Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Commun Biol ; 7(1): 871, 2024 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-39020082

RESUMO

Antibodies to Ebola virus glycoprotein (EBOV GP) represent an important correlate of the vaccine efficiency and infection survival. Both neutralization and some of the Fc-mediated effects are known to contribute the protection conferred by antibodies of various epitope specificities. At the same time, the role of the complement system remains unclear. Here, we compare complement activation by two groups of representative monoclonal antibodies (mAbs) interacting with the glycan cap (GC) or the membrane-proximal external region (MPER) of GP. Binding of GC-specific mAbs to GP induces complement-dependent cytotoxicity (CDC) in the GP-expressing cell line via C3 deposition on GP in contrast to MPER-specific mAbs. In the mouse model of EBOV infection, depletion of the complement system leads to an impairment of protection exerted by one of the GC-specific, but not MPER-specific mAbs. Our data suggest that activation of the complement system represents an important mechanism of antiviral protection by GC antibodies.


Assuntos
Anticorpos Monoclonais , Anticorpos Antivirais , Ebolavirus , Doença pelo Vírus Ebola , Polissacarídeos , Proteínas do Envelope Viral , Animais , Ebolavirus/imunologia , Anticorpos Monoclonais/imunologia , Camundongos , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/virologia , Doença pelo Vírus Ebola/prevenção & controle , Polissacarídeos/imunologia , Anticorpos Antivirais/imunologia , Humanos , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo , Ativação do Complemento , Camundongos Endogâmicos BALB C , Feminino , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Glicoproteínas/imunologia
2.
Cell Rep ; 42(10): 113254, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37858466

RESUMO

Ebola virus (EBOV) and Bundibugyo virus (BDBV) belong to the family Filoviridae and cause a severe disease in humans. We previously isolated a large panel of monoclonal antibodies from B cells of human survivors from the 2007 Uganda BDBV outbreak, 16 survivors from the 2014 EBOV outbreak in the Democratic Republic of the Congo, and one survivor from the West African 2013-2016 EBOV epidemic. Here, we demonstrate that EBOV and BDBV are capable of spreading to neighboring cells through intercellular connections in a process that depends upon actin and T cell immunoglobulin and mucin 1 protein. We quantify spread through intercellular connections by immunofluorescence microscopy and flow cytometry. One of the antibodies, BDBV223, specific to the membrane-proximal external region, induces virus accumulation at the plasma membrane. The inhibiting activity of BDBV223 depends on BST2/tetherin.


Assuntos
Anticorpos Monoclonais , Anticorpos Antivirais , Antígeno 2 do Estroma da Médula Óssea , Ebolavirus , Doença pelo Vírus Ebola , Humanos , Antígenos CD , Antígeno 2 do Estroma da Médula Óssea/imunologia , Ebolavirus/imunologia , Proteínas Ligadas por GPI , Doença pelo Vírus Ebola/virologia
3.
Cell Rep ; 42(2): 112103, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36773293

RESUMO

Retinoblastoma is a cancer of the infant retina primarily driven by loss of the Rb tumor suppressor gene, which is undruggable. Here, we report an autocrine signaling, mediated by secreted frizzled-related protein 2 (SFRP2), which suppresses nitric oxide and enables retinoblastoma growth. We show that coxsackievirus and adenovirus receptor (CXADR) is the cell-surface receptor for SFRP2 in retinoblastoma cells; that CXADR functions as a "dependence receptor," transmitting a growth-inhibitory signal in the absence of SFRP2; and that the balance between SFRP2 and CXADR determines nitric oxide production. Accordingly, high SFRP2 RNA expression correlates with high-risk histopathologic features in retinoblastoma. Targeting SFRP2 signaling by SFRP2-binding peptides or by a pharmacological inhibitor rapidly induces nitric oxide and profoundly inhibits retinoblastoma growth in orthotopic xenograft models. These results reveal a cytokine signaling pathway that regulates nitric oxide production and retinoblastoma cell proliferation and is amenable to therapeutic intervention.


Assuntos
Neoplasias da Retina , Retinoblastoma , Humanos , Óxido Nítrico , Proteínas Secretadas Relacionadas a Receptores Frizzled , Transdução de Sinais
4.
Cell Rep ; 36(1): 109254, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34233189

RESUMO

BAF chromatin remodeling complexes play important roles in chromatin regulation and cancer. Here, we report that Ewing sarcoma cells are dependent on the autocrine signaling mediated by NELL2, a secreted glycoprotein that has been characterized as an axon guidance molecule. NELL2 uses Robo3 as the receptor to transmit critical growth signaling. NELL2 signaling inhibits cdc42 and upregulates BAF complexes and EWS-FLI1 transcriptional output. We demonstrate that cdc42 is a negative regulator of BAF complexes, inducing actin polymerization and complex disassembly. Furthermore, we identify NELL2highCD133highEWS-FLI1high and NELL2lowCD133lowEWS-FLI1low populations in Ewing sarcoma, which display phenotypes consistent with high and low NELL2 signaling, respectively. We show that NELL2, CD133, and EWS-FLI1 positively regulate each other and upregulate BAF complexes and cell proliferation in Ewing sarcoma. These results reveal a signaling pathway regulating critical chromatin remodeling complexes and cancer cell proliferation.


Assuntos
Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia , Transdução de Sinais , Proteína cdc42 de Ligação ao GTP/metabolismo , Antígeno AC133/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Montagem e Desmontagem da Cromatina , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos SCID , Proteínas de Fusão Oncogênica/metabolismo , Fenótipo , Polimerização , Subunidades Proteicas/metabolismo , Proteômica , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA/metabolismo , Receptores de Superfície Celular/metabolismo , Sarcoma de Ewing/genética , Regulação para Cima
5.
Cell Rep ; 33(5): 108332, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33147457

RESUMO

We report here that the autocrine signaling mediated by growth and differentiation factor 6 (GDF6), a member of the bone morphogenetic protein (BMP) family of cytokines, maintains Ewing sarcoma growth by preventing Src hyperactivation. Surprisingly, Ewing sarcoma depends on the prodomain, not the BMP domain, of GDF6. We demonstrate that the GDF6 prodomain is a ligand for CD99, a transmembrane protein that has been widely used as a marker of Ewing sarcoma. The binding of the GDF6 prodomain to the CD99 extracellular domain results in recruitment of CSK (C-terminal Src kinase) to the YQKKK motif in the intracellular domain of CD99, inhibiting Src activity. GDF6 silencing causes hyperactivation of Src and p21-dependent growth arrest. We demonstrate that two GDF6 prodomain mutants linked to Klippel-Feil syndrome are hyperactive in CD99-Src signaling. These results reveal a cytokine signaling pathway that regulates the CSK-Src axis and cancer cell proliferation and suggest the gain-of-function activity for disease-causing GDF6 mutants.


Assuntos
Antígeno 12E7/metabolismo , Fator 6 de Diferenciação de Crescimento/metabolismo , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia , Transdução de Sinais , Quinases da Família src/metabolismo , Animais , Proteína Tirosina Quinase CSK/metabolismo , Proliferação de Células , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Fator 6 de Diferenciação de Crescimento/química , Humanos , Síndrome de Klippel-Feil/genética , Camundongos SCID , Mutação/genética , Proteínas de Fusão Oncogênica/metabolismo , Domínios Proteicos , Proteoma/metabolismo , Proteômica , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA/metabolismo , Transcrição Gênica
7.
J Virol ; 88(11): 6355-67, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24672028

RESUMO

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) establishes persistent latent infection in immunocompetent hosts. Disruption of KSHV latency results in viral lytic replication, which promotes the development of KSHV-related malignancies in immunocompromised individuals. While inhibitors of classes I and II histone deacetylases (HDACs) potently reactivate KSHV from latency, the role of class III HDAC sirtuins (SIRTs) in KSHV latency remains unclear. Here, we examined the effects of inhibitors of SIRTs, nicotinamide (NAM) and sirtinol, on KSHV reactivation from latency. Treatment of latently KSHV-infected cells with NAM or sirtinol induced transcripts and proteins of the master lytic transactivator RTA (ORF50), early lytic genes ORF57 and ORF59, and late lytic gene ORF65 and increased the production of infectious virions. NAM increased the acetylation of histones H3 and H4 as well as the level of the active histone H3 trimethyl Lys4 (H3K4me3) mark but decreased the level of the repressive histone H3 trimethyl Lys27 (H3K27me3) mark in the RTA promoter. Consistent with these results, we detected SIRT1 binding to the RTA promoter. Importantly, knockdown of SIRT1 was sufficient to increase the expression of KSHV lytic genes. Accordingly, the level of the H3K4me3 mark in the RTA promoter was increased following SIRT1 knockdown, while that of the H3K27me3 mark was decreased. Furthermore, SIRT1 interacted with RTA and inhibited RTA transactivation of its own promoter and that of its downstream target, the viral interleukin-6 gene. These results indicate that SIRT1 regulates KSHV latency by inhibiting different stages of viral lytic replication and link the cellular metabolic state with the KSHV life cycle. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causal agent of several malignancies, including Kaposi's sarcoma, commonly found in immunocompromised patients. While latent infection is required for the development of KSHV-induced malignancies, viral lytic replication also promotes disease progression. However, the mechanism controlling KSHV latent versus lytic replication remains unclear. In this study, we found that class III histone deacetylases (HDACs), also known as SIRTs, whose activities are linked to the cellular metabolic state, mediate KSHV replication. Inhibitors of SIRTs can reactivate KSHV from latency. SIRTs mediate KSHV latency by epigenetically silencing a key KSHV lytic replication activator, RTA. We found that one of the SIRTs, SIRT1, binds to the RTA promoter to mediate KSHV latency. Knockdown of SIRT1 is sufficient to induce epigenetic remodeling and KSHV lytic replication. SIRT1 also interacts with RTA and inhibits RTA's transactivation function, preventing the expression of its downstream genes. Our results indicate that SIRTs regulate KSHV latency by inhibiting different stages of viral lytic replication and link the cellular metabolic state with the KSHV life cycle.


Assuntos
Regulação Viral da Expressão Gênica/efeitos dos fármacos , Herpesvirus Humano 8/fisiologia , Sirtuína 1/antagonistas & inibidores , Ativação Viral/efeitos dos fármacos , Benzamidas/farmacologia , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Primers do DNA/genética , Técnicas de Silenciamento de Genes , Humanos , Proteínas Imediatamente Precoces/metabolismo , Imunoprecipitação , Luciferases , Microscopia Confocal , Naftóis/farmacologia , Niacinamida/farmacologia , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirtuína 1/genética , Transativadores/metabolismo , Ativação Viral/fisiologia , Latência Viral/efeitos dos fármacos
8.
Cell Cycle ; 13(5): 845-58, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24419204

RESUMO

Kaposi sarcoma-associated herpesvirus (KSHV) is a tumor virus encoding several proto-oncogenes. However, the roles of these viral genes in KSHV-induced tumorigenesis have not been defined. In this study, we used a recently developed model of KSHV-induced cellular transformation and tumorigenesis combining with a reverse genetic system to examine the role of a KSHV latent gene vCyclin (ORF72), a cellular Cyclin D2 homolog, in KSHV-induced oncogenesis. Deletion of vCyclin did not affect cell proliferation and cell cycle progression at a low-density condition, when cells were at an active proliferation state. However, vCyclin mutant cells were contact-inhibited and arrested at G 1 phase at a high-density condition. As a result, vCyclin mutant cells formed less and smaller colonies in soft agar assay. Nude mice inoculated with vCyclin mutant cells had reduced tumor incidence and extended tumor latency and survival compared with mice inoculated with wild-type (WT) virus-infected cells. WT but not mutant virus effectively induced Cyclin-dependent kinase inhibitor p27/Kip1 Ser10 phosphorylation and cytoplasmic relocalization. shRNA knockdown of p27 released the blockage of the mutant cells from cell cycle arrest at G 1 phase at a high-density condition. Together, these results indicate that vCyclin primarily functions to enhance cellular transformation and tumorigenesis by promoting cell cycle progression and cell proliferation at a contact-inhibited condition.


Assuntos
Carcinogênese/genética , Inibição de Contato/fisiologia , Ciclina D2/metabolismo , Herpesvirus Humano 8/fisiologia , Proteínas Virais/metabolismo , Animais , Apoptose , Carcinogênese/patologia , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Senescência Celular , Ciclina D2/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Camundongos Nus , Mutação , Fosforilação , Proteínas Virais/genética
9.
J Virol ; 87(11): 6326-35, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536671

RESUMO

Kaposi sarcoma-associated herpesvirus (KSHV) stimulates proliferation, angiogenesis, and inflammation to promote Kaposi sarcoma (KS) tumor growth, which involves various growth factors and cytokines. Previously, we found that KSHV infection of human umbilical vein endothelial cells (HUVECs) induces a transcriptional induction of the proangiogenic and proinflammatory cytokine angiopoietin-2 (Ang-2). Here, we report that KSHV induces rapid release of Ang-2 that is presynthesized and stored in the Weibel-Palade bodies (WPB) of endothelial cells upon binding to its integrin receptors. Blocking viral binding to integrins inhibits Ang-2 release. KSHV binding activates the integrin tyrosine kinase receptor signaling pathways, leading to tyrosine phosphorylation of focal adhesion kinase (FAK), the tyrosine kinase Src, and the Calα2 subunit of the l-type calcium channel to trigger rapid calcium (Ca(2+)) influx. Pretreatment of endothelial cells with specific inhibitors of protein tyrosine kinases inhibits KSHV-induced Ca(2+) influx and Ang-2 release. Inhibition of Ca(2+) mobilization with calcium channel blockers also inhibits Ang-2 release. Thus, the interaction between KSHV and its integrin receptors plays a key role in regulating rapid Ang-2 release from endothelial cells. This finding highlights a novel mechanism of viral induction of angiogenesis and inflammation, which might play important roles in the early event of KS tumor development.


Assuntos
Angiopoietina-2/metabolismo , Células Endoteliais/metabolismo , Herpesvirus Humano 8/fisiologia , Sarcoma de Kaposi/metabolismo , Cálcio/metabolismo , Células Endoteliais/enzimologia , Células Endoteliais/virologia , Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Herpesvirus Humano 8/genética , Humanos , Integrinas/metabolismo , Fosforilação , Sarcoma de Kaposi/enzimologia , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virologia
10.
J Biol Chem ; 287(21): 17554-17567, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22467874

RESUMO

c-Jun N-terminal kinase (JNK) is a serine/threonine phosphotransferase whose sustained activation in response to genotoxic stress promotes apoptosis. In Drosophila, the normally rapid JNK-dependent apoptotic response to genotoxic stress is significantly delayed in Dmp53 (Drosophila p53) mutants. Likewise, the extent of JNK activity after UV irradiation is dependent on p53 in murine embryonic fibroblasts with loss of p53 resulting in diminished JNK activity. Together, these results suggest that p53 potentiates the JNK-dependent response to genotoxic stress; however, the mechanism whereby p53 stimulates JNK activity remains undefined. Here, we demonstrate that both Drosophila and human p53 can directly stimulate JNK activity independently of p53-dependent gene transcription. Furthermore, we demonstrate that both the Drosophila and human p53 orthologs form a physical complex with diphosphorylated JNK ((DP)JNK) both in vivo and in vitro, suggesting that the interaction is evolutionarily conserved. Focusing on human p53, we demonstrate that the interaction maps to the DNA binding domain (hp53(DBD)). Intriguingly, binding of p53(DBD) alone to (DP)JNK prevented its inactivation by MAPK phosphatase (MKP)-5; however, JNK was still able to phosphorylate c-Jun while in a complex with the p53(DBD). Apparent dissociation constants for the p53(DBD)·(DP)JNK (274 ± 14 nm) and MKP-5·(DP)JNK (55 ± 8 nm) complexes were established; however, binding of MKP-5 and p53 to JNK was not mutually exclusive. Together, these results suggest that stress-dependent increases in p53 levels potentiate JNK activation by preventing its rapid dephosphorylation by MKPs and that the simultaneous activation of p53 and JNK may constitute a "fail-safe" switch for the JNK-dependent apoptotic response.


Assuntos
Apoptose/fisiologia , Proteínas de Drosophila/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Complexos Multiproteicos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Drosophila melanogaster , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Ativação Enzimática/fisiologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Complexos Multiproteicos/genética , Fosforilação , Estrutura Terciária de Proteína , Proteína Supressora de Tumor p53/genética
11.
J Clin Invest ; 122(3): 1076-81, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22293176

RESUMO

Infections by viruses are associated with approximately 12% of human cancer. Kaposi's sarcoma-associated herpesvirus (KSHV) is causally linked to several malignancies commonly found in AIDS patients. The mechanism of KSHV-induced oncogenesis remains elusive, due in part to the lack of an adequate experimental system for cellular transformation of primary cells. Here, we report efficient infection and cellular transformation of primary rat embryonic metanephric mesenchymal precursor cells (MM cells) by KSHV. Cellular transformation occurred at as early as day 4 after infection and in nearly all infected cells. Transformed cells expressed hallmark vascular endothelial, lymphatic endothelial, and mesenchymal markers and efficiently induced tumors in nude mice. KSHV established latent infection in MM cells, and lytic induction resulted in low levels of detectable infectious virions despite robust expression of lytic genes. Most KSHV-induced tumor cells were in a latent state, although a few showed heterogeneous expression of lytic genes. This efficient system for KSHV cellular transformation of primary cells might facilitate the study of growth deregulation mechanisms resulting from KSHV infections.


Assuntos
Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica , Herpesvirus Humano 8/metabolismo , Células-Tronco Mesenquimais/citologia , Animais , Técnicas de Cultura de Células , Linhagem Celular , Transformação Celular Viral , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Nus , Microcirculação , Fases de Leitura Aberta , Ratos , Fatores de Tempo
12.
Virus Genes ; 44(2): 225-36, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22180077

RESUMO

Kaposi's sarcoma-associated herpesvirus ORF30-33 locus encodes four genes with unknown functions. We performed transcriptional mapping of these genes. Northern-hybridization, 5'- and 3'-rapid amplification of cDNA ends, and DNA sequencing identified four transcripts of 3.7, 3.6, 2.7, and 1.4 kb, none of which has alternative splicing. While all transcripts have the same termination site, their start sites vary. All transcripts are not expressed or only weakly expressed in latent cells but can be chemically induced. The 3.7 and 3.6 kb transcripts contain all four genes and are sensitive to cycloheximide (CH) but resistant to phosphonoacetic acid (PAA), indicating that they are early lytic transcripts. The 2.7 kb transcript contains ORF32 and ORF33 genes while the 1.4 kb transcript contains the ORF33 gene. Both transcripts are sensitive to CH and PAA, indicating that they are late lytic transcripts. Furthermore, we identified four promoters with functional TATA boxes, none of which is directly transactivated by RTA. Examination of the 5' untranslated region of ORF31 failed to identify any functional internal ribosome entry sites. These results define the transcriptional patterns of the ORF30-33 locus, which should help the delineation of its function.


Assuntos
Perfilação da Expressão Gênica , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/genética , Fases de Leitura Aberta , Transcrição Gênica , Northern Blotting , DNA Complementar/genética , Humanos , Técnicas de Amplificação de Ácido Nucleico , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Sítio de Iniciação de Transcrição
13.
PLoS Pathog ; 7(5): e1002054, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21625536

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) establishes a latent infection in the host following an acute infection. Reactivation from latency contributes to the development of KSHV-induced malignancies, which include Kaposi's sarcoma (KS), the most common cancer in untreated AIDS patients, primary effusion lymphoma and multicentric Castleman's disease. However, the physiological cues that trigger KSHV reactivation remain unclear. Here, we show that the reactive oxygen species (ROS) hydrogen peroxide (H2O2) induces KSHV reactivation from latency through both autocrine and paracrine signaling. Furthermore, KSHV spontaneous lytic replication, and KSHV reactivation from latency induced by oxidative stress, hypoxia, and proinflammatory and proangiogenic cytokines are mediated by H2O2. Mechanistically, H2O2 induction of KSHV reactivation depends on the activation of mitogen-activated protein kinase ERK1/2, JNK, and p38 pathways. Significantly, H2O2 scavengers N-acetyl-L-cysteine (NAC), catalase and glutathione inhibit KSHV lytic replication in culture. In a mouse model of KSHV-induced lymphoma, NAC effectively inhibits KSHV lytic replication and significantly prolongs the lifespan of the mice. These results directly relate KSHV reactivation to oxidative stress and inflammation, which are physiological hallmarks of KS patients. The discovery of this novel mechanism of KSHV reactivation indicates that antioxidants and anti-inflammation drugs could be promising preventive and therapeutic agents for effectively targeting KSHV replication and KSHV-related malignancies.


Assuntos
Herpesvirus Humano 8/fisiologia , Peróxido de Hidrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ativação Viral , Latência Viral , Acetilcisteína/metabolismo , Animais , Western Blotting , Butadienos/farmacologia , Catalase/metabolismo , Linhagem Celular , Imunofluorescência , Glutationa/metabolismo , Células HEK293 , Infecções por Herpesviridae , Herpesvirus Humano 8/metabolismo , Humanos , Imidazóis/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/farmacologia , Estresse Oxidativo , Piridinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoma de Kaposi/virologia , Transdução de Sinais , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Arch Virol ; 156(5): 739-46, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21487663

RESUMO

Simian varicella virus (SVV) is closely related to human varicella-zoster virus and causes varicella and zoster-like disease in nonhuman primates. In this study, a mini-F replicon was inserted into a SVV cosmid, and infectious SVV was generated by co-transfection of Vero cells with overlapping SVV cosmids. The entire SVV genome, cloned as a bacterial artificial chromosome (BAC), was stably propagated upon serial passage in E. coli. Transfection of pSVV-BAC DNA into Vero cells yielded infectious SVV (rSVV-BAC). The mini-F vector sequences flanked by loxP sites were removed by co-infection of Vero cells with rSVV-BAC and adenovirus expressing Cre-recombinase. Recombinant SVV generated using the SVV-BAC genetic system has similar molecular and in vitro replication properties as wild-type SVV. To demonstrate the utility of this approach, a SVV ORF 10 deletion mutant was created using two-step Red-mediated recombination. The results indicate that SVV ORF 10, which encodes a homolog of the HSV-1 virion VP-16 transactivator protein, is not essential for in vitro replication but is required for optimal replication in cell culture.


Assuntos
Cromossomos Artificiais Bacterianos , Clonagem Molecular , Genoma Viral , Herpesviridae/genética , Animais , Chlorocebus aethiops , Escherichia coli/genética , Células Vero
15.
J Virol ; 84(22): 11709-17, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20826690

RESUMO

Rhesus rhadinovirus (RRV) is a gammaherpesvirus closely related to Kaposi's sarcoma-associated herpesvirus (KSHV), an oncogenic virus linked to the development of Kaposi's sarcoma and several other lymphoproliferative diseases, including primary effusion lymphoma and multicentric Castleman's disease. RRV naturally infects rhesus macaques and induces lymphoproliferative diseases under experimental conditions, making it an excellent model for the study of KSHV. Unlike KSHV, which grows poorly in cell culture, RRV replicates efficiently in rhesus fibroblasts (RFs). In this study, we have characterized the entry pathway of RRV in RFs. Using a luciferase-expressing recombinant RRV (RRV-luciferase), we show that the infectivity of RRV is reduced by inhibitors of endosomal acidification. RRV infectivity is also reduced by inhibitors of clathrin-mediated but not caveola-mediated endocytosis, indicating that RRV enters into RFs via clathrin-mediated endocytosis. Using a red fluorescent protein (RFP)-expressing recombinant RRV (RRV-RFP), we show that RRV particles are colocalized with markers of endocytosis (early endosome antigen 1) and clathrin-mediated endocytosis (clathrin heavy chain) during entry into RFs. RRV particles are also colocalized with transferrin, which enters cells by clathrin-mediated endocytosis, but not with cholera toxin B, which enters cells by caveola-mediated endocytosis. Inhibition of clathrin-mediated endocytosis with a dominant-negative construct of EPS15, an essential component of clathrin-coated pits, blocked the entry of RRV into RFs. Together, these results indicate that RRV entry into RFs is mediated by clathrin-mediated endocytosis.


Assuntos
Clatrina/metabolismo , Endocitose , Fibroblastos/metabolismo , Infecções por Herpesviridae/metabolismo , Macaca mulatta , Rhadinovirus/fisiologia , Animais , Células Cultivadas , Endossomos/metabolismo , Endossomos/virologia , Fibroblastos/virologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/fisiologia , Humanos , Macaca mulatta/virologia
16.
Nat Cell Biol ; 11(11): 1355-62, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19838173

RESUMO

Autophagy is an active homeostatic degradation process for the removal or turnover of cytoplasmic components wherein the LC3 ubiquitin-like protein undergoes an Atg7 E1-like enzyme/Atg3 E2-like enzyme-mediated conjugation process to induce autophagosome biogenesis. Besides its cytoprotective role, autophagy acts on cell death when it is abnormally upregulated. Thus, the autophagy pathway requires tight regulation to ensure that this degradative process is well balanced. Two death effector domains (DED1/2) containing cellular FLICE-like inhibitor protein (cFLIP) and viral FLIP (vFLIP) of Kaposi's sarcoma-associated herpesvirus (KSHV), Herpesvirus saimiri (HVS), and Molluscum contagiosum virus (MCV) protect cells from apoptosis mediated by death receptors. Here, we report that cellular and viral FLIPs suppress autophagy by preventing Atg3 from binding and processing LC3. Consequently, FLIP expression effectively represses cell death with autophagy, as induced by rapamycin, an mTor inhibitor and an effective anti-tumour drug against KSHV-induced Kaposi's sarcoma (KS) and primary effusion lymphoma (PEL). Remarkably, either a DED1 alpha2-helix ten amino-acid (alpha2) peptide or a DED2 alpha4-helix twelve amino-acid (alpha4) peptide of FLIP is individually sufficient for binding FLIP itself and Atg3, with the peptide interactions effectively suppressing Atg3-FLIP interaction without affecting Atg3-LC3 interaction, resulting in robust cell death with autophagy. Our study thus identifies a checkpoint of the autophagy pathway where cellular and viral FLIPs limit the Atg3-mediated step of LC3 conjugation to regulate autophagosome biogenesis. Furthermore, the FLIP-derived short peptides induce growth suppression and cell death with autophagy, representing biologically active molecules for potential anti-cancer therapies.


Assuntos
Apoptose/fisiologia , Autofagia/fisiologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/fisiologia , Animais , Herpesvirus Humano 8/patogenicidade , Camundongos , Células NIH 3T3 , Técnicas do Sistema de Duplo-Híbrido
17.
Virology ; 379(2): 234-44, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18684478

RESUMO

Following primary infection, KSHV establishes a lifelong persistent latent infection in the host. The mechanism of KSHV latency is not fully understood. The latent nuclear antigen (LANA or LNA) encoded by ORF73 is one of a few viral genes expressed during KSHV latency, and is consistently detected in all KSHV-related malignancies. LANA is essential for KSHV episome persistence, and regulates the expression of viral lytic genes through epigenetic silencing, and inhibition of the expression and transactivation function of the key KSHV lytic replication initiator RTA (ORF50). In this study, we used a genetic approach to examine the role of LANA in regulating KSHV lytic replication program. Deletion of LANA did not affect the expression of its adjacent genes vCyclin (ORF72) and vFLIP (ORF71). In contrast, the expression levels of viral lytic genes including immediate-early gene RTA, early genes MTA (ORF57), vIL-6 (ORF-K2) and ORF59, and late gene ORF-K8.1 were increased before and after viral lytic induction with 12-O-tetradecanoyl-phorbol-13-acetate and sodium butyrate. This enhanced expression of viral lytic genes was also observed following overexpression of RTA with or without simultaneous chemical induction. Consistent with these results, the LANA mutant cells produced more infectious virions than the wild-type virus cells did. Furthermore, genetic repair of the mutant virus reverted the phenotypes to those of wild-type virus. Together, these results have demonstrated that, in the context of viral genome, LANA contributes to KSHV latency by regulating the expression of RTA and its downstream genes.


Assuntos
Antígenos Virais/genética , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/imunologia , Proteínas Nucleares/genética , Antígenos Virais/fisiologia , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Expressão Gênica/efeitos dos fármacos , Genes Virais , Herpesvirus Humano 8/patogenicidade , Herpesvirus Humano 8/fisiologia , Humanos , Proteínas Nucleares/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Deleção de Sequência , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica , Proteínas Virais/genética , Latência Viral/genética , Latência Viral/imunologia , Replicação Viral/genética , Replicação Viral/imunologia
18.
J Virol ; 82(9): 4235-49, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18305042

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) latency is central to the evasion of host immune surveillances and induction of KSHV-related malignancies. The mechanism of KSHV latency remains unclear. Here, we show that the KSHV latent gene vFLIP promotes viral latency by inhibiting viral lytic replication. vFLIP suppresses the AP-1 pathway, which is essential for KSHV lytic replication, by activating the NF-kappaB pathway. Thus, by manipulating two convergent cellular pathways, vFLIP regulates both cell survival and KSHV lytic replication to promote viral latency. These results also indicate that the effect of the NF-kappaB pathway on KSHV replication is determined by the status of the AP-1 pathway and hence provide a mechanistic explanation for the contradictory role of the NF-kappaB pathway in KSHV replication. Since the NF-kappaB pathway is commonly activated during infection of gammaherpesviruses, these findings might have general implications for the control of gammaherpesviral latency.


Assuntos
Herpesvirus Humano 8/genética , NF-kappa B/metabolismo , Fator de Transcrição AP-1/metabolismo , Proteínas Virais/genética , Proteínas Virais/fisiologia , Latência Viral/genética , Replicação Viral , Linhagem Celular , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Transativadores/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores
19.
Cancer Treat Res ; 133: 69-127, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17672038

RESUMO

KSHV has been established as the causative agent of KS, PEL, and MCD, malignancies occurring more frequently in AIDS patients. The aggressive nature of KSHV in the context of HIV infection suggests that interactions between the two viruses enhance pathogenesis. KSHV latent infection and lytic reactivation are characterized by distinct gene expression profiles, and both latency and lytic reactivation seem to be required for malignant progression. As a sophisticated oncogenic virus, KSHV has evolved to possess a formidable repertoire of potent mechanisms that enable it to target and manipulate host cell pathways, leading to increased cell proliferation, increased cell survival, dysregulated angiogenesis, evasion of immunity, and malignant progression in the immunocompromised host. Worldwide, approximately 40.3 million people are currently living with HIV infection. Of these, a significant number are coinfected with KSHV. The complex interplay between the two viruses dramatically elevates the risk for development of KSHV-induced malignancies, KS, PEL, and MCD. Although HAART significantly reduces HIV viral load, the entire T-cell repertoire and immune function may not be completely restored. In fact, clinically significant immune deficiency is not necessary for the induction of KSHV-related malignancy. Because of variables such as lack of access to therapy noncompliance with prescribed treatment, failure to respond to treatment and the development of drug-resistant strains of HIV, KSHV-induced malignancies will continue to present as major health concerns.


Assuntos
Síndrome da Imunodeficiência Adquirida/complicações , Síndrome da Imunodeficiência Adquirida/virologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Neoplasias/complicações , Neoplasias/virologia , Animais , Herpesvirus Humano 8/química , Humanos , Sarcoma de Kaposi/complicações , Sarcoma de Kaposi/virologia
20.
J Virol ; 80(24): 12187-96, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17035322

RESUMO

Open reading frame 45 (ORF45) of Kaposi's sarcoma-associated herpesvirus (KSHV) encodes an immediate-early protein. This protein is also present in virions as a tegument protein. ORF45 protein interacts with interferon regulatory factor 7 (IRF-7) and inhibits virus-induced type I interferon production by blocking activation of IRF-7. To define further the function of ORF45 and the mechanism underlying its action, we constructed an ORF45-null recombinant virus genome (BAC-stop45) by using a bacterial artificial chromosome (BAC) system. Stable 293T cells carrying the BAC36 (wild type) and BAC-stop45 genomes were generated. When monolayers of 293T BAC36 and 293T BAC-stop45 cells were induced with 12-O-tetradecanoylphorbol-13-acetate and sodium butyrate, no significant difference was found between them in overall viral gene expression and lytic DNA replication, but induced 293T BAC-stop45 cells released 10-fold fewer virions to the medium than did 293T BAC36 cells. When ORF45-null virus was used to infect cells, lower infectivity was observed than for wild-type BAC36. These results suggest that KSHV ORF45 plays roles in both early and late stages of viral infection, probably in viral ingress and egress.


Assuntos
Expressão Gênica , Herpesvirus Humano 8/genética , Proteínas Imediatamente Precoces/genética , Proteínas Virais/genética , Vírion/fisiologia , Western Blotting , Linhagem Celular , Cromossomos Artificiais Bacterianos , Primers do DNA , Escherichia coli , Componentes do Gene , Humanos , Proteínas Imediatamente Precoces/metabolismo , Mutagênese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Virais/metabolismo , Vírion/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA