Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Nat Commun ; 14(1): 2275, 2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-37080973

RESUMO

Life-threatening bacterial infections in women after childbirth, known as puerperal sepsis, resulted in classical epidemics and remain a global health problem. While outbreaks of puerperal sepsis have been ascribed to Streptococcus pyogenes, little is known about disease mechanisms. Here, we show that the bacterial R28 protein, which is epidemiologically associated with outbreaks of puerperal sepsis, specifically targets the human receptor CEACAM1. This interaction triggers events that would favor the development of puerperal sepsis, including adhesion to cervical cells, suppression of epithelial wound repair and subversion of innate immune responses. High-resolution structural analysis showed that an R28 domain with IgI3-like fold binds to the N-terminal domain of CEACAM1. Together, these findings demonstrate that a single adhesin-receptor interaction can drive the pathogenesis of bacterial sepsis and provide molecular insights into the pathogenesis of one of the most important infectious diseases in medical history.


Assuntos
Infecção Puerperal , Sepse , Infecções Estreptocócicas , Feminino , Humanos , Gravidez , Adesinas Bacterianas/genética , Proteínas de Bactérias/genética , Infecção Puerperal/epidemiologia , Infecção Puerperal/microbiologia , Sepse/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes
3.
EMBO J ; 40(7): e106103, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33522633

RESUMO

Streptococcus agalactiae, also known as group B Streptococcus (GBS), is the major cause of neonatal sepsis in humans. A critical step to infection is adhesion of bacteria to epithelial surfaces. GBS adhesins have been identified to bind extracellular matrix components and cellular receptors. However, several putative adhesins have no host binding partner characterised. We report here that surface-expressed ß protein of GBS binds to human CEACAM1 and CEACAM5 receptors. A crystal structure of the complex showed that an IgSF domain in ß represents a novel Ig-fold subtype called IgI3, in which unique features allow binding to CEACAM1. Bioinformatic assessment revealed that this newly identified IgI3 fold is not exclusively present in GBS but is predicted to be present in adhesins from other clinically important human pathogens. In agreement with this prediction, we found that CEACAM1 binds to an IgI3 domain found in an adhesin from a different streptococcal species. Overall, our results indicate that the IgI3 fold could provide a broadly applied mechanism for bacteria to target CEACAMs.


Assuntos
Adesinas Bacterianas/química , Antígenos CD/química , Antígeno Carcinoembrionário/química , Moléculas de Adesão Celular/química , Adesinas Bacterianas/metabolismo , Animais , Antígenos CD/metabolismo , Sítios de Ligação , Células CHO , Antígeno Carcinoembrionário/metabolismo , Moléculas de Adesão Celular/metabolismo , Cricetinae , Cricetulus , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/metabolismo , Células HeLa , Humanos , Ligação Proteica , Streptococcus agalactiae/metabolismo
4.
Microbiol Mol Biol Rev ; 85(1)2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33239435

RESUMO

Vaccines work primarily by eliciting antibodies, even when recovery from natural infection depends on cellular immunity. Large efforts have therefore been made to identify microbial antigens that elicit protective antibodies, but these endeavors have encountered major difficulties, as witnessed by the lack of vaccines against many pathogens. This review summarizes accumulating evidence that subdominant protein regions, i.e., surface-exposed regions that elicit relatively weak antibody responses, are of particular interest for vaccine development. This concept may seem counterintuitive, but subdominance may represent an immune evasion mechanism, implying that the corresponding region potentially is a key target for protective immunity. Following a presentation of the concepts of immunodominance and subdominance, the review will present work on subdominant regions in several major human pathogens: the protozoan Plasmodium falciparum, two species of pathogenic streptococci, and the dengue and influenza viruses. Later sections are devoted to the molecular basis of subdominance, its potential role in immune evasion, and general implications for vaccine development. Special emphasis will be placed on the fact that a whole surface-exposed protein domain can be subdominant, as demonstrated for all of the pathogens described here. Overall, the available data indicate that subdominant protein regions are of much interest for vaccine development, not least in bacterial and protozoal systems, for which antibody subdominance remains largely unexplored.


Assuntos
Formação de Anticorpos/imunologia , Epitopos Imunodominantes/imunologia , Proteínas de Membrana/imunologia , Vacinas/imunologia , Vírus da Dengue/imunologia , Humanos , Evasão da Resposta Imune/imunologia , Orthomyxoviridae/imunologia , Plasmodium falciparum/imunologia , Domínios Proteicos/imunologia , Streptococcus agalactiae/imunologia , Streptococcus pyogenes/imunologia , Vacinação
5.
Mol Microbiol ; 112(4): 1116-1130, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31290194

RESUMO

Inhibition of cell division is critical for viability under DNA-damaging conditions. DNA damage induces the SOS response that in bacteria inhibits cell division while repairs are being made. In coccoids, such as the human pathogen, Staphylococcus aureus, this process remains poorly studied. Here, we identify SosA as the staphylococcal SOS-induced cell division inhibitor. Overproduction of SosA inhibits cell division, while sosA inactivation sensitizes cells to genotoxic stress. SosA is a small, predicted membrane protein with an extracellular C-terminal domain in which point mutation of residues that are conserved in staphylococci and major truncations abolished the inhibitory activity. In contrast, a minor truncation led to SosA accumulation and a strong cell division inhibitory activity, phenotypically similar to expression of wild-type SosA in a CtpA membrane protease mutant. This suggests that the extracellular C-terminus of SosA is required both for cell division inhibition and for turnover of the protein. Microscopy analysis revealed that SosA halts cell division and synchronizes the cell population at a point where division proteins such as FtsZ and EzrA are localized at midcell, and the septum formation is initiated but unable to progress to closure. Thus, our findings show that SosA is central in cell division regulation in staphylococci.


Assuntos
Divisão Celular/genética , Divisão Celular/fisiologia , Resposta SOS em Genética/fisiologia , Proteínas de Bactérias/metabolismo , Proteínas do Citoesqueleto/metabolismo , Dano ao DNA/genética , Dano ao DNA/fisiologia , Proteínas de Membrana/metabolismo , Resposta SOS em Genética/genética , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
7.
Microbiologyopen ; 4(5): 774-89, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26175306

RESUMO

The M protein of Streptococcus pyogenes, a major bacterial virulence factor, has an amino-terminal hypervariable region (HVR) that is a target for type-specific protective antibodies. Intriguingly, the HVR elicits a weak antibody response, indicating that it escapes host immunity by two mechanisms, sequence variability and weak immunogenicity. However, the properties influencing the immunogenicity of regions in an M protein remain poorly understood. Here, we studied the antibody response to different regions of the classical M1 and M5 proteins, in which not only the HVR but also the adjacent fibrinogen-binding B repeat region exhibits extensive sequence divergence. Analysis of antisera from S. pyogenes-infected patients, infected mice, and immunized mice showed that both the HVR and the B repeat region elicited weak antibody responses, while the conserved carboxy-terminal part was immunodominant. Thus, we identified a correlation between sequence variability and weak immunogenicity for M protein regions. A potential explanation for the weak immunogenicity was provided by the demonstration that protease digestion selectively eliminated the HVR-B part from whole M protein-expressing bacteria. These data support a coherent model, in which the entire variable HVR-B part evades antibody attack, not only by sequence variability but also by weak immunogenicity resulting from protease attack.


Assuntos
Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Variação Genética , Streptococcus pyogenes/genética , Streptococcus pyogenes/imunologia , Animais , Anticorpos Antibacterianos/sangue , Humanos , Evasão da Resposta Imune , Camundongos , Infecções Estreptocócicas/imunologia
8.
J Immunol ; 193(1): 317-26, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24850720

RESUMO

IgA nephropathy (IgAN) is characterized by mesangial cell proliferation and extracellular matrix expansion associated with immune deposits consisting of galactose-deficient polymeric IgA1 and C3. We have previously shown that IgA-binding regions of streptococcal M proteins colocalize with IgA in mesangial immune deposits in patients with IgAN. In the present study, the IgA-binding M4 protein from group A Streptococcus was found to bind to galactose-deficient polymeric IgA1 with higher affinity than to other forms of IgA1, as shown by surface plasmon resonance and solid-phase immunoassay. The M4 protein was demonstrated to bind to mesangial cells not via the IgA-binding region but rather via the C-terminal region, as demonstrated by flow cytometry. IgA1 enhanced binding of M4 to mesangial cells, but not vice versa. Costimulation of human mesangial cells with M4 and galactose-deficient polymeric IgA1 resulted in a significant increase in IL-6 secretion compared with each stimulant alone. Galactose-deficient polymeric IgA1 alone, but not M4, induced C3 secretion from the cells, and costimulation enhanced this effect. Additionally, costimulation enhanced mesangial cell proliferation compared with each stimulant alone. These results indicate that IgA-binding M4 protein binds preferentially to galactose-deficient polymeric IgA1 and that these proteins together induce excessive proinflammatory responses and proliferation of human mesangial cells. Thus, tissue deposition of streptococcal IgA-binding M proteins may contribute to the pathogenesis of IgAN.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Transporte/imunologia , Complemento C3/imunologia , Glomerulonefrite por IGA/imunologia , Imunoglobulina A/imunologia , Interleucina-6/imunologia , Células Mesangiais/imunologia , Streptococcus/imunologia , Adolescente , Feminino , Glomerulonefrite por IGA/patologia , Humanos , Masculino , Células Mesangiais/patologia , Pessoa de Meia-Idade
9.
PLoS One ; 8(11): e81303, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24278416

RESUMO

Recent studies indicate that defective activity of complement factor H (FH) is associated with several human diseases, suggesting that pure FH may be used for therapy. Here, we describe a simple method to isolate human FH, based on the specific interaction between FH and the hypervariable region (HVR) of certain Streptococcus pyogenes M proteins. Special interest was focused on the FH polymorphism Y402H, which is associated with the common eye disease age-related macular degeneration (AMD) and has also been implicated in the binding to M protein. Using a fusion protein containing two copies of the M5-HVR, we found that the Y402 and H402 variants of FH could be efficiently purified by single-step affinity chromatography from human serum containing the corresponding protein. Different M proteins vary in their binding properties, and the M6 and M5 proteins, but not the M18 protein, showed selective binding of the FH Y402 variant. Accordingly, chromatography on a fusion protein derived from the M6-HVR allowed enrichment of the Y402 protein from serum containing both variants. Thus, the exquisite binding specificity of a bacterial protein can be exploited to develop a simple and robust procedure to purify FH and to enrich for the FH variant that protects against AMD.


Assuntos
Antígenos de Bactérias/química , Proteínas da Membrana Bacteriana Externa/química , Proteínas de Transporte/química , Cromatografia de Afinidade , Peptídeos/química , Substituição de Aminoácidos , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/metabolismo , Fator H do Complemento/química , Fator H do Complemento/genética , Fator H do Complemento/isolamento & purificação , Fator H do Complemento/metabolismo , Variação Genética , Humanos , Peptídeos/metabolismo , Ligação Proteica
10.
PLoS Pathog ; 9(4): e1003323, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23637608

RESUMO

Many pathogens express a surface protein that binds the human complement regulator factor H (FH), as first described for Streptococcus pyogenes and the antiphagocytic M6 protein. It is commonly assumed that FH recruited to an M protein enhances virulence by protecting the bacteria against complement deposition and phagocytosis, but the role of FH-binding in S. pyogenes pathogenesis has remained unclear and controversial. Here, we studied seven purified M proteins for ability to bind FH and found that FH binds to the M5, M6 and M18 proteins but not the M1, M3, M4 and M22 proteins. Extensive immunochemical analysis indicated that FH binds solely to the hypervariable region (HVR) of an M protein, suggesting that selection has favored the ability of certain HVRs to bind FH. These FH-binding HVRs could be studied as isolated polypeptides that retain ability to bind FH, implying that an FH-binding HVR represents a distinct ligand-binding domain. The isolated HVRs specifically interacted with FH among all human serum proteins, interacted with the same region in FH and showed species specificity, but exhibited little or no antigenic cross-reactivity. Although these findings suggested that FH recruited to an M protein promotes virulence, studies in transgenic mice did not demonstrate a role for bound FH during acute infection. Moreover, phagocytosis tests indicated that ability to bind FH is neither sufficient nor necessary for S. pyogenes to resist killing in whole human blood. While these data shed new light on the HVR of M proteins, they suggest that FH-binding may affect S. pyogenes virulence by mechanisms not assessed in currently used model systems.


Assuntos
Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Fator H do Complemento/imunologia , Fator H do Complemento/metabolismo , Streptococcus pyogenes/imunologia , Streptococcus pyogenes/patogenicidade , Animais , Sítios de Ligação , Proteína de Ligação ao Complemento C4b/metabolismo , Fator H do Complemento/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fagocitose , Ligação Proteica , Estrutura Terciária de Proteína , Especificidade da Espécie , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/metabolismo
11.
Adv Exp Med Biol ; 946: 87-112, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21948364

RESUMO

A common site in the constant region (Fc) of immunoglobulins is recognized by host receptors and is a frequent target of proteins expressed by pathogens. This site is located at the junction of two constant domains in the antibody heavy chains and produces a large shallow cavity formed by loops of the CH2 and CH3 domains in IgG and IgA (CH3 and CH4 domains in IgM). Crystal structures have been determined for complexes of IgG-Fc and IgA-Fc with a structurally diverse set of host, pathogen and in vitro selected ligands. While pathogen proteins may directly block interactions with the immunoglobulins thereby evading host immunity, it is likely that the same pathogen molecules also interact with other host factors to carry out their primary biological function. Herein we review the structural and functional aspects of host and pathogen molecular recognition of the common site on the Fc of immunoglobulins. We also propose that some pathogen proteins may promote virulence by affecting the bridging between innate and adaptive immunity.


Assuntos
Anticorpos Antibacterianos/química , Anticorpos Antibacterianos/imunologia , Infecções Bacterianas/imunologia , Sítios de Ligação de Anticorpos/imunologia , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/imunologia , Bactérias/imunologia , Humanos , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
12.
Cell Host Microbe ; 10(2): 147-57, 2011 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-21843871

RESUMO

Sequence variation of antigenic proteins allows pathogens to evade antibody attack. The variable protein commonly includes a hypervariable region (HVR), which represents a key target for antibodies and is therefore predicted to be immunodominant. To understand the mechanism(s) of antibody evasion, we analyzed the clinically important HVR-containing M proteins of the human pathogen Streptococcus pyogenes. Antibodies elicited by M proteins were directed almost exclusively against the C-terminal part and not against the N-terminal HVR. Similar results were obtained for mice and humans with invasive S. pyogenes infection. Nevertheless, only anti-HVR antibodies protected efficiently against infection, as shown by passive immunizations. The HVR fused to an unrelated protein elicited no antibodies, implying that it is inherently weakly immunogenic. These data indicate that the M protein HVR evades antibody attack not only through antigenic variation but also by weak immunogenicity, a paradoxical observation that may apply to other HVR-containing proteins.


Assuntos
Variação Antigênica , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/metabolismo , Evasão da Resposta Imune , Streptococcus pyogenes/imunologia , Sequência de Aminoácidos , Animais , Formação de Anticorpos , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Transporte/administração & dosagem , Proteínas de Transporte/imunologia , Humanos , Soros Imunes/administração & dosagem , Soros Imunes/imunologia , Imunização Passiva , Imunização Secundária , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fagocitose , Coelhos , Proteínas Recombinantes de Fusão/imunologia , Testes Sorológicos/métodos , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/crescimento & desenvolvimento , Streptococcus pyogenes/patogenicidade , Vacinação
13.
Nephrol Dial Transplant ; 25(10): 3434-6, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20558662

RESUMO

BACKGROUND: IgA nephropathy (IgAN), the most common glomerulonephritis worldwide, is characterized by mesangial deposits containing predominantly IgA. IgAN commonly occurs or exacerbates after upper respiratory tract infections such as streptococcal pharyngitis. Certain group A streptococci express M proteins with IgA-binding regions (IgA-BRs). We have previously shown that these IgA-BRs co-localize with mesangial IgA in IgAN. METHODS: Blood samples from patients with IgAN (n = 21) and age-matched controls (n = 83) were assayed by ELISA to detect an IgG antibody response to the IgA-BRs of the M4, M22 and M60 proteins. Antibodies were assayed for each IgA-BR separately and the results were combined. RESULTS: Antibody levels to the IgA-BRs were significantly higher in IgAN patients than controls (P = 0.016), particularly in patients with recent streptococcal infection (P = 0.008). Conclusions. The results suggest that children with IgAN had a previous infection with a streptococcal strain expressing an IgA-binding M protein.


Assuntos
Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Transporte/imunologia , Glomerulonefrite por IGA/imunologia , Imunoglobulina A/metabolismo , Adolescente , Adulto , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/metabolismo , Criança , Pré-Escolar , Feminino , Humanos , Masculino
14.
Am J Pathol ; 176(2): 608-18, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20056836

RESUMO

IgA nephropathy (IgAN) and Henoch-Schönlein purpura (HSP) are diseases characterized by IgA deposits in the kidney and/or skin. Both may arise after upper respiratory tract infections, but the pathogenic mechanisms governing these diseases remain unclear. Patients with IgAN (n = 16) and HSP (n = 17) were included in this study aimed at examining whether IgA-binding M proteins of group A streptococci could be involved. As M proteins vary in sequence, the study focused on the IgA-binding-region (IgA-BR) of three different M proteins: M4, M22, and M60. Renal tissue from IgAN and HSP patients and skin from HSP patients were examined for deposits of streptococcal IgA-BR by immunohistochemistry and electron microscopy using specific antibodies, and a skin sample from a HSP patient was examined by mass spectrometry. IgA-BR deposits were detected in 10/16 IgAN kidneys and 7/13 HSP kidneys. Electron microscopy demonstrated deposits of IgA-BRs in the mesangial matrix and glomerular basement membrane, which colocalized with IgA. Skin samples exhibited IgA-BR deposits in 4/5 biopsies, a result confirmed by mass spectrometry in one patient. IgA-BR deposits were not detected in normal kidney and skin samples. Taken together, these results demonstrate IgA-BR from streptococcal M proteins in patient tissues. IgA-BR, would on gaining access to the circulation, encounter circulatory IgA and form a complex with IgA-Fc that could deposit in tissues and contribute to the pathogenesis of IgAN and HSP.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/metabolismo , Glomerulonefrite por IGA/metabolismo , Vasculite por IgA/metabolismo , Imunoglobulina A/metabolismo , Precipitinas/metabolismo , Adolescente , Adulto , Sequência de Aminoácidos , Antígenos de Bactérias/química , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/imunologia , Biópsia , Proteínas de Transporte/química , Proteínas de Transporte/imunologia , Criança , Pré-Escolar , Feminino , Glomerulonefrite por IGA/patologia , Humanos , Vasculite por IgA/patologia , Imuno-Histoquímica , Rim/metabolismo , Rim/patologia , Masculino , Microscopia Eletrônica , Dados de Sequência Molecular , Precipitinas/ultraestrutura , Ligação Proteica , Pele/metabolismo , Pele/patologia , Pele/ultraestrutura , Adulto Jovem
15.
PLoS One ; 4(10): e7279, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19794915

RESUMO

The surface-localized M protein of Streptococcus pyogenes is a major virulence factor that inhibits phagocytosis, as determined ex vivo. Because little is known about the role of M protein in vivo we analyzed the contribution of different M protein regions to virulence, using the fibrinogen (Fg)-binding M5 protein and a mouse model of acute invasive infection. This model was suitable, because M5 is required for mouse virulence and binds mouse and human Fg equally well, as shown here. Mixed infection experiments with wild type bacteria demonstrated that mutants lacking the N-terminal hypervariable region (HVR) or the Fg-binding B-repeat region were strongly attenuated, while a mutant lacking the conserved C-repeats was only slightly attenuated. Because the HVR of M5 is not required for phagocytosis resistance, our data imply that this HVR plays a major but unknown role during acute infection. The B-repeat region is required for phagocytosis resistance and specifically binds Fg, suggesting that it promotes virulence by binding Fg. However, B-repeat mutants were attenuated even in Fg-deficient mice, implying that the B-repeats may have a second function, in addition to Fg-binding. These data demonstrate that two distinct M5 regions, including the HVR, are essential to virulence during the early stages of an infection. In particular, our data provide the first in vivo evidence that the HVR of an M protein plays a major role in virulence, focusing interest on the molecular role of this region.


Assuntos
Antígenos de Bactérias/química , Proteínas da Membrana Bacteriana Externa/química , Proteínas de Transporte/química , Regiões Determinantes de Complementaridade/imunologia , Streptococcus pyogenes/metabolismo , Virulência , Animais , Cromatografia de Afinidade/métodos , Feminino , Fibrinogênio/química , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Fagocitose , Ligação Proteica , Estrutura Terciária de Proteína , Streptococcus pyogenes/imunologia
16.
J Exp Med ; 206(8): 1691-9, 2009 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-19596804

RESUMO

Group B Streptococcus (GBS) is a leading cause of invasive bacterial infections in human newborns. A key GBS virulence factor is its capsular polysaccharide (CPS), displaying terminal sialic acid (Sia) residues which block deposition and activation of complement on the bacterial surface. We recently demonstrated that GBS Sia can bind human CD33-related Sia-recognizing immunoglobulin (Ig) superfamily lectins (hCD33rSiglecs), a family of inhibitory receptors expressed on the surface of leukocytes. We report the unexpected discovery that certain GBS strains may bind one such receptor, hSiglec-5, in a Sia-independent manner, via the cell wall-anchored beta protein, resulting in recruitment of SHP protein tyrosine phosphatases. Using a panel of WT and mutant GBS strains together with Siglec-expressing cells and soluble Siglec-Fc chimeras, we show that GBS beta protein binding to Siglec-5 functions to impair human leukocyte phagocytosis, oxidative burst, and extracellular trap production, promoting bacterial survival. We conclude that protein-mediated functional engagement of an inhibitory host lectin receptor promotes bacterial innate immune evasion.


Assuntos
Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Lectinas/imunologia , Fagócitos/imunologia , Fagócitos/microbiologia , Streptococcus agalactiae/imunologia , Streptococcus agalactiae/patogenicidade , Antígenos de Bactérias/química , Antígenos de Bactérias/metabolismo , Sítios de Ligação , Adesão Celular , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Humanos , Imunidade Inata , Técnicas In Vitro , Leucócitos/imunologia , Leucócitos/metabolismo , Leucócitos/microbiologia , Mutação , Ácido N-Acetilneuramínico/metabolismo , Fagócitos/metabolismo , Fagocitose , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Tirosina Fosfatases/metabolismo , Explosão Respiratória , Streptococcus agalactiae/genética , Virulência/imunologia
17.
Eur J Immunol ; 39(4): 1147-56, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19266484

RESUMO

Here we unravel the structural features of human IgM and IgA that govern their interaction with the human Fcalpha/mu receptor (hFcalpha/muR). Ligand polymerization status was crucial for the interaction, because hFcalpha/muR binding did not occur with monomeric Ab of either class. hFcalpha/muR bound IgM with an affinity in the nanomolar range, whereas the affinity for dimeric IgA (dIgA) was tenfold lower. Panels of mutant IgM and dIgA were used to identify regions critical for hFcalpha/muR binding. IgM binding required contributions from both Cmu3 and Cmu4 Fc domains, whereas for dIgA, an exposed loop in the Calpha3 domain was crucial. This loop, comprising residues Pro440-Phe443, lies at the Fc domain interface and has been implicated in the binding of host receptors FcalphaRI and polymeric Ig receptor (pIgR), as well as IgA-binding proteins produced by certain pathogenic bacteria. Substitutions within the Pro440-Phe443 loop resulted in loss of hFcalpha/muR binding. Furthermore, secretory component (SC, the extracellular portion of pIgR) and bacterial IgA-binding proteins were shown to inhibit the dIgA-hFcalpha/muR interaction. Therefore, we have identified a motif in the IgA-Fc inter-domain region critical for hFcalpha/muR interaction, and highlighted the multi-functional nature of a key site for protein-protein interaction at the IgA Fc domain interface.


Assuntos
Afinidade de Anticorpos , Imunoglobulina A/química , Imunoglobulina M/química , Receptores Fc/imunologia , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Afinidade de Anticorpos/genética , Afinidade de Anticorpos/imunologia , Células COS , Chlorocebus aethiops , Humanos , Imunoglobulina A/genética , Imunoglobulina A/imunologia , Imunoglobulina M/genética , Imunoglobulina M/imunologia , Proteínas Mutantes/imunologia , Mutação , Domínios e Motivos de Interação entre Proteínas/imunologia , Multimerização Proteica , Estrutura Terciária de Proteína , Receptores Fc/genética
18.
Biomaterials ; 30(13): 2653-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19171378

RESUMO

The aim of this study was to evaluate a new approach to inhibit complement activation triggered by biomaterial surfaces in contact with blood. In order to inhibit complement activation initiated by the classical pathway (CP), we used streptococcal M protein-derived peptides that specifically bind human C4BP, an inhibitor of the CP. The peptides were used to coat polystyrene microtiter wells which served as a model biomaterial. The ability of coated peptides to bind C4BP and to attenuate complement activation via the CP (monitored as generation of fluid-phase C3a and binding of fragments of C3 and C4 to the surface) was investigated using diluted normal human serum, where complement activation by the AP is minimal, as well as serum from a patient lacking alternative pathway activation. Complement activation (all parameters) was significantly decreased in serum incubated in well surfaces coated with peptides. Total inhibition of complement activation was obtained at peptide coating concentrations as low as 1-5 microg/mL. Successful use of Streptococcus-derived peptides shows that it is feasible to control complement activation at a model biomaterial surface by capturing autologous complement regulatory molecules from plasma.


Assuntos
Antígenos de Bactérias/química , Proteínas da Membrana Bacteriana Externa/química , Materiais Biocompatíveis/química , Proteínas de Transporte/química , Modelos Biológicos , Fragmentos de Peptídeos/química , Proteína de Ligação ao Complemento C4b , Antígenos de Histocompatibilidade/metabolismo , Humanos , Ligação Proteica , Propriedades de Superfície
19.
Cell Host Microbe ; 2(6): 427-34, 2007 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-18078694

RESUMO

Identification of antigens that elicit protective immunity is essential for effective vaccine development. We investigated the related surface proteins of group B Streptococcus, Rib and alpha, as potential vaccine candidates. Paradoxically, nonimmunodominant regions proved to be of particular interest as vaccine components. Mouse antibodies elicited by Rib and alpha were directed almost exclusively against the C-terminal repeats and not against the N-terminal regions. However, a fusion protein derived from the nonimmunodominant N-terminal regions of Rib and alpha was much more immunogenic than one derived from the repeats and was immunogenic even without adjuvant. Moreover, antibodies to the N-terminal fusion protein protected against infection and inhibited bacterial invasion of epithelial cells. Similarly, the N-terminal region of Streptococcus pyogenes M22 protein, which is targeted by opsonic antibodies, is nonimmunodominant. These data indicate that nonimmunodominant regions of bacterial antigens could be valuable for vaccine development.


Assuntos
Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Antígenos de Superfície/imunologia , Proteínas de Bactérias/imunologia , ISCOMs/imunologia , Proteínas de Membrana/imunologia , Infecções Estreptocócicas/sangue , Infecções Estreptocócicas/prevenção & controle , Vacinas Estreptocócicas/imunologia , Streptococcus agalactiae/imunologia , Animais , Células Epiteliais/microbiologia , Epitopos/imunologia , Imunização , Camundongos , Camundongos Endogâmicos C3H , Streptococcus pyogenes/imunologia
20.
J Mol Biol ; 369(1): 69-78, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17418236

RESUMO

Glycosylation defects occur in several human diseases. In IgA nephropathy, IgA1 contains O-glycans that are galactose-deficient and consist mostly of core 1 alpha2,6 sialylated N-acetylgalactosamine, a configuration suspected to prevent beta1,3 galactosylation. We confirmed the same aberrancy in IgA1 secreted by the human DAKIKI B cell line. Biochemical assays indicated CMP-NeuAc:GalNAc-IgA1 alpha2,6-sialyltransferase activity in this cell line. However, a candidate enzyme, ST6-GalNAcI, was not transcribed in DAKIKI cells, B cells isolated from blood, or Epstein-Barr virus (EBV)-immortalized IgA1-producing cells from the blood of IgAN patients and healthy controls. Instead, ST6-GalNAcII transcription was detected at a high level. Expression of the ST6-GalNAcII gene and activity of the CMP-NeuAc:GalNAc-IgA1 alpha2,6-sialyltransferase were higher in IgA1-producing cell lines from IgAN patients than in such cells from healthy controls. These data are the first evidence that human cells that lack ST6-GalNAcI can sialylate core 1 GalNAc-Ser/Thr.


Assuntos
Imunoglobulina A/biossíntese , Sialiltransferases/metabolismo , Linhagem Celular , Linhagem Celular Transformada , Ensaio de Imunoadsorção Enzimática , Regulação Enzimológica da Expressão Gênica , Glicosilação , Células HT29 , Herpesvirus Humano 4/metabolismo , Humanos , Lectinas/metabolismo , Leucócitos Mononucleares/enzimologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Reversa/genética , Sialiltransferases/genética , beta-D-Galactosídeo alfa 2-6-Sialiltransferase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA