Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 285(31): 24290-8, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20525690

RESUMO

Human immunodeficiency virus, type 1 (HIV-1) envelope glycoprotein (gp120/gp41) plays a critical role in virus infection and pathogenesis. Three of the six monoclonal antibodies considered to have broadly neutralizing activities (2F5, 4E10, and Z13e1) bind to the membrane-proximal external region (MPER) of gp41. This makes the MPER a desirable template for developing immunogens that can elicit antibodies with properties similar to these monoclonal antibodies, with a long term goal of developing antigens that could serve as novel HIV vaccines. In order to provide a structural basis for rational antigen design, an MPER construct, HR1-54Q, was generated for x-ray crystallographic and x-ray footprinting studies to provide both high resolution atomic coordinates and verification of the solution state of the antigen, respectively. The crystal structure of HR1-54Q reveals a trimeric, coiled-coil six-helical bundle, which probably represents a postfusion form of gp41. The MPER portion extends from HR2 in continuation of a slightly bent long helix and is relatively flexible. The structures observed for the 2F5 and 4E10 epitopes agree well with existing structural data, and enzyme-linked immunosorbent assays indicate that the antigen binds well to antibodies that recognize the above epitopes. Hydroxyl radical-mediated protein footprinting of the antigen in solution reveals specifically protected and accessible regions consistent with the predictions based on the trimeric structure from the crystallographic data. Overall, the HR1-54Q antigen, as characterized by crystallography and footprinting, represents a postfusion, trimeric form of HIV gp41, and its structure provides a rational basis for gp41 antigen design suitable for HIV vaccine development.


Assuntos
Membrana Celular/metabolismo , Proteína gp41 do Envelope de HIV/química , HIV-1/metabolismo , Vacinas contra a AIDS/química , Sequência de Aminoácidos , Antígenos/química , Cristalografia por Raios X/métodos , Desenho de Fármacos , Ensaio de Imunoadsorção Enzimática/métodos , Epitopos/química , Radical Hidroxila , Espectrometria de Massas/métodos , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica
2.
J Virol ; 82(19): 9445-57, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18632854

RESUMO

Human immunodeficiency virus type 1 (HIV-1)-infected T cells form a virological synapse with noninfected CD4(+) T cells in order to efficiently transfer HIV-1 virions from cell to cell. The virological synapse is a specialized cellular junction that is similar in some respects to the immunological synapse involved in T-cell activation and effector functions mediated by the T-cell antigen receptor. The immunological synapse stops T-cell migration to allow a sustained interaction between T-cells and antigen-presenting cells. Here, we have asked whether HIV-1 envelope gp120 presented on a surface to mimic an HIV-1-infected cell also delivers a stop signal and if this is sufficient to induce a virological synapse. We demonstrate that HIV-1 gp120-presenting surfaces arrested the migration of primary activated CD4 T cells that occurs spontaneously in the presence of ICAM-1 and induced the formation of a virological synapse, which was characterized by segregated supramolecular structures with a central cluster of envelope surrounded by a ring of ICAM-1. The virological synapse was formed transiently, with the initiation of migration within 30 min. Thus, HIV-1 gp120-presenting surfaces induce a transient stop signal and supramolecular segregation in noninfected CD4(+) T cells.


Assuntos
Linfócitos T CD4-Positivos/virologia , Proteína gp120 do Envelope de HIV/fisiologia , HIV-1/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Adesão Celular , Movimento Celular , Regulação Viral da Expressão Gênica , Proteína gp120 do Envelope de HIV/metabolismo , Humanos , Processamento de Imagem Assistida por Computador , Sistema Imunitário , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos Mononucleares/virologia , Bicamadas Lipídicas/química , Camundongos , Transdução de Sinais
3.
Virology ; 372(2): 442-56, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18068750

RESUMO

Human immunodeficiency virus type 1 (HIV-1) transmembrane glycoprotein gp41 is targeted by broadly-reactive neutralizing antibodies 2F5 and 4E10, making it an attractive target for vaccine development. To better assess immunogenic properties of gp41, we generated five soluble glutathione S-transferase fusion proteins encompassing C-terminal 30, 64, 100, 142, or 172 (full-length) amino acids of gp41 ectodomain from M group consensus envelope sequence. Antibody responses in HIV-1-infected patients were evaluated using these proteins and overlapping peptides. We found (i) antibody responses against different regions of gp41 varied tremendously among individual patients, (ii) patients with stronger antibody responses against membrane-proximal external region exhibit broader and more potent neutralizing activity, and (iii) several patients mounted antibodies against epitopes that are near, or overlap with, those targeted by 2F5 or 4E10. These soluble gp41 fusion proteins could be an important source of antigens for future vaccine development efforts.


Assuntos
Sequência Consenso , Anticorpos Anti-HIV/imunologia , Antígenos HIV/imunologia , Proteína gp41 do Envelope de HIV/imunologia , Infecções por HIV/imunologia , Proteínas Recombinantes de Fusão/imunologia , Sequência de Aminoácidos , Epitopos , Regulação Viral da Expressão Gênica , Antígenos HIV/química , Proteína gp41 do Envelope de HIV/química , Infecções por HIV/virologia , Humanos , Proteínas Recombinantes de Fusão/química , Solubilidade
4.
J Virol ; 81(21): 12029-39, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17715238

RESUMO

Severe acute respiratory syndrome (SARS) is caused by a newly emerged coronavirus (CoV) designated SARS-CoV. The virus utilizes angiotensin-converting enzyme 2 (ACE2) as the primary receptor. Although the idea is less clear and somewhat controversial, SARS-CoV is thought to use C-type lectins DC-SIGN and/or L-SIGN (collectively referred to as DC/L-SIGN) as alternative receptors or as enhancer factors that facilitate ACE2-mediated virus infection. In this study, the function of DC/L-SIGN in SARS-CoV infection was examined in detail. The results of our study clearly demonstrate that both proteins serve as receptors independently of ACE2 and that there is a minimal level of synergy between DC/L-SIGN and ACE2. As expected, glycans on spike (S) glycoprotein are important for DC/L-SIGN-mediated virus infection. Site-directed mutagenesis analyses have identified seven glycosylation sites on the S protein critical for DC/L-SIGN-mediated virus entry. They include asparagine residues at amino acid positions 109, 118, 119, 158, 227, 589, and 699, which are distinct from residues of the ACE2-binding domain (amino acids 318 to 510). Amino acid sequence analyses of S proteins encoded by viruses isolated from animals and humans suggest that glycosylation sites N227 and N699 have facilitated zoonotic transmission.


Assuntos
Asparagina/química , Moléculas de Adesão Celular/química , Lectinas Tipo C/química , Receptores de Superfície Celular/química , Síndrome Respiratória Aguda Grave/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Glicosilação , Células HeLa , Humanos , Lectinas/química , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Homologia de Sequência de Aminoácidos , Células Vero
5.
Virology ; 350(1): 15-25, 2006 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-16510163

RESUMO

Severe acute respiratory syndrome (SARS) is caused by a novel coronavirus, SARS-CoV. Virus entry into cells is mediated through interactions between spike (S) glycoprotein and angiotensin-converting enzyme 2 (ACE2). Alanine scanning mutagenesis analysis was performed to identify determinants on ACE2 critical for SARS-CoV infection. Results indicated that charged amino acids between residues 22 and 57 were important, K26 and D30, in particular. Peptides representing various regions of ACE2 critical for virus infection were chemically synthesized and evaluated for antiviral activity. Two peptides (a.a. 22-44 and 22-57) exhibited a modest antiviral activity with IC50 of about 50 microM and 6 microM, respectively. One peptide comprised of two discontinuous segments of ACE2 (a.a. 22-44 and 351-357) artificially linked together by glycine, exhibited a potent antiviral activity with IC50 of about 0.1 microM. This novel peptide is a promising candidate as a therapeutic agent against this deadly emerging pathogen.


Assuntos
Antivirais/farmacologia , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Sequência de Aminoácidos , Enzima de Conversão de Angiotensina 2 , Animais , Chlorocebus aethiops , Células HeLa , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Mutagênese Sítio-Dirigida , Peptidil Dipeptidase A/genética , Conformação Proteica , Glicoproteína da Espícula de Coronavírus , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
6.
J Virol ; 80(8): 4079-87, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16571824

RESUMO

Induction of mucosal immunity may be important for preventing SARS-CoV infections. For safe and effective delivery of viral antigens to the mucosal immune system, we have developed a novel surface antigen display system for lactic acid bacteria using the poly-gamma-glutamic acid synthetase A protein (PgsA) of Bacillus subtilis as an anchoring matrix. Recombinant fusion proteins comprised of PgsA and the Spike (S) protein segments SA (residues 2 to 114) and SB (residues 264 to 596) were stably expressed in Lactobacillus casei. Surface localization of the fusion protein was verified by cellular fractionation analyses, immunofluorescence microscopy, and flow cytometry. Oral and nasal inoculations of recombinant L. casei into mice resulted in high levels of serum immunoglobulin G (IgG) and mucosal IgA, as demonstrated by enzyme-linked immunosorbent assays using S protein peptides. More importantly, these antibodies exhibited potent neutralizing activities against severe acute respiratory syndrome (SARS) pseudoviruses. Orally immunized mice mounted a greater neutralizing-antibody response than those immunized intranasally. Three new neutralizing epitopes were identified on the S protein using a peptide neutralization interference assay (residues 291 to 308, 520 to 529, and 564 to 581). These results indicate that mucosal immunization with recombinant L. casei expressing SARS-associated coronavirus S protein on its surface provides an effective means for eliciting protective immune response against the virus.


Assuntos
Anticorpos Antivirais/sangue , Lacticaseibacillus casei/genética , Glicoproteínas de Membrana/imunologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Sequência de Aminoácidos , Animais , Imunidade nas Mucosas , Imunização , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Testes de Neutralização , Glicoproteína da Espícula de Coronavírus
7.
Virology ; 326(1): 140-9, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15262502

RESUMO

The etiological agent of severe acute respiratory syndrome (SARS) has been identified as a novel coronavirus SARS-CoV. Similar to other coronaviruses, spike (S)-glycoprotein of the virus interacts with a cellular receptor and mediates membrane fusion to allow viral entry into susceptible target cells. Accordingly, S-protein plays an important role in virus infection cycle and is the primary target of neutralizing antibodies. To begin to understand its biochemical and immunological properties, we expressed both full-length and ectodomain of the protein in various primate cells. Our results show that the protein has an electrophoretic mobility of about 160-170 kDa. The protein is glycosylated with high mannose and/or hybrid oligosaccharides, which account for approximately 30 kDa of the apparent protein mass. The detection of S-protein by immunoassays was difficult using human convalescent sera, suggesting that the protein may not elicit strong humoral immune response in virus-infected patients. We were able to pseudotype murine leukemia virus particles with S-protein and produce SARS pseudoviruses. Pseudoviruses infected Vero E6 cells in a pH-independent manner and the infection could be specifically inhibited by convalescent sera. Consistent with low levels of antibodies against S-protein, neutralizing activity was weak with 50% neutralization titers ranging between 1:15 to 1:25. To facilitate quantifying pseudovirus-infected cells, which are stained blue with X-Gal, we devised an automated procedure using an ELISPOT analyzer. The high-throughput capacity of this procedure and the safety of using SARS pseudoviruses should make possible large-scale analyses of neutralizing antibody responses against SARS-CoV.


Assuntos
Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/genética , Testes de Neutralização/métodos , Síndrome Respiratória Aguda Grave/diagnóstico , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/isolamento & purificação , Proteínas do Envelope Viral/análise , Proteínas do Envelope Viral/genética , Animais , Anticorpos Antivirais/análise , Chlorocebus aethiops , Convalescença , Vetores Genéticos , Glicosilação , Células HeLa , Humanos , Vírus da Leucemia Murina/genética , Manose/química , Glicoproteínas de Membrana/química , Peso Molecular , Oligossacarídeos/química , Vírus Reordenados/isolamento & purificação , Vírus Reordenados/patogenicidade , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Sensibilidade e Especificidade , Síndrome Respiratória Aguda Grave/sangue , Especificidade da Espécie , Glicoproteína da Espícula de Coronavírus , Células Vero , Proteínas do Envelope Viral/química
8.
Virology ; 305(1): 124-37, 2003 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-12504547

RESUMO

It has been extremely difficult to elicit broadly cross-reactive neutralizing antibodies (Nabs) against human immunodeficiency virus type 1 (HIV-1). In this study, we compared the immunogenic properties of the wild-type and variable loop-deleted HIV-1 envelope glycoproteins. Mice were immunized with recombinant vaccinia viruses expressing either the wild-type or the variable loop-deleted (V1-2, V3, V4, and V1-3) HIV-1(DH12) gp160s. The animals were subsequently boosted with respective recombinant gp120s. All envelope constructs elicited similar levels of gp120-binding antibodies when analyzed by enzyme-linked immunosorbent assay (ELISA). However, the highest neutralizing activity was observed in sera from animals immunized with the wild-type envelope protein, followed by those immunized with DeltaV4 and DeltaV1-2. No neutralizing activity was detected in sera from animals immunized with DeltaV3 or DeltaV1-3. To identify immunogenic epitopes, ELISA was performed with overlapping 15-mer peptides that cover the entire length of gp120. For the wild-type gp120, the immunogenic epitopes mapped primarily to the variable loops V1-2 and to the conserved regions C1 and C5. When they were plotted onto known coordinates of gp120 core crystal structure, the epitopes in the conserved regions mapped predominantly to the inner domain of the protein. By immunizing with variable loop-deleted envelopes, the immune responses could be redirected to other regions of the protein. However, the newly targeted epitopes were neither on the exposed surface of the protein nor on the receptor binding regions. Interestingly, the removal of the V3 loop resulted in loss of immunoreactivity for both V3 and V1/V2 loops, suggesting structural interaction between the two regions. Our results suggest that obtaining broadly reactive Nabs may not be achieved simply by deleting the variable loops of gp120. However, the observation that the immune responses could be redirected by altering the protein composition might allow us to explore alternative strategies for modifying the antigenic properties of HIV-1 envelope glycoprotein.


Assuntos
Anticorpos Anti-HIV/biossíntese , Proteína gp120 do Envelope de HIV/imunologia , HIV-1/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Ensaio de Imunoadsorção Enzimática , Epitopos , Proteína gp120 do Envelope de HIV/química , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Testes de Neutralização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...