Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 15: 1273942, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38410511

RESUMO

Introduction: It is now clear that coronavirus disease 19 (COVID-19) severity is associated with a dysregulated immune response, but the relative contributions of different immune cells is still not fully understood. SARS CoV-2 infection triggers marked changes in NK cell populations, but there are contradictory reports as to whether these effector lymphocytes play a protective or pathogenic role in immunity to SARS-CoV-2. Methods: To address this question we have analysed differences in the phenotype and function of NK cells in SARS-CoV-2 infected individuals who developed either very mild, or life-threatening COVID-19 disease. Results: Although NK cells from patients with severe disease appeared more activated and the frequency of adaptive NK cells was increased, they were less potent mediators of ADCC than NK cells from patients with mild disease. Further analysis of peripheral blood NK cells in these patients revealed that a population of NK cells that had lost expression of the activating receptor NKG2D were a feature of patients with severe disease and this correlated with elevated levels of cell free NKG2D ligands, especially ULBP2 and ULBP3 in the plasma of critically ill patients. In vitro, culture in NKG2DL containing patient sera reduced the ADCC function of healthy donor NK cells and this could be blocked by NKG2DL-specific antibodies. Discussion: These observations of reduced NK function in severe disease are consistent with the hypothesis that defects in immune surveillance by NK cells permit higher levels of viral replication, rather than that aberrant NK cell function contributes to immune system dysregulation and immunopathogenicity.


Assuntos
COVID-19 , Citotoxicidade Imunológica , Humanos , COVID-19/patologia , Células Matadoras Naturais , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , SARS-CoV-2/metabolismo
2.
Cells ; 10(12)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34943787

RESUMO

Dendritic cells (DCs) are the most potent antigen-presenting cells, and their function is essential to configure adaptative immunity and avoid excessive inflammation. DCs are predicted to play a crucial role in the clinical evolution of the infection by the severe acute respiratory syndrome (SARS) coronavirus (CoV)-2. DCs interaction with the SARS-CoV-2 Spike protein, which mediates cell receptor binding and subsequent fusion of the viral particle with host cell, is a key step to induce effective immunity against this virus and in the S protein-based vaccination protocols. Here we evaluated human DCs in response to SARS-CoV-2 S protein, or to a fragment encompassing the receptor binding domain (RBD) challenge. Both proteins increased the expression of maturation markers, including MHC molecules and costimulatory receptors. DCs interaction with the SARS-CoV-2 S protein promotes activation of key signaling molecules involved in inflammation, including MAPK, AKT, STAT1, and NFκB, which correlates with the expression and secretion of distinctive proinflammatory cytokines. Differences in the expression of ACE2 along the differentiation of human monocytes to mature DCs and inter-donor were found. Our results show that SARS-CoV-2 S protein promotes inflammatory response and provides molecular links between individual variations and the degree of response against this virus.


Assuntos
Células Dendríticas/patologia , Células Dendríticas/virologia , Receptores Virais/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular , Citocinas/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Inflamação/patologia , Lectinas Tipo C/metabolismo , Domínios Proteicos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Superfície Celular/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Doadores de Tecidos
3.
Eur J Immunol ; 51(11): 2633-2640, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34358329

RESUMO

Here, we describe a new, simple, highly multiplexed serological test that generates a more complete picture of seroconversion than single antigen-based assays. Flow cytometry is used to detect multiple Ig isotypes binding to four SARS-CoV-2 antigens: the Spike glycoprotein, its RBD fragment (the main target for neutralizing antibodies), the nucleocapsid protein, and the main cysteine-like protease in a single reaction. Until now, most diagnostic serological tests measured antibodies to only one antigen and in some laboratory-confirmed patients no SARS-CoV-2-specific antibodies could be detected. Our data reveal that while most patients respond against all the viral antigens tested, others show a marked bias to make antibodies against either proteins exposed on the viral particle or those released after cellular infection. With this assay, it was possible to discriminate between patients and healthy controls with 100% confidence. Analysing the response of multiple Ig isotypes to the four antigens in combination may also help to establish a correlation with the severity degree of disease. A more detailed description of the immune responses of different patients to SARS-CoV-2 virus might provide insight into the wide array of clinical presentations of COVID-19.


Assuntos
Anticorpos Antivirais/sangue , Teste Sorológico para COVID-19/métodos , COVID-19/diagnóstico , Citometria de Fluxo/métodos , Antígenos Virais/imunologia , COVID-19/imunologia , Ensaios de Triagem em Larga Escala , Humanos , SARS-CoV-2 , Sensibilidade e Especificidade , Testes Sorológicos
4.
Front Immunol ; 12: 824728, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35154086

RESUMO

We generated an optimized COVID-19 vaccine candidate based on the modified vaccinia virus Ankara (MVA) vector expressing a full-length prefusion-stabilized SARS-CoV-2 spike (S) protein, termed MVA-CoV2-S(3P). The S(3P) protein was expressed at higher levels (2-fold) than the non-stabilized S in cells infected with the corresponding recombinant MVA viruses. One single dose of MVA-CoV2-S(3P) induced higher IgG and neutralizing antibody titers against parental SARS-CoV-2 and variants of concern than MVA-CoV2-S in wild-type C57BL/6 and in transgenic K18-hACE2 mice. In immunized C57BL/6 mice, two doses of MVA-CoV2-S or MVA-CoV2-S(3P) induced similar levels of SARS-CoV-2-specific B- and T-cell immune responses. Remarkably, a single administration of MVA-CoV2-S(3P) protected all K18-hACE2 mice from morbidity and mortality caused by SARS-CoV-2 infection, reducing SARS-CoV-2 viral loads, histopathological lesions, and levels of pro-inflammatory cytokines in the lungs. These results demonstrated that expression of a novel full-length prefusion-stabilized SARS-CoV-2 S protein by the MVA poxvirus vector enhanced immunogenicity and efficacy against SARS-CoV-2 in animal models, further supporting MVA-CoV2-S(3P) as an optimized vaccine candidate for clinical trials.


Assuntos
Vacinas contra COVID-19/imunologia , COVID-19/prevenção & controle , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Idoso , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , COVID-19/mortalidade , Vacinas contra COVID-19/genética , Linhagem Celular Tumoral , Embrião de Galinha , Chlorocebus aethiops , Citocinas/análise , Feminino , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasmídeos/genética , Glicoproteína da Espícula de Coronavírus/genética , Eficácia de Vacinas , Vacinas de DNA/genética , Vaccinia virus/imunologia , Células Vero , Vacinas Virais/genética
5.
J Immunol ; 205(11): 3130-3140, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33148714

RESUMO

Currently, there is a need for reliable tests that allow identification of individuals that have been infected with SARS-CoV-2 even if the infection was asymptomatic. To date, the vast majority of the serological tests for SARS-CoV-2-specific Abs are based on serum detection of Abs to either the viral spike glycoprotein (the major target for neutralizing Abs) or the viral nucleocapsid protein that is known to be highly immunogenic in other coronaviruses. Conceivably, exposure of Ags released from infected cells could stimulate Ab responses that might correlate with tissue damage and, hence, they may have some value as a prognostic indicator. We addressed whether other nonstructural viral proteins, not incorporated into the infectious viral particle, specifically the viral cysteine-like protease, might also be potent immunogens. Using ELISA tests, coating several SARS-CoV-2 proteins produced in vitro, we describe that COVID-19 patients make high titer IgG, IgM, and IgA Ab responses to the Cys-like protease from SARS-CoV-2, also known as 3CLpro or Mpro, and it can be used to identify individuals with positive serology against the coronavirus. Higher Ab titers in these assays associated with more-severe disease, and no cross-reactive Abs against prior betacoronavirus were found. Remarkably, IgG Abs specific for Mpro and other SARS-CoV-2 Ags can also be detected in saliva. In conclusion, Mpro is a potent Ag in infected patients that can be used in serological tests, and its detection in saliva could be the basis for a rapid, noninvasive test for COVID-19 seropositivity.


Assuntos
Anticorpos Antivirais/sangue , Betacoronavirus/metabolismo , Infecções por Coronavirus/sangue , Cisteína Proteases/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Pneumonia Viral/sangue , Saliva/metabolismo , Adulto , Idoso , COVID-19 , Feminino , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Pandemias , SARS-CoV-2
6.
Virus Res ; 236: 30-43, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28465158

RESUMO

Measles virus (MV) remains a leading cause of vaccine-preventable deaths in children. Protection against MV is associated with neutralizing antibodies that preferentially recognize the viral hemagglutinin (MV-H), and to a lesser extent, the fusion protein (MV-F). Although MV is serologically monotypic, 24 genotypes have been identified. Here we report three neutralization epitopes conserved in the more prevalent circulating MV genotypes, two located in the MV-H receptor binding site (RBS) (antigenic site III) and a third in MV-H/MV-F interphase (antigenic site Ia) which are essential for MV multiplication. In contrast, two MV-H neutralization epitopes, showed a genotype-specific neutralization escape due to a single amino acid change, that we mapped in the "noose" antigenic site, or an enhanced neutralization epitope (antigenic site IIa). The monoclonal antibody (mAb) neutralization potency correlated with its binding affinity and was mainly driven by kinetic dissociation rate (koff). We developed an immunoassay for mAb binding to MV-H in its native hetero-oligomeric structure with MV-F on the surface of a MV productive steady-state persistently infected (p.i.) human cell lines, and a competitive-binding assay with serum from individuals with past infection by different MV genotypes. Binding assays revealed that a broad neutralization epitope, in RBS antigenic site, a genotype specific neutralization epitopes, in noose and IIa sites, were immunogenic in natural infection and vaccination and may elicit long-lasting humoral immunity that might contribute to explain MV immunogenic stability. These results support the design of improved measles vaccines, broad-spectrum prophylactic or therapeutic antibodies and MV-used in oncolytic therapies.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Hemaglutininas Virais/imunologia , Vírus do Sarampo/imunologia , Sarampo/imunologia , Anticorpos Neutralizantes/imunologia , Epitopos/administração & dosagem , Epitopos/imunologia , Genótipo , Hemaglutininas Virais/administração & dosagem , Hemaglutininas Virais/genética , Humanos , Sarampo/prevenção & controle , Sarampo/virologia , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/imunologia , Vírus do Sarampo/classificação , Vírus do Sarampo/genética , Vírus do Sarampo/isolamento & purificação , Testes de Neutralização , Vacinação
7.
Sci Rep ; 7: 46045, 2017 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-28393915

RESUMO

Cell surface aminopeptidase N (APN) is a membrane-bound ectoenzyme that hydrolyzes proteins and peptides and regulates numerous cell functions. APN participates in tumor cell expansion and motility, and is a target for cancer therapies. Small drugs that bind to the APN active site inhibit catalysis and suppress tumor growth. APN is also a major cell entry receptor for coronavirus, which binds to a region distant from the active site. Three crystal structures that we determined of human and pig APN ectodomains defined the dynamic conformation of the protein. These structures offered snapshots of closed, intermediate and open APN, which represent distinct functional states. Coronavirus envelope proteins specifically recognized the open APN form, prevented ectodomain progression to the closed form and substrate hydrolysis. In addition, drugs that bind the active site inhibited both coronavirus binding to cell surface APN and infection; the drugs probably hindered APN transition to the virus-specific open form. We conclude that allosteric inhibition of APN functions occurs by ligand suppression of ectodomain motions necessary for catalysis and virus cell entry, as validated by locking APN with disulfides. Blocking APN dynamics can thus be a valuable approach to development of drugs that target this ectoenzyme.


Assuntos
Antígenos CD13/antagonistas & inibidores , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Coronavirus/fisiologia , Neoplasias/patologia , Regulação Alostérica/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Biocatálise/efeitos dos fármacos , Antígenos CD13/química , Antígenos CD13/metabolismo , Células CHO , Domínio Catalítico , Proliferação de Células/efeitos dos fármacos , Coronavirus/efeitos dos fármacos , Cricetinae , Cricetulus , Dissulfetos/metabolismo , Humanos , Modelos Moleculares , Domínios Proteicos , Sus scrofa
8.
PLoS Pathog ; 8(8): e1002859, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22876187

RESUMO

The coronaviruses (CoVs) are enveloped viruses of animals and humans associated mostly with enteric and respiratory diseases, such as the severe acute respiratory syndrome and 10-20% of all common colds. A subset of CoVs uses the cell surface aminopeptidase N (APN), a membrane-bound metalloprotease, as a cell entry receptor. In these viruses, the envelope spike glycoprotein (S) mediates the attachment of the virus particles to APN and subsequent cell entry, which can be blocked by neutralizing antibodies. Here we describe the crystal structures of the receptor-binding domains (RBDs) of two closely related CoV strains, transmissible gastroenteritis virus (TGEV) and porcine respiratory CoV (PRCV), in complex with their receptor, porcine APN (pAPN), or with a neutralizing antibody. The data provide detailed information on the architecture of the dimeric pAPN ectodomain and its interaction with the CoV S. We show that a protruding receptor-binding edge in the S determines virus-binding specificity for recessed glycan-containing surfaces in the membrane-distal region of the pAPN ectodomain. Comparison of the RBDs of TGEV and PRCV to those of other related CoVs, suggests that the conformation of the S receptor-binding region determines cell entry receptor specificity. Moreover, the receptor-binding edge is a major antigenic determinant in the TGEV envelope S that is targeted by neutralizing antibodies. Our results provide a compelling view on CoV cell entry and immune neutralization, and may aid the design of antivirals or CoV vaccines. APN is also considered a target for cancer therapy and its structure, reported here, could facilitate the development of anti-cancer drugs.


Assuntos
Anticorpos Neutralizantes/química , Anticorpos Antivirais/química , Antígenos CD13/química , Coronavirus Respiratório Porcino/química , Proteínas do Envelope Viral/química , Ligação Viral , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Antígenos CD13/imunologia , Cristalografia por Raios X , Humanos , Dados de Sequência Molecular , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/imunologia
9.
Gastroenterology ; 142(7): 1516-25.e3, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22430395

RESUMO

BACKGROUND & AIMS: CD4+ T-regulatory (Treg) cells suppress immune responses and control self-tolerance and immunity to pathogens, cancer, and alloantigens. Most pathogens activate Treg cells to minimize immune-mediated tissue damage and prevent clearance, which promotes chronic infections. However, hepatitis A virus (HAV) temporarily inhibits Treg-cell functions. We investigated whether the interaction of HAV with its cellular receptor 1 (HAVCR1), a T-cell co-stimulatory molecule, inhibits the function of Treg cells to control HAV infection. METHODS: We studied the effects of HAV interaction with HAVCR1 on human T cells using binding, signal transduction, apoptosis, activation, suppression, cytokine production, and confocal microscopy analyses. Cytokines were analyzed in sera from 14 patients with HAV infection using bead arrays. RESULTS: Human Treg cells constitutively express HAVCR1. Binding of HAV to HAVCR1 blocked phosphorylation of Akt, prevented activation of the T-cell receptor, and inhibited function of Treg cells. At the peak viremia, patients with acute HAV infection had no Treg-cell suppression function, produced low levels of transforming growth factor-ß , which limited leukocyte recruitment and survival, and produced high levels of interleukin-22, which prevented liver damage. CONCLUSIONS: Interaction between HAV and its receptor HAVCR1 inhibits Treg-cell function, resulting in an immune imbalance that allows viral expansion with limited hepatocellular damage during early stages of infection-a characteristic of HAV pathogenesis. The mechanism by which HAV is cleared in the absence of Treg-cell function could be used as a model to develop anticancer therapies, modulate autoimmune and allergic responses, and prevent transplant rejection.


Assuntos
Vírus da Hepatite A/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Virais/metabolismo , Linfócitos T Reguladores/imunologia , Ligação Viral , Linhagem Celular , Hepatite A/imunologia , Hepatite A/metabolismo , Receptor Celular 1 do Vírus da Hepatite A , Humanos , Interleucinas/biossíntese , Proteínas Proto-Oncogênicas c-akt , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/virologia , Fator de Crescimento Transformador beta1/sangue , Interleucina 22
10.
J Gen Virol ; 92(Pt 5): 1117-1126, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21228126

RESUMO

The N-terminal S1 region of the transmissible gastroenteritis virus (TGEV) spike (S) glycoprotein contains four antigenic sites (C, B, D and A, from the N- to the C-terminal end) and is engaged in host-cell receptor recognition. The most N-terminal portion of the S1 region, which comprises antigenic sites C and B, is needed for the enteric tropism of TGEV, whereas the major antigenic site A at the C-terminal moiety is required for both respiratory and enteric cell tropism, and is engaged in recognition of the aminopeptidase N (APN) receptor. This study determined the kinetics for binding of a soluble S1 protein to the APN protein. Moreover, the S1 region of the TGEV S protein was dissected, with the aim of identifying discrete modules displaying unique antigenic sites and receptor-binding functions. Following protease treatments and mammalian cell expression methods, four modules or domains (D1-D4) were defined at the S1 region. Papain treatment identified an N-terminal domain (D1) resistant to proteolysis, whereas receptor binding defined a soluble and functional APN receptor-binding domain (D3). This domain was recognized by neutralizing antibodies belonging to the antigenic site A and therefore could be used as an immunogen for the prevention of viral infection. The organization of the four modules in the S1 region of the TGEV S glycoprotein is discussed.


Assuntos
Antígenos CD13/metabolismo , Epitopos/imunologia , Glicoproteínas de Membrana/imunologia , Receptores Virais/metabolismo , Vírus da Gastroenterite Transmissível/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Linhagem Celular , Humanos , Glicoproteínas de Membrana/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Glicoproteína da Espícula de Coronavírus , Proteínas do Envelope Viral/metabolismo
11.
PLoS Pathog ; 6(9): e1001122, 2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20941397

RESUMO

The human membrane cofactor protein (MCP, CD46) is a central component of the innate immune system. CD46 protects autologous cells from complement attack by binding to complement proteins C3b and C4b and serving as a cofactor for their cleavage. Recent data show that CD46 also plays a role in mediating acquired immune responses, and in triggering autophagy. In addition to these physiologic functions, a significant number of pathogens, including select adenoviruses, measles virus, human herpes virus 6 (HHV-6), Streptococci, and Neisseria, use CD46 as a cell attachment receptor. We have determined the crystal structure of the extracellular region of CD46 in complex with the human adenovirus type 11 fiber knob. Extracellular CD46 comprises four short consensus repeats (SCR1-SCR4) that form an elongated structure resembling a hockey stick, with a long shaft and a short blade. Domains SCR1, SCR2 and SCR3 are arranged in a nearly linear fashion. Unexpectedly, however, the structure reveals a profound bend between domains SCR3 and SCR4, which has implications for the interactions with ligands as well as the orientation of the protein at the cell surface. This bend can be attributed to an insertion of five hydrophobic residues in a SCR3 surface loop. Residues in this loop have been implicated in interactions with complement, indicating that the bend participates in binding to C3b and C4b. The structure provides an accurate framework for mapping all known ligand binding sites onto the surface of CD46, thereby advancing an understanding of how CD46 acts as a receptor for pathogens and physiologic ligands of the immune system.


Assuntos
Adenovírus Humanos/metabolismo , Complemento C3b/metabolismo , Complemento C4b/metabolismo , Proteína Cofatora de Membrana/química , Proteína Cofatora de Membrana/metabolismo , Adenovírus Humanos/classificação , Sítios de Ligação , Cristalografia por Raios X , Glicosilação , Humanos , Modelos Moleculares , Polissacarídeos/metabolismo , Conformação Proteica , Receptores Virais/metabolismo
12.
World J Gastroenterol ; 16(21): 2648-56, 2010 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-20518087

RESUMO

AIM: To evaluate the prophylactic properties of integrin CD18-betaA peptide in a murine model of abdominal polymicrobial peritonitis and sepsis. METHODS: Bacterial sepsis was induced in Institute of Cancer Research (ICR) mice by cecal ligation and puncture (CLP) surgery. Inflicted mice were then injected with either sterile saline or CD18-betaA peptide intraperitoneally at 2 h after surgery, and were sacrificed at 12 and 24 h after surgery. Blood samples were immediately collected, and analyzed for endotoxin activity and tumor necrosis factor (TNF)-alpha and interleukin (IL)-6. Lungs and liver were studied for CD45+ leukocyte and CD3 mRNA content. Pulmonary expression of intercellular adhesion molecule (ICAM)-1, vascular cell adhesion molecule (VCAM) and E-selectin was also determined. RESULTS: Intraperitoneal injection of CD18-betaA peptide significantly suppressed circulating endotoxin activity (P < 0.01) at 24 h, as well as serum levels of TNF-alpha (P < 0.05 at 12 and 24 h) and IL-6 (P < 0.01 at 12 h, P < 0.05 at 24 h) in CLP-inflicted mice. CD18-betaA peptide also abrogated leukocyte infiltration into liver and lungs as unveiled by reduced CD45+ leukocyte and CD3 mRNA contents. Furthermore, the peptide significantly reduced pulmonary expression of VCAM (P < 0.01 at 12 h, P < 0.001 at 24 h), E-selectin (P < 0.01 at 12 and 24 h), and ICAM-1 (P < 0.01 at 12 h, P < 0.001 at 24 h). These actions of CD18-betaA peptide collectively protected septic mice against lethality (P < 0.01). CONCLUSION: CD18-betaA peptide is a potent endotoxin antagonist that can protect surgical patients against sepsis-associated lethality.


Assuntos
Antígenos CD18/uso terapêutico , Peptídeos/uso terapêutico , Peritonite/microbiologia , Peritonite/prevenção & controle , Sepse/microbiologia , Sepse/prevenção & controle , Sequência de Aminoácidos , Animais , Complexo CD3/genética , Complexo CD3/metabolismo , Modelos Animais de Doenças , Selectina E/genética , Selectina E/metabolismo , Endotoxinas/antagonistas & inibidores , Endotoxinas/sangue , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-6/sangue , Interleucina-6/imunologia , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Peritonite/sangue , Peritonite/mortalidade , Sepse/sangue , Sepse/mortalidade , Taxa de Sobrevida , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/imunologia , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
13.
Infect Immun ; 78(4): 1426-36, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20100861

RESUMO

Innate immunity to Candida albicans depends upon the recognition of molecular patterns on the fungal cell wall. However, the masking of major components such as beta-glucan seems to be a mechanism that fungi have evolved to avoid immune cell recognition through the dectin-1 receptor. Although the role of C. albicans mitogen-activated protein kinase (MAPK) pathways as virulence determinants has been established previously with animal models, the mechanism involved in this behavior is largely unknown. In this study we demonstrate that a disruption of the C. albicans extracellular signal-regulated kinase (ERK)-like 1 (CEK1)-mediated MAPK pathway causes enhanced cell wall beta-glucan exposure, triggering immune responses more efficiently than the wild type, as measured by dectin-1-mediated specific binding and human dendritic cell (hDC)- and macrophage-mediated phagocytosis, killing, and activation of intracellular signaling pathways. At the molecular level, the disruption of CEK1 resulted in altered spleen tyrosine kinase (Syk), Raf-1, and ERK1/2 activations together with IkappaB degradation on hDCs and increased dectin-1-dependent activator protein 1 (AP-1) activation on transfected cells. In addition, concurring with these altered pathways, we detected increased reactive oxygen species production and cytokine secretion. In conclusion, the CEK1-mediated MAPK pathway is involved in beta-glucan exposure in a fungal pathogen, hence influencing dectin-1-dependent immune cell recognition, thus establishing this fungal intracellular signaling route as a promising novel therapeutic target.


Assuntos
Candida albicans/imunologia , Candida albicans/fisiologia , Proteínas Fúngicas/fisiologia , Regulação Fúngica da Expressão Gênica , Proteínas de Membrana/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Proteínas do Tecido Nervoso/metabolismo , beta-Glucanas/metabolismo , Candida albicans/genética , Adesão Celular , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Proteínas Fúngicas/genética , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Lectinas Tipo C , Macrófagos/imunologia , Macrófagos/microbiologia , Proteínas de Membrana/imunologia , Viabilidade Microbiana , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteínas do Tecido Nervoso/imunologia , Fagocitose , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo , Baço/imunologia , beta-Glucanas/imunologia
14.
J Immunol ; 184(4): 1918-30, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20083673

RESUMO

T cell/transmembrane, Ig, and mucin (TIM) proteins, identified using a congenic mouse model of asthma, critically regulate innate and adaptive immunity. TIM-1 and TIM-4 are receptors for phosphatidylserine (PtdSer), exposed on the surfaces of apoptotic cells. Herein, we show with structural and biological studies that TIM-3 is also a receptor for PtdSer that binds in a pocket on the N-terminal IgV domain in coordination with a calcium ion. The TIM-3/PtdSer structure is similar to that of TIM-4/PtdSer, reflecting a conserved PtdSer binding mode by TIM family members. Fibroblastic cells expressing mouse or human TIM-3 bound and phagocytosed apoptotic cells, with the BALB/c allelic variant of mouse TIM-3 showing a higher capacity than the congenic C.D2 Es-Hba-allelic variant. These functional differences were due to structural differences in the BC loop of the IgV domain of the TIM-3 polymorphic variants. In contrast to fibroblastic cells, T or B cells expressing TIM-3 formed conjugates with but failed to engulf apoptotic cells. Together these findings indicate that TIM-3-expressing cells can respond to apoptotic cells, but the consequence of TIM-3 engagement of PtdSer depends on the polymorphic variants of and type of cell expressing TIM-3. These findings establish a new paradigm for TIM proteins as PtdSer receptors and unify the function of the TIM gene family, which has been associated with asthma and autoimmunity and shown to modulate peripheral tolerance.


Assuntos
Alelos , Apoptose/imunologia , Variação Genética/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mucina-3/genética , Fagocitose/imunologia , Fosfatidilserinas/metabolismo , Sequência de Aminoácidos , Animais , Apoptose/genética , Linhagem Celular , Linhagem Celular Tumoral , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mucina-3/metabolismo , Mucina-3/fisiologia , Família Multigênica/imunologia , Células NIH 3T3 , Fagocitose/genética
15.
Curr Opin Struct Biol ; 19(2): 181-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19342221

RESUMO

Several structures of complexes between viral attachment proteins and their cellular receptors have been determined recently, enhancing our understanding of the molecular recognition processes that guide formation of virus-receptor complexes. Moreover, these structures also highlight strategies by which highly similar viral proteins within a single virus family can adapt to engage different receptors. Consequences of such differences are altered tropism and pathogenicity. An improved understanding of the molecular details of this specificity switching in receptor binding will help to establish links between receptor tropism, spread, and disease. Moreover, it also has relevance for the design and use of viruses as gene delivery vehicles with altered properties as well as for the identification of target viral epitopes of new vaccines.


Assuntos
Receptores Virais/metabolismo , Vírus/metabolismo , Adenoviridae/química , Adenoviridae/metabolismo , Sequência de Aminoácidos , Animais , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Epitopos , Técnicas de Transferência de Genes , Humanos , Proteína Cofatora de Membrana/química , Proteína Cofatora de Membrana/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Paramyxoviridae/química , Paramyxoviridae/metabolismo , Conformação Proteica , Receptores Virais/química , Alinhamento de Sequência , Vírus/química
16.
Immunity ; 27(6): 927-40, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18082433

RESUMO

The T cell immunoglobulin mucin (TIM) proteins regulate T cell activation and tolerance. Here we showed that TIM-4 is expressed on human and mouse macrophages and dendritic cells, and both TIM-4 and TIM-1 specifically bound phosphatidylserine (PS) on the surface of apoptotic cells but not any other phospholipid tested. TIM-4(+) peritoneal macrophages, TIM-1(+) kidney cells, and TIM-4- or TIM-1-transfected cells efficiently phagocytosed apoptotic cells, and phagocytosis could be blocked by TIM-4 or TIM-1 monoclonal antibodies. Mutations in the unique cavity of TIM-4 eliminated PS binding and phagocytosis. TIM-4 mAbs that blocked PS binding and phagocytosis mapped to epitopes in this binding cavity. These results show that TIM-4 and TIM-1 are immunologically restricted members of the group of receptors whose recognition of PS is critical for the efficient clearance of apoptotic cells and prevention of autoimmunity.


Assuntos
Apoptose , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Fagocitose , Fosfatidilserinas/metabolismo , Receptores Virais/metabolismo , Animais , Células Dendríticas/metabolismo , Receptor Celular 1 do Vírus da Hepatite A , Humanos , Ativação Linfocitária , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas de Membrana/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Fosfatidilserinas/química , Linfócitos T/imunologia
17.
Immunity ; 26(3): 299-310, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17363299

RESUMO

The T cell immunoglobulin mucin (TIM) receptors are involved in the regulation of immune responses, autoimmunity, and allergy. Structures of the N-terminal ligand binding domain of the murine mTIM-1 and mTIM-2 receptors revealed an immunoglobulin (Ig) fold, with four Cys residues bridging a distinctive CC' loop to the GFC beta-sheet. The structures showed two ligand-recognition modes in the TIM family. The mTIM-1 structure identified a homophilic TIM-TIM adhesion interaction, whereas the mTIM-2 domain formed a dimer that prevented homophilic binding. Biochemical, mutational, and cell adhesion analyses confirmed the divergent ligand-binding modes revealed by the structures. Structural features characteristic of mTIM-1 appear conserved in human TIM-1, which also mediated homophilic interactions. The extracellular mucin domain enhanced binding through the Ig domain, modulating TIM receptor functions. These results explain the divergent immune functions described for the murine receptors and the role of TIM-1 as a cell adhesion receptor in renal regeneration and cancer.


Assuntos
Proteínas de Membrana/química , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Dimerização , Receptor Celular 1 do Vírus da Hepatite A , Humanos , Imunidade , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Dados de Sequência Molecular , Conformação Proteica , Estrutura Terciária de Proteína , Receptores Imunológicos/química
18.
Nat Struct Mol Biol ; 14(2): 164-6, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17220899

RESUMO

Adenoviruses (Ads) are important human pathogens and valuable gene delivery vehicles. We report here the crystal structure of the species B Ad11 knob complexed with the Ad11-binding region of its receptor CD46. The conformation of bound CD46 differs profoundly from its unbound state, with the bent surface structure straightened into an elongated rod. This mechanism of interaction is likely to be conserved among many pathogens that target CD46 or related molecules.


Assuntos
Adenovírus Humanos/metabolismo , Receptores Virais/química , Proteínas Virais/química , Sítios de Ligação , Cristalografia por Raios X , Humanos , Modelos Moleculares , Conformação Proteica , Receptores Virais/metabolismo , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA