Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
PLoS Pathog ; 11(9): e1005148, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26325270

RESUMO

Vaccinia virus A33 is an extracellular enveloped virus (EEV)-specific type II membrane glycoprotein that is essential for efficient EEV formation and long-range viral spread within the host. A33 is a target for neutralizing antibody responses against EEV. In this study, we produced seven murine anti-A33 monoclonal antibodies (MAbs) by immunizing mice with live VACV, followed by boosting with the soluble A33 homodimeric ectodomain. Five A33 specific MAbs were capable of neutralizing EEV in the presence of complement. All MAbs bind to conformational epitopes on A33 but not to linear peptides. To identify the epitopes, we have adetermined the crystal structures of three representative neutralizing MAbs in complex with A33. We have further determined the binding kinetics for each of the three antibodies to wild-type A33, as well as to engineered A33 that contained single alanine substitutions within the epitopes of the three crystallized antibodies. While the Fab of both MAbs A2C7 and A20G2 binds to a single A33 subunit, the Fab from MAb A27D7 binds to both A33 subunits simultaneously. A27D7 binding is resistant to single alanine substitutions within the A33 epitope. A27D7 also demonstrated high-affinity binding with recombinant A33 protein that mimics other orthopoxvirus strains in the A27D7 epitope, such as ectromelia, monkeypox, and cowpox virus, suggesting that A27D7 is a potent cross-neutralizer. Finally, we confirmed that A27D7 protects mice against a lethal challenge with ectromelia virus.


Assuntos
Anticorpos Neutralizantes/metabolismo , Glicoproteínas de Membrana/antagonistas & inibidores , Modelos Moleculares , Orthopoxvirus/fisiologia , Infecções por Poxviridae/virologia , Proteínas do Envelope Viral/antagonistas & inibidores , Tropismo Viral , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/uso terapêutico , Afinidade de Anticorpos , Especificidade de Anticorpos , Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/genética , Complexo Antígeno-Anticorpo/metabolismo , Chlorocebus aethiops , Feminino , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos BALB C , Mutação , Orthopoxvirus/imunologia , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/prevenção & controle , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Vacinas Sintéticas/química , Vacinas Sintéticas/genética , Vacinas Sintéticas/metabolismo , Vacinas Sintéticas/uso terapêutico , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Vacinas Virais/química , Vacinas Virais/genética , Vacinas Virais/metabolismo , Vacinas Virais/uso terapêutico
2.
J Virol ; 87(3): 1569-85, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23152530

RESUMO

The extracellular virion form (EV) of vaccinia virus (VACV) is essential for viral pathogenesis and is difficult to neutralize with antibodies. Why this is the case and how the smallpox vaccine overcomes this challenge remain incompletely understood. We previously showed that high concentrations of anti-B5 antibodies are insufficient to directly neutralize EV (M. R. Benhnia, et al., J. Virol. 83:1201-1215, 2009). This allowed for at least two possible interpretations: covering the EV surface is insufficient for neutralization, or there are insufficient copies of B5 to allow anti-B5 IgG to cover the whole surface of EV and another viral receptor protein remains active. We endeavored to test these possibilities, focusing on the antibody responses elicited by immunization against smallpox. We tested whether human monoclonal antibodies (MAbs) against the three major EV antigens, B5, A33, and A56, could individually or together neutralize EV. While anti-B5 or anti-A33 (but not anti-A56) MAbs of appropriate isotypes were capable of neutralizing EV in the presence of complement, a mixture of anti-B5, anti-A33, and anti-A56 MAbs was incapable of directly neutralizing EV, even at high concentrations. This remained true when neutralizing the IHD-J strain, which lacks a functional version of the fourth and final known EV surface protein, A34. These immunological data are consistent with the possibility that viral proteins may not be the active component of the EV surface for target cell binding and infectivity. We conclude that the protection afforded by the smallpox vaccine anti-EV response is predominantly mediated not by direct neutralization but by isotype-dependent effector functions, such as complement recruitment for antibodies targeting B5 and A33.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vacina Antivariólica/imunologia , Vaccinia virus/imunologia , Vírion/imunologia , Anticorpos Monoclonais/imunologia , Antígenos Virais/imunologia , Proteínas do Sistema Complemento/imunologia , Humanos , Testes de Neutralização
3.
Int J Biol Macromol ; 263(Pt 2): 129858, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38423911

RESUMO

Tissue engineering (TE) demands scaffolds that have the necessary resistance to withstand the mechanical stresses once implanted in our body, as well as excellent biocompatibility. Hydrogels are postulated as interesting materials for this purpose, especially those made from biopolymers. In this study, the microstructure and rheological performance, as well as functional and biological properties of chitosan and collagen hydrogels (CH/CG) crosslinked with different coupling agents, both natural such as d-Fructose (F), genipin (G) and transglutaminase (T) and synthetic, using a combination of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride with N-hydroxysuccinimide (EDC/NHS) will be assessed. FTIR tests were carried out to determine if the proposed crosslinking reactions for each crosslinking agent occurred as expected, obtaining positive results in this aspect. Regarding the characterization of the properties of each system, two main trends were observed, from which it could be established that crosslinking with G and EDC-NHS turned out to be more effective and beneficial than with the other two crosslinking agents, producing significant improvements with respect to the base CH/CG hydrogel. In addition, in vitro tests demonstrated the potential application in TE of these systems, especially for those crosslinked with G, T and EDC-NHS.


Assuntos
Quitosana , Engenharia Tecidual , Engenharia Tecidual/métodos , Quitosana/química , Hidrogéis/farmacologia , Hidrogéis/química , Colágeno/química , Biopolímeros , Reagentes de Ligações Cruzadas/química , Alicerces Teciduais/química , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/química
4.
J Virol ; 86(15): 8050-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22623786

RESUMO

Smallpox vaccine is considered a gold standard of vaccines, as it is the only one that has led to the complete eradication of an infectious disease from the human population. B cell responses are critical for the protective immunity induced by the vaccine, yet their targeted epitopes recognized in humans remain poorly described. Here we describe the biochemical and structural characterization of one of the immunodominant vaccinia virus (VACV) antigens, D8, and its binding to the monoclonal antibody LA5, which is capable of neutralizing VACV in the presence of complement. The full-length D8 ectodomain was found to form a tetramer. We determined the crystal structure of the LA5 Fab-monomeric D8 complex at a resolution of 2.1 Å, as well as the unliganded structures of D8 and LA5-Fab at resolutions of 1.42 Å and 1.6 Å, respectively. D8 features a carbonic anhydrase (CAH) fold that has evolved to bind to the glycosaminoglycan (GAG) chondroitin sulfate (CS) on host cells. The central positively charged crevice of D8 was predicted to be the CS binding site by automated docking experiments. Furthermore, sequence alignment of various poxvirus D8 orthologs revealed that this crevice is structurally conserved. The D8 epitope is formed by 23 discontinuous residues that are spread across 80% of the D8 protein sequence. Interestingly, LA5 binds with a high-affinity lock-and-key mechanism above this crevice with an unusually large antibody-antigen interface, burying 2,434 Å(2) of protein surface.


Assuntos
Anticorpos Monoclonais Murinos/química , Anticorpos Antivirais/química , Fragmentos Fab das Imunoglobulinas/química , Vaccinia virus/química , Proteínas do Envelope Viral/química , Animais , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Antivirais/imunologia , Sítios de Ligação , Chlorocebus aethiops , Cristalografia por Raios X , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Camundongos , Estrutura Quaternária de Proteína , Vaccinia virus/imunologia , Células Vero , Proteínas do Envelope Viral/imunologia
5.
Pharmaceutics ; 14(12)2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36559299

RESUMO

The research on flavonoids has exponentially grown since their first therapeutic evidence, in 1937. They are effective in vitro in a wide range of human diseases, particularly those mediated by free radicals, such as cancer, atherosclerosis, AIDS, or neuronal diseases. However, their applications have been reduced due to their low solubility, poor absorption, and rapid metabolism. Flavonoid encapsulation in nanocarriers significantly improves their oral absorption, protects the drug against degradation, decreases the first-pass hepatic effect, and makes absorption through the lymphatic system easier. In this work, carbon nanotubes were used as nanocarriers of 7-hydroxyflavone, 7-HF. The encapsulation of 7-HF into pristine single- and multi-walled carbon nanotubes, and into -COOH functionalized single-walled carbon nanotubes has been investigated. The equilibrium association constants were estimated. The structural backbone of 7-HF, two benzene rings linked through three carbon atoms that form a pyran heterocyclic ring containing a keto group, seems to play a key role in the 7-HF/CNT interactions, although other types of interactions are also at work. The in vitro release of 7-HF was studied at three pHs, 2.0, 7.4, and 9.2, mimicking the different biological barriers of the human organism.

6.
Proc Natl Acad Sci U S A ; 105(50): 19863-8, 2008 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19060201

RESUMO

Borrelia burgdorferi is the etiologic agent of Lyme disease, a multisystem inflammatory disorder that principally targets the skin, joints, heart, and nervous system. The role of T lymphocytes in the development of chronic inflammation resulting from B. burgdorferi infection has been controversial. We previously showed that natural killer T (NKT) cells with an invariant (i) TCR alpha chain (iNKT cells) recognize glycolipids from B. burgdorferi, but did not establish an in vivo role for iNKT cells in Lyme disease pathogenesis. Here, we evaluate the importance of iNKT cells for host defense against these pathogenic spirochetes by using Valpha14i NKT cell-deficient (Jalpha18(-/-)) BALB/c mice. On tick inoculation with B. burgdorferi, Jalpha18(-/-) mice exhibited more severe and prolonged arthritis as well as a reduced ability to clear spirochetes from infected tissues. Valpha14i NKT cell deficiency also resulted in increased production of antibodies directed against both B. burgdorferi protein antigens and borrelial diacylglycerols; the latter finding demonstrates that anti-glycolipid antibody production does not require cognate help from Valpha14i NKT cells. Valpha14i NKT cells in infected wild-type mice expressed surface activation markers and produced IFNgamma in vivo after infection, suggesting a participatory role for this unique population in cellular immunity. Our data are consistent with the hypothesis that the antigen-specific activation of Valpha14i NKT cells is important for the prevention of persistent joint inflammation and spirochete clearance, and they counter the long-standing notion that humoral rather than cellular immunity is sufficient to facilitate Lyme disease resolution.


Assuntos
Artrite Infecciosa/imunologia , Borrelia burgdorferi/imunologia , Articulações/imunologia , Doença de Lyme/complicações , Células T Matadoras Naturais/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Artrite Infecciosa/microbiologia , Doença Crônica , Interferon gama/imunologia , Articulações/microbiologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Células T Matadoras Naturais/microbiologia
7.
J Virol ; 83(3): 1201-15, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19019965

RESUMO

Antibody neutralization is an important component of protective immunity against vaccinia virus (VACV). Two distinct virion forms, mature virion and enveloped virion (MV and EV, respectively), possess separate functions and nonoverlapping immunological properties. In this study we examined the mechanics of EV neutralization, focusing on EV protein B5 (also called B5R). We show that neutralization of EV is predominantly complement dependent. From a panel of high-affinity anti-B5 monoclonal antibodies (MAbs), the only potent neutralizer in vitro (90% at 535 ng/ml) was an immunoglobulin G2a (IgG2a), and neutralization was complement mediated. This MAb was the most protective in vivo against lethal intranasal VACV challenge. Further studies demonstrated that in vivo depletion of complement caused a >50% loss of anti-B5 IgG2a protection, directly establishing the importance of complement for protection against the EV form. However, the mechanism of protection is not sterilizing immunity via elimination of the inoculum as the viral inoculum consisted of a purified MV form. The prevention of illness in vivo indicated rapid control of infection. We further demonstrate that antibody-mediated killing of VACV-infected cells expressing surface B5 is a second protective mechanism provided by complement-fixing anti-B5 IgG. Cell killing was very efficient, and this effector function was highly isotype specific. These results indicate that anti-B5 antibody-directed cell lysis via complement is a powerful mechanism for clearance of infected cells, keeping poxvirus-infected cells from being invisible to humoral immune responses. These findings highlight the importance of multiple mechanisms of antibody-mediated protection against VACV and point to key immunobiological differences between MVs and EVs that impact the outcome of infection.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Vaccinia virus/imunologia , Vírion/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Chlorocebus aethiops , Ensaio de Imunoadsorção Enzimática , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Células Vero
8.
J Virol ; 83(23): 12355-67, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19793826

RESUMO

Antibodies against the extracellular virion (EV or EEV) form of vaccinia virus are an important component of protective immunity in animal models and likely contribute to the protection of immunized humans against poxviruses. Using fully human monoclonal antibodies (MAbs), we now have shown that the protective attributes of the human anti-B5 antibody response to the smallpox vaccine (vaccinia virus) are heavily dependent on effector functions. By switching Fc domains of a single MAb, we have definitively shown that neutralization in vitro--and protection in vivo in a mouse model--by the human anti-B5 immunoglobulin G MAbs is isotype dependent, thereby demonstrating that efficient protection by these antibodies is not simply dependent on binding an appropriate vaccinia virion antigen with high affinity but in fact requires antibody effector function. The complement components C3 and C1q, but not C5, were required for neutralization. We also have demonstrated that human MAbs against B5 can potently direct complement-dependent cytotoxicity of vaccinia virus-infected cells. Each of these results was then extended to the polyclonal human antibody response to the smallpox vaccine. A model is proposed to explain the mechanism of EV neutralization. Altogether these findings enhance our understanding of the central protective activities of smallpox vaccine-elicited antibodies in immunized humans.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Isotipos de Imunoglobulinas/imunologia , Varíola/prevenção & controle , Vaccinia virus/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Peso Corporal , Complemento C1q/imunologia , Complemento C3/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Testes de Neutralização , Análise de Sobrevida
9.
J Virol ; 82(7): 3751-68, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18234801

RESUMO

The smallpox vaccine is widely considered the gold standard for human vaccines, yet the key antibody targets in humans remain unclear. We endeavored to identify a stereotypic, dominant, mature virion (MV) neutralizing antibody target in humans which could be used as a diagnostic serological marker of protective humoral immunity induced by the smallpox vaccine (vaccinia virus [VACV]). We have instead found that diversity is a defining characteristic of the human antibody response to the smallpox vaccine. We show that H3 is the most immunodominant VACV neutralizing antibody target, as determined by correlation analysis of immunoglobulin G (IgG) specificities to MV neutralizing antibody titers. It was determined that purified human anti-H3 IgG is sufficient for neutralization of VACV; however, depletion or blockade of anti-H3 antibodies revealed no significant reduction in neutralization activity, showing anti-H3 IgG is not required in vaccinated humans (or mice) for neutralization of MV. Comparable results were obtained for human (and mouse) anti-L1 IgG and even for anti-H3 and anti-L1 IgG in combination. In addition to H3 and L1, human antibody responses to D8, A27, D13, and A14 exhibited statistically significant correlations with virus neutralization. Altogether, these data indicate the smallpox vaccine succeeds in generating strong neutralizing antibody responses not by eliciting a stereotypic response to a single key antigen but instead by driving development of neutralizing antibodies to multiple viral proteins, resulting in a "safety net" of highly redundant neutralizing antibody responses, the specificities of which can vary from individual to individual. We propose that this is a fundamental attribute of the smallpox vaccine.


Assuntos
Anticorpos Antivirais/sangue , Vacina Antivariólica/imunologia , Vaccinia virus/imunologia , Proteínas de Transporte/imunologia , Humanos , Imunoglobulina G/sangue , Testes de Neutralização , Proteínas do Envelope Viral/imunologia , Proteínas Virais/imunologia , Vírion/imunologia
10.
Structure ; 22(4): 646-57, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24631463

RESUMO

Antibody epitope mapping is crucial for understanding B cell-mediated immunity and required for characterizing therapeutic antibodies. In contrast to T cell epitope mapping, no computational tools are in widespread use for prediction of B cell epitopes. Here, we show that, utilizing the sequence of an antibody, it is possible to identify discontinuous epitopes on its cognate antigen. The predictions are based on residue-pairing preferences and other interface characteristics. We combined these antibody-specific predictions with results of cross-blocking experiments that identify groups of antibodies with overlapping epitopes to improve the predictions. We validate the high performance of this approach by mapping the epitopes of a set of antibodies against the previously uncharacterized D8 antigen, using complementary techniques to reduce method-specific biases (X-ray crystallography, peptide ELISA, deuterium exchange, and site-directed mutagenesis). These results suggest that antibody-specific computational predictions and simple cross-blocking experiments allow for accurate prediction of residues in conformational B cell epitopes.


Assuntos
Anticorpos Monoclonais/química , Complexo Antígeno-Anticorpo/química , Antígenos Virais/química , Epitopos de Linfócito B/química , Peptídeos/química , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Complexo Antígeno-Anticorpo/imunologia , Antígenos Virais/imunologia , Cristalografia por Raios X , Medição da Troca de Deutério , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Epitopos de Linfócito B/imunologia , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Peptídeos/imunologia , Vaccinia virus/química
11.
Antiviral Res ; 111: 26-32, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25173576

RESUMO

Novel strategies are necessary to decrease inflammatory parameters in successfully treated HIV-infected patients. Our aim was to evaluate the maintenance of viral suppression and potential changes in inflammatory, immune-activation and coagulation biomarkers in virologically suppressed HIV-infected patients switched to a nucleoside reverse transcriptase inhibitor-sparing (NRTI) and maraviroc (MVC)-containing combined antiretroviral therapy (cART). Fifty-eight HIV-infected patients were observed after their treatment regimens were changed to MVC 150mg/once daily plus ritonavir-boosted protease inhibitor therapy. Activation-, inflammation- and coagulation-associated biomarkers and mitochondrial (mt)DNA were analyzed after a median of 24weeks of follow-up. We observed that after changing to an NRTI-sparing regimen, 96.6% of HIV-patients on viral suppressive cART maintained viral suppression and their CD4+ T cell counts did not change significantly (median of 31weeks of follow-up). This cART switch reduced soluble CD40 ligand (p=0.002), beta-2 microglobulin (p=0.025), and soluble CD14 (p=0.009) in patients with higher baseline levels of these inflammation biomarkers after a median of 24weeks of follow-up. The results of our study show that changing to NRTI-sparing dual therapy decreased the levels of inflammatory biomarkers and maintained the immune-virologic efficacy. The potential benefits of this regimen warrant further investigation to uncover the association of this therapy with the potential decrease in the morbidity and mortality of HIV-infected patients from non-AIDS-defining illnesses.


Assuntos
Ligante de CD40/sangue , Cicloexanos/administração & dosagem , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Receptores de Lipopolissacarídeos/sangue , Inibidores da Transcriptase Reversa/administração & dosagem , Triazóis/administração & dosagem , Microglobulina beta-2/sangue , Adulto , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Quimioterapia Combinada , Feminino , Infecções por HIV/sangue , Infecções por HIV/virologia , HIV-1/genética , HIV-1/isolamento & purificação , HIV-1/fisiologia , Humanos , Masculino , Maraviroc , Pessoa de Meia-Idade
12.
Antivir Ther ; 15(4): 661-75, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20587859

RESUMO

BACKGROUND: Treatment of rare severe side effects of vaccinia virus (VACV) immunization in humans is currently very challenging. VACV possesses two immunologically distinct virion forms in vivo - intracellular mature virion (MV, IMV) and extracellular virion (EV, EEV). METHODS: Antibody-mediated therapeutic efficacy was determined against VACV infection in a small animal model of progressive vaccinia. The model consisted of severe combined immunodeficiency mice infected with VACV New York City Board of Health vaccine strain and treated with monoclonal antibodies (mAbs). RESULTS: Here, we show that combination therapy with two fully human mAbs against an immunodominant MV antigen, H3 (H3L), and an EV antigen, B5 (B5R), provides significantly better protection against disease and death than either single human monoclonal or human vaccinia immune globulin, the currently licensed therapeutic for side effects of smallpox vaccination. CONCLUSIONS: The preclinical studies validate that this combination of mAbs against H3 and B5 is a promising approach as a poxvirus infection treatment for use in humans.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Proteínas de Transporte/imunologia , Vaccinia virus/imunologia , Vacínia/tratamento farmacológico , Proteínas do Envelope Viral/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/uso terapêutico , Chlorocebus aethiops , Modelos Animais de Doenças , Quimioterapia Combinada , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Testes de Neutralização , Resultado do Tratamento , Vacínia/imunologia , Células Vero
13.
Nat Immunol ; 7(9): 978-86, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16921381

RESUMO

Natural killer T (NKT) cells recognize glycosphingolipids presented by CD1d molecules and have been linked to defense against microbial infections. Previously defined foreign glycosphingolipids recognized by NKT cells are uniquely found in nonpathogenic sphingomonas bacteria. Here we show that mouse and human NKT cells also recognized glycolipids, specifically a diacylglycerol, from Borrelia burgdorferi, which causes Lyme disease. The B. burgdorferi-derived, glycolipid-induced NKT cell proliferation and cytokine production and the antigenic potency of this glycolipid was dependent on acyl chain length and saturation. These data indicate that NKT cells recognize categories of glycolipids beyond those in sphingomonas and suggest that NKT cell responses driven by T cell receptor-mediated glycolipid recognition may provide protection against diverse pathogens.


Assuntos
Antígenos de Bactérias/imunologia , Borrelia burgdorferi/imunologia , Glicolipídeos/imunologia , Células Matadoras Naturais/imunologia , Saponinas/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Antígenos de Bactérias/química , Antígenos de Bactérias/farmacologia , Antígenos CD1/imunologia , Antígenos CD1d , Células Cultivadas , Diglicerídeos/química , Diglicerídeos/metabolismo , Diglicerídeos/farmacologia , Glicolipídeos/química , Glicolipídeos/farmacologia , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Conformação Proteica , Saponinas/química , Saponinas/farmacologia , Receptores Toll-Like/metabolismo
14.
J Immunol ; 174(3): 1539-48, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15661914

RESUMO

Lyme disease is a chronic inflammatory disorder caused by the spirochetal bacterium, Borrelia burgdorferi. In vitro evidence suggests that binding of spirochetal lipoproteins to CD14, a pattern recognition receptor expressed on monocytes/macrophages and polymorphonuclear cells, is a critical requirement for cellular activation and the subsequent release of proinflammatory cytokines that most likely contribute to symptomatology and clinical manifestations. To test the validity of this notion, we assessed the impact of CD14 deficiency on Lyme disease in C3H/HeN mice. Contrary to an anticipated diminution in pathology, CD14(-/-) mice exhibited more severe and persistent inflammation than did CD14(+/+) mice. This disparity reflects altered gene regulation within immune cells that may engender the higher bacterial burden and serum cytokine levels observed in CD14(-/-) mice. Comparing their in vitro stimulatory activity, live spirochetes, but not lysed organisms, were a potent CD14-independent stimulus of cytokine production, triggering an exaggerated response by CD14(-/-) macrophages. Collectively, our in vivo and in vitro findings support the provocative notion that: 1) pattern recognition by CD14 is entirely dispensable for elaboration of an inflammatory response to B. burgdorferi, and 2) CD14-independent signaling pathways are inherently more destructive than CD14-dependent pathways. Continued study of CD14-independent signaling pathways may provide mechanistic insight into the inflammatory processes that underlie development of chronic inflammation.


Assuntos
Borrelia burgdorferi/imunologia , Mediadores da Inflamação/fisiologia , Receptores de Lipopolissacarídeos/fisiologia , Doença de Lyme/imunologia , Doença de Lyme/patologia , Transdução de Sinais/imunologia , Animais , Citocinas/biossíntese , Citocinas/sangue , Citocinas/genética , Imunidade Inata/genética , Mediadores da Inflamação/sangue , Receptores de Lipopolissacarídeos/genética , Doença de Lyme/genética , Doença de Lyme/microbiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , RNA Mensageiro/biossíntese , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/fisiologia , Índice de Gravidade de Doença , Transdução de Sinais/genética , Membrana Sinovial/imunologia , Membrana Sinovial/microbiologia , Membrana Sinovial/patologia , Receptores Toll-Like , Transcrição Gênica/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA