Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Gen Virol ; 104(10)2023 10.
Article in English | MEDLINE | ID: mdl-37831061

ABSTRACT

Peste des petits ruminants virus (PPRV) is known to induce transient immunosuppression in infected small ruminants by modulating several cellular pathways involved in the antiviral immune response. Our study shows that the PPRV-coded non-structural proteins C and V can interact with the cellular NF-κB p65 subunit. The PPRV-C protein interacts with the transactivation domain (TAD) while PPRV-V interacts with the Rel homology domain (RHD) of the NF-κB p65 subunit. Both viral proteins can suppress the NF-κB transcriptional activity and NF-κB-mediated transcription of cellular genes. PPRV-V protein expression can significantly inhibit the nuclear translocation of NF-κB p65 upon TNF-α stimulation, whereas PPRV-C does not affect it. The NF-κB-mediated pro-inflammatory cytokine gene expression is significantly downregulated in cells expressing PPRV-C or PPRV-V protein. Our study provides evidence suggesting a role of PPRV non-structural proteins V and C in the modulation of NF-κB signalling through interaction with the NF-κB p65 subunit.


Subject(s)
Peste-des-Petits-Ruminants , Peste-des-petits-ruminants virus , Animals , Humans , Peste-des-petits-ruminants virus/genetics , Peste-des-Petits-Ruminants/metabolism , Cytokines/genetics , Cytokines/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Ruminants , Gene Expression , Goats/genetics
2.
Viruses ; 15(9)2023 08 24.
Article in English | MEDLINE | ID: mdl-37766213

ABSTRACT

Autophagy is an essential and highly conserved catabolic process in cells, which is important in the battle against intracellular pathogens. Viruses have evolved several ways to alter the host defense mechanisms. PPRV infection is known to modulate the components of a host cell's defense system, resulting in enhanced autophagy. In this study, we demonstrate that the N protein of PPRV interacts with the core components of the class III phosphatidylinositol-3-kinase (PI3K) complex-I and results in the induction of autophagy in the host cell over, thereby expressing this viral protein. Our data shows the interaction between PPRV-N protein and different core components of the autophagy pathway, i.e., VPS34, VPS15, BECN1 and ATG14L. The PPRV-N protein can specifically interact with VPS34 of the PI3K complex-I and colocalize with the proteins of PI3K complex-I in the same sub-cellular compartment, that is, in the cytoplasm. These interactions do not affect the intracellular localization of the different host proteins. The autophagy-related genes were transcriptionally modulated in PPRV-N-expressing cells. The expression of LC3B and SQSTM1/p62 was also modulated in PPRV-N-expressing cells, indicating the induction of autophagic activity. The formation of typical autophagosomes with double membranes was visualized by transmission electron microscopy in PPRV-N-expressing cells. Taken together, our findings provide evidence for the critical role of the N protein of the PPR virus in the induction of autophagy, which is likely to be mediated by PI3K complex-I of the host.


Subject(s)
Nucleocapsid Proteins , Peste-des-petits-ruminants virus , Phosphatidylinositol 3-Kinases , Autophagy , Phosphatidylinositols
3.
Virusdisease ; 33(1): 65-75, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35493756

ABSTRACT

The vaccination of the susceptible animal population against FMDV remains the most important measure to control the virus and prevent economic loss. Occurrence of infection in vaccinated animals is well-known in some diseases and is termed as breakthrough infection. The reasons include host genetic factors which can play an important role resulting in differences in susceptibility of animals to virus infection even with vaccine induced protective immune response. The Major Histocompatibility Complex (MHC) of bovines i.e. Bovine Leukocyte Antigen (BoLA) is important for antigen presentation. The BoLA DRB3 allele, which codes for the beta chain in Class II antigen, has been extensively studied and numerous reports have previously shown association of polymorphism in the gene with resistance/ susceptibility to several bacterial and viral diseases. In addition, previous studies have shown relationship between BoLA Class I and resistance or susceptibility to different diseases in cattle. The present study investigated the polymorphism in BoLA DRB3 and BoLA gene sequences of host and their relation with breakthrough FMDV infection in vaccinated animals. The study has identified three polymorphic sites each in both the genes which correlate with evidence of recent infection indicating their role in determining susceptibility of vaccinated animals to FMDV infection. Our limited study was performed on a relatively small samples size collected from one region of country. Further validation would require more detailed investigations on larger sample size. Supplementary Information: The online version contains supplementary material available at 10.1007/s13337-021-00754-8.

4.
J Gen Virol ; 102(10)2021 10.
Article in English | MEDLINE | ID: mdl-34704922

ABSTRACT

Members of the family Herpesviridae have enveloped, spherical virions with characteristic complex structures consisting of symmetrical and non-symmetrical components. The linear, double-stranded DNA genomes of 125-241 kbp contain 70-170 genes, of which 43 have been inherited from an ancestral herpesvirus. In general, herpesviruses have coevolved with and are highly adapted to their hosts, which comprise many mammalian, avian and reptilian species. Following primary infection, they are able to establish lifelong latent infection, during which there is limited viral gene expression. Severe disease is usually observed only in the foetus, the very young, the immunocompromised or following infection of an alternative host. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Herpesviridae, which is available at ictv.global/report/herpesviridae.


Subject(s)
Genome, Viral , Herpesviridae , Animals , Evolution, Molecular , Herpesviridae/classification , Herpesviridae/genetics , Herpesviridae/physiology , Herpesviridae/ultrastructure , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology , Host Adaptation , Virion/chemistry , Virion/ultrastructure , Virus Latency , Virus Replication
6.
Front Immunol ; 12: 630307, 2021.
Article in English | MEDLINE | ID: mdl-33912160

ABSTRACT

The study was aimed at developing an accessible laboratory animal model to elucidate protective and pathological roles of immune mediators during Peste des petits ruminants virus (PPRV) infection. It is because of the critical roles of type I IFNs in anti-viral defense, we assessed the susceptibility of IFN receptor knock out (IFNR KO) mice to PPRV infection. IFNR KO mice were exceedingly susceptible to the infection but WT animals efficiently controlled PPRV. Accordingly, the PPRV infected IFNR KO mice gradually reduced their body weights and succumbed to the infection within 10 days irrespective of the dose and route of infection. The lower infecting doses predominantly induced immunopathological lesions. The viral antigens as well as the replicating PPRV were abundantly present in most of the critical organs such as brain, lungs, heart and kidneys of IFNR KO mice infected with high dose of the virus. Neutrophils and macrophages transported the replicating virus to central nervous system (CNS) and contributed to pathology while the elevated NK and T cell responses directly correlated with the resolution of PPRV infection in WT animals. Using an array of fluorescently labeled H-2Kb tetramers, we discovered four immunogenic epitopes of PPRV. The PPRV-peptides interacted well with H-2Kb in acellular and cellular assay as well as expanded the virus-specific CD8+ T cells in immunized or infected mice. Adoptively transferred CD8+ T cells helped control PPRV in infected mice. Our study therefore established and employed a mouse model for investigating the pathogenesis of PPRV. The model could be useful for elucidating the contribution of immune cells in disease progression as well as to test anti-viral agents.


Subject(s)
Peste-des-Petits-Ruminants/immunology , Animals , Brain/virology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , H-2 Antigens/immunology , Immunity, Innate , Immunization , Lung/virology , Mice , Mice, Inbred C57BL , Peste-des-Petits-Ruminants/mortality , Peste-des-Petits-Ruminants/pathology , Peste-des-petits-ruminants virus/immunology , Receptors, Interferon/physiology , Viral Vaccines/immunology
7.
Virusdisease ; 31(2): 75-79, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32368569

ABSTRACT

CoV-2 which is the causative agent of COVID-19 belongs to genus betacoronaviruses. The sequence analysis of S protein of CoV-2 has shown that it has acquired a 'polybasic cleavage site' consisting of 12 aminoacids that has been predicted to enable its cleavage by other cellular proteases possibly increasing its transmissibility. The aminoacids present in receptor binding domain of S protein of SARS CoV which are critical for its binding to cellular receptor are different in CoV-2. The presence of heptanucleotide slippery sequence in ORF1 resulting in ribosomal frameshifting, and presence of transcription regulatory sequences between ORFs resulting in discontinuous transcription, are peculiar features of Coronavirus infection cycle. The exonuclease activity of nsp14 provides possible proofreading ability to RNA polymerase makes coronaviruses different from other RNA viruses allowing coronaviruses to maintain their relatively large genome size. This mini-review summarizes the peculiar features of Coronaviruses genome and the critical events during the infection cycle with focus on CoV-2.

8.
PLoS One ; 14(4): e0215394, 2019.
Article in English | MEDLINE | ID: mdl-30998737

ABSTRACT

The oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV) has two distinct life cycles with lifelong latent/non-productive and a sporadic lytic-reactivating/productive phases in the infected immune compromised human hosts. The virus reactivates from latency in response to various chemical or environmental stimuli, which triggers the lytic cascade and leads to the expression of immediate early gene, i.e. Replication and Transcription Activator (K-RTA). K-RTA, the latent-to-lytic switch protein, activates the expression of early (E) and late (L) lytic genes by transactivating multiple viral promoters. Expression of K-RTA is shown to be sufficient and essential to switch the latent virus to enter into the lytic phase of infection. Similarly, the virus-encoded bZIP family of protein, K8 also plays an important role in viral lytic DNA replication. Although, both K-RTA and K8 are found to be the ori-Lyt binding proteins and are required for lytic DNA replication, the detailed DNA-binding profile of these proteins in the KSHV and host genomes remains uncharacterized. In this study, using chromatin immunoprecipitation combined with high-throughput sequencing (ChIP-seq) assay, we performed a comprehensive analysis of K-RTA and K8 binding sites in the KSHV and human genomes in order to identify specific DNA binding sequences/motifs. We identified two novel K-RTA binding motifs, (i.e. AGAGAGAGGA/motif RB and AGAAAAATTC/motif RV) and one K8 binding motif (i.e. AAAATGAAAA/motif KB), respectively. The binding of K-RTA/K8 proteins with these motifs and resulting transcriptional modulation of downstream genes was further confirmed by DNA electrophoretic gel mobility shift assay (EMSA), reporter promoter assay, Chromatin Immunoprecipitation (ChIP) assay and mRNA quantitation assay. Our data conclusively shows that K-RTA/K8 proteins specifically bind to these motifs on the host/viral genomes to modulate transcription of host/viral genes during KSHV lytic reactivation.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Chromatin/metabolism , Gene Expression Regulation, Viral/physiology , Herpesvirus 8, Human/physiology , Promoter Regions, Genetic , Repressor Proteins/metabolism , Viral Proteins/metabolism , Virus Activation/physiology , Basic-Leucine Zipper Transcription Factors/genetics , Chromatin/genetics , Chromatin/virology , HEK293 Cells , Humans , Repressor Proteins/genetics , Viral Proteins/genetics
9.
Arch Virol ; 164(5): 1271-1285, 2019 May.
Article in English | MEDLINE | ID: mdl-30859475

ABSTRACT

Hepatitis C virus (HCV) is the major etiological agent of hepatocellular carcinoma (HCC), which is the fourth most common cause of cancer-related deaths worldwide and second in terms of deaths of males (Bray et al. in CA Cancer J Clin 68(6):394-424, 2018). HCV-induced HCC is a multi-step process that involves alteration of several host regulatory pathways. One of the key features of HCV-associated hepatocellular carcinoma is the metastasis of cancer cells to different organs. Human Nm23-H1 is one of the best-studied metastasis suppressor proteins, and it has been shown to be modulated in many human cancers. Our study shows that the core protein of HCV genotype 2a can co-localize and interact directly with Nm23-H1 within cancer cells, resulting in modulation of the anti-metastasis properties of Nm23-H1. The HCV core protein promotes SUMOylation and degradation of the Nm23-H1 protein, as well as transcriptional downregulation. This study provides evidence that the HCV core protein is a pro-metastatic protein that can interact directly with and modulate the functions of cellular metastasis suppressor Nm23-H1.


Subject(s)
Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Hepacivirus/pathogenicity , Liver Neoplasms/pathology , NM23 Nucleoside Diphosphate Kinases/metabolism , Viral Core Proteins/metabolism , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Cell Movement/physiology , Down-Regulation , HEK293 Cells , Hepatitis C/pathology , Humans , Liver Neoplasms/virology , Neoplasm Invasiveness/pathology , Protein Processing, Post-Translational/physiology
10.
Arch Virol ; 164(1): 91-104, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30284629

ABSTRACT

Kaposi's sarcoma-associated herpes virus (KSHV) is a gammaherpesvirus associated with Kaposi's sarcoma and various lymphoproliferative diseases. Epithelial-to-mesenchymal transition (EMT) is an important step in the metastasis of cancer cells. Previous studies have shown an important role for EMT markers in B-cell malignancies. In the present study, we investigated the role of the KSHV latent protein LANA in the progression of EMT. Our data suggest that expression of LANA results in an increase in the migration and invasion potential of cancer cells, which is concurrent with modulation of transcriptional regulation and protein expression of several cellular genes associated with EMT. LANA expression results in upregulation of the cellular intermediate filament protein vimentin and transcription factor TCF8/ZEB1 and downregulation of tight junction protein ZO1 and adhesion protein E-cadherin. LANA co-localizes with TCF8/ZEB1, a major contributor in EMT, further suggesting an important role for LANA in epithelial-to-mesenchymal transition of KSHV-infected cancer cells.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Herpesvirus 8, Human , Viral Proteins/metabolism , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Humans , Neoplasm Invasiveness , Real-Time Polymerase Chain Reaction , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
11.
Curr Drug Metab ; 19(3): 224-235, 2018.
Article in English | MEDLINE | ID: mdl-29380693

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common causes of cancer associated deaths. Prognosis is relatively poor in cases where Hepatitis C Virus (HCV) is associated as causative agent, mainly due to increased risk of metastasis. Metastasis is the major cause of all cancer related deaths. METHODS: We reviewed reports linking expression of HCV encoded proteins with changes in cellular functions. We also compared reports on HCV-induced HCC with those on non-viral and Hepatitis B Virus (HBV) induced HCC. Novel therapeutic approaches for handling metastatic HCC were also reviewed. RESULTS: HCV infection is associated with expression of multiple pro-metastatic factors in HCC patients. HCV encoded proteins can directly induce pro-metastasis cellular functions. HCV-induced HCC has a greater chance of recurrence than any non-viral and Hepatitis B Virus (HBV) induced HCC. Recent advances in understanding of evolutionary dynamics of tumor argue that trying to prevent spreading of cancer may ultimately prove to be a better approach than striving to cure it. Inhibiting the metastasis can thereby substantially increase the survival period in patients. Host cell protein Nm23-H1 is a known suppressor of tumor metastasis and has been shown to be modulated by proteins encoded by different viruses associated with cancers. CONCLUSION: Nm23-H1 is an important therapeutic target for virus mediated malignancies. This review is an attempt to summarize the current state of understanding of cancer cell metastasis in HCV induced tumors, and argues for approaches based on targeting host and viral factors critical for cancer metastasis as therapeutic targets.


Subject(s)
Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Hepatitis C/drug therapy , Liver Neoplasms/drug therapy , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Hepacivirus/metabolism , Hepatitis C/metabolism , Hepatitis C/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/virology , NM23 Nucleoside Diphosphate Kinases/metabolism , Neovascularization, Pathologic/drug therapy , Viral Proteins/metabolism
12.
Sci Rep ; 7(1): 12048, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28935957

ABSTRACT

Cyclic nucleotide signaling pathway plays a significant role in various biological processes such as cell growth, transcription, inflammation, in microbial pathogenesis, etc. Modulation of cyclic nucleotide levels by optogenetic tools has overcome certain limitations of studying transduction cascade by pharmacological agents and has allowed several ways to modulate biological processes in a spatiotemporal manner. Here, we have shown the optogenetic modulation of the cyclooxygenase 2 (Cox-2) gene expression and their downstream effector molecule (PGE2) in HEK-293T cells and the development process of Dictyostelium discoideum via modulating the cyclic nucleotide (cAMP) signaling pathway utilizing photoactivated adenylyl cyclases (PACs) as an optogenetic tool. Light-induced activation of PACs in HEK-293T cells increases the cAMP level that leads to activation of cAMP response element-binding protein (CREB) transcription factor and further upregulates downstream Cox-2 gene expression and their downstream effector molecule prostaglandin E2. In D. discoideum, the light-regulated increase in cAMP level affects the starvation-induced developmental process. These PACs could modulate the cAMP levels in a light-dependent manner and have a potential to control gene expression and their downstream effector molecules with varying magnitude. It would enable one to utilize PAC as a tool to decipher cyclic nucleotide mediated signaling pathway regulations and their mechanism.


Subject(s)
Adenylyl Cyclases/metabolism , Cyclic AMP/metabolism , Light , Optogenetics/methods , Signal Transduction/radiation effects , Adenylyl Cyclases/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dictyostelium/genetics , Dictyostelium/growth & development , Dictyostelium/metabolism , Dinoprostone/metabolism , Enzyme Activation/radiation effects , Gene Expression Regulation/radiation effects , HEK293 Cells , Humans , Microscopy, Confocal , Signal Transduction/genetics
13.
Front Microbiol ; 8: 538, 2017.
Article in English | MEDLINE | ID: mdl-28400769

ABSTRACT

Chronic inflammation is recognized as a threat factor for cancer progression. Release of inflammatory molecules generates microenvironment which is highly favorable for development of tumor, cancer progression and metastasis. In cases of latent viral infections, generation of such a microenvironment is one of the major predisposing factors related to virus mediated tumorigenesis. Among various inflammatory mediators implicated in pathological process associated with cancer, the cyclooxygenase (COX) and its downstream effector molecules are of greater significance. Though the role of infectious agents in causing inflammation leading to transformation of cells has been more or less well established, however, the mechanism by which inflammation in itself modulates the events in life cycle of infectious agent is not very much clear. This is specifically important for gammaherpesviruses infections where viral life cycle is characterized by prolonged periods of latency when the virus remains hidden, immunologically undetectable and expresses only a very limited set of genes. Therefore, it is important to understand the mechanisms for role of inflammation in virus life cycle and tumorigenesis. This review is an attempt to summarize the latest findings highlighting the significance of COX-2 and its downstream signaling effectors role in life cycle events of gammaherpesviruses leading to progression of cancer.

14.
Virology ; 506: 110-120, 2017 06.
Article in English | MEDLINE | ID: mdl-28376369

ABSTRACT

Hepatocellular carcinoma (HCC) is the most prevalent primary liver cancer and its incidence is on the rise largely attributed to Hepatitis C virus (HCV) related liver cancer. A distinct feature of HCV associated HCC is the substantially increased incidence of metastasis compared to non-viral or HBV associated HCC. Nm23-H1 is the first reported human metastasis suppressor down-regulated in many human metastatic cancers. Nm23-H1 functions are modulated in several virus associated cancers. Our study now shows that HCV E1 protein expression as well as HCV infection induces pro-metastatic effect on cancer cells which is simultaneous to Nm23-H1 transcriptional down-regulation and Nm23-H1 protein degradation. Moreover, Nm23-H1 intracellular localization is significantly altered in cells expressing HCV E1 protein. Importantly, overexpression of Nm23-H1 can rescue the cancer cells from pro-metastatic effects of HCV E1 and HCV infection. Our limited study provides evidence for role for Nm23-H1 in HCV mediated cancer metastasis.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/physiopathology , Hepacivirus/metabolism , Liver Neoplasms/physiopathology , NM23 Nucleoside Diphosphate Kinases/metabolism , Viral Envelope Proteins/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Movement , Down-Regulation , Gene Expression Regulation, Neoplastic , Hepacivirus/genetics , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Liver Neoplasms/virology , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasm Invasiveness , Viral Envelope Proteins/genetics
15.
Virology ; 484: 1-14, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26057147

ABSTRACT

Inflammation is one of the predisposing factors known to be associated with Epstein Barr Virus (EBV) mediated tumorigenesis. However it is not well understood whether inflammation in itself plays a role in regulating the life cycle of this infectious agent. COX-2, a key mediator of the inflammatory processes is frequently over-expressed in EBV positive cancer cells. In various tumors, PGE2 is the principle COX-2 regulated downstream product which exerts its effects on cellular processes through the EP1-4 receptors. In this study, we further elucidated how upregulated COX-2 levels can modulate the events in EBV life cycle related to latency-lytic reactivation. Our data suggest a role for upregulated COX-2 on modulation of EBV latency through its downstream effector PGE2. This study demonstrates a role for increased COX-2 levels in modulation of EBV latency. This is important for understanding the pathogenesis of EBV-associated cancers in people with chronic inflammatory conditions.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Herpesvirus 4, Human/physiology , Receptors, Prostaglandin E/metabolism , Signal Transduction , Virus Latency , Virus Replication , Cells, Cultured , Healthy Volunteers , Humans , Leukocytes, Mononuclear/virology
16.
Tumour Biol ; 36(4): 3051-60, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25501510

ABSTRACT

Epithelial-mesenchymal transition is an important mechanism in cancer invasiveness and metastasis. We had previously reported that cancer cells expressing Epstein-Barr virus (EBV) latent viral antigens EBV nuclear antigen EBNA3C and/ or EBNA1 showed higher motility and migration potential and had a propensity for increased metastases when tested in nude mice model. We now show that both EBNA3C and EBNA1 can modulate cellular pathways critical for epithelial to mesenchymal transition of cancer cells. Our data confirms that presence of EBNA3C or EBNA1 result in upregulation of transcriptional repressor Slug and Snail, upregulation of intermediate filament of mesenchymal origin vimentin, upregulation of transcription factor TCF8/ZEB1, downregulation as well as disruption of tight junction zona occludens protein ZO-1, downregulation of cell adhesion molecule E-cadherin, and nuclear translocation of ß-catenin. We further show that the primary tumors as well as metastasized lesions derived from EBV antigen-expressing cancer cells in nude mice model display EMT markers expression pattern suggesting their greater propensity to mesenchymal transition.


Subject(s)
Epithelial-Mesenchymal Transition , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/genetics , Neoplasms/genetics , Animals , Cadherins/biosynthesis , Epstein-Barr Virus Nuclear Antigens/immunology , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/pathogenicity , Homeodomain Proteins/biosynthesis , Humans , Mice , Neoplasm Metastasis , Neoplasms/pathology , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Vimentin/biosynthesis , Zinc Finger E-box-Binding Homeobox 1 , Zonula Occludens-1 Protein/biosynthesis , beta Catenin/biosynthesis
17.
Virusdisease ; 25(4): 493-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25674628

ABSTRACT

Virus-associated cancers account for more than 12 % of all the cancers. Hepatitis C virus (HCV) infects nearly 3 % of the population worldwide and has emerged as a major causative agent of liver disease with a big impact on public health. The HCV non-structural protein NS4A is a 54-amino-acid polypeptide that serves as an essential co-factor for the NS3 serine protease. We report here on a proteomic study to identify cellular proteins associated with NS4A. The results of this study show an association of three host cellular proteins with NS4A including two novel NS4A interacting partners. Our data provide evidence for complex involving NS4A with previously unreported cellular proteins including housekeeping protein GAPDH, and PI3P-5 K which is involved in cellular protein trafficking to nucleus. These novel associations add to the diversity of NS4A functions in relation to the virus infection and subsequent pathogenesis.

18.
Virology ; 410(1): 64-75, 2011 Feb 05.
Article in English | MEDLINE | ID: mdl-21093004

ABSTRACT

Resistance to apoptosis is an important component of the overall mechanism which drives the tumorigenic process. EBV is a ubiquitous human gamma-herpesvirus which preferentially establishes latent infection in viral infected B-lymphocytes. EBNA1 is typically expressed in most forms of EBV-positive malignancies and is important for replication of the latent episome in concert with replication of the host cells. Here, we investigate the effects of EBNA1 on survivin up-regulation in EBV-infected human B-lymphoma cells. We present evidence which demonstrates that EBNA1 forms a complex with Sp1 or Sp1-like proteins bound to their cis-element at the survivin promoter. This enhances the activity of the complex and up-regulates survivin. Knockdown of survivin and EBNA1 showed enhanced apoptosis in infected cells and thus supports a role for EBNA1 in suppressing apoptosis in EBV-infected cells. Here, we suggest that EBV encoded EBNA1 can contribute to the oncogenic process by up-regulating the apoptosis suppressor protein, survivin in EBV-associated B-lymphoma cells.


Subject(s)
Apoptosis/physiology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/immunology , Microtubule-Associated Proteins/metabolism , Cell Line, Tumor , Epstein-Barr Virus Nuclear Antigens/genetics , Herpesvirus 4, Human/genetics , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA Interference , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Survivin , Transcriptional Activation , Up-Regulation/drug effects , Up-Regulation/physiology
19.
Cancer Biol Ther ; 10(10): 961-78, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21084867

ABSTRACT

Tumor viruses have provided relatively simple genetic systems, which can be manipulated for understanding the molecular mechanisms of the cellular transformation process. A growing body of information in the tumor virology field provides several prospects for rationally targeted therapies. However, further research is needed to better understand the multiple mechanisms utilized by these viruses in cancer progression in order to develop therapeutic strategies. Initially viruses were believed to be associated with cancers as causative agents only in animals. It was almost half a century before the first human tumor virus, Epstein-Barr virus (EBV), was identified in 1964. Subsequently, several human tumor viruses have been identified including Kaposi sarcoma associated herpesvirus (KSHV), human Papillomaviruses (HPV), Hepatitis B virus (HBV), Hepatitis C virus (HCV), Human T lymphotropic virus (HTLV-1) and recently identified Merkel cell Polyomavirus (MCPyV). Tumor viruses are sub-categorized as either DNA viruses, which include EBV, KSHV, HPV, HBV, and MCPyV, or RNA viruses such as HCV and HTLV-1. Tumor-viruses induce oncogenesis through manipulating an array of different cellular pathways. These viruses initiate a series of cellular events, which lead to immortalization and proliferation of the infected cells by disrupting the mitotic checkpoint upon infection of the host cell. This is often accomplished by functional inhibition or proteasomal degradation of many tumor suppressor proteins by virally encoded gene products. The virally infected cells can either be eliminated via cell-mediated apoptosis or persist in a state of chronic infection. Importantly, the chronic persistence of infection by tumor viruses can lead to oncogenesis. This review discusses the major human tumor associated viruses and their ability to modulate numerous cell signaling pathways, which can be targeted for potential therapeutic approaches.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/etiology , Neoplasms/prevention & control , Oncogenic Viruses/pathogenicity , Signal Transduction/drug effects , Tumor Virus Infections/complications , Humans , Tumor Virus Infections/virology
20.
J Virol ; 84(19): 9718-32, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20660191

ABSTRACT

The latency-associated nuclear antigen (LANA) encoded by Kaposi's sarcoma-associated herpesvirus (KSHV) is critical for segregation of viral episomes to progeny nuclei and allows for maintenance of the viral genome in newly divided daughter cells. LANA binds to KSHV terminal repeat (TR) DNA and simultaneously associates with chromatin-bound cellular proteins. This process tethers the viral episomes to host chromosomes. However, the mechanism of tethering is complex and involves multiple protein-protein interactions. Our previous proteomics studies which showed the association of LANA with centromeric protein F (CENP-F) prompted us to further study whether LANA targets centromeric proteins for persistence of KSHV episomes during cell division. Here we show that LANA colocalized with CENP-F as speckles, some of which are paired at centromeric regions of a subset of chromosomes in KSHV-infected JSC-1 cells. We also confirm that both the amino and carboxy termini of LANA can bind to CENP-F. Moreover, LANA associated with another kinetochore protein, Bub1 (budding uninhibited by benzimidazole 1), which is known to form a complex with CENP-F. Importantly, we demonstrated the dynamic association of LANA and Bub1/CENP-F and the colocalization between Bub1, LANA, and the KSHV episome tethered to the host chromosome using fluorescence in situ hybridization (FISH). Knockdown of Bub1 expression by lentivirus-delivered short hairpin RNA (shRNA) dramatically reduced the number of KSHV genome copies, whereas no dramatic effect was seen with CENP-F knockdown. Therefore, the interaction between LANA and the kinetochore proteins CENP-F and Bub1 is important for KSHV genome tethering and its segregation to new daughter cells, with Bub1 potentially playing a more critical role in the long-term persistence of the viral genome in the infected cell.


Subject(s)
Antigens, Viral/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/physiology , Kinetochores/virology , Microfilament Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Antigens, Viral/chemistry , Antigens, Viral/genetics , Base Sequence , Cell Line , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , Gene Knockdown Techniques , Genome, Viral , Herpesvirus 8, Human/pathogenicity , Host-Pathogen Interactions , Humans , In Vitro Techniques , Kinetochores/metabolism , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Mitosis , Models, Biological , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Plasmids/genetics , Protein Interaction Mapping , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...