Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 202
Filtrar
1.
PLoS One ; 8(3): e58572, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23484037

RESUMO

Human metapneumovirus (HMPV) encodes a small hydrophobic (SH) protein of unknown function. HMPV from which the SH open reading frame was deleted (HMPVΔSH) was viable and displayed similar replication kinetics, cytopathic effect and plaque size compared with wild type HMPV in several cell-lines. In addition, no differences were observed in infection efficiency or cell-to-cell spreading in human primary bronchial epithelial cells (HPBEC) cultured at an air-liquid interphase. Host gene expression was analyzed in A549 cells infected with HMPV or HMPVΔSH using microarrays and mass spectrometry (MS) based techniques at multiple time points post infection. Only minor differences were observed in mRNA or protein expression levels. A possible function of HMPV SH as apoptosis blocker, as proposed for several members of the family Paramyxoviridae, was rejected based on this analysis. So far, a clear phenotype of HMPV SH deletion mutants in vitro at the virus and host levels is absent.


Assuntos
Regulação da Expressão Gênica/genética , Metapneumovirus/genética , Metapneumovirus/fisiologia , Proteínas Oncogênicas de Retroviridae/fisiologia , Replicação Viral/genética , Western Blotting , Brônquios/citologia , Linhagem Celular , Células Epiteliais/virologia , Deleção de Genes , Humanos , Espectrometria de Massas , Análise em Microsséries , Proteínas Oncogênicas de Retroviridae/deficiência
2.
J Virol ; 83(22): 11467-76, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19726513

RESUMO

Human T-lymphotropic virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia/lymphoma, and it encodes a number of nonstructural proteins that are involved in virus replication and immune evasion. The viral protein p12 previously has been characterized to interfere with major histocompatibility complex class, ICAM-1, and ICAM-2 expression, and it activates STAT5. Using a previously established T-cell line immortalized with an HTLV-1 molecular clone deleted for p12, we assessed the role of p12 in regulating cellular growth and virus transmission. These cells were complemented for p12 expression by the transduction of a lentivirus vector expressing p12. We report that p12 conferred a selective growth advantage in vitro and increased the colony formation of human T cells in soft-agar assays. Consistently with previous studies, p12- and p12+ cell lines produced similar amounts of virus particles released into the supernatant of cultured cells, although we found that p12 expression greatly enhanced virus transmission. Moreover, we found that interleukin-2 (IL-2) stimulation also increased HTLV-1 transmission whether p12 was expressed or not, and inversely, that the inhibition of Jak signaling significantly reduced HTLV-1 transmission. Intriguingly, IL-2/Jak signaling was not associated with changes in viral gene expression, viral RNA encapsidation, the maturation of the virus particle, cell-cell adherence, or Gag polarization and virological synapse formation. We do demonstrate, however, that IL-2 stimulation and p12 expression significantly increased the rate of syncytium formation, revealing a novel role for IL-2 signaling and Jak activation in HTLV-1 virus transmission.


Assuntos
Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Janus Quinases/fisiologia , Receptores de Interleucina-2/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Transformação Celular Viral/fisiologia , Humanos , Interleucina-2/fisiologia , Proteínas Oncogênicas de Retroviridae/fisiologia , Fatores de Transcrição STAT/fisiologia , Transdução de Sinais/fisiologia , Replicação Viral/fisiologia
3.
Cell Mol Life Sci ; 65(21): 3422-32, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18818869

RESUMO

Sheep betaretroviruses offer a unique model system to study the complex interaction between retroviruses and their host. Jaagsiekte sheep retrovirus (JSRV) is a pathogenic exogenous retrovirus and the causative agent of ovine pulmonary adenocarcinoma. The sheep genome contains at least 27 copies of endogenous retroviruses (enJSRVs) highly related to JSRV. enJSRVs have played several roles in the evolution of the domestic sheep as they are able to block the JSRV replication cycle and play a critical role in sheep conceptus development and placental morphogenesis. Available data strongly suggest that some dominant negative enJSRV proviruses (i.e. able to block JSRV replication) have been positively selected during evolution. Interestingly, viruses escaping the transdominant enJSRV loci have recently emerged (less than 200 years ago). Thus, endogenization of these retroviruses may still be occurring today. Therefore, sheep provide an exciting and unique system to study retrovirus-host coevolution. (Part of a multi-author review).


Assuntos
Betaretrovirus/fisiologia , Interações Hospedeiro-Patógeno , Infecções por Retroviridae/veterinária , Doenças dos Ovinos/virologia , Ovinos/virologia , Sequência de Aminoácidos , Animais , Betaretrovirus/genética , Betaretrovirus/patogenicidade , Transformação Celular Viral/genética , Transformação Celular Viral/fisiologia , Desenvolvimento Embrionário/fisiologia , Evolução Molecular , Feminino , Regulação Viral da Expressão Gênica , Genes Virais , Interações Hospedeiro-Patógeno/genética , Modelos Moleculares , Dados de Sequência Molecular , Morfogênese , Placenta/virologia , Placentação , Gravidez , Conformação Proteica , Provírus/genética , Provírus/fisiologia , Adenomatose Pulmonar Ovina/virologia , Infecções por Retroviridae/virologia , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/fisiologia , Seleção Genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Ovinos/embriologia , Especificidade da Espécie , Infecções Tumorais por Vírus/veterinária , Infecções Tumorais por Vírus/virologia , Interferência Viral
4.
Cell Mol Life Sci ; 65(21): 3366-82, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18818873

RESUMO

The genomes of vertebrates contain sequences that are similar to present-day exogenous retroviruses. Such sequences, called endogenous retroviruses (ERVs), have resulted from ancestral germ line infections by exogenous retroviruses which have thereafter been transmitted in a Mendelian fashion. By analogy to exogenous tumorigenic retroviruses, ERVs have been implicated in the pathogenesis of cancer. Cumulative evidence from animal models indicates that ERVs may participate in the process of malignant transformation or promote tumor growth, e.g. through insertional mutagenesis or via counteracting tumor immunosurveillance. Here, we review the role of ERVs in tumorigenesis with focus on human ERVs (HERVs) in human cancer. Although available data suggest a potential role of HERVs in human cancers, in particular germ cell tumors, the contributions of HERVs to human tumorigenesis warrant further elucidation. (Part of a multi-author review).


Assuntos
Transformação Celular Viral , Retrovirus Endógenos/patogenicidade , Interações Hospedeiro-Patógeno/fisiologia , Neoplasias/virologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Animais , Cocarcinogênese , Retrovirus Endógenos/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Vigilância Imunológica , Masculino , Mamíferos/genética , Mamíferos/virologia , Modelos Biológicos , Neoplasias/genética , Neoplasias/imunologia , Proto-Oncogenes/genética , Provírus/genética , Provírus/fisiologia , Infecções por Retroviridae/genética , Infecções por Retroviridae/veterinária , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/fisiologia , Evasão Tumoral , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/veterinária , Integração Viral/genética
5.
J Virol ; 82(16): 8224-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550666

RESUMO

Human metapneumovirus, a leading cause of respiratory tract infections in infants, encodes a small hydrophobic (SH) protein of unknown function. In this study, we showed that infection of airway epithelial cells or mice with recombinant human metapneumovirus lacking SH expression (rhMPV-DeltaSH) enhanced secretion of proinflammatory mediators, including interleukin 6 (IL-6) and IL-8, encoded by two NF-kB-dependent genes, compared to infection with wild-type rhMPV. RhMPV-DeltaSH infection resulted in enhanced NF-kB-dependent gene transcription and in increased levels of phosphorylated and acetylated NF-kB without affecting its nuclear translocation, identifying a possible novel mechanism by which paramyxovirus SH proteins modulate NF-kB activation.


Assuntos
Regulação Viral da Expressão Gênica , Metapneumovirus/metabolismo , NF-kappa B/metabolismo , Proteínas Oncogênicas de Retroviridae/fisiologia , Transcrição Gênica , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular Tumoral , Citocinas/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Macaca mulatta , Camundongos , Fosforilação , Proteínas Oncogênicas de Retroviridae/metabolismo
6.
Virology ; 370(2): 273-84, 2008 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-17945326

RESUMO

The receptor-binding domain (RBD) in the surface (SU) subunit of gammaretrovirus envelope glycoprotein is critical for determining the host receptor specificity of the virus. This domain is separated from the carboxy terminal C domain (Cdom) of SU by a proline-rich region. In this study, we show that the Cdom region in the SU from subgroup C feline leukemia virus (FeLV-C) forms a second receptor-binding domain that is distinct from its RBD, and which can independently bind to its host receptor FLVCR1, in the absence of RBD. Furthermore, our results suggest that residues located in the C2 disulfide-bonded loop in FeLV-C Cdom are critical for SU binding to FLVCR1 and for virus infection. We propose that binding of FeLV-C SU to FLVCR1 involves interaction of two receptor-binding domains (RBD and Cdom) with FLVCR1, and that this mechanism of interaction is conserved for other gammaretroviruses. Our results could have important implications for designing gammaretrovirus vectors that can efficiently infect specific target cells.


Assuntos
Produtos do Gene env/química , Produtos do Gene env/fisiologia , Vírus da Leucemia Felina/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação/genética , Gatos , Linhagem Celular , Primers do DNA/genética , DNA Viral/genética , Produtos do Gene env/genética , Humanos , Vírus da Leucemia Felina/classificação , Vírus da Leucemia Felina/genética , Vírus da Leucemia Felina/patogenicidade , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/fisiologia , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Receptores Virais/genética , Receptores Virais/fisiologia , Proteínas Oncogênicas de Retroviridae/química , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/fisiologia , Homologia de Sequência de Aminoácidos , Virulência
7.
J Virol ; 80(4): 1619-28, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16439518

RESUMO

Gammaretroviruses, including the subgroups A, B, and C of feline leukemia virus (FeLV), use a multiple-membrane-spanning transport protein as a receptor. In some cases, such as FeLV-T, a nonclassical receptor that includes both a transport protein (Pit1) and a soluble cofactor (FeLIX) is required for entry. To define which regions confer specificity to classical versus nonclassical receptor pathways, we engineered mutations found in either FeLV-A/T or FeLV-T, individually and in combination, into the backbone of the transmissible form of the virus, FeLV-A. The receptor specificities of these viruses were tested by measuring infection and binding to cells expressing the FeLV-A receptor or the FeLV-T receptors. FeLV-A receptor specificity was maintained when changes at amino acid position 6, 7, or 8 of the mature envelope glycoprotein were introduced, although differences in infection efficiency were observed. When these N-terminal mutations were introduced together with a C-terminal 4-amino-acid insertion and an adjacent amino acid change, the resulting viruses acquired FeLV-T receptor specificity. Additionally, a W-->L change at amino acid position 378, although not required, enhanced infectivity for some viruses. Thus, we have found that determinants in the N and C termini of the envelope surface unit can direct entry via the nonclassical FeLV-T receptor pathway. The region that has been defined as the receptor binding domain of gammaretroviral envelope proteins determined entry via the FeLV-A receptor independently of the presence of the N- and C-terminal FeLV-T receptor determinants.


Assuntos
Vírus da Leucemia Felina/genética , Vírus da Leucemia Felina/fisiologia , Receptores Virais/fisiologia , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/fisiologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia , Substituição de Aminoácidos , Animais , Sítios de Ligação/genética , Linhagem Celular , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Mutação
8.
J Virol ; 80(4): 1700-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16439527

RESUMO

Mumps virus (MuV), a rubulavirus of the paramyxovirus family, causes acute infections in humans. MuV has seven genes including a small hydrophobic (SH) gene, which encodes a type I membrane protein of 57 amino acid residues. The function of the SH protein is not clear, although its expression is not necessary for growth of MuV in tissue culture cells. It is speculated that MuV SH plays a role in viral pathogenesis. Simian virus 5 (SV5), a closely related rubulavirus, encodes a 44-amino-acid-residue SH protein. Recombinant SV5 lacking the SH gene (rSV5DeltaSH) is viable and has no growth defect in tissue culture cells. However, rSV5DeltaSH induces apoptosis in tissue culture cells and is attenuated in vivo. Neutralizing antibodies against tumor necrosis factor alpha (TNF-alpha) and TNF-alpha receptor 1 block rSV5DeltaSH-induced apoptosis, suggesting that SV5 SH plays an essential role in blocking the TNF-alpha-mediated apoptosis pathway. Because MuV is closely related to SV5, we hypothesize that the SH protein of MuV has a function similar to that of SV5, even though there is no sequence homology between them. To test this hypothesis and to study the function of MuV SH, we have replaced the open reading frame (ORF) of SV5 SH with the ORF of MuV SH in a SV5 genome background. The recombinant SV5 (rSV5DeltaSH+MuV-SH) was analyzed in comparison with SV5. It was found that rSV5DeltaSH+MuV-SH was viable and behaved like wild-type SV5, suggesting that MuV SH has a function similar to that of SV5 SH. Furthermore, both ectopically expressed SV5 SH and MuV SH blocked activation of NF-kappaB by TNF-alpha in a reporter gene assay, suggesting that both SH proteins can inhibit TNF-alpha signaling.


Assuntos
Vírus da Caxumba/fisiologia , Vírus da Parainfluenza 5/fisiologia , Proteínas Oncogênicas de Retroviridae/fisiologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Proteínas Virais/fisiologia , Animais , Apoptose , Bovinos , Linhagem Celular , Chlorocebus aethiops , Efeito Citopatogênico Viral , Humanos , Camundongos , Modelos Biológicos , Vírus da Caxumba/genética , Vírus da Parainfluenza 5/genética , Recombinação Genética , Proteínas Oncogênicas de Retroviridae/genética , Ensaio de Placa Viral , Proteínas Virais/genética
9.
Neurochem Res ; 30(3): 355-61, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16018579

RESUMO

We investigated the importance of the phosphoinositide3-kinase (PI3K) pathway in CA1 and dentate gyrus (DG) areas of hippocampus by exposing organotypic cultures to LY294002, a PI3K inhibitor, or to oxygen and glucose deprivation (OGD) for up to 21 hours. LY294002 induced increased propidium iodide (PI) uptake and caspase 3/7 activity in both regions, with a faster onset in DG. In contrast, cultures exposed to 60 min of OGD showed a PI uptake only in the CA1 area, beginning 13 h after the insult and increasing until 21 h. We did not observe any significant changes in AKT phosphorylation and immunocontent in CA1 or DG areas of organotypic cultures exposed to OGD, suggesting that the phosphorylation of this protein at Ser-473 is unrelated to the cellular damage induced by ischemia. Our results suggest that the inhibition of the PI3K pathway does not mimic the cell death profile observed with an ischemic model.


Assuntos
Inibidores Enzimáticos/farmacologia , Glucose/deficiência , Hipocampo/patologia , Hipóxia/patologia , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Cromonas/farmacologia , Ativação Enzimática/efeitos dos fármacos , Morfolinas/farmacologia , Proteína Oncogênica v-akt , Fosforilação , Ratos , Ratos Wistar , Proteínas Oncogênicas de Retroviridae/fisiologia
10.
J Biol Chem ; 280(32): 29025-9, 2005 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-15955807

RESUMO

The glucose transporter GLUT1, a member of the multimembrane-spanning facilitative nutrient transporter family, serves as a receptor for human T cell leukemia virus (HTLV) infection. Here, we show that the 7 amino acids of the extracellular loop 6 of GLUT1 (ECL6) placed in the context of the related GLUT3 transporter were sufficient for HTLV envelope binding. Glutamate residue 426 in ECL6 was identified as critical for binding. However, binding to ECL6 was not sufficient for HTLV envelope-driven infection. Infection required two additional determinants located in ECL1 and ECL5, which otherwise did not influence HTLV envelope binding. Moreover the single N-glycosylation chain located in ECL1 was not required for HTLV infection. Therefore, binding involves a discrete determinant in the carboxyl terminal ECL6, whereas post-binding events engage extracellular sequences in the amino and carboxyl terminus of GLUT1.


Assuntos
Deltaretrovirus/metabolismo , Produtos do Gene env/fisiologia , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Oncogênicas de Retroviridae/fisiologia , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular , Produtos do Gene env/química , Glucose/metabolismo , Transportador de Glucose Tipo 1 , Ácido Glutâmico/química , Glicosilação , Humanos , Óperon Lac , Dados de Sequência Molecular , Mutação Puntual , Ligação Proteica , Estrutura Terciária de Proteína , Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/química , Homologia de Sequência de Aminoácidos
11.
J Cereb Blood Flow Metab ; 25(9): 1150-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15815584

RESUMO

The intracellular mechanisms that regulate neurogenesis remain unclear. Using neurospheres isolated from the subventricular zone (SVZ) of the adult rat, we investigated the effect of cyclic guanosine monophosphate (cGMP) and its signaling pathway on the induction of neurogenesis. Neurospheres expressed phosphodiesterase 5 (PDE5) and treatment of neurospheres with Sildenafil, a specific inhibitor of PDE5, significantly increased cGMP levels and neurogenesis. In addition, incubation of neurospheres with Sildenafil significantly phosphorylated Akt, which was associated with an increase of phosphorylation of glycogen synthase kinase 3 (GSK-3), a downstream target of Akt. Coincubation of neurospheres with Sildenafil and LY 294002, a pharmacological inhibitor of PI3-K/Akt, abolished Sildenafil-induced phosphorylated Akt and GSK-3. Furthermore, LY 294002 blocked Sildenafil-increased SVZ cell proliferation. These data suggest that Sildenafil-enhanced neurogenesis likely occurs through activation of the PI3-K/Akt/GSK-3 pathway.


Assuntos
Ventrículos Cerebrais/citologia , AMP Cíclico/fisiologia , Neurônios/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Oncogênicas de Retroviridae/fisiologia , Transdução de Sinais/fisiologia , 3',5'-GMP Cíclico Fosfodiesterases , Animais , Western Blotting , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Ventrículos Cerebrais/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5 , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Masculino , Proteína Oncogênica v-akt , Inibidores de Fosfodiesterase/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Piperazinas/farmacologia , Purinas , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Citrato de Sildenafila , Células-Tronco/fisiologia , Sulfonas
12.
J Cereb Blood Flow Metab ; 25(9): 1111-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15829918

RESUMO

Delayed administration of vascular endothelial growth factor (VEGF) promotes functional recovery after focal cerebral ischemia. However, early intravenous injection of VEGF increases blood-brain barrier (BBB) leakage, hemorrhagic transformation and infarct volume whereas its application to cortical surface is neuroprotective. We have investigated whether or not early intracerebroventricular administration of VEGF could replicate the neuroprotective effect observed with topical application and the mechanism of action of this protection. Mice were subjected to 90 mins middle cerebral artery (MCA) occlusion and 24 h of reperfusion. Vascular endothelial growth factor (8 ng, intracerebroventricular) was administered 1 or 3 h after reperfusion. Compared with the vehicle-treated (intracerebroventricular) group, VEGF decreased the infarct volume along with BBB leakage in both treatment groups. Neurologic disability scores improved in parallel to the changes in infarct volume. Independently of the decrease in infarct size, VEGF also reduced the number of TUNEL-positive apoptotic neurons. Phospo-Akt levels were significantly higher in ischemic hemispheres of the VEGF-treated mice. Contrary to intracerebroventricular route, intravenous administration of VEGF (15 microg/kg) enhanced the infarct volume as previously reported for the rat. In conclusion, single intracerebroventricular injection of VEGF protects brain against ischemia without adversely affecting BBB permeability, and has a relatively long therapeutic time window. This early neuroprotective action, observed well before recovery-promoting actions such as angiogenesis, possibly involves activation of the PI-3-Akt pathway.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Isquemia Encefálica/prevenção & controle , Fármacos Neuroprotetores , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Western Blotting , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Infarto Cerebral/patologia , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Hemodinâmica/efeitos dos fármacos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Injeções Intraventriculares , Camundongos , Artéria Cerebral Média/fisiologia , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/prevenção & controle , Proteína Oncogênica v-akt , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle , Proteínas Oncogênicas de Retroviridae/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/administração & dosagem
13.
J Virol ; 79(7): 4533-9, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15767455

RESUMO

Retroviral transmembrane proteins (TMs) contain an N-terminal fusion peptide that initiates virus-cell membrane fusion. The fusion peptide is linked to the coiled-coil core through a conserved sequence that is often rich in glycines. We investigated the functional role of the glycine-rich segment, Met-326 to Ser-337, of the human T-cell leukemia virus type 1 (HTLV-1) TM, gp21, by alanine and proline scanning mutagenesis. Alanine substitution for the hydrophobic residue Ile-334 caused an approximately 90% reduction in cell-cell fusion activity without detectable effects on the lipid-mixing and pore formation phases of fusion. Alanine substitutions at other positions had smaller effects (Gly-329, Val-330, and Gly-332) or no effect on fusion function. Proline substitution for glycine residues inhibited cell-cell fusion function with position-dependent effects on the three phases of fusion. Retroviral glycoprotein fusion function thus appears to require flexibility within the glycine-rich segment and hydrophobic contacts mediated by this segment.


Assuntos
Produtos do Gene env/fisiologia , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Fusão de Membrana , Estrutura Terciária de Proteína , Proteínas Oncogênicas de Retroviridae/fisiologia , Substituição de Aminoácidos , Produtos do Gene env/química , Produtos do Gene env/genética , Vírus Linfotrópico T Tipo 1 Humano/genética , Mutagênese Sítio-Dirigida , Mutação , Proteínas Oncogênicas de Retroviridae/química , Proteínas Oncogênicas de Retroviridae/genética , Produtos do Gene env do Vírus da Imunodeficiência Humana
14.
Leuk Res ; 29(3): 307-16, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15661267

RESUMO

Friend leukemia virus (FLV) infection strongly enhances gamma-irradiation-induced apoptosis of hematopoietic cells of C3H hosts leading to a lethal anemia. Experiments using p53 knockout mice with the C3H background have clarified that the apoptosis is p53-dependent and would not be associated with changes of cell populations caused by the infection with FLV. In bone marrow cells of FLV + total body irradiation (TBI)-treated C3H mice, the p53 protein was prominently activated to overexpress p21 and bax suggesting that apoptosis-enhancing mechanisms lay upstream of p53 protein in the signaling pathway. Neither of DNA-dependent protein kinase (DNA-PK)-deficient SCID mice nor ataxia telangiectasia mutated (ATM) gene knockout mice with the C3H background exhibited a remarkable enhancement of apoptosis or p53 activation on FLV + TBI-treatment indicating that DNA-PK and ATM were both essential. ATM appeared necessary for introducing DNA damage-induced apoptosis, while DNA-PK enhanced p53-dependent apoptosis under FLV-infection. Surprisingly, viral envelope protein, gp70, was co-precipitated with DNA-PK but not with ATM in FLV + TBI-treated C3H mice. These results indicated that FLV-infection enhances DNA damage-induced apoptosis via p53 activation and that DNA-PK, in association with gp70, might play critical roles in modulating the signaling pathway.


Assuntos
Dano ao DNA/efeitos da radiação , Proteínas de Ligação a DNA/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Oncogênicas de Retroviridae/fisiologia , Proteínas do Envelope Viral/fisiologia , Animais , Apoptose/efeitos da radiação , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteína Quinase Ativada por DNA , Proteínas de Ligação a DNA/genética , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Leucemia Experimental/enzimologia , Leucemia Experimental/virologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Camundongos SCID , Proteínas Serina-Treonina Quinases/genética , Infecções por Retroviridae/radioterapia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Infecções Tumorais por Vírus/radioterapia , Irradiação Corporal Total
15.
Biochemistry ; 43(49): 15494-502, 2004 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-15581361

RESUMO

Cbl is phosphorylated by the insulin receptor and reportedly functions within the flotillin/CAP/Cbl/Crk/C3G/TC10 complex during insulin-stimulated glucose transport in 3T3/L1 adipocytes. Cbl, via pYXXM motifs at tyrosine-371 and tyrosine-731, also activates phosphatidylinositol (PI) 3-kinase, which is required to activate atypical protein kinase C (aPKC) and glucose transport during thiazolidinedione action in 3T3/L1 and human adipocytes [Miura et al. (2003) Biochemistry 42, 14335-14341]. Presently, we have examined the importance of Cbl in activating PI 3-kinase and aPKC during insulin action in 3T3/L1 adipocytes by expressing Y371F and Y731F Cbl mutants, which nullify pYXXM binding of Cbl to SH2 domains of downstream effectors. Interestingly, these mutants inhibited insulin-induced increases in (a) binding of Cbl to both Crk and the p85 subunit of PI 3-kinase, (b) activation of Cbl-dependent PI 3-kinase, (c) activation and translocation of aPKC to the plasma membrane, (d) translocation of Glut4 to the plasma membrane, (e) and glucose transport. Importantly, coexpression of wild-type Cbl reversed the inhibitory effects of Cbl mutants. In contrast to Cbl-dependent PI 3-kinase, Cbl mutants did not significantly inhibit the activation of PI 3-kinase by IRS-1, which is also required during insulin action. Our findings suggest that (a) Cbl uses pYXXM motifs to simultaneously activate PI 3-kinase and Crk/C3G/TC10 pathways and (b) Cbl, along with IRS-1, functions upstream of PI 3-kinase and aPKCs during insulin-stimulated glucose transport in 3T3/L1 adipocytes.


Assuntos
Glucose/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/metabolismo , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Oncogênicas de Retroviridae/fisiologia , Células 3T3-L1 , Motivos de Aminoácidos/genética , Animais , Transporte Biológico/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Humanos , Insulina/farmacologia , Antagonistas da Insulina/metabolismo , Antagonistas da Insulina/farmacologia , Proteínas Substratos do Receptor de Insulina , Isoenzimas/metabolismo , Camundongos , Proteína Oncogênica v-cbl , Fenilalanina/genética , Fosfoproteínas/fisiologia , Fosforilação , Ligação Proteica , Transporte Proteico , Proteínas Proto-Oncogênicas c-crk , Proteínas Oncogênicas de Retroviridae/biossíntese , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/metabolismo , Tirosina/genética , Tirosina/metabolismo
16.
Biochemistry ; 43(49): 15503-9, 2004 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-15581362

RESUMO

Phosphatidylinositol 3-kinase (PI3K)-dependent activation of atypical protein kinase C (aPKC) is required for insulin-stimulated glucose transport. Although insulin receptor substrate-1 (IRS-1) and IRS-2, among other factors, activate PI3K, there is little information on the relative roles of IRS-1and IRS-2 during aPKC activation by insulin action in specific cell types. Presently, we have used immortalized brown adipocytes in which either IRS-1 or IRS-2 has been knocked out by recombinant methods to examine IRS-1 and IRS-2 requirements for activation of aPKC. We have also used these adipocytes to see if IRS-1 and IRS-2 are required for activation of Cbl, which is required for insulin-stimulated glucose transport and has been found to function upstream of both PI3K/aPKC and Crk during thiazolidinedione action in 3T3/L1 adipocytes [Miura et al. (2003) Biochemistry 42, 14335]. In brown adipocytes in which either IRS-1 or IRS-2 was knocked out, insulin-induced increases in aPKC activity and glucose transport were markedly diminished. These effects of insulin on aPKC and glucose transport were fully restored by retroviral-mediated expression of IRS-1 or IRS-2 in their respective knockout cells. Knockout of IRS-1 or IRS-2 also inhibited insulin-induced increases in Cbl binding to the p85 subunit of PI3K, which, along with IRS-1/2, may be required for activation of PI3K, aPKC, and glucose transport during insulin action in 3T3/L1 adipocytes. These findings provide evidence that directly links both IRS-1 and IRS-2 to aPKC activation in immortalized brown adipocytes, and further suggest that IRS-1 and IRS-2 are required for the activation of Cbl/PI3K during insulin action in these cells.


Assuntos
Tecido Adiposo Marrom/metabolismo , Insulina/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/fisiologia , Proteína Quinase C/metabolismo , Proteínas Oncogênicas de Retroviridae/fisiologia , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/enzimologia , Animais , Linhagem Celular Transformada , Desoxiglucose/metabolismo , Sinergismo Farmacológico , Ativação Enzimática , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Proteína Oncogênica v-cbl , Fosfatidilinositol 3-Quinases/fisiologia , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Fosforilação , Proteína Quinase C/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Trítio/metabolismo
17.
Biochemistry ; 43(46): 14584-93, 2004 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-15544329

RESUMO

Activation of many Rho family GTPase pathways involves the signaling module consisting of the Dbl-like guanine nucleotide exchange factors (GEFs), the Rho GTPases, and the Rho GTPase specific effectors. The current biochemical model postulates that the GEF-stimulated GDP/GTP exchange of Rho GTPases leads to the active Rho-GTP species, and subsequently the active Rho GTPases interact with and activate the effectors. Here we report an unexpected finding that the Dbl oncoprotein, Cdc42 GTPase, and PAK1 can form a complex through their minimum functional motifs, i.e., the Dbl-homolgy (DH) and Pleckstrin-homology domains of Dbl, Cdc42, and the PBD domain of PAK1. The Dbl-Cdc42-PAK1 complex is sensitive to the nucleotide-binding state of Cdc42 since either dominant negative or constitutively active Cdc42 readily disrupts the ternary binding interaction. The complex formation depends on the interactions between the DH domain of Dbl and Cdc42 and between Cdc42 and the PBD domain of PAK1 and can be reconstituted in vitro by using the purified components. Furthermore, the Dbl-Cdc42-PAK1 ternary complex is active in generating signaling output through the activated PAK1 kinase in the complex. The GEF-Rho-effector ternary intermediate is also found in other Dbl-like GEF, Rho GTPase, and effector interactions. Finally, PAK1, through the PDB domain, is able to accelerate the GEF-induced GTP loading onto Cdc42. These results suggest that signal transduction through Cdc42 and possibly other Rho family GTPases could involve tightly coupled guanine nucleotide exchange and effector activation mechanisms and that Rho GTPase effector may have a feedback regulatory role in the Rho GTPase activation.


Assuntos
Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Oncogênicas de Retroviridae/fisiologia , Transdução de Sinais/fisiologia , Proteína cdc42 de Ligação ao GTP/fisiologia , Animais , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Células COS , Chlorocebus aethiops , Sinergismo Farmacológico , Ativação Enzimática , Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Modelos Moleculares , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Ligação Proteica , Conformação Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas Oncogênicas de Retroviridae/química , Proteínas Oncogênicas de Retroviridae/metabolismo , Homologia Estrutural de Proteína , Frações Subcelulares/química , Frações Subcelulares/metabolismo , Proteína cdc42 de Ligação ao GTP/química , Proteína cdc42 de Ligação ao GTP/metabolismo , Quinases Ativadas por p21 , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/fisiologia
18.
Front Biosci ; 9: 2556-76, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15358581

RESUMO

Human T-cell lymphotropic virus type 1 (HTLV-1), causes adult T cell leukemia/lymphoma (ATLL), and initiates a variety of immune mediated disorders. The viral genome encodes common structural and enzymatic proteins characteristic of all retroviruses and utilizes alternative splicing and alternate codon usage to make several regulatory and accessory proteins encoded in the pX region (pX ORF I to IV). Recent studies indicate that the accessory proteins p12I, p27I, p13II, and p30II, encoded by pX ORF I and II, contribute to viral replication and the ability of the virus to maintain typical in vivo expression levels. Proviral clones that are mutated in either pX ORF I or II, while fully competent in cell culture, are severely limited in their replicative capacity in a rabbit model. These HTLV-1 accessory proteins are critical for establishment of viral infectivity, enhance T-lymphocyte activation and potentially alter gene transcription and mitochondrial function. HTLV-1 pX ORF I expression is critical to the viral infectivity in resting primary lymphocytes suggesting a role for the calcineurin-binding protein p12I in lymphocyte activation. The endoplasmic reticulum and cis-Golgi localizing p12I activates NFAT, a key T cell transcription factor, through calcium-mediated signaling pathways and may lower the threshold of lymphocyte activation via the JAK/STAT pathway. In contrast p30II localizes to the nucleus and represses viral promoter activity, but may regulate cellular gene expression through p300/CBP or related co-activators of transcription. The mitochondrial localizing p13II induces morphologic changes in the organelle and may influence energy metabolism infected cells. Future studies of the molecular details HTLV-1 "accessory" proteins interactions will provide important new directions for investigations of HTLV-1 and related viruses associated with lymphoproliferative diseases. Thus, the accessory proteins of HTLV-1, once thought to be dispensable for viral replication, have proven to be directly involved in viral spread in vivo and represent potential targets for therapeutic intervention against HTLV-1 infection and disease.


Assuntos
Regulação Viral da Expressão Gênica , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Proteínas Oncogênicas de Retroviridae/fisiologia , Fatores de Transcrição/fisiologia , Replicação Viral , Processamento Alternativo , Animais , Adesão Celular , Códon , DNA Complementar/metabolismo , Retículo Endoplasmático/metabolismo , Biblioteca Gênica , Complexo de Golgi/metabolismo , Humanos , Linfócitos/virologia , Potenciais da Membrana , Mitocôndrias/metabolismo , Fases de Leitura Aberta , Coelhos , Transcrição Gênica , Proteínas Virais Reguladoras e Acessórias
19.
Neurotoxicology ; 25(5): 793-802, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15288510

RESUMO

Epigallocatechin gallate (EGCG) is one of most famous compounds of green tea. EGCG suppresses apoptosis induced by oxidative radical stress through several mechanisms. This study was designed to investigate whether EGCG plays a cytoprotective role by activating phosphatidylinositol-3 kinase (PI3K)/Akt-dependent anti-apoptotic pathway and inhibiting glycogen synthase kinase-3 (GSK-3) activity in oxidative stressed N18D3 neural cells. N18D3 cells, mouse neuroblastoma X dorsal root ganglion hybrid cell line, were pre-treated with EGCG or z-VAD-fmk, non-selective caspase inhibitor used as a control substance, for 2 h. The N18D3 cells were then exposed to low concentration of H(2)O(2) (100 microM) for 30 min, and further incubated for 24 h. MTT (3,[4,5-dimethylthiazol]-2-yl) assay and trypan blue staining were used to identify cell viability. Immunoreactivity (IR) of PI3K, Akt, and GSK-3 beta were measured by Western blotting. MTT assay and trypan blue staining showed that EGCG and z-VAD-fmk significantly increased cell viability, and IR of PI3K, phospho-Akt and phospho-GSK-3 beta was significantly increased in the cells treated with EGCG, but not in z-VAD-fmk treated. These results imply that EGCG has neuroprotective effect by increasing PI3K/Akt-dependent anti-apoptotic signals.


Assuntos
Catequina/análogos & derivados , Catequina/farmacologia , Quinase 3 da Glicogênio Sintase/fisiologia , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Oncogênicas de Retroviridae/fisiologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Inibidores de Caspase , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Proteína Oncogênica v-akt , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
20.
Am J Physiol Endocrinol Metab ; 287(4): E686-95, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15165993

RESUMO

Peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha) participates in control of expression of genes involved in adaptive thermogenesis, muscle fiber type differentiation, and fuel homeostasis. The objective of the present study was to evaluate the participation of cold-induced PGC-1alpha expression in muscle fiber type-specific activity of proteins that belong to the insulin-signaling pathway. Rats were exposed to 4 degrees C for 4 days and acutely treated with insulin in the presence or absence of an antisense oligonucleotide to PGC-1alpha. Cold exposure promoted a significant increase of PGC-1alpha and uncoupling protein-3 protein expression in type I and type II fibers of gastrocnemius muscle. In addition, cold exposure led to higher glucose uptake during a hyperinsulinemic clamp, which was accompanied by higher expression and membrane localization of GLUT4 in both muscle fiber types. Cold exposure promoted significantly lower insulin-induced tyrosine phosphorylation of the insulin receptor (IR) and Ser473 phosphorylation of acute transforming retrovirus thymoma (Akt) and an insulin-independent increase of Thr172 phosphorylation of adenosine 5'-monophosphate-activated protein kinase (AMPK). Inhibition of PGC-1alpha expression in cold-exposed rats by antisense oligonucleotide treatment diminished glucose clearance rates during a hyperinsulinemic clamp and reduced expression and membrane localization of GLUT4. Reduction of PGC-1alpha expression resulted in no modification of insulin-induced tyrosine phosphorylation of the IR and Ser473 phosphorylation of Akt. Finally, reduction of PGC-1alpha resulted in lower Thr172 phosphorylation of AMPK. Thus cold-induced hyperexpression of PGC-1alpha participates in control of skeletal muscle glucose uptake through a mechanism that controls GLUT4 expression and subcellular localization independent of the IR and Akt activities but dependent on AMPK.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Glucose/metabolismo , Proteínas de Choque Térmico/biossíntese , Músculo Esquelético/metabolismo , Receptor de Insulina/fisiologia , Fatores de Transcrição/biossíntese , Animais , Antimetabólitos/farmacologia , Proteínas de Transporte/metabolismo , Temperatura Baixa , Proteína Quinase Tipo II Dependente de AMP Cíclico , Desoxiglucose/farmacologia , Transportador de Glucose Tipo 4 , Hormônios/sangue , Insulina/farmacologia , Canais Iônicos , Masculino , Mitocôndrias Musculares/efeitos dos fármacos , Mitocôndrias Musculares/metabolismo , Proteínas Mitocondriais , Proteínas de Transporte de Monossacarídeos/metabolismo , Fibras Musculares de Contração Rápida/metabolismo , Fibras Musculares de Contração Lenta/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/citologia , Oligonucleotídeos Antissenso/farmacologia , Proteína Oncogênica v-akt , Consumo de Oxigênio/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosforilação , Ratos , Ratos Wistar , Proteínas Oncogênicas de Retroviridae/fisiologia , Transdução de Sinais/fisiologia , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Proteína Desacopladora 3
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA