Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Mucosal Immunol ; 10(2): 385-394, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27579859

RESUMO

The ability of pneumococcal conjugate vaccine (PCV) to decrease transmission by blocking the acquisition of colonization has been attributed to herd immunity. We describe the role of mucosal immunoglobulin G (IgG) to capsular polysaccharide (CPS) in mediating protection from carriage, translating our findings from a murine model to humans. We used a flow cytometric assay to quantify antibody-mediated agglutination demonstrating that hyperimmune sera generated against an unencapsulated mutant was poorly agglutinating. Passive immunization with this antiserum was ineffective to block acquisition of colonization compared to agglutinating antisera raised against the encapsulated parent strain. In the human challenge model, samples were collected from PCV and control-vaccinated adults. In PCV-vaccinated subjects, IgG levels to CPS were increased in serum and nasal wash (NW). IgG to the inoculated strain CPS dropped in NW samples after inoculation suggesting its sequestration by colonizing pneumococci. In post-vaccination NW samples pneumococci were heavily agglutinated compared with pre-vaccination samples in subjects protected against carriage. Our results indicate that pneumococcal agglutination mediated by CPS-specific antibodies is a key mechanism of protection against acquisition of carriage. Capsule may be the only vaccine target that can elicit strong agglutinating antibody responses, leading to protection against carriage acquisition and generation of herd immunity.


Assuntos
Aglutinação , Anticorpos Antibacterianos/metabolismo , Infecções Pneumocócicas/imunologia , Vacinas Pneumocócicas/imunologia , Streptococcus pneumoniae/imunologia , Adolescente , Adulto , Animais , Cápsulas Bacterianas/imunologia , Portador Sadio , Feminino , Humanos , Imunização Passiva , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Infecções Pneumocócicas/prevenção & controle , Vacinação , Vacinas Conjugadas , Adulto Jovem
2.
Mucosal Immunol ; 10(1): 250-259, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27118490

RESUMO

Pneumonia caused by Streptococcus pneumoniae (Sp) remains a leading cause of serious illness and death worldwide. Immunization with conjugated pneumococcal vaccine has lowered the colonization rate and consequently invasive diseases by inducing serotype-specific antibodies. However, many of the current pneumonia cases result from infection by serotype strains not included in the vaccine. In this study, we asked if cross-protection against lung infection by heterologous strains can be induced, and investigated the underlying immune mechanism. We found that immune mice recovered from a prior infection were protected against heterologous Sp strains in the pneumonia challenge model, as evident by accelerated bacterial clearance, reduced pathology, and apoptosis of lung epithelial cells. Sp infection in the lung induced strong T-helper type 17 (Th17) responses at the lung mucosal site. Transfer of CD4+ T cells from immune mice provided heterologous protection against pneumonia, and this protection was abrogated by interleukin-17A (IL-17A) blockade. Transfer of memory CD4+ T cells from IL-17A-knockout mice failed to provide protection. These results indicate that memory Th17 cells had a key role in providing protection against pneumonia in a serotype-independent manner and suggest the feasibility of developing a broadly protective vaccine against bacterial pneumonia by targeting mucosal Th17 T cells.


Assuntos
Reações Cruzadas , Interleucina-17/metabolismo , Vacinas Pneumocócicas/imunologia , Pneumonia/imunologia , Mucosa Respiratória/imunologia , Streptococcus pneumoniae/imunologia , Células Th17/imunologia , Animais , Carga Bacteriana , Células Cultivadas , Feminino , Humanos , Imunidade nas Mucosas , Memória Imunológica , Interleucina-17/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucosa Respiratória/microbiologia , Especificidade da Espécie , Células Th17/microbiologia
3.
Mucosal Immunol ; 8(1): 176-85, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24962092

RESUMO

Invasive infection often begins with asymptomatic colonization of mucosal surfaces. A murine model of bacterial colonization with Streptococcus pneumoniae was used to study the mechanism for mucosal protection by immunoglobulin. In previously colonized immune mice, bacteria were rapidly sequestered within large aggregates in the nasal lumen. To further examine the role of bacterial agglutination in protection by specific antibodies, mice were passively immunized with immunoglobulin G (IgG) purified from antipneumococcal sera or pneumococcal type-specific monoclonal human IgA (hIgA1 or hIgA2). Systemically delivered IgG accessed the mucosal surface and blocked acquisition of colonization and transmission between littermates. Optimal protection by IgG was independent of Fc fragment and complement and, therefore, did not involve an opsonophagocytic mechanism. Enzymatic digestion or reduction of IgG before administration showed that protection required divalent binding that maintained its agglutinating effect. Divalent hIgA1 is cleaved by the pneumococcal member of a family of bacterial proteases that generate monovalent Fabα fragments. Thus, passive immunization with hIgA1 blocked colonization by an IgA1-protease-deficient mutant (agglutinated) but not the protease-producing wild-type parent (not agglutinated), whereas protease-resistant hIgA2 agglutinated and blocked colonization by both. Our findings highlight the importance of agglutinating antibodies in mucosal defense and reveal how successful pathogens evade this effect.


Assuntos
Anticorpos Monoclonais/metabolismo , Imunoglobulina A/metabolismo , Imunoglobulina G/metabolismo , Mucosa Nasal/imunologia , Infecções Pneumocócicas/imunologia , Streptococcus pneumoniae/fisiologia , Aglutinação/genética , Aglutinação/imunologia , Animais , Proteínas de Bactérias/genética , Processos de Crescimento Celular/imunologia , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Humanos , Evasão da Resposta Imune , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Mucosa Nasal/microbiologia , Peptídeo Hidrolases/genética , Infecções Pneumocócicas/transmissão , Streptococcus pneumoniae/crescimento & desenvolvimento
4.
Mucosal Immunol ; 7(2): 249-56, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23820749

RESUMO

Bacterial immunoglobulin A1 (IgA1) proteases may sabotage the protective effects of IgA. In vitro, both exogenous and endogenously produced IgA1 protease inhibited phagocytic killing of Streptococcus pneumoniae by capsule-specific IgA1 human monoclonal antibodies (hMAbs) but not IgA2. These IgA1 proteases cleaved and reduced binding of the the effector Fcα1 heavy chain but not the antigen-binding F(ab)/light chain to pneumococcal surfaces. In vivo, IgA1 protease-resistant IgA2, but not IgA1 protease-sensitive IgA1, supported 60% survival in mice infected with wild-type S. pneumoniae. IgA1 hMAbs protected mice against IgA1 protease-deficient but not -producing pneumococci. Parallel mouse sera with human IgA2 showed more efficient complement-mediated reductions in pneumococci with neutrophils than did IgA1, particularly with protease-producing organisms. After natural human pneumococcal bacteremia, purified serum IgG inhibited IgA1 protease activity in 7 of 11 patients (64%). These observations provide the first evidence in vivo that IgA1 protease can circumvent killing of S. pneumoniae by human IgA. Acquisition of IgA1 protease-neutralizing IgG after infection directs attention to IgA1 protease both as a determinant of successful colonization and infection and as a potential vaccine candidate.


Assuntos
Imunoglobulina A/imunologia , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/metabolismo , Serina Endopeptidases/metabolismo , Streptococcus pneumoniae/enzimologia , Streptococcus pneumoniae/imunologia , Animais , Modelos Animais de Doenças , Humanos , Imunoglobulina A/metabolismo , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Camundongos , Fagocitose/imunologia
5.
J Bacteriol ; 187(12): 3969-79, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15937159

RESUMO

Activation of the CiaRH two-component signaling system prevents the development of competence for genetic transformation in Streptococcus pneumoniae through a previously unknown mechanism. Earlier studies have shown that CiaRH controls the expression of htrA, which we show encodes a surface-expressed serine protease. We found that mutagenesis of the putative catalytic serine of HtrA, while not impacting the competence of a ciaRH+ strain, restored a normal competence profile to a strain having a mutation that constitutively activates the CiaH histidine kinase. This result implies that activity of HtrA is necessary for the CiaRH system to inhibit competence. Consistent with this finding, recombinant HtrA (rHtrA) decreased the competence of pneumococcal cultures. The rHtrA-mediated decline in transformation efficiency could not be corrected with excess competence-stimulating peptide (CSP), suggesting that HtrA does not act through degradation of this signaling molecule. The inhibitory effects of rHtrA and activated CiaH, however, were largely overcome in a strain having constitutive activation of the competence pathway through a mutation in the cytoplasmic domain of the ComD histidine kinase. Although these results suggested that HtrA might act through degradation of the extracellular portion of the ComD receptor, Western immunoblots for ComD did not reveal changes in protein levels attributable to HtrA. We therefore postulate that HtrA may act on an unknown protein target that potentiates the activation of the ComDE system by CSP. These findings suggest a novel regulatory role for pneumococcal HtrA in modulating the activity of a two-component signaling system that controls the development of genetic competence.


Assuntos
Serina Endopeptidases/metabolismo , Streptococcus pneumoniae/enzimologia , Streptococcus pneumoniae/genética , Transformação Bacteriana , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/fisiologia , Expressão Gênica , Proteínas de Choque Térmico/química , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Proteínas Periplásmicas/química , Proteínas Quinases/fisiologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Serina Endopeptidases/química , Serina Endopeptidases/genética , Transdução de Sinais
6.
Infect Immun ; 70(8): 4059-67, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12117912

RESUMO

Nasopharyngeal carriage is the reservoir from which most disease with Streptococcus pneumoniae arises. Survival as a commensal in this environment is likely to require a set of adaptations distinct from those needed to cause disease, some of which may be mediated by two-component signal transduction systems (TCSTS). We examined the contributions of nine pneumococcal TCSTS to the process of nasopharyngeal colonization by using an infant rat model. Whereas deletions in all but one of these systems have been associated previously with a high degree of attenuation in a murine model of pneumonia, only the CiaRH system was necessary for efficient carriage. Transcriptional analysis by using microarray hybridization identified a locus consisting of two adjacent genes, htrA and spoJ, that was specifically and strongly downregulated in a DeltaciaRH-null mutant. A S. pneumoniae strain lacking the htrA gene encoding a putative serine protease, but not one lacking spoJ, showed decreased fitness in a competitive model of colonization, a finding consistent with this gene mediating a portion of the carriage deficit observed with the DeltaciaRH strain.


Assuntos
Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Proteínas de Choque Térmico , Proteínas Periplásmicas , Proteínas Quinases/metabolismo , Serina Endopeptidases/genética , Transdução de Sinais , Streptococcus pneumoniae/enzimologia , Animais , Modelos Animais de Doenças , Regulação para Baixo , Genes Bacterianos , Histidina Quinase , Nasofaringe/microbiologia , Análise de Sequência com Séries de Oligonucleotídeos , Infecções Pneumocócicas/microbiologia , Proteínas Quinases/genética , Ratos , Ratos Sprague-Dawley , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/crescimento & desenvolvimento
7.
Mol Microbiol ; 41(5): 1029-36, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11555284

RESUMO

Haemophilus influenzae incorporates choline obtained from environmental sources onto its lipopolysaccharide as phosphorylcholine (ChoP). The decoration of the bacterial surface with ChoP contributes to pathogenesis by allowing for mimicry of the host. As the main reservoir for choline in the host is phosphatidylcholine, we tested whether other choline-containing molecules associated with eukaryotic membranes could provide an alternative source of choline. H. influenzae was able to use glycerophosphorylcholine (GPC), an abundant degradation product of phospholipids, as efficiently as free choline. Utilization of GPC required glpQ, which expresses an enzyme with glycerophosphodiester phosphodiesterase activity. In the absence of free choline, this gene was required for adherent H. influenzae to obtain choline directly from epithelial cells in culture. GlpQ therefore allows choline to be transferred from the host to the bacterial cell surface.


Assuntos
Proteínas de Bactérias/metabolismo , Colina/metabolismo , Haemophilus influenzae/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Adulto , Animais , Proteínas de Bactérias/genética , Feminino , Glicerilfosforilcolina/metabolismo , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/genética , Haemophilus influenzae/isolamento & purificação , Humanos , Nasofaringe/microbiologia , Neoplasias Faríngeas , Diester Fosfórico Hidrolases/genética , Gravidez , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas
8.
Cell Microbiol ; 3(8): 525-36, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11488814

RESUMO

Non-typeable Haemophilus influenzae (NTHi) invades host cells by binding of the platelet-activating factor (PAF) receptor via lipooligosaccharide (LOS) glycoforms containing phosphorylcholine (ChoP). The effect of NTHi infection on host cell signalling and its role in NTHi invasion was examined. The infection of human bronchial epithelial cells with NTHi 2019 increased cytosolic Ca2+ levels, and the invasion of bronchial cells by NTHi 2019 was inhibited by pretreatment with the cell-permeant intracellular Ca2+ chelator BAPTA-AM (P = 0.022) or thapsigargin (P = 0.016). Cytosolic inositol phosphate (IP) levels were also increased after infection with NTHi 2019 (P < 0.001), but not after infection with isogenic mutants expressing altered LOS glycoforms lacking ChoP. PAF receptor antagonist reduced NTHi 2019-stimulated IP production in a dose-dependent manner. NTHi 2019 invasion was inhibited by pertussis toxin (PTX) and the phosphatidylinositol-3-kinase inhibitors wortmannin and LY294002. The less invasive strain NTHi 7502 also initiated IP production, but was unaffected by PAF receptor antagonist or PTX. These data demonstrate that the binding of the PAF receptor by NTHi initiates receptor coupling to a PTX-sensitive heterotrimeric G protein complex, resulting in a multifactorial host cell signal cascade and bacterial invasion. Moreover, the data suggest that NTHi strains initiate cell signalling and invade by different mechanisms, and that invasion mediated by PAF receptor activation is more efficient than macropinocytosis.


Assuntos
Brônquios/microbiologia , Sinalização do Cálcio , Haemophilus influenzae/patogenicidade , Lipopolissacarídeos/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Mucosa Respiratória/microbiologia , Aderência Bacteriana , Técnicas de Tipagem Bacteriana , Brônquios/citologia , Brônquios/metabolismo , Linhagem Celular , Citosol/metabolismo , Haemophilus influenzae/classificação , Humanos , Fosfatos de Inositol/biossíntese , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo
9.
Infect Immun ; 69(9): 5430-9, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11500414

RESUMO

Most isolates of Streptococcus pneumoniae are mixed populations of transparent (T) and opaque (O) colony phenotypes. Differences in the production of capsular polysaccharide (CPS) between O and T variants were accentuated by changes in the environmental concentration of oxygen. O variants demonstrated a 5.2- to 10.6-fold increase in amounts of CPS under anaerobic compared to atmospheric growth conditions, while CPS production remained low under all conditions for T variants. Increased amounts of CPS in O compared to T pneumococci were associated with increased expression of cps-encoded proteins. The inhibitory effect of oxygen on expression of CPS in O variants correlated with decreased tyrosine phosphorylation of CpsD, a tyrosine kinase and regulator of CPS synthesis. Modulation of CpsD expression and its activity by tyrosine phosphorylation may allow the pneumococcus to adapt to the requirements of both colonization, where decreased CPS allows for adherence, and bacteremia, where increased CPS may be required to escape from opsonic clearance. In patients with invasive infection, paired isolates from the same patient were shown to have predominantly a T colony phenotype without phosphotyrosine on CpsD when cultured from the nasopharynx, and an O phenotype that phosphorylates CpsD in response to oxygen when cultured from the blood. Differences in the availability of oxygen, therefore, may be a key factor in allowing for the selection of distinct phenotypes in these two host environments.


Assuntos
Cápsulas Bacterianas/efeitos dos fármacos , Cápsulas Bacterianas/metabolismo , Proteínas de Bactérias , Regulação Bacteriana da Expressão Gênica , Oxigênio/farmacologia , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/metabolismo , Bacteriemia/microbiologia , Portador Sadio/microbiologia , Galactosiltransferases/genética , Galactosiltransferases/metabolismo , Humanos , Dados de Sequência Molecular , Fenótipo , Fosforilação , Streptococcus pneumoniae/classificação , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/isolamento & purificação , Transcrição Gênica , Tirosina/metabolismo
10.
Infect Immun ; 69(3): 1747-54, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11179352

RESUMO

C-reactive protein (CRP) is a normal constituent of human sera synthesized by hepatocytes and induced by proinflammatory cytokines. The function of this acute-phase reactant includes activation of complement and enhancement of opsonophagocytosis. CRP binds to phosphorylcholine (ChoP), a constituent of eukaryotic membranes that is also found on the cell surface of major bacterial pathogens of the human respiratory tract, including Streptococcus pneumoniae and Haemophilus influenzae. The presence of CRP on mucosal surfaces and role in innate immunity in the human respiratory tract where ChoP-containing organisms reside have not been previously studied. We have shown using a monoclonal antibody to CRP that CRP is present in inflamed (0.17 to 42 microg/ml) and uninflamed (<0.05 to 0.88 microg/ml) secretions from the human respiratory tract in sufficient quantities for an antimicrobial effect. In addition, the CRP gene was expressed in epithelial cells of the human respiratory tract using in situ hybridization on nasal polyps and reverse transcriptase PCR of pharyngeal cells in culture. The complement-dependent bactericidal activity of normal nasal airway surface fluid and sputum against ChoP-expressing H. influenzae was abolished when the secretions were pretreated to remove CRP. In summary, the results indicate that CRP is present in secretions of the human respiratory tract, that human respiratory epithelial cells are capable of CRP expression, and that this protein may contribute to bacterial clearance in the human respiratory tract.


Assuntos
Proteína C-Reativa/análise , Sistema Respiratório/imunologia , Adulto , Antibacterianos/análise , Bronquite , Haemophilus influenzae/imunologia , Humanos , Imunidade Inata , Faringe/citologia , Fosforilcolina , Mucosa Respiratória , Escarro , Células Tumorais Cultivadas
11.
Infect Immun ; 69(1): 602-6, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11119562

RESUMO

Phase variation in the colonial opacity of Streptococcus pneumoniae has been implicated as a factor in bacterial adherence, colonization, and invasion in the pathogenesis of pneumococcal disease. Additionally, the synergistic effects of influenza A virus and S. pneumoniae in the development of otitis media (OM) have been reported. This study examined the ability of opaque or transparent S. pneumoniae from the same strain in combination with an antecedent influenza A virus infection to colonize the nasopharynx and invade the middle ear in the chinchilla model. Our data indicated that there was no significant difference in the level of nasopharyngeal colonization and induction of OM between the opaque and transparent variants unless there was a prior challenge with influenza A virus. Subsequent to influenza A virus infection, there was a significant difference between the variants in the ability to colonize and persist in the nasopharynx and middle ear. The concentrations of the opaque variant in nasopharyngeal-lavage samples and middle-ear fluid remained consistently higher than those of the transparent variant for 10 days postinoculation. Data from this study indicate that the effects of influenza A virus on the pathogenesis of experimental S. pneumoniae-induced OM differ depending on the opacity phenotype involved.


Assuntos
Vírus da Influenza A/fisiologia , Nasofaringe/microbiologia , Infecções por Orthomyxoviridae/microbiologia , Otite Média/etiologia , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/isolamento & purificação , Animais , Chinchila , Modelos Animais de Doenças , Fenótipo , Streptococcus pneumoniae/classificação
12.
J Clin Microbiol ; 38(12): 4412-9, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11101573

RESUMO

The lipooligosaccharide (LOS) of Haemophilus somnus undergoes antigenic phase variation, which may facilitate evasion from the bovine host immune response and/or colonization and dissemination. However, LOS antigenic diversity in H. somnus has not been adequately investigated. In this study, monoclonal antibodies (MAbs) specific to various LOS epitopes were used to investigate antigenic variation and stability in LOS from H. somnus strains and phase variants. Clinical isolates of H. somnus exhibited intrastrain, as well as interstrain, antigenic heterogeneity in LOS when probed with MAbs to outer core oligosaccharide epitopes in an enzyme-linked immunosorbent assay (ELISA). However, epitopes reactive with MAbs directed predominately to the inner core heptose region were highly conserved. At least one epitope, which was expressed in few strains, was identified. One LOS component affected by phase variation was identified as phosphorylcholine (PCho), which is linked to the primary glucose residue. Inhibition ELISA, immunoblotting, and electrospray-mass spectrometry were used to confirm that MAb 5F5.9 recognized PCho. LOS reactivity with MAb 5F5.9 was associated with loss of most of the outer core oligosaccharide, indicating that reactivity with PCho was affected by phase variation of the glucose residues in this region. Our results indicate that outer core epitopes of H. somnus LOS exhibit a high degree of random, phase-variable antigenic heterogeneity and that such heterogeneity must be considered in the design of vaccines and diagnostic tests.


Assuntos
Haemophilus/imunologia , Lipopolissacarídeos/imunologia , Fosforilcolina/imunologia , Animais , Anticorpos Monoclonais/imunologia , Bovinos , Ensaio de Imunoadsorção Enzimática , Epitopos , Camundongos
14.
Mol Microbiol ; 37(1): 13-27, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10931302

RESUMO

Adherence and invasion are thought to be key events in the pathogenesis of non-typeable Haemophilus influenzae (NTHi). The role of NTHi lipooligosaccharide (LOS) in adherence was examined using an LOS-coated polystyrene bead adherence assay. Beads coated with NTHi 2019 LOS adhered significantly more to 16HBE14 human bronchial epithelial cells than beads coated with truncated LOS isolated from an NTHi 2019 pgmB:ermr mutant (P = 0.037). Adherence was inhibited by preincubation of cell monolayers with NTHi 2019 LOS (P = 0.0009), but not by preincubation with NTHi 2019 pgmB:ermr LOS. Competitive inhibition studies with a panel of compounds containing structures found within NTHi LOS suggested that a phosphorylcholine (ChoP) moiety was involved in adherence. Further experiments revealed that mutations affecting the oligosaccharide region of LOS or the incorporation of ChoP therein caused significant decreases in the adherence to and invasion of bronchial cells by NTHi 2019 (P < 0.01). Analysis of infected monolayers by confocal microscopy showed that ChoP+ NTHi bacilli co-localized with the PAF receptor. Pretreatment of bronchial cells with a PAF receptor antagonist inhibited invasion by NTHi 2109 and two other NTHi strains expressing ChoP+ LOS glycoforms exhibiting high reactivity with an anti-ChoP antibody on colony immunoblots. These data suggest that a particular subset of ChoP+ LOS glycoforms could mediate NTHi invasion of bronchial cells by means of interaction with the PAF receptor.


Assuntos
Aderência Bacteriana , Brônquios/microbiologia , Haemophilus influenzae/patogenicidade , Lipopolissacarídeos/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores de Superfície Celular , Receptores Acoplados a Proteínas G , Mucosa Respiratória/microbiologia , Ligação Competitiva , Brônquios/citologia , Células Cultivadas , Haemophilus influenzae/classificação , Haemophilus influenzae/fisiologia , Humanos , Microscopia Confocal , Microesferas , Fosfoglucomutase/genética , Fosfoglucomutase/metabolismo , Fosforilcolina/metabolismo , Poliestirenos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
15.
Infect Immun ; 68(8): 4604-10, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10899862

RESUMO

Streptococcus pneumoniae undergoes spontaneous phase variation resulting in opaque and transparent colony forms. Differences in colony opacity correlate with differences in virulence: the transparent variants are more capable of colonizing the nasopharynx, whereas the opaque variants show increased virulence during systemic infections. To gain insight into the pathogenesis of pneumococcal disease at the molecular level, protein expression patterns of the phenotypic variants of two pneumococcal strains were compared by high-resolution two-dimensional protein electrophoresis. In comparison with transparent variants, the opaque variants reduced the expression of two proteins and overexpressed one protein. The proteins were identified by mass spectrometric analysis. The protein overexpressed in the opaque phenotype revealed significant homology to elongation factor Ts of Helicobacter pylori. One of the two proteins that were underexpressed in the opaque variants revealed significant homology to the proteinase maturation protein PrtM of Lactocobacillus paracasei, a member of the family of peptidyl-prolyl cis/trans isomerases. A consensus lipoprotein signal sequence suggests that the putative proteinase maturation protein A, designated PpmA, is located at the surface of the pneumococcus and may play a role in the maturation of surface or secreted proteins. The second underexpressed protein was identified as pyruvate oxidase, SpxB. The lower SpxB expression in opaque variants most probably explains the reduced production of hydrogen peroxide, a reaction product of SpxB, in this variant. Since a spxB-defective pneumococcal mutant has decreased ability to colonize the nasopharynx (B. Spellerberg, D. R. Cundell, J. Sandros, B. J. Pearce, I. Idanpaan-Heikkila, C. Rosenow, and H. R. Masure, 1996. Mol. Microbiol. 19:803-813, 1996), our data suggest that SpxB plays an important role in enhancing the ability of transparent variants to efficiently colonize the nasopharynx.


Assuntos
Proteínas de Bactérias/isolamento & purificação , Variação Genética , Proteínas de Membrana , Streptococcus pneumoniae/citologia , Streptococcus pneumoniae/genética , Sequência de Aminoácidos , Eletroforese em Gel Bidimensional , Endopeptidases/metabolismo , Perfilação da Expressão Gênica , Dados de Sequência Molecular , Fatores de Alongamento de Peptídeos/isolamento & purificação , Fenótipo , Processamento de Proteína Pós-Traducional , Piruvato Oxidase/isolamento & purificação , Análise de Sequência de Proteína
16.
Infect Immun ; 68(7): 3990-7, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10858213

RESUMO

An inverse correlation between colonization of the human nasopharynx by Streptococcus pneumoniae and Haemophilus influenzae, both common upper respiratory pathogens, has been reported. Studies were undertaken to determine if either of these organisms produces substances which inhibit growth of the other. Culture supernatants from S. pneumoniae inhibited growth of H. influenzae, whereas culture supernatants from H. influenzae had no effect on the growth of S. pneumoniae. Moreover, coculture of S. pneumoniae and H. influenzae led to a rapid decrease in viable counts of H. influenzae. The addition of purified catalase prevented killing of H. influenzae in coculture experiments, suggesting that hydrogen peroxide may be responsible for this bactericidal activity. H. influenzae was killed by concentrations of hydrogen peroxide similar to that produced by S. pneumoniae. Hydrogen peroxide is produced by the pneumococcus through the action of pyruvate oxidase (SpxB) under conditions of aerobic growth. Both an spxB mutant and a naturally occurring variant of S. pneumoniae, which is downregulated in SpxB expression, were unable to kill H. influenzae. A catalase-reversible inhibitory effect of S. pneumoniae on the growth of the respiratory tract pathogens Moraxella catarrhalis and Neisseria meningitidis was also observed. Elevated hydrogen peroxide production, therefore, may be a means by which S. pneumoniae is able to inhibit a variety of competing organisms in the aerobic environment of the upper respiratory tract.


Assuntos
Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Sistema Respiratório/microbiologia , Streptococcus pneumoniae/metabolismo , Streptococcus pneumoniae/patogenicidade , Técnicas de Cocultura , Contagem de Colônia Microbiana , Resistência Microbiana a Medicamentos , Variação Genética , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/crescimento & desenvolvimento , Bactérias Gram-Negativas/patogenicidade , Bactérias Gram-Positivas/efeitos dos fármacos , Bactérias Gram-Positivas/crescimento & desenvolvimento , Bactérias Gram-Positivas/patogenicidade , Haemophilus influenzae/efeitos dos fármacos , Haemophilus influenzae/crescimento & desenvolvimento , Haemophilus influenzae/patogenicidade , Humanos , Moraxella catarrhalis/efeitos dos fármacos , Moraxella catarrhalis/crescimento & desenvolvimento , Moraxella catarrhalis/patogenicidade , Mutação , Neisseria meningitidis/efeitos dos fármacos , Neisseria meningitidis/crescimento & desenvolvimento , Neisseria meningitidis/patogenicidade , Infecções Pneumocócicas/microbiologia , Piruvato Oxidase/genética , Piruvato Oxidase/metabolismo , Infecções Respiratórias/microbiologia , Streptococcus pneumoniae/genética , Superinfecção/prevenção & controle
17.
Eur J Biochem ; 267(12): 3902-13, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10849010

RESUMO

Haemophilus influenzae expresses heterogeneous populations of short-chain lipopolysaccharide (LPS) which exhibit extensive antigenic diversity among multiple oligosaccharide epitopes. These LPS oligosaccharide epitopes can carry phosphocholine (PCho) substituents, the expression of which is subject to high frequency phase variation mediated by genes in the lic1 genetic locus. The location and site of attachment of PCho substituents were determined by structural analysis of LPS from two type b H. influenzae strains, Eagan and RM7004. The lic2 locus is involved in phase variation of oligosaccharide expression. LPS obtained from the parent strains, from mutants generated by insertion of antibiotic resistance cassettes in the lic2 genetic locus, and from phase-variants showing high levels of PCho expression was characterized by electrospray ionization-mass spectrometry (ESI-MS) and 1H NMR spectroscopy of derived O-deacylated samples. ESI-MS of O-deacylated LPS from wild-type strains revealed mixtures of related glycoform structures differing in the number of hexose residues. Analysis of LPS from PCho-expressing phase-variants revealed similar mixtures of glycoforms, each containing a single PCho substituent. O-Deacylated LPS preparations from the lic2 mutants were much less complex than their respective parent strains, consisting only of Hex3 and/or Hex2 glycoforms, were examined in detail by high-field NMR techniques. It was found that the LPS samples contain the phosphoethanolamine (PEtn) substituted inner-core element, L-alpha-D-Hepp-(1-->2)-[PEtn-->6]-L-alpha-D-Hepp-(1--> 3)-L-alpha-D-He pp-(1-->5)-alpha-Kdo in which the major glycoforms carry a beta-D-Glcp or beta-D-Glcp-(1-->4)-beta-D-Glcp at the O-4 position of the 3-substituted heptose (HepI) and a beta-D-Galp at the O-2 position of the terminal heptose (HepIII). LPS from the lic2 mutants of both type b strains were found to carry PCho groups at the O-6 position of the terminal beta-D-Galp residue attached to HepIII. In the parent strains, the central heptose (HepII) of the LPS inner-core element is also substituted by hexose containing oligosaccharides. The expression of the galabiose epitope in LPS of H. influenzae type b strains has previously been linked to genes comprising the lic2 locus. The present study provides definitive evidence for the role of lic2 genes in initiating chain extension from HepII. From the analysis of core oligosaccharide samples, LPS from the lic2 mutant strain of RM7004 was also found to carry O-acetyl substituents. Mono-, di-, and tri-O-acetylated LPS oligosaccharides were identified. The major O-acetylated glycoforms were found to be substituted at the O-3 position of HepIII. A di-O-acetylated species was characterized which was also substituted at the O-6 postion of the terminal beta-D-Glc in the Hex3 glycoform. This is the first report pointing to the occurrence of O-acetyl groups in the inner-core region of H. influenzae LPS. We have previously shown that in H. influenzae strain Rd, a capsule-deficient type d strain, PCho groups are expressed in a different molecular environment, being attached at the O-6 position of a beta-D-Glcp, which is in turn attached to HepI.


Assuntos
Haemophilus influenzae tipo b/química , Lipopolissacarídeos/química , Acetilação , Sequência de Carboidratos , Epitopos , Haemophilus influenzae tipo b/genética , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/isolamento & purificação , Estrutura Molecular , Mutação , Fosforilcolina/química , Fosforilcolina/imunologia
18.
Infect Immun ; 68(5): 2804-7, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10768976

RESUMO

The effect of phase variation of lipopolysaccharide (LPS) structure on the susceptibility of Haemophilus influenzae to complement-dependent killing by normal human sera and normal rat sera has been described previously. The phase-variable structure phosphorylcholine (ChoP) confers susceptibility to human serum, since ChoP on the bacterial cell surface binds to serum C-reactive protein and activates complement. In contrast, expression of galalpha1,4gal, a second phase-variable epitope that is also found on human glycoconjugates, confers resistance to human serum. We studied the role of phase variation of these structures in the susceptibilities of H. influenzae KW20 (Rd) and a clinical isolate of nontypeable H. influenzae to killing by rabbit sera, which often possess naturally acquired complement-dependent bactericidal activity for unencapsulated H. influenzae. Expression of ChoP increased the resistance of strain KW20 to killing by bactericidal rabbit sera. In contrast, the serum resistance of a clinical isolate, H233, was unaffected by ChoP expression but was reduced by galalpha1,4gal expression. The rabbit sera with bactericidal activity (but not the nonbactericidal sera) all contained immunoglobulin M (IgM) antibodies able to bind to the surface of H. influenzae bacteria, as detected by flow cytometry, and contained IgM antibodies to LPS purified from strain KW20. Preincubation of sera with LPS reduced their bactericidal activity. Bactericidal activity was recovered quantitatively in an IgM-enriched fraction of sera. It is concluded that naturally occurring bactericidal activity for unencapsulated H. influenzae is largely due to IgM antibodies directed against phase-variable structures of the LPS.


Assuntos
Anticorpos Antibacterianos/imunologia , Epitopos de Linfócito B/imunologia , Haemophilus influenzae/imunologia , Lipopolissacarídeos/imunologia , Animais , Humanos , Imunoglobulina M/imunologia , Lipopolissacarídeos/química , Fosforilcolina/imunologia , Coelhos , Ratos
19.
Infect Immun ; 68(3): 1664-71, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10678986

RESUMO

A number of pathogens of the upper respiratory tract express an unusual prokaryotic structure, phosphorylcholine (ChoP), on their cell surface. We tested the hypothesis that ChoP, also found on host membrane lipids in the form of phosphatidylcholine, acts so as to decrease killing by antimicrobial peptides that target differences between bacterial and host membranes. In Haemophilus influenzae, ChoP is a phase-variable structure on the oligosaccharide portion of the lipopolysaccharide (LPS). There was a bactericidal effect of the peptide LL-37/hCAP18 on a nontypeable H. influenzae strain, with an increasing selection for the ChoP(+) phase as the concentration of the peptide was raised from 0 to 10 microgram/ml. Moreover, constitutive ChoP-expressing mutants of unrelated strains showed up to 1,000-fold-greater survival compared to mutants without ChoP. The effect of ChoP on resistance to killing by LL-37/hCAP18 was dependent on the salt concentration and was observed only when bacteria were grown in the presence of environmental choline, a requirement for the expression of ChoP on the LPS. Further studies established that there is transcription of the LL-37/hCAP18 gene on the epithelial surface of the human nasopharynx in situ and inducible transcription in epithelial cells derived from the upper airway. The presence of highly variable amounts of LL-37/hCAP18 in normal nasal secretions (<1.2 to >80 microgram/ml) was demonstrated with an antibody against this peptide. It was concluded that ChoP alters the bacterial cell surface so as mimic host membrane lipids and decrease killing by LL-37/hCAP18, an antimicrobial peptide that may be expressed on the mucosal surface of the nasopharynx in bactericidal concentrations.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos , Proteínas de Transporte/farmacologia , Haemophilus influenzae/imunologia , Nasofaringe/imunologia , Fosforilcolina/farmacologia , Antibacterianos/análise , Proteínas de Transporte/análise , Catelicidinas , Haemophilus influenzae/efeitos dos fármacos , Humanos , Nasofaringe/química
20.
Antimicrob Agents Chemother ; 44(3): 602-7, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10681325

RESUMO

The ability of cecropin A to permeabilize and depolarize the membranes of Escherichia coli ML-35p bacteria has been compared to its bactericidal activity in an extension of earlier studies performed on synthetic lipid vesicle membranes (L. Silvestro, K. Gupta, J. H. Weiser, and P. H. Axelsen, Biochemistry 36:11452-11460, 1997). Our results indicate that differences in the concentration dependences of membrane permeabilization and depolarization seen in synthetic vesicles are not manifested in whole bacteria. The concentration dependences of both phenomena roughly correlate with bactericidal activity, suggesting that the bactericidal mechanism of cecropin A is related to membrane permeabilization.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos , Escherichia coli/efeitos dos fármacos , Peptídeos/farmacologia , Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Potenciais da Membrana/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...