Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Ther ; 41(9): 1737-1746, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31311668

RESUMO

PURPOSE: Temelimab/GNbAC1 is a humanized immunoglobulin G4 monoclonal antibody antagonist of the human endogenous retrovirus W envelope protein, which is associated with multiple sclerosis (MS) pathophysiology and possibly with other autoimmune disorders. Human endogenous retrovirus W envelope protein is expressed in the central nervous system of patients with MS, and sufficient amount of temelimab must reach the target. The safety of very high dosages of temelimab should be tested to support further clinical trials in MS. METHODS: This randomized, placebo-controlled, dose-escalation study evaluated the safety and pharmacokinetic profile of temelimab in 24 healthy volunteers after a single intravenous infusion at doses of 36, 60, 85, and 110 mg/kg administered sequentially. FINDINGS: Temelimab was well tolerated, with no particular adverse drug reactions at any dose. The maximal dose of 110 mg/kg could be administered, and no antidrug antibodies were induced. After administration of 36-110 mg/kg, mean temelimab Cmax increased from 859 to 2450 µg/mL, and AUC values increased from 319,900 to 1,030,000 µg·h/mL. There was an approximate dose-proportional increase in exposure, similar to observations at lower doses. IMPLICATIONS: The favorable data in terms of safety and pharmacokinetic variables support temelimab use at high doses in future MS trials to optimally neutralize the temelimab target in the central nervous system. ClinicalTrials.gov identifier: NCT03574428.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Produtos do Gene env/antagonistas & inibidores , Imunoglobulina G , Proteínas da Gravidez/antagonistas & inibidores , Adulto , Anticorpos Monoclonais Humanizados/efeitos adversos , Método Duplo-Cego , Retrovirus Endógenos , Voluntários Saudáveis , Humanos , Infusões Intravenosas , Masculino , Esclerose Múltipla/tratamento farmacológico , Adulto Jovem
2.
Mol Diagn Ther ; 19(5): 255-65, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26376649

RESUMO

Human endogenous retroviruses (HERV) represent about 8 % of the human genome. Some of these genetic elements are expressed in pathological circumstances. A HERV protein, the multiple sclerosis-associated retrovirus (MSRV) envelope protein (MSRV-Env), is expressed in the blood and active brain lesions of multiple sclerosis (MS) patients. It possesses pro-inflammatory and myelinotoxic properties. The patterns of expression and pathogenic properties of MSRV-Env make it a relevant drug target for MS therapeutics-in particular for preventing neurodegeneration, a key component of progressive forms of MS. An immunoglobulin G4 monoclonal antibody (mAb), called GNbAC1, has been developed to neutralize this pathogenic target. After showing neutralizing effects in vitro and in mouse models of MS, GNbAC1 is now in phase II clinical development. MSRV-related biomarkers such as MSRV-Env and MSRV polymerase (MSRV-Pol) gene transcripts are overexpressed in the blood and cerebrospinal fluid of patients with MS. These biomarkers may have prognostic value for long-term MS evolution, and their transcription levels in blood decline during treatments with GNbAC1, which has also been reported in patients administered reference MS drugs such as natalizumab or interferon-ß. GNbAC1 as a new MSRV-Env-antagonist mAb could be a specific and causal treatment for MS, with a particular application for progressive forms of the disease. For possible use in companion diagnostic tests, MSRV-associated biomarkers could open the door to a personalized therapeutic approach for MS.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Retrovirus Endógenos/efeitos dos fármacos , Produtos do Gene env/antagonistas & inibidores , Esclerose Múltipla/tratamento farmacológico , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Ensaios Clínicos Fase II como Assunto , Modelos Animais de Doenças , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Produtos do Gene env/sangue , Produtos do Gene env/líquido cefalorraquidiano , Produtos do Gene env/genética , Humanos , Camundongos , Esclerose Múltipla/virologia , Medicina de Precisão , RNA Viral/sangue , RNA Viral/líquido cefalorraquidiano , RNA Viral/efeitos dos fármacos
3.
Mult Scler ; 21(7): 885-93, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25392325

RESUMO

BACKGROUND: GNbAC1 is an immunoglobulin (IgG4) humanised monoclonal antibody against multiple sclerosis-associated retrovirus (MSRV)-Env, a protein of endogenous retroviral origin, expressed in multiple sclerosis (MS) lesions, which is pro-inflammatory and inhibits oligodendrocyte precursor cell differentiation. OBJECTIVE: This is a randomised, double-blind placebo-controlled dose-escalation study followed by a six-month open-label phase to test GNbAC1 in MS patients. The primary objective was to assess GNbAC1 safety in MS patients, and the other objectives were pharmacokinetic and pharmacodynamic assessments. METHODS: Ten MS patients were randomised into two cohorts to receive a single intravenous infusion of GNbAC1/placebo at doses of 2 or 6 mg/kg. Then all patients received five infusions of GNbAC1 at 2 or 6 mg/kg at four-week intervals in an open-label setting. Safety, brain magnetic resonance imaging (MRI), pharmacokinetics, immunogenicity, cytokines and MSRV RNA expression were studied. RESULTS: All patients completed the study. GNbAC1 was well tolerated in all patients. GNbAC1 pharmacokinetics is dose-linear with mean elimination half-life of 27-37 d. Anti-GNbAC1 antibodies were not detected. Cytokine analysis did not indicate an adverse effect. MSRV-transcripts showed a decline after the start of treatment. Nine patients had stable brain lesions at MRI. CONCLUSION: The safety, pharmacokinetic profile, and pharmacodynamic responses to GNbAC1 are favourable in MS patients over a six-month treatment period.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Produtos do Gene env/antagonistas & inibidores , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/patologia , Adulto , Anticorpos Monoclonais Humanizados/farmacocinética , Encéfalo/patologia , Método Duplo-Cego , Retrovirus Endógenos , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/virologia , Reação em Cadeia da Polimerase , RNA Viral/análise
4.
Cell Signal ; 25(4): 1027-35, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23333240

RESUMO

Placental syncytiotrophoblasts formed by the fusion of cytotrophoblasts constitute the interface between maternal and fetal circulations. The syncytium, composed of a continuous layer of syncytiotrophoblasts, assumes the fetal-maternal nutrient exchange, placental barrier, and endocrine functions important for the maintenance of normal pregnancy. Syncytin-1, an endogenous retroviral gene product, mediates the fusion of cytotrophoblasts. While the fusogenic function of syncytin-1 has been well established, little is known regarding its nonfusogenic activities. This study investigates the role of syncytin-1 in trophoblast proliferation. We found that syncytin-1 knockdown significantly inhibited BeWo cell growth and DNA synthesis. Moreover, time course studies on key cell cycle regulators demonstrated an upregulation of p15 and downregulation of CDK4, E2F1, PCNA, and c-Myc, which consequently led to a reduced level of CDK1. These results, together with those from flow cytometry analysis, indicated that syncytin-1 knockdown blocked the G1/S transition phase of the cell cycle. Moreover, syncytin-1 overexpression promoted CHO cell proliferation and led to changes opposite to those observed in syncytin-1 knockdown experiments, confirming the critical role of syncytin-1 for G1/S transition. Thus, syncytin-1, through both nonfusogenic and fusogenic, functions, may co-regulate the input (proliferation) and output (fusion) of the cytotrophoblast "pool". Such co-regulation could be an efficient way to achieve the balance between these two opposing processes, which is required for syncytium homeostasis. Since decreased syncytin-1 expression has been shown to be associated with preeclamptic and hypoxic condition, insufficient replenishing of the cytotrophoblast "pool" may contribute to syncytium deficiency, a critical pathological change frequently found in preeclamptic placentas.


Assuntos
Produtos do Gene env/metabolismo , Proteínas da Gravidez/metabolismo , Animais , Proteína Quinase CDC2/metabolismo , Células CHO , Linhagem Celular , Proliferação de Células , Cricetinae , Cricetulus , Quinase 4 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Fator de Transcrição E2F1/metabolismo , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular , Produtos do Gene env/antagonistas & inibidores , Produtos do Gene env/genética , Humanos , Placenta/citologia , Placenta/metabolismo , Gravidez , Proteínas da Gravidez/antagonistas & inibidores , Proteínas da Gravidez/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transfecção , Trofoblastos/citologia , Trofoblastos/metabolismo
5.
Immunol Rev ; 250(1): 180-98, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23046130

RESUMO

Human immunodeficiency virus-1 (HIV-1) envelope protein (Env) and influenza hemagglutinin (HA) are the surface glycoproteins responsible for viral entry into host cells, the first step in the virus life cycle necessary to initiate infection. These glycoproteins exhibit a high degree of sequence variability and glycosylation, which are used as strategies to escape host immune responses. Nonetheless, antibodies with broadly neutralizing activity against these viruses have been isolated that have managed to overcome these barriers. Here, we review recent advances in the structural characterization of these antibodies with their viral antigens that defines a few sites of vulnerability on these viral spikes. These broadly neutralizing antibodies tend to focus their recognition on the sites of similar function between the two viruses: the receptor-binding site and membrane fusion machinery. However, some sites of recognition are unique to the virus neutralized, such as the dense shield of oligomannose carbohydrates on HIV-1 Env. These observations are discussed in the context of structure-based design strategies to aid in vaccine design or development of antivirals.


Assuntos
Anticorpos Neutralizantes/química , Produtos do Gene env/química , Anticorpos Anti-HIV/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Receptores Virais/antagonistas & inibidores , Vacinas contra a AIDS/química , Vacinas contra a AIDS/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Variação Antigênica , Produtos do Gene env/antagonistas & inibidores , Produtos do Gene env/imunologia , Anticorpos Anti-HIV/imunologia , Anticorpos Anti-HIV/farmacologia , HIV-1/efeitos dos fármacos , HIV-1/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/imunologia , Vacinas contra Influenza/química , Vacinas contra Influenza/imunologia , Modelos Moleculares , Receptores Virais/imunologia , Especificidade da Espécie , Homologia Estrutural de Proteína , Internalização do Vírus/efeitos dos fármacos
6.
J Natl Cancer Inst ; 104(3): 189-210, 2012 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-22247020

RESUMO

BACKGROUND: The envelope (env) protein of the human endogenous retrovirus type K (HERV-K) family is commonly expressed on the surface of breast cancer cells. We assessed whether HERV-K env is a potential target for antibody-based immunotherapy of breast cancer. METHODS: We examined the expression of HERV-K env protein in various malignant (MDA-MB-231, MCF-7, SKBR3, MDA-MB-453, T47D, and ZR-75-1) and nonmalignant (MCF-10A and MCF-10AT) human breast cell lines by immunoblot, enzyme-linked immunosorbent assay, immunofluorescence staining, and flow cytometry. Anti-HERV-K env monoclonal antibodies (mAbs; 6H5, 4D1, 4E11, 6E11, and 4E6) were used to target expression of HERV-K, and antitumor effects were assessed by quantifying growth and apoptosis of breast cancer cells in vitro, and tumor growth in vivo in mice (n = 5 per group) bearing xenograft tumors. The mechanisms responsible for 6H5 mAb-mediated effects were investigated by microarray assays, flow cytometry, immunoblot, and immunofluorescence staining. The expression of HERV-K env protein was assessed in primary breast tumors (n = 223) by immunohistochemistry. All statistical tests were two-sided. RESULTS: The expression of HERV-K env protein in malignant breast cancer cell lines was substantially higher than nonmalignant breast cells. Anti-HERV-K-specific mAbs inhibited growth and induced apoptosis of breast cancer cells in vitro. Mice treated with 6H5 mAb showed statistically significantly reduced growth of xenograft tumors compared with mice treated with control immunoglobulin (control [mIgG] vs 6H5 mAb, for tumors originating from MDA-MB-231 cells, mean size = 1448.33 vs 475.44 mm(3); difference = 972.89 mm(3), 95% CI = 470.17 to 1475.61 mm(3); P < .001). Several proteins involved in the apoptotic signaling pathways were overexpressed in vitro in 6H5 mAb-treated malignant breast cells compared with mIgG-treated control. HERV-K expression was detected in 148 (66%) of 223 primary breast tumors, and a higher rate of lymph node metastasis was associated with HERV-K-positive compared with HERV-K-negative tumors (43% vs 23%, P = .003). CONCLUSION: Monoclonal antibodies against HERV-K env protein show potential as novel immunotherapeutic agents for breast cancer therapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/virologia , Retrovirus Endógenos , Produtos do Gene env/antagonistas & inibidores , Imunoterapia/métodos , Proteínas dos Retroviridae/antagonistas & inibidores , Adulto , Idoso , Animais , Anticorpos Anti-Idiotípicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Bromodesoxiuridina/metabolismo , Carcinoma Ductal de Mama/tratamento farmacológico , Carcinoma Ductal de Mama/virologia , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Estudos de Viabilidade , Feminino , Citometria de Fluxo , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Produtos do Gene env/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Antígeno Ki-67/análise , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Gradação de Tumores , Estadiamento de Neoplasias , Projetos Piloto , Análise Serial de Proteínas , Distribuição Aleatória , Proteínas dos Retroviridae/metabolismo , Transdução de Sinais , Transplante Heterólogo , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
7.
J Virol Methods ; 173(1): 92-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21277903

RESUMO

Since the discovery of RNAi technology, several functional genomic and disease therapy studies have been conducted using this technique in the field of oncology and virology. RNAi-based antiviral therapies are being studied for the treatment of retroviruses such as HIV-1. These studies include the silencing of regulatory, infectivity and structural genes. The HTLV-1 structural genes are responsible for the synthesis of proteins involved in the entry, assembly and release of particles during viral infection. To examine the possibility of silencing HTLV-1 genes gag and env by RNA interference technology, these genes were cloned into reporter plasmids. These vectors expressed the target mRNAs fused to EGFP reporter genes. Three small interference RNAs (siRNAs) corresponding to gag and three corresponding to env were designed to analyze the effect of silencing by RNAi technology. The plasmids and siRNAs were co-transfected into HEK 293 cells. The results demonstrated that the expression of the HTLV-1 gag and env genes decreased significantly in vitro. Thus, siRNAs can be used to inhibit HTLV-1 structural genes in transformed cells, which could provide a tool for clarifying the roles of HTLV-1 structural genes, as well as a therapy for this infection.


Assuntos
Produtos do Gene env/antagonistas & inibidores , Produtos do Gene gag/antagonistas & inibidores , Inativação Gênica , Vírus Linfotrópico T Tipo 1 Humano/genética , RNA Interferente Pequeno/genética , Fusão Gênica Artificial , Produtos do Gene env/genética , Produtos do Gene gag/genética , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos
8.
Pharmazie ; 65(9): 702-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21038850

RESUMO

Adult T-cell leukemia (ATL) is a severe chemotherapy-resistant malignancy associated with prolonged infection by the human T cell-lymphotropic virus 1 (HTLV-1) retrovirus. Epidemiology studies strongly indicate that an increase in HTLV-1 virus load is an important factor during the onset of ATL. Therefore, inhibition of the growth/transmission of HTLV-1 infected cells is a promising strategy in preventing the disease. In our previous study, we revealed that arsenic trioxide (As2O3), a drug used to treat acute promyelocytic leukemia (APL), exerts an inhibitory effect on syncytium formation between HTLV-1 infected cells and HeLa cells via suppression of HTLV-1 envelope protein gp46 expression at low concentrations. In this study, we analyze the mechanism of action of As2O3 using a proteomics approach. Our results suggest that down-regulation of gp46 might be related to As2O3-induced oxidation of the 71-kDa heat shock cognate protein (HSC70) and the 78-kDa glucose-regulated protein (BiP/GRP78). We postulate that AS2O3 exerts an inhibitory effect on HTLV-1 virus transmission via down-regulation of gp46-production, which might be caused by oxidative modification of various proteins such as chaperones.


Assuntos
Arsenicais/farmacologia , Produtos do Gene env/biossíntese , Infecções por HTLV-I/metabolismo , Óxidos/farmacologia , Proteínas Oncogênicas de Retroviridae/biossíntese , Trióxido de Arsênio , Fusão Celular , Regulação para Baixo/efeitos dos fármacos , Eletroforese em Gel Bidimensional , Chaperona BiP do Retículo Endoplasmático , Géis , Produtos do Gene env/antagonistas & inibidores , Células HeLa , Humanos , Hidrólise , Imunoprecipitação , Oxirredução , Proteômica , Proteínas Oncogênicas de Retroviridae/antagonistas & inibidores , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Tripsina/química
9.
Retrovirology ; 7: 4, 2010 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-20102634

RESUMO

BACKGROUND: The gibbon ape leukemia virus (GaLV) Env protein mediates entry into a wide range of human cells and is frequently used to pseudotype retroviral vectors. However, an incompatibility exists between GaLV Env and lentiviral vectors that results in decreased steady-state levels of the mature GaLV Env in cells and prevents its incorporation into lentiviral vector particles. RESULTS: We identified the HIV-1 Vpu protein as the major cause of the depletion in GaLV Env levels that occurs when lentiviral vector components are present. This activity of Vpu targeted the mature (cleaved) form of the GaLV Env that exists within or beyond the trans-Golgi. The activity required two conserved phospho-serines in the cytoplasmic tail of Vpu that are known to recruit beta TrCP, a substrate adaptor for an SCF E3 ubiquitin ligase complex, and could be blocked by mutation of lysine 618 in the GaLV Env tail. Moreover, the Vpu-mediated decrease of GaLV Env levels was inhibited by the lysosomal inhibitor, bafilomycin A1. Interestingly, this activity of Vpu was only observed in the presence of other lentiviral vector components. CONCLUSIONS: Similar to the mechanism whereby Vpu targets BST-2/tetherin for degradation, these findings implicate beta-TrCP-mediated ubiquitination and the endo-lysosomal pathway in the degradation of the GaLV Env by lentiviral vector components. Possibly, the cytoplasmic tail of the GaLV Env contains features that mimic bona fide targets of Vpu, important to HIV-1 replication. Furthermore, the lack of effect of Vpu on GaLV Env in the absence of other HIV-1 proteins, suggests that a more complex interaction may exist between Vpu and its target proteins, with the additional involvement of one or more component(s) of the HIV-1 replication machinery.


Assuntos
Produtos do Gene env/antagonistas & inibidores , Produtos do Gene env/genética , HIV-1/genética , Proteínas do Vírus da Imunodeficiência Humana/genética , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Vírus da Leucemia do Macaco Gibão/genética , Mapeamento de Interação de Proteínas , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo , Linhagem Celular , Humanos , Proteínas Contendo Repetições de beta-Transducina/metabolismo
10.
Biol Pharm Bull ; 32(7): 1286-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19571400

RESUMO

Adult T-cell leukemia (ATL) is a severe chemotherapy-resistant malignancy associated with prolonged infection by the human T cell-lymphotropic virus 1 (HTLV-1). One approach to prevent the onset of ATL is to inhibit the growth/transmission of HTLV-1 infected cells using arsenic trioxide (As(2)O(3)). However, there are no reports on the transmission inhibitory effect of As(2)O(3). In this study, we reveal that As(2)O(3) exerts an inhibitory effect on syncytium formation between HTLV-1 infected MT-2 and HeLa cells. In addition, Western blot analysis revealed that the HTLV-1 derived envelope protein gp46 was down regulated by As(2)O(3) treatment, suggesting that As(2)O(3) may inhibit HTLV-1 virus transmission via down-regulation of gp46. These results suggest that As(2)O(3) may be a promising drug to treat refractory HTLV-1-related diseases.


Assuntos
Antineoplásicos/farmacologia , Arsenicais/farmacologia , Produtos do Gene env/antagonistas & inibidores , Células Gigantes/efeitos dos fármacos , Vírus Linfotrópico T Tipo 1 Humano/efeitos dos fármacos , Óxidos/farmacologia , Proteínas Oncogênicas de Retroviridae/antagonistas & inibidores , Linfócitos T/efeitos dos fármacos , Trióxido de Arsênio , Western Blotting , Técnicas de Cocultura , Regulação para Baixo , Produtos do Gene env/biossíntese , Células Gigantes/metabolismo , Células HeLa , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Proteínas Oncogênicas de Retroviridae/biossíntese , Linfócitos T/virologia
11.
J Mol Med (Berl) ; 85(1): 23-38, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17066266

RESUMO

Endometrial carcinomas (EnCa) predominantly represent a steroid hormone-driven tumor initiated from prestages. The human endogenous retrovirus HERV-W envelope gene Syncytin-1 was significantly increased at the mRNA and protein levels in EnCa and prestages compared to controls. Steroid hormone treatment of primary EnCa cells and cell lines induced Syncytin-1 due to a new HERV-W estrogen response element and resulted in increased proliferation. Activation of the cAMP-pathway also resulted in Syncytin-1 upregulation, but in contrast to proliferation, classic cell-cell fusions similar to placental syncytiotrophoblasts occurred. Cell-cell fusions were also histologically identified in endometrioid EnCa tumors in vivo. Clonogenic soft agar experiments showed that Syncytin-1 is also involved in anchorage-independent colony growth as well as in colony fusions depending on steroid hormones or cAMP-activation. The posttranscriptional silencing of Syncytin-1 gene expression and a concomitant functional block of induced cell proliferation and cell-cell fusion with siRNAs proved the essential role of Syncytin-1 in these cellular processes. TGF-beta1 and TGF-beta3 were identified as main regulative factors, due to the finding that steroid hormone inducible TGF-beta1 and TGF-beta3 inhibited cell-cell fusion, whereas antibody-mediated TGF-beta neutralization induced cell-cell fusions. These results showed that induced TGF-beta could override Syncytin-1-mediated cell-cell fusions. Interactions between Syncytin-1 and TGF-beta may contribute to the etiology of EnCa progression and also help to clarify the regulation of cell-cell fusions occurring in development and in other syncytial cell tumors.


Assuntos
Proliferação de Células , Neoplasias do Endométrio/patologia , Produtos do Gene env/metabolismo , Proteínas da Gravidez/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Northern Blotting , Southern Blotting , Western Blotting , Fusão Celular , Neoplasias do Endométrio/metabolismo , Feminino , Perfilação da Expressão Gênica , Produtos do Gene env/antagonistas & inibidores , Produtos do Gene env/genética , Inativação Gênica/fisiologia , Humanos , Immunoblotting , Pessoa de Meia-Idade , Proteínas da Gravidez/antagonistas & inibidores , Proteínas da Gravidez/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Tumorais Cultivadas/efeitos dos fármacos
12.
Exp Cell Res ; 312(7): 1011-20, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16427621

RESUMO

Syncytin-1 is a captive envelope glycoprotein encoded by one of human endogenous retroviruses W. It is expressed exclusively in the placental trophoblast where it participates in cell-to-cell fusion during differentiation of syncytiotrophobast. In other tissues, however, syncytin-1 expression must be kept in check because inadvertent cell fusion might be dangerous for tissue organization and integrity. We describe here an inverse correlation between CpG methylation of syncytin-1 5' long terminal repeat and its expression. Hypomethylation of the syncytin-1 5' long terminal repeat in the placenta and in the choriocarcinoma-derived cell line BeWo was detected. However, other analyzed primary cells and cell lines non-expressing syncytin-1 contain proviruses heavily methylated in this sequence. CpG methylation of syncytin-1 is resistant to the effect of the demethylating agent 5-azacytidine. The inhibitory role of CpG methylation is further confirmed by transient transfection of in-vitro-methylated syncytin-1 promoter-driven reporter construct. Altogether, we conclude that CpG methylation plays a principal role in the transcriptional suppression of syncytin-1 in non-placental tissues, and, in contrast, demethylation of the syncytin-1 promoter in trophoblast is a prerequisite for its expression and differentiation of multinucleated syncytiotrophoblast.


Assuntos
Ilhas de CpG/fisiologia , Metilação de DNA , Produtos do Gene env/antagonistas & inibidores , Produtos do Gene env/genética , Proteínas da Gravidez/antagonistas & inibidores , Proteínas da Gravidez/genética , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Linhagem Celular , Regulação para Baixo/genética , Produtos do Gene env/biossíntese , Células HeLa , Humanos , Placenta/metabolismo , Proteínas da Gravidez/biossíntese , Regiões Promotoras Genéticas , Sequências Repetidas Terminais , Fatores de Transcrição/biossíntese , Trofoblastos/metabolismo
13.
J Immunol ; 173(7): 4618-26, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15383596

RESUMO

During HIV-1 infection or vaccination, HIV-1 envelope spikes elicit Ab responses. Neutralizing Abs block viral entry by recognizing epitopes on spikes critical for their interaction with receptor, coreceptors or fusion. In contrast, nonneutralizing Abs fail to do so because they recognize epitopes either buried or exposed but not critical for viral entry. Previously, we produced a high-affinity human mAb against the cluster II determinant of gp41. This Ab or its recombinant Fab and single-chain Fv have been repeatedly shown to bind to HIV-1 gp160 or gp41, but fail to block viral entry. We report that, surprisingly, expression of this nonneutralizing anti-HIV-1 gp41 single-chain Fv on the surface of human CD4 T cells markedly inhibits HIV-1 replication and cell-cell fusion. The inhibition targets the HIV-1 envelope at the level of viral entry, regardless of HIV-1 tropism. Although this bona fide nonneutralizing Ab does not neutralize HIV-1 entry when produced as a soluble protein, it acts as a neutralizing Ab when expressed on the cell surface. Expressing Abs on the surface of HIV-1-susceptible cells can be a new way to fight HIV-1.


Assuntos
Antivirais/fisiologia , Anticorpos Anti-HIV/fisiologia , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , HIV-1/imunologia , HIV-1/patogenicidade , Receptores de Antígenos de Linfócitos B/fisiologia , Antivirais/biossíntese , Linhagem Celular Tumoral , Membrana Celular/imunologia , Membrana Celular/virologia , Suscetibilidade a Doenças/imunologia , Produtos do Gene env/antagonistas & inibidores , Vetores Genéticos , Células Gigantes/imunologia , Células Gigantes/virologia , Proteínas de Fluorescência Verde , Anticorpos Anti-HIV/biossíntese , Inibidores da Fusão de HIV/imunologia , HIV-1/fisiologia , Humanos , Fragmentos de Imunoglobulinas/biossíntese , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/fisiologia , Proteínas Luminescentes/genética , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Testes de Neutralização , Receptores de Antígenos de Linfócitos B/biossíntese , Transdução Genética , Transgenes , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/imunologia , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/genética , Replicação Viral/imunologia
14.
J Virol ; 78(13): 7079-88, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15194784

RESUMO

We have constructed a human immunodeficiency virus type 1 (HIV-1)-based lentiviral vector expressing a 937-base antisense sequence against the HIV-1 envelope gene. Transduction of CD4(+) T lymphocytes with this vector results in expression of the therapeutic antisense sequence and subsequent inhibition of productive HIV-1 replication. In this report, we examined the effect of antisense-mediated suppression on the potential development of virus escape mutants using a permissive T-cell line cultured under conditions that over serial passages specifically allowed for generation and amplification of mutants selected for by antisense pressure. In the resulting virus clones, we found a significant increase in the number of deletions at the envelope target region (91% compared to 27.5% in wild-type HIV). Deletions were most often greater than 1 kb in length. These data demonstrate for the first time that during antisense-mediated suppression of HIV, mutants develop as a direct result of selective pressure on the HIV genomic RNA. Interestingly, in clones where deletions were not observed, there was a high rate of A-G transitions in mutants at the antisense target region but not outside this region, which is consistent with those mutations that are predicted as a result of antisense-mediated modification of double-stranded RNA by the enzyme double-stranded RNA-specific adenosine deaminase. These clones were not found to be escape mutants, as their replicative ability was severely attenuated, and they did not replicate in the presence of vector.


Assuntos
Vetores Genéticos , HIV-1/fisiologia , Lentivirus/genética , RNA Antissenso/farmacologia , Replicação Viral/efeitos dos fármacos , Sequência de Bases , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Células Cultivadas , Produtos do Gene env/antagonistas & inibidores , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , HIV-1/genética , Humanos , Dados de Sequência Molecular , Mutação , RNA Antissenso/metabolismo , Deleção de Sequência , Transdução Genética
16.
Nat Rev Microbiol ; 1(1): 25-34, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15040177

RESUMO

The worldwide infection rate for HIV-1 is estimated to be 14,000 per day, but only now, more than 20 years into the epidemic, are the immediate events between exposure to infectious virus and the establishment of infection becoming clear. Defining the mechanisms of HIV-1 transmission, the target cells involved and how the virus attaches to and fuses with these cells, could reveal ways to block the sexual spread of the virus. In this review, we will discuss how our increasing knowledge of the ways in which HIV-1 is transmitted is shaping the development of new, more sophisticated intervention strategies based on the application of vaginal or rectal microbicides.


Assuntos
Transmissão de Doença Infecciosa/prevenção & controle , Inibidores da Fusão de HIV/administração & dosagem , Infecções por HIV/prevenção & controle , Infecções por HIV/transmissão , HIV-1 , Administração Intravaginal , Administração Retal , Animais , Fármacos Anti-HIV/administração & dosagem , Anti-Infecciosos/administração & dosagem , Sistemas de Liberação de Medicamentos , Células Epiteliais/virologia , Feminino , Produtos do Gene env/antagonistas & inibidores , Humanos , Masculino , Reto/virologia , Vagina/virologia
17.
Hum Gene Ther ; 13(9): 1027-37, 2002 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-12067436

RESUMO

We have developed optimized versions of a conditionally replicating human immunodeficiency virus type 1 (HIV-1)-based lentiviral vector for gene therapy of HIV-1 infection. These vectors target HIV-1 RNAs containing sequences of the envelope gene by expressing a 1-kb fragment of the HIV-1 Tat/Rev intron in the antisense orientation. Expression of the envelope antisense gene (envAS) was evaluated under the control of different internal promoters such as the human phosphoglycerate kinase (PGK) promoter, the human EF1-alpha promoter, and the U3 region of the SL3 murine leukemia virus. The U3-SL3 promoter transactivates transcription from the vector HIV-1 LTR and drives higher expression levels of envAS-containing RNAs than other promoters in T-cell lines. The effect of other vector structural features was also evaluated. We found that the central polypurine tract and central termination sequence (cPPT) produce a small increase in vector infectivity of 2-fold to 3-fold and results in a 10-fold higher inhibition of wild-type viral replication in challenge experiments. The woodchuck hepatitis posttranscriptional regulatory element (WPRE) does not increase the cytoplasmic levels of envAS mRNA in T-cell lines. We observed that SupT1 and primary CD4(+) T cells transduced with these vectors showed high inhibition of HIV-1 replication, suppression of syncitium formation, and increased cell viability when infected with several HIV-1 laboratory strains. Our results suggest that higher vector copy number and increased levels of envAS RNA expression contribute to block replication of divergent strains of HIV-1.


Assuntos
Produtos do Gene env/genética , Vetores Genéticos , HIV-1/genética , RNA Antissenso , Replicação Viral/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Produtos do Gene env/antagonistas & inibidores , Terapia Genética , Infecções por HIV/terapia , Infecções por HIV/virologia , HIV-1/fisiologia , Células HeLa , Vírus da Hepatite B da Marmota/genética , Humanos , Regiões Promotoras Genéticas , Processamento Pós-Transcricional do RNA/genética
19.
Virus Res ; 50(2): 119-27, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9282777

RESUMO

Calmodulin, an EF-hand protein, inhibited the fusion between CD4+ human cells and cells stably expressing HIV-1 envelope proteins. Fusion was also inhibited when HIV-1, HIV-2 or SIV envelope glycoproteins were expressed by vaccinia virus (VV) recombinants, but calmodulin did not inhibit syncytia formation induced by measles virus glycoproteins. Calmodulin also inhibited fusion induced by vPE17, a VV-recombinant expressing a truncated form of HIV-1gp160 which lacks the two known calmodulin-binding sites located in the cytoplasmic domain of gp41. The inhibitory activity was specific to calmodulin among the EF-hand proteins. These observations may be important in understanding the mechanism of retroviral envelope glycoprotein-mediated cell fusion. Several possible mechanisms of action are discussed.


Assuntos
Fármacos Anti-HIV/farmacologia , Calmodulina/farmacologia , Produtos do Gene env/antagonistas & inibidores , Produtos do Gene env/fisiologia , Lentivirus de Primatas/efeitos dos fármacos , Fusão Celular/efeitos dos fármacos , Linhagem Celular , Proteína gp41 do Envelope de HIV/efeitos dos fármacos , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , HIV-2/efeitos dos fármacos , HIV-2/fisiologia , Humanos , Lentivirus de Primatas/fisiologia , Vírus do Sarampo/efeitos dos fármacos , Vírus do Sarampo/fisiologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vírus da Imunodeficiência Símia/fisiologia
20.
Nat Biotechnol ; 15(4): 343-8, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9094135

RESUMO

The identification of fusin and other chemokine receptors as coreceptors for HIV-1 has renewed the interest in agents that may prevent viral entry. Polyanionic compounds such as dextran sulfate, curdian sulfate, and suramin act on the V3 loop of the viral envelope and may prevent its interaction with fusin. These agents show activity against a wide range of HIV-1 strains, but have undesirable circulating half-life, bioavailability, and toxicity. We have developed a small molecule inhibitor of HIV-1 that has several advantages over these other agents. FP-21399 is a novel compound of the bis(disulfonaphthalene) dimethoxybenzene class that blocks entry of HIV into CD4+ cells and blocks fusion of infected and noninfected CD4+ cells. This compound only weakly inhibits binding of CD4 and gp120, at concentrations much greater than are required to block viral entry. Furthermore, FP-21399 can block the interaction between gp120 and antibodies directed against the V3 loop, but does not block binding of antibodies directed against the V4 loop. Animal studies demonstrate that FP-21399 is concentrated in lymph nodes, making it a promising compound for anti-HIV therapy. In SCID mice reconstituted with human immune cells, maintenance of HIV-1 infection was blocked by a 5-day treatment with low doses of FP-21399, suggesting that lymph node accumulation may contribute to antiviral activity. Finally, attempts to generate drug-resistant virus in cell culture resulted in only weakly resistant variants with IC90 values that are much lower than concentrations of FP-21399 found in lymph nodes.


Assuntos
Fármacos Anti-HIV/farmacologia , Clorobenzenos/farmacologia , HIV-1/efeitos dos fármacos , Fusão de Membrana/efeitos dos fármacos , Naftalenos/farmacologia , Animais , Biotecnologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Clorobenzenos/farmacocinética , Cães , Resistência Microbiana a Medicamentos/genética , Produtos do Gene env/antagonistas & inibidores , Produtos do Gene env/fisiologia , Variação Genética , Proteína gp120 do Envelope de HIV/efeitos dos fármacos , Proteína gp120 do Envelope de HIV/fisiologia , Infecções por HIV/prevenção & controle , HIV-1/genética , HIV-1/fisiologia , Humanos , Linfonodos/metabolismo , Masculino , Camundongos , Camundongos SCID , Naftalenos/farmacocinética , Fragmentos de Peptídeos/efeitos dos fármacos , Fragmentos de Peptídeos/fisiologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA