Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(9): 3758-3763, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808762

RESUMO

Echoviruses are amongst the most common causative agents of aseptic meningitis worldwide and are particularly devastating in the neonatal population, where they are associated with severe hepatitis, neurological disease, including meningitis and encephalitis, and even death. Here, we identify the neonatal Fc receptor (FcRn) as a pan-echovirus receptor. We show that loss of expression of FcRn or its binding partner beta 2 microglobulin (ß2M) renders cells resistant to infection by a panel of echoviruses at the stage of virus attachment, and that a blocking antibody to ß2M inhibits echovirus infection in cell lines and in primary human intestinal epithelial cells. We also show that expression of human, but not mouse, FcRn renders nonpermissive human and mouse cells sensitive to echovirus infection and that the extracellular domain of human FcRn directly binds echovirus particles and neutralizes infection. Lastly, we show that neonatal mice expressing human FcRn are more susceptible to echovirus infection by the enteral route. Our findings thus identify FcRn as a pan-echovirus receptor, which may explain the enhanced susceptibility of neonates to echovirus infections.


Assuntos
Enterovirus Humano B/genética , Antígenos de Histocompatibilidade Classe I/genética , Receptores Fc/genética , Receptores Virais/genética , Microglobulina beta-2/genética , Animais , Infecções por Echovirus/genética , Infecções por Echovirus/imunologia , Infecções por Echovirus/virologia , Enterovirus Humano B/patogenicidade , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Camundongos , Ligação Proteica , Microglobulina beta-2/imunologia
2.
Oncotarget ; 6(42): 44289-305, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26517239

RESUMO

ATR and ATM are DNA damage signaling kinases that phosphorylate several thousand substrates. ATR kinase activity is increased at damaged replication forks and resected DNA double-strand breaks (DSBs). ATM kinase activity is increased at DSBs. ATM has been widely studied since ataxia telangiectasia individuals who express no ATM protein are the most radiosensitive patients identified. Since ATM is not an essential protein, it is widely believed that ATM kinase inhibitors will be well-tolerated in the clinic. ATR has been widely studied, but advances have been complicated by the finding that ATR is an essential protein and it is widely believed that ATR kinase inhibitors will be toxic in the clinic. We describe AZD6738, an orally active and bioavailable ATR kinase inhibitor. AZD6738 induces cell death and senescence in non-small cell lung cancer (NSCLC) cell lines. AZD6738 potentiates the cytotoxicity of cisplatin and gemcitabine in NSCLC cell lines with intact ATM kinase signaling, and potently synergizes with cisplatin in ATM-deficient NSCLC cells. In contrast to expectations, daily administration of AZD6738 and ATR kinase inhibition for 14 consecutive days is tolerated in mice and enhances the therapeutic efficacy of cisplatin in xenograft models. Remarkably, the combination of cisplatin and AZD6738 resolves ATM-deficient lung cancer xenografts.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia/deficiência , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Cisplatino/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Pirimidinas/administração & dosagem , Sulfóxidos/administração & dosagem , Administração Oral , Animais , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Disponibilidade Biológica , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Humanos , Indóis , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos Nus , Morfolinas , Inibidores de Proteínas Quinases/farmacocinética , Pirimidinas/farmacocinética , Interferência de RNA , Sulfonamidas , Sulfóxidos/farmacocinética , Transfecção , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
3.
Cancer Biol Ther ; 10(10): 1033-40, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20818173

RESUMO

Primary effusion lymphoma (PEL) is an aggressive form of lymphoma that is associated with infection by Kaposi's sarcoma-associated herpesvirus (KSHV). One of the KSHV genes expressed in PEL cells is K13, a potent activator of the NF-κB pathway. K13 transgenic mice develop lymphomas, but after a long period of latency. A possible candidate that could cooperate with K13 in the development of PEL is c-Myc, whose expression is frequently dysregulated in PEL cells. To study the cooperative interaction between K13 and c-Myc in the pathogenesis of PEL, we crossed the K13 transgenic mice to iMyc(Eµ) transgenic mice that overexpress Myc. We report that lymphomas in the K13/iMyc(Eµ) double transgenic mice developed with shorter latency and were histologically distinct from those observed in the iMyc(Eµ) mice. Lymphomas in the K13/iMyc(Eµ) mice also lacked the expression of B- and T-cell markers, thus resembling the immunophenotype of PEL. The accelerated development of lymphoma in the K13/iMyc(Eµ) mice was associated with increased expression of K13, elevated NF-κB activity and decrease in apoptosis. Taken collectively, our results demonstrate a cooperative interaction between the NF-κB and Myc pathways in lymphomagenesis.


Assuntos
Apoptose , Herpesvirus Humano 8/genética , Linfoma de Efusão Primária/metabolismo , Linfoma de Efusão Primária/patologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Proteínas Virais/fisiologia , Animais , Western Blotting , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Citometria de Fluxo , Técnicas Imunoenzimáticas , Linfoma de Efusão Primária/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/genética , NF-kappa B/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Transfecção
4.
BMC Med Genomics ; 2: 50, 2009 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-19660139

RESUMO

BACKGROUND: Kaposi's sarcoma (KS) associated herpesvirus (KSHV) is the etiological agent of KS, a neoplasm characterized by proliferating spindle cells, extensive neoangiogenesis and a prominent inflammatory infiltrate. Infection of blood vascular endothelial cells with KSHV in vitro results in their spindle cell transformation, which is accompanied by increased expression of inflammatory chemokines and cytokines, and acquisition of lymphatic endothelial markers. Mimicking the effect of viral infection, ectopic expression of KSHV-encoded latent protein vFLIP K13 is sufficient to induce spindle transformation of vascular endothelial cells. However, the effect of K13 expression on global gene expression and induction of lymphatic endothelial markers in vascular endothelial cells has not been studied. METHODS: We used gene array analysis to determine change in global gene expression induced by K13 in human vascular endothelial cells (HUVECs). Results of microarray analysis were validated by quantitative RT-PCR, immunoblotting and a multiplex cytokine array. RESULTS: K13 affected the expression of several genes whose expression is known to be modulated by KSHV infection, including genes involved in immune and inflammatory responses, anti-apoptosis, stress response, and angiogenesis. The NF-kappaB pathway was the major signaling pathway affected by K13 expression, and genetic and pharmacological inhibitors of this pathway effectively blocked K13-induced transcriptional activation of the promoter of CXCL10, one of the chemokines whose expression was highly upregulated by K13. However, K13, failed to induce expression of lymphatic markers in blood vascular endothelial cells. CONCLUSION: While K13 may account for change in the expression of a majority of genes observed following KSHV infection, it is not sufficient for inducing lymphatic reprogramming of blood vascular endothelial cells.

5.
Blood ; 113(22): 5660-8, 2009 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-19324905

RESUMO

Kaposi sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8, is the etiologic agent of Kaposi sarcoma (KS), an angioproliferative lesion characterized by dramatic angiogenesis and inflammatory infiltration. In this study, we report that expression of chemokine CCL20, a potent chemoattractant of dendritic cells and lymphocytes, is strongly induced in cultured cells either by KSHV infection or on ectopic expression of viral FLICE inhibitory protein K13. This induction is caused by transcriptional activation of CCL20 gene, which is mediated by binding of the p65, p50, and c-Rel subunits of the transcription factor nuclear factor-kappaB (NF-kappaB) to an atypical NF-kappaB-binding site present in the CCL20 gene promoter. The CCL20 gene induction is defective in K13 mutants that lack NF-kappaB activity, and can be blocked by specific genetic and pharmacologic inhibitors of the NF-kappaB pathway. CCR6, the specific receptor for CCL20, is also induced in cultured cells either by KSHV infection or on K13 expression. Finally, expression of CCL20 and CCR6 is increased in clinical samples of KS. These results suggest that KSHV and K13-mediated induction of CCL20 and CCR6 may contribute to the recruitment of dendritic cells and lymphocytes into the KS lesions, and to tumor growth and metastases.


Assuntos
Quimiocina CCL20/genética , Herpesvirus Humano 8/fisiologia , NF-kappa B/metabolismo , Proteínas Virais/fisiologia , Sítios de Ligação , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/antagonistas & inibidores , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/fisiologia , Células Cultivadas , Quimiocina CCL20/metabolismo , Células Dendríticas/patologia , Herpesvirus Humano 8/genética , Humanos , Células K562 , Linfócitos/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Regiões Promotoras Genéticas , Ligação Proteica , RNA Interferente Pequeno/farmacologia , Receptores CCR6/genética , Receptores CCR6/metabolismo , Sarcoma de Kaposi/imunologia , Sarcoma de Kaposi/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/genética
6.
PLoS One ; 2(10): e1067, 2007 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-17957251

RESUMO

BACKGROUND: Accumulating evidence suggests that dysregulated expression of lytic genes plays an important role in KSHV (Kaposi's sarcoma associated herpesvirus) tumorigenesis. However, the molecular events leading to the dysregulation of KSHV lytic gene expression program are incompletely understood. METHODOLOGY/PRINCIPAL FINDINGS: We have studied the effect of KSHV-encoded latent protein vFLIP K13, a potent activator of the NF-kappaB pathway, on lytic reactivation of the virus. We demonstrate that K13 antagonizes RTA, the KSHV lytic-regulator, and effectively blocks the expression of lytic proteins, production of infectious virions and death of the infected cells. Induction of lytic replication selects for clones with increased K13 expression and NF-kappaB activity, while siRNA-mediated silencing of K13 induces the expression of lytic genes. However, the suppressive effect of K13 on RTA-induced lytic genes is not uniform and it fails to block RTA-induced viral IL6 secretion and cooperates with RTA to enhance cellular IL-6 production, thereby dysregulating the lytic gene expression program. CONCLUSIONS/SIGNIFICANCE: Our results support a model in which ongoing KSHV lytic replication selects for clones with progressively higher levels of K13 expression and NF-kappaB activity, which in turn drive KSHV tumorigenesis by not only directly stimulating cellular survival and proliferation, but also indirectly by dysregulating the viral lytic gene program and allowing non-lytic production of growth-promoting viral and cellular genes. Lytic Replication-Induced Clonal Selection (LyRICS) may represent a general mechanism in viral oncogenesis.


Assuntos
Herpesvirus Humano 8/metabolismo , Interleucina-6/metabolismo , Neoplasias/virologia , Replicação Viral , Linhagem Celular , Replicação do DNA , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Inativação Gênica , Genes Virais , Humanos , Proteínas Imediatamente Precoces/química , RNA Interferente Pequeno/metabolismo , Proteínas Virais/química
7.
J Biol Chem ; 282(34): 24858-65, 2007 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-17597077

RESUMO

Kaposi's sarcoma herpesvirus oncoprotein vFLIP K13 is a potent activator of NF-kappaB and plays a key role in viral pathogenesis. K13 contains a putative TRAF-interacting motif, which is reportedly required for its interaction with TRAF2. The K13-TRAF2 interaction is believed to be essential for the recruitment of K13 to the I-kappaB kinase (IKK) complex and for K13-induced NF-kappaB and JNK activation. In addition, TRAF3 has been reported to be required for K13-induced NF-kappaB and JNK activation. We have re-examined the role of the TRAFs in K13 signaling and report that mutations in the putative TRAF-interacting motif of K13 have no deleterious effect on its ability to interact with the IKK complex or activation of the NF-kappaB pathway. Furthermore, endogenously expressed TRAF2 and TRAF3 do not interact with K13 and play no role in K13-induced NF-kappaB activation or its interaction with the IKK complex. Finally, K13 does not activate the JNK pathway. Our results support a model in which K13 bypasses the upstream components of the tumor necrosis factor receptor signaling pathway and directly interacts with the IKK complex to selectively activate the NF-kappaB pathway without affecting the JNK pathway. Selective NF-kappaB activation by K13 might represent a novel strategy employed by the virus to promote latency.


Assuntos
Herpesvirus Humano 8/metabolismo , Quinase I-kappa B/metabolismo , MAP Quinase Quinase 4/metabolismo , Proteínas Virais/química , Proteínas Virais/fisiologia , Motivos de Aminoácidos , Linhagem Celular , Humanos , Imunoprecipitação , Modelos Moleculares , NF-kappa B/metabolismo , Plasmídeos/metabolismo , Conformação Proteica , Interferência de RNA , Transdução de Sinais , Células U937
8.
Proc Natl Acad Sci U S A ; 102(36): 12885-90, 2005 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-16120683

RESUMO

Human herpesvirus 8 (HHV-8, also called Kaposi's sarcoma-associated herpes virus) has been linked to Kaposi's sarcoma and primary effusion lymphoma. HHV-8-encoded viral Fas-associated death domain-like IL-1-converting enzyme inhibitory protein (vFLIP) is one of the few viral proteins to be expressed in latently infected cells and plays a key role in the survival and proliferation of primary effusion lymphoma cells. Two main functions have been ascribed to HHV-8 vFLIP, inhibition of caspase 8/Fas-associated death domain-like IL-1-converting enzyme and activation of NF-kappaB. In this article, we demonstrate that vFLIP-expressing transgenic mice lack any of the features seen in mice deficient in caspase 8 or Fas-associated death domain protein and are not resistant to Fas-induced apoptosis. On the other hand, these mice display constitutive activation of classical and alternative NF-kappaB pathways, enhanced response to mitogenic stimuli, and increased incidence of lymphoma. Collectively, our results demonstrate that HHV-8 vFLIP is an oncogenic protein that mimics the signaling activities of caspase 8 during antigen receptor signaling and could contribute to the development of lymphoproliferative disorders via constitutive NF-kappaB activation independent of inhibition of Fas-induced apoptosis.


Assuntos
Apoptose , Linfoma/metabolismo , Linfoma/patologia , NF-kappa B/metabolismo , Proteínas Virais/metabolismo , Receptor fas/metabolismo , Animais , Linfócitos B/metabolismo , Linfócitos B/patologia , Transformação Celular Neoplásica , Células Cultivadas , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/fisiologia , Homeostase , Linfoma/genética , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Baço/citologia , Baço/metabolismo , Taxa de Sobrevida , Linfócitos T/metabolismo , Transgenes/genética , Proteínas Virais/genética
9.
Cancer Res ; 65(14): 6394-400, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16024643

RESUMO

DNA-alkylating agents have a central role in the curative therapy of many human tumors; yet, resistance to these agents limits their effectiveness. The efficacy of the alkylating agent temozolomide has been attributed to the induction of O6-MeG, a DNA lesion repaired by the protein O6-methylguanine-DNA methyltransferase (MGMT). Resistance to temozolomide has been ascribed to elevated levels of MGMT and/or reduced mismatch repair. However, >80% of the DNA lesions induced by temozolomide are N-methylated bases that are recognized by DNA glycosylases and not by MGMT, and so resistance to temozolomide may also be due, in part, to robust base excision repair (BER). We used isogenic cells deficient in the BER enzymes DNA polymerase-beta (pol-beta) and alkyladenine DNA glycosylase (Aag) to determine the role of BER in the cytotoxic effect of temozolomide. Pol-beta-deficient cells were significantly more susceptible to killing by temozolomide than wild-type or Aag-deficient cells, a hypersensitivity likely caused by accumulation of BER intermediates. RNA interference-mediated pol-beta suppression was sufficient to increase temozolomide efficacy, whereas a deficiency in pol-iota or pol-lambda did not increase temozolomide-mediated cytotoxicity. Overexpression of Aag (the initiating BER enzyme) triggered a further increase in temozolomide-induced cytotoxicity. Enhanced Aag expression, coupled with pol-beta knockdown, increased temozolomide efficacy up to 4-fold. Furthermore, loss of pol-beta coupled with temozolomide treatment triggered the phosphorylation of H2AX, indicating the activation of the DNA damage response pathway as a result of unrepaired lesions. Thus, the BER pathway is a major contributor to cellular resistance to temozolomide and its efficacy depends on specific BER gene expression and activity.


Assuntos
Reparo do DNA/fisiologia , Dacarbazina/análogos & derivados , Animais , Antineoplásicos Alquilantes/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Transformada , DNA Glicosilases/biossíntese , DNA Glicosilases/deficiência , DNA Glicosilases/metabolismo , DNA Polimerase beta/deficiência , DNA Polimerase beta/genética , DNA Polimerase beta/metabolismo , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Histonas/metabolismo , Camundongos , Fosforilação , RNA Interferente Pequeno/genética , Temozolomida , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...