Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 525
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 16(1): e1008231, 2020 01.
Article in English | MEDLINE | ID: mdl-31905227

ABSTRACT

Ebola (EBOV) and Marburg (MARV) are members of the Filoviridae family, which continue to emerge and cause sporadic outbreaks of hemorrhagic fever with high mortality rates. Filoviruses utilize their VP40 matrix protein to drive virion assembly and budding, in part, by recruitment of specific WW-domain-bearing host proteins via its conserved PPxY Late (L) domain motif. Here, we screened an array of 115 mammalian, bacterially expressed and purified WW-domains using a PPxY-containing peptide from MARV VP40 (mVP40) to identify novel host interactors. Using this unbiased approach, we identified Yes Associated Protein (YAP) and Transcriptional co-Activator with PDZ-binding motif (TAZ) as novel mVP40 PPxY interactors. YAP and TAZ function as downstream transcriptional effectors of the Hippo signaling pathway that regulates cell proliferation, migration and apoptosis. We demonstrate that ectopic expression of YAP or TAZ along with mVP40 leads to significant inhibition of budding of mVP40 VLPs in a WW-domain/PPxY dependent manner. Moreover, YAP colocalized with mVP40 in the cytoplasm, and inhibition of mVP40 VLP budding was more pronounced when YAP was localized predominantly in the cytoplasm rather than in the nucleus. A key regulator of YAP nuclear/cytoplasmic localization and function is angiomotin (Amot); a multi-PPxY containing protein that strongly interacts with YAP WW-domains. Interestingly, we found that expression of PPxY-containing Amot rescued mVP40 VLP egress from either YAP- or TAZ-mediated inhibition in a PPxY-dependent manner. Importantly, using a stable Amot-knockdown cell line, we found that expression of Amot was critical for efficient egress of mVP40 VLPs as well as egress and spread of authentic MARV in infected cell cultures. In sum, we identified novel negative (YAP/TAZ) and positive (Amot) regulators of MARV VP40-mediated egress, that likely function in part, via competition between host and viral PPxY motifs binding to modular host WW-domains. These findings not only impact our mechanistic understanding of virus budding and spread, but also may impact the development of new antiviral strategies.


Subject(s)
Filoviridae/physiology , Marburgvirus/physiology , Molecular Mimicry , Proto-Oncogene Proteins c-yes/metabolism , Viral Matrix Proteins/physiology , Virus Release , Angiomotins , Binding Sites , Cell Membrane/metabolism , Gene Knockout Techniques , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Microfilament Proteins/metabolism , Models, Molecular , PDZ Domains , Protein Domains , Recombinant Fusion Proteins/metabolism
2.
PLoS Pathog ; 16(12): e1009023, 2020 12.
Article in English | MEDLINE | ID: mdl-33382850

ABSTRACT

Extracellular vesicles (EV) mediate intercellular communication events and alterations in normal vesicle content contribute to function and disease initiation or progression. The ability to package a variety of cargo and transmit molecular information between cells renders EVs important mediators of cell-to-cell crosstalk. Latent membrane protein 1 (LMP1) is a chief viral oncoprotein expressed in most Epstein-Barr virus (EBV)-associated cancers and is released from cells at high levels in EVs. LMP1 containing EVs have been demonstrated to promote cell growth, migration, differentiation, and regulate immune cell function. Despite these significant changes in recipient cells induced by LMP1 modified EVs, the mechanism how this viral oncogene modulates the recipient cells towards these phenotypes is not well understood. We hypothesize that LMP1 alters EV content and following uptake of the LMP1-modified EVs by the recipient cells results in the activation of cell signaling pathways and increased gene expression which modulates the biological properties of recipient cell towards a new phenotype. Our results show that LMP1 expression alters the EV protein and microRNA content packaged into EVs. The LMP1-modified EVs also enhance recipient cell adhesion, proliferation, migration, invasion concomitant with the activation of ERK, AKT, and NF-κB signaling pathways. The LMP1 containing EVs induced transcriptome reprogramming in the recipient cells by altering gene expression of different targets including cadherins, matrix metalloproteinases 9 (MMP9), MMP2 and integrin-α5 which contribute to extracellular matrix (ECM) remodeling. Altogether, our data demonstrate the mechanism in which LMP1-modified EVs reshape the tumor microenvironment by increasing gene expression of ECM interaction proteins.


Subject(s)
Epstein-Barr Virus Infections/metabolism , Extracellular Vesicles/metabolism , Viral Matrix Proteins/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Epstein-Barr Virus Infections/physiopathology , Extracellular Vesicles/virology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/pathogenicity , Humans , MicroRNAs/metabolism , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Neoplasms/virology , Neoplasm Invasiveness/genetics , Signal Transduction , Tumor Microenvironment , Viral Matrix Proteins/physiology
3.
J Virol ; 94(4)2020 01 31.
Article in English | MEDLINE | ID: mdl-31776277

ABSTRACT

The strongest evidence of the oncogenicity of Epstein-Barr virus (EBV) in vitro is its ability to immortalize human primary B lymphocytes into lymphoblastoid cell lines (LCLs). Yet the underlying mechanisms explaining how the virus tempers the growth program of the host cells have not been fully elucidated. The mitogen-activated protein kinases (MAPKs) are implicated in many cellular processes and are constitutively activated in LCLs. We questioned the expression and regulation of the dual-specificity phosphatases (DUSPs), the main negative regulator of MAPKs, during EBV infection and immortalization. Thirteen DUSPs, including 10 typical and 3 atypical types of DUSPs, were tested. Most of them were downregulated after EBV infection. Here, a role of viral oncogene latent membrane protein 1 (LMP1) in limiting DUSP6 and DUSP8 expression was identified. Using MAPK inhibitors, we found that LMP1 activates extracellular signal-regulated kinase (ERK) or p38 to repress the expression of DUSP6 and DUSP8, with corresponding substrate specificity. Morphologically, overexpression of DUSP6 and DUSP8 attenuates the ability of EBV-immortalized LCL cells to clump together. Mechanistically, apoptosis induced by restoring DUSP6 and DUSP8 in LCLs indicated a novel mechanism for LMP1 to provide a survival signal during EBV immortalization. Collectively, this report provides the first description of the interplay between EBV genes and DUSPs and contributes considerably to the interpretation of MAPK regulation in EBV immortalization.IMPORTANCE Infections by the ubiquitous Epstein-Barr virus (EBV) are associated with a wide spectrum of lymphomas and carcinomas. It has been well documented that activation levels of MAPKs are found in cancer cells to translate various external or intrinsic stimuli into cellular responses. Physiologically, the dual-specificity phosphates (DUSPs) exhibit great ability in regulating MAPK activities with respect to their capability of dephosphorylating MAPKs. In this study, we found that DUSPs were generally downregulated after EBV infection. EBV oncogenic latent membrane protein 1 (LMP1) suppressed DUSP6 and DUSP8 expression via MAPK pathway. In this way, LMP1-mediated MAPK activation was a continuous process. Furthermore, DUSP downregulation was found to contribute greatly to prevent apoptosis of EBV-infected cells. To sum up, this report sheds light on a novel molecular mechanism explaining how EBV maintains the unlimited proliferation status of the immortalized cells and provides a new link to understand EBV-induced B cell survival.


Subject(s)
Dual-Specificity Phosphatases/genetics , Herpesvirus 4, Human/metabolism , Viral Matrix Proteins/metabolism , Apoptosis/genetics , B-Lymphocytes/virology , Cell Line, Tumor , Dual-Specificity Phosphatases/metabolism , Epstein-Barr Virus Infections/virology , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, Viral/genetics , Humans , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Primary Cell Culture , Viral Matrix Proteins/physiology , Viral Proteins/metabolism , Virus Latency/genetics , Virus Latency/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
4.
PLoS Pathog ; 15(2): e1007615, 2019 02.
Article in English | MEDLINE | ID: mdl-30779794

ABSTRACT

Cytomegalovirus (CMV) infection causes birth defects and life-threatening complications in immunosuppressed patients. Lack of vaccine and need for more effective drugs have driven widespread ongoing therapeutic development efforts against human CMV (HCMV), mostly using murine CMV (MCMV) as the model system for preclinical animal tests. The recent publication (Yu et al., 2017, DOI: 10.1126/science.aam6892) of an atomic model for HCMV capsid with associated tegument protein pp150 has infused impetus for rational design of novel vaccines and drugs, but the absence of high-resolution structural data on MCMV remains a significant knowledge gap in such development efforts. Here, by cryoEM with sub-particle reconstruction method, we have obtained the first atomic structure of MCMV capsid with associated pp150. Surprisingly, the capsid-binding patterns of pp150 differ between HCMV and MCMV despite their highly similar capsid structures. In MCMV, pp150 is absent on triplex Tc and exists as a "Λ"-shaped dimer on other triplexes, leading to only 260 groups of two pp150 subunits per capsid in contrast to 320 groups of three pp150 subunits each in a "Δ"-shaped fortifying configuration. Many more amino acids contribute to pp150-pp150 interactions in MCMV than in HCMV, making MCMV pp150 dimer inflexible thus incompatible to instigate triplex Tc-binding as observed in HCMV. While pp150 is essential in HCMV, our pp150-deletion mutant of MCMV remained viable though with attenuated infectivity and exhibiting defects in retaining viral genome. These results thus invalidate targeting pp150, but lend support to targeting capsid proteins, when using MCMV as a model for HCMV pathogenesis and therapeutic studies.


Subject(s)
Capsid Proteins/ultrastructure , Phosphoproteins/metabolism , Phosphoproteins/physiology , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/physiology , Animals , Capsid , Capsid Proteins/metabolism , Cryoelectron Microscopy/methods , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/metabolism , Genome, Viral/genetics , Humans , Mice , Muromegalovirus/metabolism , Muromegalovirus/pathogenicity , Phosphoproteins/ultrastructure , Sequence Deletion/genetics , Viral Matrix Proteins/ultrastructure , Virion , Virus Assembly
5.
Fish Shellfish Immunol ; 113: 24-34, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33757800

ABSTRACT

An increasing important area in immunology is the process cell death mechanism, enabling the immune system triggered thru extrinsic or intrinsic signals to effectively remove unwanted or virus infected cells called apoptosis. A recently isolated infectious Snakehead fish vesiculovirus (SHVV), comprising negative strand RNA and encoded viral matrix (M) proteins, is responsible for causing cytopathic effects in infected fish cells. However, the mechanism by which viral M protein mediates apoptosis has not been elucidated. Therefore, in the present experiments, it was investigated the regulatory potential of apoptosis signals during SHVV infection. By employing the model of SHVV infection in SSN-1 cells, the accelerated apoptosis pathway involves an intrinsic pathway requiring the activation of caspase-9 but not caspase-3 or -8. In the groups of infection (SHVV) or treatment (hydrogen peroxide) were induced apoptotic morphological changes and indicated the activation of the main caspases, i.e.; executioner caspase-3, initiators caspase-8 and caspase-9 using colorimetric assays. Turning to the role of viral M protein when it was overexpressed in SSN-1 cells, it was indicated that the viral M gene alone has the ability to induce apoptosis. To elucidate the mechanism of apoptosis in SSN-1 cells, the activation inhibitors of main caspases were used showing that inhibiting of caspase-3 or caspase-8 activation did not seize induction of apoptosis in virus-infected SSN-1 cells. However, the inhibiting of caspase-9 activation reduced significantly the apoptosis initiation process and sharply the expression of viral M gene, suggesting that SHVV plays a major role in the early induction of apoptosis by caspase-9. Interestingly, there were also differences in the mitochondrial membrane potential after the apoptotic induction of caspases, which confirm that caspase-9 is primarily responsible for the cleavage of caspases during apoptosis. Taken together, these findings can therefore be assumed that viral M protein induces apoptosis via the intrinsic apoptotic pathway in SHVV infecting SSN-1 cells.


Subject(s)
Apoptosis , Fish Diseases/immunology , Fishes , Rhabdoviridae Infections/veterinary , Signal Transduction/immunology , Vesiculovirus/physiology , Viral Matrix Proteins/physiology , Animals , Cell Line , Fish Diseases/virology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/virology
6.
Proc Natl Acad Sci U S A ; 115(37): E8595-E8603, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150411

ABSTRACT

The influenza A matrix 2 (M2) transmembrane protein facilitates virion release from the infected host cell. In particular, M2 plays a role in the induction of membrane curvature and/or in the scission process whereby the envelope is cut upon virion release. Here we show using coarse-grained computer simulations that various M2 assembly geometries emerge due to an entropic driving force, resulting in compact clusters or linearly extended aggregates as a direct consequence of the lateral membrane stresses. Conditions under which these protein assemblies will cause the lipid membrane to curve are explored, and we predict that a critical cluster size is required for this to happen. We go on to demonstrate that under the stress conditions taking place in the cellular membrane as it undergoes large-scale membrane remodeling, the M2 protein will, in principle, be able to both contribute to curvature induction and sense curvature to line up in manifolds where local membrane line tension is high. M2 is found to exhibit linactant behavior in liquid-disordered-liquid-ordered phase-separated lipid mixtures and to be excluded from the liquid-ordered phase, in near-quantitative agreement with experimental observations. Our findings support a role for M2 in membrane remodeling during influenza viral budding both as an inducer and a sensor of membrane curvature, and they suggest a mechanism by which localization of M2 can occur as the virion assembles and releases from the host cell, independent of how the membrane curvature is produced.


Subject(s)
Cell Membrane/virology , Viral Matrix Proteins/physiology , Virus Assembly , Virus Release , Algorithms , Animals , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Dogs , Entropy , Host-Pathogen Interactions , Madin Darby Canine Kidney Cells , Membrane Lipids/chemistry , Microscopy, Electron , Models, Biological , Molecular Dynamics Simulation
7.
PLoS Pathog ; 14(1): e1006867, 2018 01.
Article in English | MEDLINE | ID: mdl-29377960

ABSTRACT

Interferon-γ (IFN-γ) represents one of the most important innate immunity responses in a host to combat infections of many human viruses including human herpesviruses. Human N-myc interactor (Nmi) protein, which has been shown to interact with signal transducer and activator of transcription (STAT) proteins including STAT1, is important for the activation of IFN-γ induced STAT1-dependent transcription of many genes responsible for IFN-γ immune responses. However, no proteins encoded by herpesviruses have been reported to interact with Nmi and inhibit Nmi-mediated activation of IFN-γ immune responses to achieve immune evasion from IFN-γ responses. In this study, we show strong evidence that the UL23 protein of human cytomegalovirus (HCMV), a human herpesvirus, specifically interacts with Nmi. This interaction was identified through a yeast two-hybrid screen and co-immunoprecipitation in human cells. We observed that Nmi, when bound to UL23, was not associated with STAT1, suggesting that UL23 binding of Nmi disrupts the interaction of Nmi with STAT1. In cells overexpressing UL23, we observed (a) significantly reduced levels of Nmi and STAT1 in the nuclei, the sites where these proteins act to induce transcription of IFN-γ stimulated genes, and (b) decreased levels of the induction of the transcription of IFN-γ stimulated genes. UL23-deficient HCMV mutants induced higher transcription of IFN-γ stimulated genes and exhibited lower titers than parental and control revertant viruses expressing functional UL23 in IFN-γ treated cells. Thus, UL23 appears to interact directly with Nmi and inhibit nuclear translocation of Nmi and its associated protein STAT1, leading to a decrease of IFN-γ induced responses and an increase of viral resistance to IFN-γ. Our results further highlight the roles of UL23-Nmi interactions in facilitating viral immune escape from IFN-γ responses and enhancing viral resistance to IFN antiviral effects.


Subject(s)
Cytomegalovirus/physiology , Immune Evasion , Immunity, Innate/drug effects , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Viral Matrix Proteins/physiology , Cells, Cultured , Cytomegalovirus/immunology , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Immune Evasion/drug effects , Immune Evasion/genetics , Immunity, Innate/genetics , Protein Binding , Signal Transduction/genetics , Signal Transduction/immunology
8.
Hematol Oncol ; 38(5): 705-714, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32822067

ABSTRACT

Alterations of B-cell lymphoma 2 (BCL-2) family proteins contribute to the survival of B-cell malignancies. Recently, venetoclax, a BCL-2 inhibitor, was approved for B-cell chronic lymphocytic leukemia therapy and is being investigated in clinical trials for a variety of hematologic cell malignancies. Furthermore, combination therapy with other molecularly targeted drugs was reported to be more effective than monotherapy. However, combining venetoclax with immunotherapy based on T-cells has not been tested. Because both venetoclax and granzyme B activate the mitochondrial apoptosis pathway by targeting different BCL-2 family molecules, it is possible that combinations of venetoclax with immunotherapy will be effective treatments. We examined the effect of combining venetoclax with immunotherapy using an in vitro model system involving cytomegalovirus (CMV) pp65 antigen-specific cytotoxic T-cells (CMV-CTLs) as the effector cells and CMVpp65 antigen-expressing B-cell lines as the target cells. Cytotoxicity of CMV-CTLs to the target B-cell lines was enhanced by venetoclax with combination index values of 0.47-0.83. This suggests that venetoclax synergizes with T-cell-based immunotherapy to affect B-cell malignancies. Interestingly, venetoclax synergized not only with antigen-specific cytotoxicity but also with nonspecific cytotoxicity. Importantly, CMV-CTLs could be expanded in the presence of venetoclax at the maximum concentration (5 µM) that induced apoptosis in resting CMV-CTLs. B-cell lymphoma-extra large (BCL-xL) expression in CMV-CTLs increased transiently after activation by CMVpp65-transfected B-cell lines, indicating that the expression of BCL-xL was important for the effectiveness of combination treatment with venetoclax. These findings suggest that T-cell-based immunotherapy combined with venetoclax is effective against B-cell malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Gene Expression , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunohistochemistry , Immunophenotyping , Immunotherapy , Mice , T-Cell Antigen Receptor Specificity , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Viral Matrix Proteins/physiology
9.
Proc Natl Acad Sci U S A ; 114(5): 1147-1152, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096411

ABSTRACT

Hepatitis E virus (HEV) is the leading cause of enterically transmitted viral hepatitis globally. Of HEV's three ORFs, the function of ORF3 has remained elusive. Here, we demonstrate that via homophilic interactions ORF3 forms multimeric complexes associated with intracellular endoplasmic reticulum (ER)-derived membranes. HEV ORF3 shares several structural features with class I viroporins, and the function of HEV ORF3 can be maintained by replacing it with the well-characterized viroporin influenza A virus (IAV) matrix-2 protein. ORF3's ion channel function is further evidenced by its ability to mediate ionic currents when expressed in Xenopus laevis oocytes. Furthermore, we identified several positions in ORF3 critical for its formation of multimeric complexes, ion channel activity, and, ultimately, release of infectious particles. Collectively, our data demonstrate a previously undescribed function of HEV ORF3 as a viroporin, which may serve as an attractive target in developing direct-acting antivirals.


Subject(s)
Hepatitis E virus/physiology , Ion Channels/physiology , Viral Proteins/physiology , Virus Release/physiology , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Animals , Endoplasmic Reticulum/metabolism , Gene Deletion , HEK293 Cells , Hep G2 Cells , Humans , Ion Channels/chemistry , Ion Transport , Oocytes , Patch-Clamp Techniques , Protein Domains , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Viral Matrix Proteins/physiology , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication , Xenopus laevis
10.
J Gen Virol ; 99(6): 805-817, 2018 06.
Article in English | MEDLINE | ID: mdl-29580369

ABSTRACT

The human gamma herpes virus Epstein-Barr virus (EBV) exploits multiple routes to evade the cellular immune response. During the EBV lytic replication cycle, viral proteins are expressed that provide excellent targets for recognition by cytotoxic T cells. This is countered by the viral BNLF2a gene. In B cells during latency, where BNLF2a is not expressed, we show that its regulatory region is embedded in repressive chromatin. The expression of BNLF2a mirrors the expression of a viral lytic cycle transcriptional regulator, Zta (BZLF1, EB1, ZEBRA), in B cells and we propose that Zta plays a role in up-regulating BNLF2a. In cells undergoing EBV lytic replication, we identified two distinct regions of interaction of Zta with the chromatin-associated BNLF2a promoter. We identify five potential Zta-response elements (ZREs) in the promoter that are highly conserved between virus isolates. Zta binds to these elements in vitro and activates the expression of the BNLF2a promoter in both epithelial and B cells. We also found redundancy amongst the ZREs. The EBV genome undergoes a biphasic DNA methylation cycle during its infection cycle. One of the ZREs contains an integral CpG motif. We show that this can be DNA methylated during EBV latency and that both Zta binding and promoter activation are enhanced by its methylation. In summary, we find that the BNLF2a promoter is directly targeted by Zta and that DNA methylation within the proximal ZRE aids activation. The implications for regulation of this key viral gene during the reactivation of EBV from latency are discussed.


Subject(s)
Herpesvirus 4, Human/immunology , Immune Evasion , Trans-Activators/physiology , Viral Matrix Proteins/physiology , Virus Latency/genetics , B-Lymphocytes/virology , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Viral , Genome, Viral , HEK293 Cells , HeLa Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Promoter Regions, Genetic , Trans-Activators/genetics , Transcriptional Activation , Viral Matrix Proteins/genetics , Virus Replication/genetics
11.
J Virol ; 91(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28077657

ABSTRACT

Epstein-Barr virus (EBV) infection is associated with B cell lymphomas in humans. The ability of EBV to convert human B cells into long-lived lymphoblastoid cell lines (LCLs) in vitro requires the collaborative effects of EBNA2 (which hijacks Notch signaling), latent membrane protein 1 (LMP1) (which mimics CD40 signaling), and EBV-encoded nuclear antigen 3A (EBNA3A) and EBNA3C (which inhibit oncogene-induced senescence and apoptosis). However, we recently showed that an LMP1-deleted EBV mutant induces B cell lymphomas in a newly developed cord blood-humanized mouse model that allows EBV-infected B cells to interact with CD4 T cells (the major source of CD40 ligand). Here we examined whether the EBV LMP2A protein, which mimics constitutively active B cell receptor signaling, is required for EBV-induced lymphomas in this model. We find that the deletion of LMP2A delays the onset of EBV-induced lymphomas but does not affect the tumor phenotype or the number of tumors. The simultaneous deletion of both LMP1 and LMP2A results in fewer tumors and a further delay in tumor onset. Nevertheless, the LMP1/LMP2A double mutant induces lymphomas in approximately half of the infected animals. These results indicate that neither LMP1 nor LMP2A is absolutely essential for the ability of EBV to induce B cell lymphomas in the cord blood-humanized mouse model, although the simultaneous loss of both LMP1 and LMP2A decreases the proportion of animals developing tumors and increases the time to tumor onset. Thus, the expression of either LMP1 or LMP2A may be sufficient to promote early-onset EBV-induced tumors in this model.IMPORTANCE EBV causes human lymphomas, but few models are available for dissecting how EBV causes lymphomas in vivo in the context of a host immune response. We recently used a newly developed cord blood-humanized mouse model to show that EBV can cooperate with human CD4 T cells to cause B cell lymphomas even when a major viral transforming protein, LMP1, is deleted. Here we examined whether the EBV protein LMP2A, which mimics B cell receptor signaling, is required for EBV-induced lymphomas in this model. We find that the deletion of LMP2A alone has little effect on the ability of EBV to cause lymphomas but delays tumor onset. The deletion of both LMP1 and LMP2A results in a smaller number of lymphomas in infected animals, with an even more delayed time to tumor onset. These results suggest that LMP1 and LMP2A collaborate to promote early-onset lymphomas in this model, but neither protein is absolutely essential.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/physiology , Lymphoma, Large B-Cell, Diffuse/virology , Viral Matrix Proteins/physiology , Animals , Cell Transformation, Neoplastic , Cells, Cultured , Epstein-Barr Virus Infections/immunology , Gene Knockout Techniques , Humans , Lymphocytes, Tumor-Infiltrating/physiology , Lymphoma, Large B-Cell, Diffuse/immunology , Mice, Inbred NOD , Mice, SCID
12.
Biochemistry ; 56(44): 5955-5963, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29034683

ABSTRACT

Influenza A M2 is a membrane-associated protein with a C-terminal amphipathic helix that plays a cholesterol-dependent role in viral budding. An M2 mutant with alanine substitutions in the C-terminal amphipathic helix is deficient in viral scission. With the goal of providing atomic-level understanding of how the wild-type protein functions, we used a multipronged site-directed spin labeling electron paramagnetic resonance spectroscopy (SDSL-EPR) approach to characterize the conformational properties of the alanine mutant. We spin-labeled sites in the transmembrane (TM) domain and the C-terminal amphipathic helix (AH) of wild-type (WT) and mutant M2, and collected information on line shapes, relaxation rates, membrane topology, and distances within the homotetramer in membranes with and without cholesterol. Our results identify marked differences in the conformation and dynamics between the WT and the alanine mutant. Compared to WT, the dominant population of the mutant AH is more dynamic, shallower in the membrane, and has altered quaternary arrangement of the C-terminal domain. While the AH becomes more dynamic, the dominant population of the TM domain of the mutant is immobilized. The presence of cholesterol changes the conformation and dynamics of the WT protein, while the alanine mutant is insensitive to cholesterol. These findings provide new insight into how M2 may facilitate budding. We propose the AH-membrane interaction modulates the arrangement of the TM helices, effectively stabilizing a conformational state that enables M2 to facilitate viral budding. Antagonizing the properties of the AH that enable interdomain coupling within M2 may therefore present a novel strategy for anti-influenza drug design.


Subject(s)
Mutation , Protein Domains/physiology , Viral Matrix Proteins/genetics , Virus Release/genetics , Cell Membrane/metabolism , Cholesterol/pharmacology , Electron Spin Resonance Spectroscopy , Humans , Influenza A virus , Protein Conformation , Protein Structural Elements , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/physiology
13.
Int J Cancer ; 140(6): 1233-1245, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27750386

ABSTRACT

The pathogenesis of classical Hodgkin lymphoma (cHL) is still enigmatic, largely because its tumor cells, the so-called Hodgkin and Reed-Stenberg (HRS) cells, invariably reside in a prominent reactive microenvironment, are rare and therefore difficult to analyze. On the other hand, the broadly investigated cHL-derived cell lines are not unequivocally considered as suitable and representative models for this puzzling disease. Based on current knowledge, it appears that the cross talk between the tumor cells and the reactive infiltrate of the microenvironment is complex and that multiple mechanisms occur, making cHL a very heterogeneous disease. In 20-40% of cHL cases, HRS cells carry a monoclonal infection by Epstein Barr virus (EBV), which is considered a tumor-initiating factor. In these cases, EBV shows a latency type II infection pattern with the expression of latent membrane protein-1 (LMP-1), a viral oncoprotein that mimics CD40 activation. This scenario is particularly intriguing for the pathogenesis of cHL arising in HIV-infected patients, which, for still obscure reasons, is invariably EBV-associated with LMP-1 expression in HRS cells. Recent evidences are consistent with the occurrence of different pathogenic pathways variably triggered by virus infections (EBV and HIV), genetic alterations, and interactions with critical microenvironmental components. This review focuses on the different microenvironmental niches that characterize cHL of the general population as well as cases of HIV-infected patients. A more comprehensive understanding of the complex interplay existing between HRS and tumor microenvironment is pivotal for the development of more effective treatments, particularly for relapsed or refractory diseases.


Subject(s)
Epstein-Barr Virus Infections/physiopathology , Hodgkin Disease/virology , Lymphoma, AIDS-Related/virology , Tumor Microenvironment , Viral Matrix Proteins/physiology , Antigens, CD/biosynthesis , Antigens, CD/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/virology , Comparative Genomic Hybridization , Cytokines/physiology , Discoidin Domain Receptor 1/physiology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Fibroblasts/physiology , Gene Expression Regulation, Neoplastic , Hodgkin Disease/classification , Hodgkin Disease/etiology , Hodgkin Disease/immunology , Hodgkin Disease/pathology , Humans , Immunocompetence , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, AIDS-Related/etiology , Lymphoma, AIDS-Related/immunology , Lymphoma, AIDS-Related/pathology , Macrophages/physiology , Models, Biological , Neoplasm Proteins/physiology , Reed-Sternberg Cells/virology , Signal Transduction , Virus Latency
14.
PLoS Pathog ; 11(10): e1005220, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26513362

ABSTRACT

Hemorrhagic fever viruses, including the filoviruses (Ebola and Marburg) and arenaviruses (Lassa and Junín viruses), are serious human pathogens for which there are currently no FDA approved therapeutics or vaccines. Importantly, transmission of these viruses, and specifically late steps of budding, critically depend upon host cell machinery. Consequently, strategies which target these mechanisms represent potential targets for broad spectrum host oriented therapeutics. An important cellular signal implicated previously in EBOV budding is calcium. Indeed, host cell calcium signals are increasingly being recognized to play a role in steps of entry, replication, and transmission for a range of viruses, but if and how filoviruses and arenaviruses mobilize calcium and the precise stage of virus transmission regulated by calcium have not been defined. Here we demonstrate that expression of matrix proteins from both filoviruses and arenaviruses triggers an increase in host cytoplasmic Ca2+ concentration by a mechanism that requires host Orai1 channels. Furthermore, we demonstrate that Orai1 regulates both VLP and infectious filovirus and arenavirus production and spread. Notably, suppression of the protein that triggers Orai activation (Stromal Interaction Molecule 1, STIM1) and genetic inactivation or pharmacological blockade of Orai1 channels inhibits VLP and infectious virus egress. These findings are highly significant as they expand our understanding of host mechanisms that may broadly control enveloped RNA virus budding, and they establish Orai and STIM1 as novel targets for broad-spectrum host-oriented therapeutics to combat these emerging BSL-4 pathogens and potentially other enveloped RNA viruses that bud via similar mechanisms.


Subject(s)
Arenavirus/physiology , Filoviridae/physiology , Virus Release , Animals , Calcium/metabolism , Calcium Channels/physiology , HEK293 Cells , HeLa Cells , Humans , ORAI1 Protein , Vero Cells , Viral Matrix Proteins/physiology , Virion/physiology
15.
Blood ; 125(13): 2101-10, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25568351

ABSTRACT

Hodgkin lymphoma (HL) and Burkitt lymphoma are both germinal center-derived B-cell lymphomas. To assess the consequences of permanent latent membrane protein 1 (LMP1) expression as observed in tumor cells of Epstein-Barr virus (EBV) -associated HL, we analyzed 3-dimensional (3D) telomere dynamics and measured the expression of shelterin proteins at the transcriptional and translational level and their topographic distribution in the EBV-negative Burkitt cell line BJAB stably transfected with an inducible LMP1 system. Stable LMP1 expression led to a highly significant increase of multinucleated cells, nuclear volume, and 3D telomeric aggregates when compared with the LMP1-suppressed BJAB controls. Most importantly, LMP1 induced a significant downregulation of the shelterin components TRF1, TRF2, and POT1 at the transcriptional and translational level, and this downregulation was reversed after resuppression of LMP1. In addition, as revealed by spectral karyotyping, LMP1 induced "outré" giant cells and hypoploid "ghost" cells. This LMP1-induced multinucleation was blocked upon LMP1-independent TRF2 expression. These results show that LMP1-dependent deregulation of telomere stability and nuclear organization via shelterin downregulation, in particular TRF2, favors chromosomal rearrangements. We speculate that telomeric aggregates and ongoing breakage-bridge-fusion cycles lead to disturbed cytokinesis and finally to multinuclearity, as observed in EBV-associated HL.


Subject(s)
Cell Nucleus , Giant Cells/metabolism , Telomere-Binding Proteins/genetics , Telomere/metabolism , Viral Matrix Proteins/physiology , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Transformation, Viral/genetics , Down-Regulation , Giant Cells/pathology , Hodgkin Disease/genetics , Hodgkin Disease/pathology , Hodgkin Disease/virology , Humans , Protein Aggregates/genetics , Shelterin Complex , Telomere-Binding Proteins/metabolism
16.
J Virol ; 89(10): 5200-3, 2015 May.
Article in English | MEDLINE | ID: mdl-25740980

ABSTRACT

Exosomes are small vesicles secreted from cells that participate in intercellular communication events. Accumulating evidence demonstrates that host exosome pathways are hijacked by viruses and that virally modified exosomes contribute to virus spread and immune evasion. In the case of tumor viruses, recent findings suggest that alterations in normal exosome biology may promote the development and progression of cancer. These studies will be discussed in the context of our current knowledge of Epstein-Barr virus (EBV)-modified exosomes.


Subject(s)
Cell Communication/physiology , Exosomes/physiology , Exosomes/virology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/physiopathology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/pathogenicity , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , Humans , Immune Evasion , Models, Biological , Viral Matrix Proteins/physiology
17.
PLoS Pathog ; 10(5): e1004125, 2014 May.
Article in English | MEDLINE | ID: mdl-24809689

ABSTRACT

The DOK1 tumor suppressor gene encodes an adapter protein that acts as a negative regulator of several signaling pathways. We have previously reported that DOK1 expression is up-regulated upon cellular stress, via the transcription factor E2F1, and down-regulated in a variety of human malignancies due to aberrant hypermethylation of its promoter. Here we show that Epstein Barr virus (EBV) infection of primary human B-cells leads to the down-regulation of DOK1 gene expression via the viral oncoprotein LMP1. LMP1 alone induces recruitment to the DOK1 promoter of at least two independent inhibitory complexes, one containing E2F1/pRB/DNMT1 and another containing at least EZH2. These events result in tri-methylation of histone H3 at lysine 27 (H3K27me3) of the DOK1 promoter and gene expression silencing. We also present evidence that the presence of additional EBV proteins leads to further repression of DOK1 expression with an additional mechanism. Indeed, EBV infection of B-cells induces DNA methylation at the DOK1 promoter region including the E2F1 responsive elements that, in turn, lose the ability to interact with E2F complexes. Treatment of EBV-infected B-cell-lines with the methyl-transferase inhibitor 5-aza-2'-deoxycytidine rescues DOK1 expression. In summary, our data show the deregulation of DOK1 gene expression by EBV and provide novel insights into the regulation of the DOK1 tumor suppressor in viral-related carcinogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/physiology , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Transformation, Viral/genetics , Cells, Cultured , DNA Methylation , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Epstein-Barr Virus Infections/immunology , Gene Expression Regulation , Gene Silencing , Genes, Tumor Suppressor , Humans , Phosphoproteins/metabolism , Primary Cell Culture , RNA-Binding Proteins/metabolism , Viral Matrix Proteins/physiology
18.
FASEB J ; 29(7): 2712-25, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25795456

ABSTRACT

We sought to determine the mechanisms by which influenza infection of human epithelial cells decreases cystic fibrosis transmembrane conductance regulator (CFTR) expression and function. We infected human bronchial epithelial (NHBE) cells and murine nasal epithelial (MNE) cells with various strains of influenza A virus. Influenza infection significantly reduced CFTR short circuit currents (Isc) and protein levels at 8 hours postinfection. We then infected CFTR expressing human embryonic kidney (HEK)-293 cells (HEK-293 CFTRwt) with influenza virus encoding a green fluorescent protein (GFP) tag and performed whole-cell and cell-attached patch clamp recordings. Forskolin-stimulated, GlyH-101-sensitive CFTR conductances, and CFTR open probabilities were reduced by 80% in GFP-positive cells; Western blots also showed significant reduction in total and plasma membrane CFTR levels. Knockdown of the influenza matrix protein 2 (M2) with siRNA, or inhibition of its activity by amantadine, prevented the decrease in CFTR expression and function. Lysosome inhibition (bafilomycin-A1), but not proteasome inhibition (lactacystin), prevented the reduction in CFTR levels. Western blots of immunoprecipitated CFTR from influenza-infected cells, treated with BafA1, and probed with antibodies against lysine 63-linked (K-63) or lysine 48-linked (K-48) polyubiquitin chains supported lysosomal targeting. These results highlight CFTR damage, leading to early degradation as an important contributing factor to influenza infection-associated ion transport defects.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Influenza A virus/physiology , Influenza A virus/pathogenicity , Viral Matrix Proteins/physiology , Animals , Apoptosis , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Gene Expression , Gene Knockdown Techniques , HEK293 Cells , Humans , Influenza A virus/genetics , Influenza, Human/metabolism , Influenza, Human/pathology , Influenza, Human/virology , Ion Transport , Lysosomes/metabolism , Mice , Necrosis , Patch-Clamp Techniques , Proteolysis , Transfection , Viral Matrix Proteins/antagonists & inhibitors , Viral Matrix Proteins/genetics
19.
J Pathol ; 235(3): 456-65, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25294670

ABSTRACT

Undifferentiated nasopharyngeal carcinoma (NPC) is a highly metastatic disease that is consistently associated with Epstein-Barr virus (EBV) infection. In this study, we have investigated the contribution of lysophosphatidic acid (LPA) signalling to the pathogenesis of NPC. Here we demonstrate two distinct functional roles for LPA in NPC. First, we show that LPA enhances the migration of NPC cells and second, that it can inhibit the activity of EBV-specific cytotoxic T cells. Focusing on the first of these phenotypes, we show that one of the LPA receptors, LPA receptor 5 (LPAR5), is down-regulated in primary NPC tissues and that this down-regulation promotes the LPA-induced migration of NPC cell lines. Furthermore, we found that EBV infection or ectopic expression of the EBV-encoded LMP2A was sufficient to down-regulate LPAR5 in NPC cell lines. Our data point to a central role for EBV in mediating the oncogenic effects of LPA in NPC and identify LPA signalling as a potential therapeutic target in this disease.


Subject(s)
Down-Regulation/physiology , Epstein-Barr Virus Infections/physiopathology , Gene Expression Regulation, Neoplastic/physiology , Lysophospholipids/physiology , Nasopharyngeal Neoplasms/physiopathology , Receptors, Lysophosphatidic Acid/physiology , Signal Transduction/physiology , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Carcinoma , Cell Line, Tumor , Cell Movement/physiology , Herpesvirus 4, Human/physiology , Humans , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Phosphoric Diester Hydrolases/physiology , Receptors, Lysophosphatidic Acid/genetics , T-Lymphocytes, Cytotoxic/pathology , Viral Matrix Proteins/physiology
20.
Biochim Biophys Acta ; 1838(4): 1088-95, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23906728

ABSTRACT

In addition to a set of canonical genes, coronaviruses encode additional accessory proteins. A locus located between the spike and envelope genes is conserved in all coronaviruses and contains a complete or truncated open reading frame (ORF). Previously, we demonstrated that this locus, which contains the gene for accessory protein 3a from severe acute respiratory syndrome coronavirus (SARS-CoV), encodes a protein that forms ion channels and regulates virus release. In the current study, we explored whether the ORF4a protein of HCoV-229E has similar functions. Our findings revealed that the ORF4a proteins were expressed in infected cells and localized at the endoplasmic reticulum/Golgi intermediate compartment (ERGIC). The ORF4a proteins formed homo-oligomers through disulfide bridges and possessed ion channel activity in both Xenopus oocytes and yeast. Based on the measurement of conductance to different monovalent cations, the ORF4a was suggested to form a non-selective channel for monovalent cations, although Li(+) partially reduced the inward current. Furthermore, viral production decreased when the ORF4a protein expression was suppressed by siRNA in infected cells. Collectively, this evidence indicates that the HCoV-229E ORF4a protein is functionally analogous to the SARS-CoV 3a protein, which also acts as a viroporin that regulates virus production. This article is part of a Special Issue entitled: Viral Membrane Proteins - Channels for Cellular Networking.


Subject(s)
Coronavirus 229E, Human/physiology , Viral Matrix Proteins/physiology , Amino Acid Sequence , Coronavirus 229E, Human/chemistry , HEK293 Cells , Humans , Molecular Sequence Data , Protein Multimerization , Viral Matrix Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL