Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Virology ; 600: 110212, 2024 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-39232265

RESUMO

Viruses enter the host cell, and various strategies are employed to evade the host immune system. These include overcoming the various components of the immune system, including modulation of the physical and chemical barriers, non-specific innate response and specific adaptive immune response. Morbilliviruses impose immune modulation by utilizing various approaches including hindering antigen presentation to T-Helper (TH) cells, hematopoiesis and suppression of effector molecule activities. These viruses can also impede the early stages of T cell activation. Despite the availability of effective vaccines, morbilliviruses are still a significant threat to mankind. After infection, they also induce a state of immune suppression in the host. The molecular mechanisms employed by morbilliviruses to induce the state of immune suppression in the infected host are still being investigated. This review is an attempt to summarize insights into some of the strategies adopted by morbilliviruses to mediate immune modulation in the host.

2.
Front Microbiol ; 15: 1427606, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38966393

RESUMO

Peste des petits ruminants (PPR), a disease of socioeconomic importance has been a serious threat to small ruminants. The causative agent of this disease is PPR virus (PPRV) which belongs to the genus Morbillivirus. Hemagglutinin (H) is a PPRV coded transmembrane protein embedded in the viral envelope and plays a vital role in mediating the entry of virion particle into the cell. The infected host mounts an effective humoral response against H protein which is important for host to overcome the infection. In the present study, we have investigated structural, physiological and functional properties of hemagglutinin protein using various computational tools. The sequence analysis and structure prediction analysis show that hemagglutinin protein comprises of beta sheets as the predominant secondary structure, and may lack neuraminidase activity. PPRV-H consists of several important domains and motifs that form an essential scaffold which impart various critical roles to the protein. Comparative modeling predicted the protein to exist as a homo-tetramer that binds to its cognate cellular receptors. Certain amino acid substitutions identified by multiple sequence alignment were found to alter the predicted structure of the protein. PPRV-H through its predicted interaction with TLR-2 molecule may drive the expression of CD150 which could further propagate the virus into the host. Together, our study provides new insights into PPRV-H protein structure and its predicted functions.

3.
J Gen Virol ; 104(10)2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37831061

RESUMO

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.


Assuntos
Peste dos Pequenos Ruminantes , Vírus da Peste dos Pequenos Ruminantes , Animais , Humanos , Vírus da Peste dos Pequenos Ruminantes/genética , Peste dos Pequenos Ruminantes/metabolismo , Citocinas/genética , Citocinas/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ruminantes , Expressão Gênica , Cabras/genética
4.
Viruses ; 15(9)2023 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-37766213

RESUMO

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.


Assuntos
Proteínas do Nucleocapsídeo , Vírus da Peste dos Pequenos Ruminantes , Fosfatidilinositol 3-Quinases , Autofagia , Fosfatidilinositóis
5.
Virusdisease ; 33(1): 65-75, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35493756

RESUMO

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.

6.
J Gen Virol ; 102(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34704922

RESUMO

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.


Assuntos
Genoma Viral , Herpesviridae , Animais , Evolução Molecular , Herpesviridae/classificação , Herpesviridae/genética , Herpesviridae/fisiologia , Herpesviridae/ultraestrutura , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Adaptação ao Hospedeiro , Vírion/química , Vírion/ultraestrutura , Latência Viral , Replicação Viral
8.
Front Immunol ; 12: 630307, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33912160

RESUMO

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.


Assuntos
Peste dos Pequenos Ruminantes/imunologia , Animais , Encéfalo/virologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Antígenos H-2/imunologia , Imunidade Inata , Imunização , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Peste dos Pequenos Ruminantes/mortalidade , Peste dos Pequenos Ruminantes/patologia , Vírus da Peste dos Pequenos Ruminantes/imunologia , Receptores de Interferon/fisiologia , Vacinas Virais/imunologia
9.
Virusdisease ; 31(2): 75-79, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32368569

RESUMO

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.

10.
PLoS One ; 14(4): e0215394, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30998737

RESUMO

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.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Cromatina/metabolismo , Regulação Viral da Expressão Gênica/fisiologia , Herpesvirus Humano 8/fisiologia , Regiões Promotoras Genéticas , Proteínas Repressoras/metabolismo , Proteínas Virais/metabolismo , Ativação Viral/fisiologia , Fatores de Transcrição de Zíper de Leucina Básica/genética , Cromatina/genética , Cromatina/virologia , Células HEK293 , Humanos , Proteínas Repressoras/genética , Proteínas Virais/genética
11.
Arch Virol ; 164(5): 1271-1285, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30859475

RESUMO

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.


Assuntos
Carcinogênese/patologia , Carcinoma Hepatocelular/patologia , Hepacivirus/patogenicidade , Neoplasias Hepáticas/patologia , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Proteínas do Core Viral/metabolismo , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Regulação para Baixo , Células HEK293 , Hepatite C/patologia , Humanos , Neoplasias Hepáticas/virologia , Invasividade Neoplásica/patologia , Processamento de Proteína Pós-Traducional/fisiologia
12.
Arch Virol ; 164(1): 91-104, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30284629

RESUMO

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.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Herpesvirus Humano 8 , Proteínas Virais/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Humanos , Invasividade Neoplásica , Reação em Cadeia da Polimerase em Tempo Real , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
13.
Curr Drug Metab ; 19(3): 224-235, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29380693

RESUMO

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.


Assuntos
Antivirais/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Hepatite C/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Animais , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Hepacivirus/metabolismo , Hepatite C/metabolismo , Hepatite C/patologia , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neovascularização Patológica/tratamento farmacológico , Proteínas Virais/metabolismo
14.
Sci Rep ; 7(1): 12048, 2017 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-28935957

RESUMO

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.


Assuntos
Adenilil Ciclases/metabolismo , AMP Cíclico/metabolismo , Luz , Optogenética/métodos , Transdução de Sinais/efeitos da radiação , Adenilil Ciclases/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dictyostelium/genética , Dictyostelium/crescimento & desenvolvimento , Dictyostelium/metabolismo , Dinoprostona/metabolismo , Ativação Enzimática/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Células HEK293 , Humanos , Microscopia Confocal , Transdução de Sinais/genética
15.
Virology ; 506: 110-120, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28376369

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/fisiopatologia , Hepacivirus/metabolismo , Neoplasias Hepáticas/fisiopatologia , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Proteínas do Envelope Viral/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Movimento Celular , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Hepacivirus/genética , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Nucleosídeo NM23 Difosfato Quinases/genética , Invasividade Neoplásica , Proteínas do Envelope Viral/genética
16.
Front Microbiol ; 8: 538, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28400769

RESUMO

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.

17.
Virology ; 484: 1-14, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26057147

RESUMO

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.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Herpesvirus Humano 4/fisiologia , Receptores de Prostaglandina E/metabolismo , Transdução de Sinais , Latência Viral , Replicação Viral , Células Cultivadas , Voluntários Saudáveis , Humanos , Leucócitos Mononucleares/virologia
18.
Tumour Biol ; 36(4): 3051-60, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25501510

RESUMO

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.


Assuntos
Transição Epitelial-Mesenquimal , Antígenos Nucleares do Vírus Epstein-Barr/genética , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/genética , Neoplasias/genética , Animais , Caderinas/biossíntese , Antígenos Nucleares do Vírus Epstein-Barr/imunologia , Regulação Neoplásica da Expressão Gênica , Herpesvirus Humano 4/imunologia , Herpesvirus Humano 4/patogenicidade , Proteínas de Homeodomínio/biossíntese , Humanos , Camundongos , Metástase Neoplásica , Neoplasias/patologia , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese , Vimentina/biossíntese , Homeobox 1 de Ligação a E-box em Dedo de Zinco , Proteína da Zônula de Oclusão-1/biossíntese , beta Catenina/biossíntese
19.
Virusdisease ; 25(4): 493-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25674628

RESUMO

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.

20.
Virology ; 410(1): 64-75, 2011 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-21093004

RESUMO

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.


Assuntos
Apoptose/fisiologia , Linfócitos B/metabolismo , Linfócitos B/virologia , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/imunologia , Proteínas Associadas aos Microtúbulos/metabolismo , Linhagem Celular Tumoral , Antígenos Nucleares do Vírus Epstein-Barr/genética , Herpesvirus Humano 4/genética , Humanos , Proteínas Inibidoras de Apoptose , Proteínas Associadas aos Microtúbulos/genética , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Interferência de RNA , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Survivina , Ativação Transcricional , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA