigens, Nuclear/genetics , Autoantigens/genetics , Cell Line , Gene Expression Regulation, Viral , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesvirus 2, Saimiriine/genetics , Humans , Immunity , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Protein Binding , Viral Proteins/genetics
18.
J Virol ; 86(3): 1862-73, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22130528

ABSTRACT

Herpesviruses establish latency in suitable cells of the host organism after a primary lytic infection. Subgroup C strains of herpesvirus saimiri (HVS), a primate gamma-2 herpesvirus, are able to transform human and other primate T lymphocytes to stable growth in vitro. The viral genomes persist as nonintegrated, circular, and histone-associated episomes in the nuclei of those latently infected T cells. Epigenetic modifications of episomes are essential to restrict the transcription during latency to selected viral genes, such as the viral oncogenes stpC/tip and the orf73/LANA. In this study, we describe a genome-wide chromatin immunoprecipitation-on-chip (ChIP-on-chip) analysis to profile the occupancy of CTCF on the latent HVS genome. We then focused on two distinct, conserved CTCF binding sites (CBS) within the orf73/LANA promoter region. Analysis of recombinant viruses harboring deletions or mutations within the CBS indicated that the lytic replication of such viruses is not substantially influenced by CTCF. However, T cells latently infected with CBS mutants were impaired in their proliferation abilities and showed a significantly reduced episomal maintenance. We detected a reduced transcription of the orf73/LANA gene in the T cells, corresponding to the reduced viral genomes; this might contribute to the loss of HVS episomes, as LANA is central in the maintenance of viral episomes in the dividing T cell populations. These data demonstrate that the episomal stability of HVS genomes in latently infected human T cells is dependent on CTCF.


Subject(s)
Herpesvirus 2, Saimiriine/genetics , Plasmids/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , T-Lymphocytes/virology , Viral Proteins/genetics , Virus Latency , Base Sequence , Binding Sites , CCCTC-Binding Factor , Cell Line , Chromatin Immunoprecipitation , Chromosomes, Artificial, Bacterial , DNA Primers , Herpesvirus 2, Saimiriine/physiology , Humans , Open Reading Frames , Polymerase Chain Reaction , Viral Proteins/metabolism
19.
J Virol ; 85(11): 5456-64, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21430050

ABSTRACT

Herpesviruses establish latency in suitable host cells after primary infection and persist in their host organisms for life. Most of the viral genes are silenced during latency, also enabling the virus to escape from an immune response. This study addresses the control of viral gene silencing by epigenetic mechanisms, using Herpesvirus saimiri (HVS) as a model system. Strain C488 of this gamma-2-herpesvirus can transform human T cells to stable growth in vitro, and it persists in the nuclei of those latently infected T cells as a nonintegrating, circular, and histone-associated episome. The whole viral genome was probed for histone acetylation at high resolution by chromatin immunoprecipitation-on-chip (ChIP-on-chip) with a custom tiling microarray. Corresponding to their inactive status in human T cells, the lytic promoters consistently revealed a heterochromatic phenotype. In contrast, the left terminal region of the genome, which encodes the stably expressed oncogenes stpC and tip as well as the herpesvirus U RNAs, was associated with euchromatic histone acetylation marks representing "open" chromatin. Although HVS latency in human T lymphocytes is considered a stable and irreversible state, incubation with the histone deacetylase inhibitor trichostatin A resulted in changes reminiscent of the induction of early lytic replication. However, infectious viral particles were not produced, as the majority of cells went into apoptosis. These data show that epigenetic mechanisms are involved in both rhadinoviral latency and transition into lytic replication.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 2, Saimiriine/pathogenicity , Histones/metabolism , Host-Pathogen Interactions , T-Lymphocytes/virology , Virus Latency , Acetylation , Cells, Cultured , Histone Deacetylase Inhibitors/metabolism , Humans
20.
J Virol ; 84(8): 4083-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20147393

ABSTRACT

Cytomegalovirus (CMV) infection in patients receiving hematopoietic stem cell transplants (HSCT) is associated with morbidity and mortality. Adoptive T cell immunotherapy has been used to treat viral reactivation but is hardly feasible in high-risk constellations of CMV-positive HSCT patients and CMV-negative stem cell donors. We endowed human effector T cells with a chimeric immunoreceptor (cIR) directed against CMV glycoprotein B. These cIR-engineered primary T cells mediated antiviral effector functions such as cytokine production and cytolysis. This first description of cIR-redirected CMV-specific T cells opens up a new perspective for HLA-independent immunotherapy of CMV infection in high-risk patients.


Subject(s)
Cytomegalovirus/immunology , Cytomegalovirus/physiology , Receptors, Immunologic/genetics , T-Lymphocytes/immunology , T-Lymphocytes/virology , Cells, Cultured , Cytomegalovirus Infections/therapy , Humans , Immunotherapy/methods , Receptors, Immunologic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...