Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Hematol ; 88: 15-27, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32721504

RESUMO

Thrombopoietin (TPO), through activation of its cognate receptor Mpl, is the major regulator of platelet production. However, residual platelets observed in TPO- and Mpl-loss-of-function (LOF) mice suggest the existence of an additional factor to TPO in platelet production. As erythropoietin (EPO) exhibited both in vitro megakaryocytic potential, in association with other early-acting cytokines, and in vivo platelet activation activity, we sought to investigate its role in this setting. Here, we used multiple LOF models to decipher the reciprocal role of EPO and TPO in the regulation of platelet production in TPO-LOF and Mpl-LOF mice and of platelet size heterogeneity in wild-type mice. We first identified EPO as the major thrombopoietic factor in the absence of the TPO-Mpl pathway. Based on the study of several mouse models we found that the EPO-EPO receptor pathway acts on late-stage megakaryopoiesis and is responsible for large-sized platelet production, while the TPO-Mpl pathway promotes small-sized platelet production. On the basis of our data, EPO might be used for thrombocytopenia supportive therapy in congenital amegakaryocytopoiesis. Furthermore, as a distribution skewed toward large platelets is an independent risk factor and a poor prognosis indicator in atherothrombosis, the characterization of EPO's role in the production of large-sized platelets, if confirmed in humans, may open new perspectives in the understanding of the role of EPO-induced platelets in atherothrombosis.


Assuntos
Plaquetas/metabolismo , Eritropoetina/metabolismo , Megacariócitos/microbiologia , Trombopoese , Trombopoetina/metabolismo , Animais , Eritropoetina/genética , Feminino , Camundongos , Camundongos Knockout , Trombopoetina/genética
3.
J Virol ; 82(16): 8022-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18524827

RESUMO

Immunization with recombinant proteins may provide a safer alternative to live vaccinia virus for prophylaxis of poxvirus infections. Although antibody protects against vaccinia virus infection, the mechanism is not understood and the selection of immunogens is daunting as there are dozens of surface proteins and two infectious forms known as the mature virion (MV) and the enveloped virion (EV). Our previous studies showed that mice immunized with soluble forms of EV membrane proteins A33 and B5 and MV membrane protein L1 or passively immunized with antibodies to these proteins survived an intranasal challenge with vaccinia virus. The present study compared MV protein A27, which has a role in virus attachment to glycosaminoglycans on the cell surface, to L1 with respect to immunogenicity and protection. Although mice developed similar levels of neutralizing antibody after immunizations with A27 or L1, A27-immunized mice exhibited more severe disease upon an intranasal challenge with vaccinia virus. In addition, mice immunized with A27 and A33 were not as well protected as mice receiving L1 and A33. Polyclonal rabbit anti-A27 and anti-L1 IgG had equivalent MV-neutralizing activities when measured by the prevention of infection of human or mouse cells or cells deficient in glycosaminoglycans or by adding antibody prior to or after virus adsorption. Nevertheless, the passive administration of antibody to A27 was poorly protective compared to the antibody to L1. These studies raise questions regarding the basis for antibody protection against poxvirus disease and highlight the importance of animal models for the early evaluation of vaccine candidates.


Assuntos
Vaccinia virus/metabolismo , Vacinas Virais/química , Administração Intranasal , Animais , Proteínas de Transporte/metabolismo , Feminino , Glicosaminoglicanos/metabolismo , Células HeLa , Humanos , Cinética , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Vacinas Sintéticas/química , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo
4.
J Virol ; 81(15): 8131-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17522205

RESUMO

Immunization against smallpox (variola virus) with Dryvax, a live vaccinia virus (VV), was effective, but now safety is a major concern. To overcome this issue, subunit vaccines composed of VV envelope proteins from both forms of infectious virions, including the extracellular enveloped virion (EV) protein B5, are being developed. However, since B5 has 23 amino acid differences compared with its B6 variola virus homologue, B6 might be a better choice for such a strategy. Therefore, we compared the properties of both proteins using a panel of monoclonal antibodies (MAbs) to B5 that we had previously characterized and grouped according to structural and functional properties. The B6 gene was obtained from the Centers for Disease Control and Prevention, and the ectodomain was cloned and expressed in baculovirus as previously done with B5, allowing us to compare the antigenic properties of the proteins. Polyclonal antibodies to B5 or B6 cross-reacted with the heterologous protein, and 16 of 26 anti-B5 MAbs cross-reacted with B6. Importantly, 10 anti-B5 MAbs did not cross-react with B6. Of these, three have important anti-VV biologic properties, including their ability to neutralize EV infectivity and block comet formation. Here, we found that one of these three MAbs protected mice from a lethal VV challenge by passive immunization. Thus, epitopes that are present on B5 but not on B6 would generate an antibody response that would not recognize B6. Assuming that B6 contains similar variola virus-specific epitopes, our data suggest that a subunit vaccine using the variola virus homologues might exhibit improved protective efficacy against smallpox.


Assuntos
Glicoproteínas/imunologia , Vacina Antivariólica/imunologia , Vaccinia virus/imunologia , Vírus da Varíola/imunologia , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Linhagem Celular , Ensaio Cometa , Glicoproteínas/química , Glicoproteínas/genética , Camundongos , Dados de Sequência Molecular , Testes de Neutralização , Coelhos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
5.
J Immunol ; 178(10): 6374-86, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17475867

RESUMO

Vaccination with replication-competent vaccinia protects against heterologous orthopoxvirus challenge. CD4 T cells have essential roles helping functionally important Ab and CD8 antiviral responses, and contribute to the durability of vaccinia-specific memory. Little is known about the specificity, diversity, or dominance hierarchy of orthopoxvirus-specific CD4 T cell responses. We interrogated vaccinia-reactive CD4 in vitro T cell lines with vaccinia protein fragments expressed from an unbiased genomic library, and also with a panel of membrane proteins. CD4 T cells from three primary vaccinees reacted with 44 separate antigenic regions in 35 vaccinia proteins, recognizing 8 to 20 proteins per person. The integrated responses to the Ags that we defined accounted for 49 to 81% of the CD4 reactivity to whole vaccinia Ag. Individual dominant Ags drove up to 30% of the total response. The gene F11L-encoded protein was immunodominant in two of three subjects and is fragmented in a replication-incompetent vaccine candidate. The presence of protein in virions was strongly associated with CD4 antigenicity. These findings are consistent with models in which exogenous Ag drives CD4 immunodominance, and provides tools to investigate the relationship between Ab and CD4 T cell specificity for complex pathogens.


Assuntos
Anticorpos Antivirais/biossíntese , Diversidade de Anticorpos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Epitopos de Linfócito T/imunologia , Epitopos Imunodominantes/imunologia , Vaccinia virus/imunologia , Replicação Viral/imunologia , Adulto , Sequência de Aminoácidos , Apresentação de Antígeno/imunologia , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Células Clonais , Epitopos de Linfócito T/metabolismo , Humanos , Epitopos Imunodominantes/metabolismo , Dados de Sequência Molecular , Vaccinia virus/fisiologia
6.
Vaccine ; 25(7): 1214-24, 2007 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-17098336

RESUMO

The heightened concern about the intentional release of variola virus has led to the need to develop safer smallpox vaccines. While subunit vaccine strategies are safer than live virus vaccines, subunit vaccines have been hampered by the need for multiple boosts to confer optimal protection. Here we developed a protein-based subunit vaccine strategy that provides rapid protection in mouse models of orthopoxvirus infections after a prime and single boost. Mice vaccinated with vaccinia virus envelope proteins from the mature virus (MV) and extracellular virus (EV) adjuvanted with CpG ODN and alum were protected from lethal intranasal challenge with vaccinia virus and the mouse-specific ectromelia virus. Organs from mice vaccinated with three proteins (A33, B5 and L1) and then sacrificed after challenge contained significantly lower titers of virus when compared to control groups of mice that were not vaccinated or that received sub-optimal formulations of the vaccine. Sera from groups of mice obtained prior to challenge had neutralizing activity against the MV and also inhibited comet formation indicating anti-EV activity. Long-term partial protection was also seen in mice challenged with vaccinia virus 6 months after initial vaccinations. Thus, this work represents a step toward the development of a practical subunit smallpox vaccine.


Assuntos
Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/imunologia , Ectromelia Infecciosa/prevenção & controle , Vacina Antivariólica/imunologia , Vaccinia virus/imunologia , Vacínia/imunologia , Vacínia/prevenção & controle , Proteínas Virais/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Química Farmacêutica , Ensaio Cometa , Feminino , Esquemas de Imunização , Imunização Secundária , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sobrevida , Proteínas Virais/biossíntese , Proteínas Virais/genética , Redução de Peso/efeitos dos fármacos
7.
Virology ; 341(1): 59-71, 2005 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-16083934

RESUMO

We reported that immunization with recombinant proteins derived from vaccinia virus (VV) particles could provide protection against infection. Here we describe the physical and antigenic properties of the L1R membrane protein. The recombinant protein (L1R(185t)) was secreted as a monomer and correct folding was suggested by the presence of three intramolecular disulfide bonds and binding to conformation-specific monoclonal antibodies (MAbs). Furthermore, anti-L1R(185t) rabbit antisera exhibited potent virus-neutralizing activity against the IMV form of VV. We raised six MAbs against L1R(185t). Three recognized linear epitopes (residues 118--128) and neutralized IMV infectivity. These MAbs blocked binding of each other to L1R(185t) but failed to block binding of two previously described neutralizing anti-L1R MAbs, 7D11 and 2D5. The latter two antibodies blocked each other in binding L1R(185t). Thus, two antigenic sites on L1R overlap functional domains and based on recent structural studies these are found in accessible regions of the IMV L1R protein.


Assuntos
Genes Virais , Vaccinia virus/genética , Vaccinia virus/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Anticorpos Antivirais , Antígenos Virais/genética , Sequência de Bases , DNA Viral/genética , Epitopos/genética , Glicosilação , Técnicas In Vitro , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Testes de Neutralização , Coelhos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vaccinia virus/patogenicidade , Proteínas do Envelope Viral/imunologia
8.
J Virol ; 79(10): 6260-71, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15858010

RESUMO

Vaccinia extracellular enveloped virus (EEV) is critical for cell-to-cell and long-range virus spread both in vitro and in vivo. The B5R gene encodes an EEV-specific type I membrane protein that is essential for efficient EEV formation. The majority of the B5R ectodomain consists of four domains with homology to short consensus repeat domains followed by a stalk. Previous studies have shown that polyclonal antibodies raised against the B5R ectodomain inhibit EEV infection. In this study, our goal was to elucidate the antigenic structure of B5R and relate this to its function. To do this, we produced multimilligram quantities of vaccinia virus B5R as a soluble protein [B5R(275t)] using a baculovirus expression system. We then selected and characterized a panel of 26 monoclonal antibodies (MAbs) that recognize B5R(275t). Five of these MAbs neutralized EEV and inhibited comet formation. Two other MAbs were able only to neutralize EEV, while five others were able only to inhibit comet formation. This suggests that the EEV neutralization and comet inhibition assays measure different viral functions and that at least two different antigenic sites on B5R are important for these activities. We further characterized the MAbs and the antigenic structure of B5R(275t) by peptide mapping and by reciprocal MAb blocking studies using biosensor analysis. The epitopes recognized by neutralizing MAbs were localized to SCR1-SCR2 and/or the stalk of B5R(275t). Furthermore, the peptide and blocking data support the concept that SCR1 and the stalk may be in juxtaposition and may be part of the same functional domain.


Assuntos
Mapeamento de Epitopos , Glicoproteínas de Membrana/imunologia , Vaccinia virus/imunologia , Proteínas do Envelope Viral/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos , Antígenos Virais/biossíntese , Antígenos Virais/química , Antígenos Virais/imunologia , Baculoviridae/metabolismo , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/química , Testes de Neutralização , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/imunologia , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/química
9.
Nucleic Acids Res ; 31(2): 734-42, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12527783

RESUMO

Initiation of protein synthesis on the hepatitis C virus (HCV) mRNA involves a structured element corresponding to the 5' untranslated region and constituting an internal ribosome entry site (IRES). The domain IIId of the HCV IRES, an imperfect RNA hairpin extending from nucleotides 253 to 279 of the viral mRNA, has been shown to be essential for translation and for the binding of the 40S ribosomal subunit. We investigated the properties of a series of antisense 2'-O-methyloligoribonucleotides targeted to various portions of the domain IIId. Several oligomers, 14-17 nt in length, selectively inhibited in vitro translation of a bicistronic RNA construct in rabbit reticulocyte lysate with IC(50)s <10 nM. The effect was restricted to the second cistron (the Renilla luciferase) located downstream of the HCV IRES; no effect was observed on the expression of the first cistron (the firefly luciferase) which was translated in a cap-dependent manner. Moreover, antisense 2'-O-methyloligoribonucleotides specifically competed with the 40S ribosomal subunit for binding to the IRES RNA in a filter- retention assay. The antisense efficiency of the oligonucleotides was nicely correlated to their affinity for the IIId subdomain and to their ability to displace 40S ribosomal subunit, making this process a likely explanation for in vitro inhibition of HCV-IRES-dependent translation.


Assuntos
Hepacivirus/genética , Oligonucleotídeos Antissenso/genética , Biossíntese de Proteínas/genética , Ribossomos/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Ligação Competitiva , Sistema Livre de Células , Relação Dose-Resposta a Droga , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Luciferases/genética , Luciferases/metabolismo , Oligonucleotídeos Antissenso/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Plasmídeos/genética , Biossíntese de Proteínas/efeitos dos fármacos , RNA/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Células Tumorais Cultivadas
10.
Biochemistry ; 41(18): 5883-93, 2002 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-11980492

RESUMO

We performed in vitro selection of oligoribonucleotides in order to identify high-affinity motifs recognizing RNA hairpins located at the 3' end (SL1) and at the 5' end (domain IV of the internal ribosome entry site) of the hepatitis C virus mRNA. We selected aptamers constituted by an internal loop complementary to the SL1 apical loop, flanked by G-C-rich double-stranded regions, able to form complexes with a K(d) of 70 nM, at 37 degrees C under ionic conditions close to intracellular ones. The complex involves selective apical loop (SL1)-internal loop (aptamer) interactions. Similar structurally organized aptamers were independently identified against domain IV and were shown to also give rise to such complexes. Apical loop-internal loop interaction could constitute a new recognition motif allowing specific intra- or intermolecular RNA-RNA association.


Assuntos
Hepacivirus/genética , Conformação de Ácido Nucleico , RNA Viral/química , RNA Viral/metabolismo , Sequência de Bases , Ensaio de Desvio de Mobilidade Eletroforética , Dados de Sequência Molecular , Oligorribonucleotídeos/química , Oligorribonucleotídeos/genética , Oligorribonucleotídeos/metabolismo , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Líder para Processamento/química , RNA Líder para Processamento/genética , RNA Líder para Processamento/metabolismo , RNA Viral/genética , Ribonucleases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...