Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Mol Microbiol ; 99(2): 407-24, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26434356

RESUMO

Borrelia (B.) bavariensis exhibits a marked tropism for nervous tissues and frequently causes neurological manifestations in humans. The molecular mechanism by which B. bavariensis overcomes innate immunity, in particular, complement remains elusive. In contrast to other serum-resistant spirochetes, none of the B. bavariensis isolates investigated bound complement regulators of the alternative (AP) and classical pathway (CP) or proteolytically inactivated complement components. Focusing on outer surface proteins BGA66 and BGA71, we demonstrated that both molecules either inhibit AP, CP and terminal pathway (TP) activation, or block activation of the CP and TP respectively. Both molecules bind complement components C7, C8 and C9, and thereby prevent assembly of the terminal complement complex. This inhibitory activity was confirmed by the introduction of the BGA66 and BGA71 encoding genes into a serum-sensitive B. garinii strain. Transformed spirochetes producing either BGA66 or BGA71 overcome complement-mediated killing, thus indicating that both proteins independently facilitate serum resistance of B. bavariensis. The generation of C-terminally truncated proteins as well as a chimeric BGA71 protein lead to the localization of the complement-interacting binding site within the N-terminus. Collectively, our data reveal a novel immune evasion strategy of B. bavariensis that is directed against the activation of the TP.


Assuntos
Proteínas de Bactérias/imunologia , Borrelia burgdorferi/imunologia , Proteínas do Sistema Complemento/imunologia , Doença de Lyme/imunologia , Animais , Proteínas de Bactérias/genética , Borrelia burgdorferi/genética , Complexo de Ataque à Membrana do Sistema Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Humanos , Doença de Lyme/microbiologia , Camundongos
2.
Int J Med Microbiol ; 306(1): 59-68, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26679571

RESUMO

Allergies are an increasing issue in human health and can, eventually, cause severe anaphylactic shock. Aspergillus fumigatus and Candida albicans are leading causes of life-threatening invasive fungal infections in immunocompromised patients, but can also cause severe allergic responses in otherwise healthy individuals. The glycolytic enzyme enolase is known as a major allergen despite its function in intracellular metabolism. Therefore, its presentation on surfaces of different fungal species was investigated by using antibodies raised against recombinant enolases from A. fumigatus and C. albicans. Examination of antibody specificity revealed cross-reactivity to cell-free extracts from Aspergillus terreus, Aspergillus flavus, Aspergillus nidulans and Candida glabrata, but not against any of the three human enolases. Antibody specificity was further confirmed by hybridization with other recombinant fungal enolases, where the antibodies recognized different subsets of fungal enolases. When surface presentation of enolase was tested on intact fungal cells, a positive staining was obtained with those antibodies that also recognized the enzyme from the respective cell-free extract. This implies a general surface presentation of this glycolytic enzyme among fungal species and provides hints for its predominant recognition as an allergen. Additionally, A. fumigatus and C. albicans enolase bound to human plasminogen, which remained accessible for the plasminogen activator uPA. This implies a potential role of enolase in the invasion and dissemination process during fungal infections.


Assuntos
Alérgenos/análise , Aspergillus/enzimologia , Candida/enzimologia , Proteínas de Transporte/análise , Proteínas de Membrana/análise , Fosfopiruvato Hidratase/análise , Animais , Anticorpos Antifúngicos/metabolismo , Reações Cruzadas , Epitopos/análise , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Plasminogênio/metabolismo , Ligação Proteica , Fatores de Virulência/análise
3.
J Immunol ; 192(12): 5913-23, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24835392

RESUMO

Haemophilus influenzae is a Gram-negative human pathogen that resides in the upper respiratory tract. Encapsulated H. influenzae type b (Hib) and type f (Hif) are the most common serotypes associated with invasive disease. H. influenzae displays various strategies to circumvent the host innate immune response, including the bactericidal effect of the complement system. In this study, we identified an H. influenzae lipoprotein having the ability to bind factor H (FH), the major regulator of the alternative pathway of complement activation. This protein, named protein H (PH), was surface exposed and was found in all clinical Hib and Hif isolates tested. Deletion of the gene encoding for PH (lph) in Hib and Hif significantly reduced the interaction between bacteria and FH. When Hib and Hif PH variants were separately expressed in nontypeable (unencapsulated) H. influenzae, which did not bind FH, an increased FH affinity was observed. We recombinantly expressed the two PH variants in Escherichia coli, and despite sharing only 56% identical amino acids, both FH-binding Haemophilus proteins similarly interacted with the complement regulator FH short consensus repeats 7 and 18-20. Importantly, Hib and Hif resistance against the bactericidal effect of human serum was significantly reduced when bacterial mutants devoid of PH were tested. In conclusion, we have characterized a hitherto unknown bacterial protein that is crucial for mediating an interaction between the human pathogen H. influenzae and FH. This novel interaction is important for H. influenzae resistance against complement activation and will consequently promote bacterial pathogenesis.


Assuntos
Proteínas de Bactérias/imunologia , Atividade Bactericida do Sangue/imunologia , Fator H do Complemento/imunologia , Haemophilus influenzae/imunologia , Lipoproteínas/imunologia , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Feminino , Haemophilus influenzae/química , Haemophilus influenzae/genética , Humanos , Lipoproteínas/química , Lipoproteínas/genética , Masculino , Mutação , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
4.
J Biol Chem ; 288(22): 15614-27, 2013 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-23603906

RESUMO

Adherence of Streptococcus pneumoniae is directly mediated by interactions of adhesins with eukaryotic cellular receptors or indirectly by exploiting matrix and serum proteins as molecular bridges. Pneumococci engage vitronectin, the human adhesive glycoprotein and complement inhibitor, to facilitate attachment to epithelial cells of the mucosal cavity, thereby modulating host cell signaling. In this study, we identified PspC as a vitronectin-binding protein interacting with the C-terminal heparin-binding domain of vitronectin. PspC is a multifunctional surface-exposed choline-binding protein displaying various adhesive properties. Vitronectin binding required the R domains in the mature PspC protein, which are also essential for the interaction with the ectodomain of the polymeric immunoglobulin receptor and secretory IgA. Consequently, secretory IgA competitively inhibited binding of vitronectin to purified PspC and to PspC-expressing pneumococci. In contrast, Factor H, which binds to the N-terminal part of mature PspC molecules, did not interfere with the PspC-vitronectin interaction. Using a series of vitronectin peptides, the C-terminal heparin-binding domain was shown to be essential for the interaction of soluble vitronectin with PspC. Binding experiments with immobilized vitronectin suggested a region N-terminal to the identified heparin-binding domain as an additional binding region for PspC, suggesting that soluble, immobilized, as well as cellularly bound vitronectin possesses different conformations. Finally, vitronectin bound to PspC was functionally active and inhibited the deposition of the terminal complement complex. In conclusion, this study identifies and characterizes (on the molecular level) the interaction between the pneumococcal adhesin PspC and the human glycoprotein vitronectin.


Assuntos
Proteínas de Bactérias/metabolismo , Streptococcus pneumoniae/metabolismo , Vitronectina/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Humanos , Ligação Proteica , Estrutura Terciária de Proteína , Streptococcus pneumoniae/química , Streptococcus pneumoniae/genética , Vitronectina/química , Vitronectina/genética
5.
Infect Immun ; 82(1): 380-92, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24191298

RESUMO

CspA of the Lyme disease spirochete Borrelia burgdorferi represents a key molecule in immune evasion, protecting borrelial cells from complement-mediated killing. As previous studies focused almost exclusively on CspA of B. burgdorferi, here we investigate the different binding capacities of CspA orthologs of Borrelia burgdorferi, B. afzelii, and B. spielmanii for complement regulator factor H and plasminogen and their ability to inhibit complement activation by either binding these host-derived plasma proteins or independently by direct interaction with components involved in formation of the lethal, pore-like terminal complement complex. To further examine their function in serum resistance in vivo, a serum-sensitive B. garinii strain was used to generate spirochetes, ectopically producing functional CspA orthologs. Irrespective of their species origin, all three CspA orthologs impart resistance to complement-mediated killing when produced in a serum-sensitive B. garinii surrogate strain. To analyze the inhibitory effect on complement activation and to assess the potential to inactivate C3b by binding of factor H and plasminogen, recombinant CspA orthologs were also investigated. All three CspA orthologs simultaneously bound factor H and plasminogen but differed in regard to their capacity to inactivate C3b via bound plasmin(ogen) and inhibit formation of the terminal complement complex. CspA of B. afzelii binds plasmin(ogen) and inhibits the terminal complement complex more efficiently than CspA of B. burgdorferi and B. spielmanii. Taken together, CspA orthologs of serum-resistant Lyme disease spirochetes act as multifunctional evasion molecules that inhibit complement on two central activation levels, C3b generation and assembly of the terminal complement complex.


Assuntos
Proteínas de Bactérias/fisiologia , Borrelia burgdorferi/fisiologia , Proteínas do Sistema Complemento/metabolismo , Doença de Lyme/microbiologia , Análise de Variância , Bacteriólise/fisiologia , Atividade Bactericida do Sangue , Borrelia/fisiologia , Células Cultivadas , Complemento C3b/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Doença de Lyme/imunologia , Plasminogênio/metabolismo , Ligação Proteica/fisiologia
6.
Infect Immun ; 82(6): 2378-89, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24664511

RESUMO

Haemophilus influenzae type b (Hib) escapes the host immune system by recruitment of the complement regulator vitronectin, which inhibits the formation of the membrane attack complex (MAC) by inhibiting C5b-C7 complex formation and C9 polymerization. We reported previously that Hib acquires vitronectin at the surface by using Haemophilus surface fibrils (Hsf). Here we studied in detail the interaction between Hsf and vitronectin and its role in the inhibition of MAC formation and the invasion of lung epithelial cells. The vitronectin-binding region of Hsf was defined at the N-terminal region comprising Hsf amino acids 429 to 652. Moreover, the Hsf recognition site on vitronectin consisted of the C-terminal amino acids 352 to 374. H. influenzae was killed more rapidly in vitronectin-depleted serum than in normal human serum (NHS), and increased MAC deposition was observed at the surface of an Hsf-deficient H. influenzae mutant. In parallel, Hsf-expressing Escherichia coli selectively acquired vitronectin from serum, resulting in significant inhibition of the MAC. Moreover, when vitronectin was bound to Hsf, increased bacterial adherence and internalization into epithelial cells were observed. Taking our findings together, we have defined a fine-tuned protein-protein interaction between Hsf and vitronectin that may contribute to increased Hib virulence.


Assuntos
Adesinas Bacterianas/fisiologia , Atividade Bactericida do Sangue/imunologia , Adesão Celular/fisiologia , Haemophilus influenzae tipo b/imunologia , Vitronectina/metabolismo , Análise de Variância , Ligação Competitiva , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Relação Dose-Resposta Imunológica , Haemophilus influenzae tipo b/patogenicidade , Heparina/fisiologia , Humanos , Soro/imunologia , Soro/microbiologia
7.
J Immunol ; 189(10): 4939-50, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23071278

RESUMO

The opportunistic human pathogen Pseudomonas aeruginosa causes a wide range of diseases. To cross host innate immune barriers, P. aeruginosa has developed efficient strategies to escape host complement attack. In this study, we identify the 57-kDa dihydrolipoamide dehydrogenase (Lpd) as a surface-exposed protein of P. aeruginosa that binds the four human plasma proteins, Factor H, Factor H-like protein-1 (FHL-1), complement Factor H-related protein 1 (CFHR1), and plasminogen. Factor H contacts Lpd via short consensus repeats 7 and 18-20. Factor H, FHL-1, and plasminogen when bound to Lpd were functionally active. Factor H and FHL-1 displayed complement-regulatory activity, and bound plasminogen, when converted to the active protease plasmin, cleaved the chromogenic substrate S-2251 and the natural substrate fibrinogen. The lpd of P. aeruginosa is a rather conserved gene; a total of 22 synonymous and 3 nonsynonymous mutations was identified in the lpd gene of the 5 laboratory strains and 13 clinical isolates. Lpd is surface exposed and contributes to survival of P. aeruginosa in human serum. Bacterial survival was reduced when Lpd was blocked on the surface prior to challenge with human serum. Similarly, bacterial survival was reduced up to 84% when the bacteria was challenged with complement active serum depleted of Factor H, FHL-1, and CFHR1, demonstrating a protective role of the attached human regulators from complement attack. In summary, Lpd is a novel surface-exposed virulence factor of P. aeruginosa that binds Factor H, FHL-1, CFHR1, and plasminogen, and the Lpd-attached regulators are relevant for innate immune escape and most likely contribute to tissue invasion.


Assuntos
Proteínas de Bactérias/imunologia , Proteínas Inativadoras do Complemento C3b/metabolismo , Fator H do Complemento/imunologia , Di-Hidrolipoamida Desidrogenase/imunologia , Evasão da Resposta Imune , Plasminogênio/imunologia , Pseudomonas aeruginosa/imunologia , Fatores de Virulência/imunologia , Proteínas de Bactérias/genética , Atividade Bactericida do Sangue/genética , Atividade Bactericida do Sangue/imunologia , Ativação do Complemento/genética , Ativação do Complemento/imunologia , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/genética , Di-Hidrolipoamida Desidrogenase/genética , Humanos , Mutação , Plasminogênio/genética , Ligação Proteica/genética , Ligação Proteica/imunologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Fatores de Virulência/genética
8.
J Immunol ; 189(10): 4900-8, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23071281

RESUMO

Yersinia adhesin A (YadA) is a major virulence factor of Yersinia enterocolitica. YadA mediates host cell binding and autoaggregation and protects the pathogen from killing by the complement system. Previous studies demonstrated that YadA is the most important single factor mediating serum resistance of Y. enterocolitica, presumably by binding C4b binding protein (C4BP) and factor H, which are both complement inhibitors. Factor H acts as a cofactor for factor I-mediated cleavage of C3b into the inactive form iC3b and thus prevents formation of inflammatory effector compounds and the terminal complement complex. In this study, we challenged the current direct binding model of factor H to YadA and show that Y. enterocolitica YadA recruits C3b and iC3b directly, without the need of an active complement cascade or additional serum factors. Enhanced binding of C3b does not decrease survival of YadA-expressing Yersiniae because C3b becomes readily inactivated by factor H and factor I. Binding of factor H to YadA is greatly reduced in the absence of C3. Experiments using Yersinia lacking YadA or expressing YadA with reduced trimeric stability clearly demonstrate that both the presence and full trimeric stability of YadA are essential for complement resistance. A novel mechanism of factor H binding is presented in which YadA exploits recruitment of C3b or iC3b to attract large amounts of factor H. As a consequence, formation of the terminal complement complex is limited and bacterial survival is enhanced. These findings add a new aspect of how Y. enterocolitica effectively evades the host complement system.


Assuntos
Adesinas Bacterianas/imunologia , Ativação do Complemento , Complemento C3/imunologia , Evasão da Resposta Imune , Yersinia enterocolitica/imunologia , Adesinas Bacterianas/genética , Complemento C3/antagonistas & inibidores , Complemento C3/genética , Proteína de Ligação ao Complemento C4b , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Antígenos de Histocompatibilidade/genética , Antígenos de Histocompatibilidade/imunologia , Humanos , Ligação Proteica , Yersinia enterocolitica/genética
9.
Blood ; 118(10): 2774-83, 2011 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-21757614

RESUMO

The human plasma protein ß(2)-glycoprotein I (ß(2)-GPI) is the major target of autoantibodies associated with antiphospholipid syndrome. However, the biologic function of this abundant protein is still unclear. Here we identify ß(2)-GPI as a complement regulator. ß(2)-GPI circulates in the plasma in an inactive circular form. On surface binding, such as to apoptotic cells, ß(2)-GPI changes conformation to an elongated form that acquires C3/C3b binding activities. ß(2)-GPI apparently changes conformation of C3, so that the regulator factor H attaches and induces subsequent degradation by the protease factor I. ß(2)-GPI also mediates further cleavage of C3/C3b compared with factor H alone. Our data provide important insights into innate immune regulation by plasma protein ß(2)-GPI, which may be exploited in the prevention and therapy of autoimmune disease antiphospholipid syndrome.


Assuntos
Síndrome Antifosfolipídica/imunologia , Autoanticorpos/imunologia , Complemento C3/imunologia , Complemento C3b/imunologia , beta 2-Glicoproteína I/metabolismo , Síndrome Antifosfolipídica/fisiopatologia , Apoptose , Western Blotting , Ativação do Complemento , Complemento C3/metabolismo , Convertases de Complemento C3-C5/metabolismo , Complemento C3b/metabolismo , Fator H do Complemento/metabolismo , Humanos , Imunoprecipitação , Conformação Molecular , Ligação Proteica , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , beta 2-Glicoproteína I/imunologia
10.
J Immunol ; 186(5): 3120-9, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21270401

RESUMO

The complement system plays an important role in eliminating invading pathogens. Activation of complement results in C3b deposition (opsonization), phagocytosis, anaphylatoxin (C3a, C5a) release, and consequently cell lysis. Moraxella catarrhalis is a human respiratory pathogen commonly found in children with otitis media and in adults with chronic obstructive pulmonary disease. The species has evolved multiple complement evasion strategies, which among others involves the ubiquitous surface protein (Usp) family consisting of UspA1, A2, and A2 hybrid. In the present study, we found that the ability of M. catarrhalis to bind C3 correlated with UspA expression and that C3 binding contributed to serum resistance in a large number of clinical isolates. Recombinantly expressed UspA1 and A2 inhibit both the alternative and classical pathways, C3b deposition, and C3a generation when bound to the C3 molecule. We also revealed that the M. catarrhalis UspA-binding domain on C3b was located to C3d and that the major bacterial C3d-binding domains were within UspA1(299-452) and UspA2(165-318). The interaction with C3 was not species specific since UspA-expressing M. catarrhalis also bound mouse C3 that resulted in inhibition of the alternative pathway of mouse complement. Taken together, the binding of C3 to UspAs is an efficient strategy of Moraxella to block the activation of complement and to inhibit C3a-mediated inflammation.


Assuntos
Antígenos de Bactérias/fisiologia , Antígenos de Superfície/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Complemento C3d/metabolismo , Evasão da Resposta Imune/imunologia , Moraxella catarrhalis/imunologia , Adulto , Animais , Antígenos de Bactérias/metabolismo , Antígenos de Superfície/metabolismo , Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas da Membrana Bacteriana Externa/metabolismo , Criança , Ativação do Complemento/imunologia , Complemento C3a/antagonistas & inibidores , Complemento C3a/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Via Alternativa do Complemento/imunologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Camundongos , Moraxella catarrhalis/isolamento & purificação , Moraxella catarrhalis/patogenicidade , Infecções por Moraxellaceae/imunologia , Infecções por Moraxellaceae/microbiologia , Infecções por Moraxellaceae/patologia , Ligação Proteica/imunologia , Coelhos , Ovinos
11.
Clin Dev Immunol ; 2012: 349657, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22400034

RESUMO

Borrelia burgdorferi evades complement-mediated killing by interacting with complement regulators through distinct complement regulator-acquiring surface proteins (CRASPs). Here, we extend our analyses to the contribution of CRASP-4 in mediating complement resistance of B. burgdorferi and its interaction with human complement regulators. CRASP-4 (also known as ErpC) was immobilized onto magnetic beads and used to capture proteins from human serum. Following Western blotting, factor H (CFH), CFH-related protein 1 (CFHR1), CFHR2, and CFHR5 were identified as ligands of CRASP-4. To analyze the impact of native CRASP-4 on mediating survival of serum-sensitive cells in human serum, a B. garinii strain was generated that ectopically expresses CRASP-4. CRASP-4-producing bacteria bound CFHR1, CFHR2, and CFHR5 but not CFH. In addition, transformed spirochetes deposited significant amounts of lethal complement components on their surface and were susceptible to human serum, thus indicating that CRASP-4 plays a subordinate role in complement resistance of B. burgdorferi.


Assuntos
Proteínas de Bactérias/imunologia , Borrelia burgdorferi/imunologia , Evasão da Resposta Imune , Doença de Lyme/imunologia , Proteínas de Membrana/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Western Blotting , Borrelia burgdorferi/genética , Proteínas Inativadoras do Complemento C3b/imunologia , Proteínas Inativadoras do Complemento C3b/metabolismo , Fator H do Complemento/imunologia , Fator H do Complemento/metabolismo , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Farmacorresistência Bacteriana , Humanos , Proteínas Imobilizadas/genética , Proteínas Imobilizadas/imunologia , Proteínas Imobilizadas/metabolismo , Doença de Lyme/metabolismo , Doença de Lyme/microbiologia , Magnetismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Plasmídeos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Transformação Genética
12.
J Infect Dis ; 204(7): 1065-74, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21881122

RESUMO

Nontypeable Haemophilus influenzae (NTHi) causes otitis media and is commonly found in patients with chronic obstructive pulmonary disease (COPD). Adhesins are important for bacterial attachment and colonization. Protein E (PE) is a recently characterized ubiquitous 16 kDa adhesin with vitronectin-binding capacity that results in increased survival in serum. In addition to PE, NTHi utilizes Haemophilus adhesion protein (Hap) that binds to the basement-membrane glycoprotein laminin. We show that most clinical isolates bind laminin and that both Hap and PE are crucial for the NTHi-dependent interaction with laminin as revealed with different mutants. The laminin-binding region is located at the N-terminus of PE, and PE binds to the heparin-binding C-terminal globular domain of laminin. PE simultaneously attracts vitronectin and laminin at separate binding sites, proving the multifunctional nature of the adhesin. This previously unknown PE-dependent interaction with laminin may contribute to NTHi colonization, particularly in smokers with COPD.


Assuntos
Adesinas Bacterianas/química , Matriz Extracelular/química , Haemophilus influenzae/metabolismo , Laminina/química , Vitronectina/química , Adesinas Bacterianas/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Sítios de Ligação , Criança , Pré-Escolar , Ensaio de Imunoadsorção Enzimática , Feminino , Fibrinogênio/química , Fibronectinas/química , Haemophilus influenzae/fisiologia , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Ressonância de Plasmônio de Superfície , Adulto Jovem
13.
J Immunol ; 183(4): 2593-601, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19635912

RESUMO

Nontypeable Haemophilus influenzae (NTHi) commonly causes local disease in the upper and lower respiratory tract and has recently been shown to interfere with both the classical and alternative pathways of complement activation. The terminal pathway of the complement system is regulated by vitronectin that is a component of both plasma and the extracellular matrix. In this study, we identify protein E (PE; 16 kDa), which is a recently characterized ubiquitous outer membrane protein, as a vitronectin-binding protein of NTHi. A PE-deficient NTHi mutant had a markedly reduced survival in serum compared with the PE-expressing isogenic NTHi wild type. Moreover, the PE-deficient mutant showed a significantly decreased binding to both soluble and immobilized vitronectin. In parallel, PE-expressing Escherichia coli bound soluble vitronectin and adhered to immobilized vitronectin compared with controls. Surface plasmon resonance technology revealed a K(D) of 0.4 microM for the interaction between recombinant PE and immobilized vitronectin. Moreover, the PE-dependent vitronectin-binding site was located at the heparin-binding domains of vitronectin and the major vitronectin-binding domain was found in the central core of PE (aa 84-108). Importantly, vitronectin bound to the surface of NTHi 3655 reduced membrane attack complex-induced hemolysis. In contrast to incubation with normal human serum, NTHi 3655 showed a reduced survival in vitronectin-depleted human serum, thus demonstrating that vitronectin mediates a protective role at the bacterial surface. Our findings show that PE, by binding vitronectin, may play an important role in NTHi pathogenesis.


Assuntos
Aderência Bacteriana/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Atividade Bactericida do Sangue/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Esterases/metabolismo , Haemophilus influenzae/imunologia , Lipoproteínas/metabolismo , Vitronectina/metabolismo , Sequência de Aminoácidos , Aderência Bacteriana/genética , Proteínas da Membrana Bacteriana Externa/fisiologia , Atividade Bactericida do Sangue/genética , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Esterases/deficiência , Esterases/fisiologia , Haemophilus influenzae/genética , Haemophilus influenzae/crescimento & desenvolvimento , Hemólise/imunologia , Humanos , Lipoproteínas/deficiência , Lipoproteínas/fisiologia , Dados de Sequência Molecular , Fragmentos de Peptídeos/deficiência , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/fisiologia , Ligação Proteica/genética , Ligação Proteica/imunologia , Sorotipagem , Solubilidade , Vitronectina/fisiologia
14.
J Infect Dis ; 202(3): 490-8, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20565259

RESUMO

Lyme disease-causing Borrelia burgdorferi spirochetes express up to 5 complement regulator-acquiring surface proteins (CRASPs). To better define how CRASP-1 contributes to infection, we aimed to identify novel CRASP-1-binding host proteins. Here, we identified a number of novel human CRASP-1-binding proteins, including bone morphogenic protein 2, collagen I, collagen III, collagen IV, fibronectin, laminin, and plasminogen. The plasminogen-binding regions were located in 2 separate regions of CRASP-1. Our results demonstrated that plasminogen-bound CRASP-1 can be converted to plasmin by the urokinase-type plasminogen activator and that proteolytically active plasmin cleaves the synthetic chromogenic substrate S-2251 and the natural substrate fibrinogen. In conclusion, CRASP-1 is a multifunctional protein of B. burgdorferi that binds to several human extracellular matrix proteins and plasminogen. These interactions may contribute to adhesion, bacterial colonization, and organ tropism and may allow dissemination of B. burgdorferi in the host.


Assuntos
Proteínas de Bactérias/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Borrelia burgdorferi/patogenicidade , Proteínas da Matriz Extracelular/metabolismo , Interações Hospedeiro-Patógeno , Proteínas de Membrana/metabolismo , Plasminogênio/metabolismo , Fatores de Virulência/metabolismo , Citometria de Fluxo , Técnica Direta de Fluorescência para Anticorpo , Humanos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Ressonância de Plasmônio de Superfície
15.
J Clin Microbiol ; 48(3): 921-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20089757

RESUMO

The aim of the present study was to analyze the importance of nontypeable Haemophilus influenzae (NTHi) isolated from patients with sepsis (invasive isolates) compared to nasopharyngeal isolates from patients with upper respiratory tract infection for resistance to complement-mediated attack in human serum and to correlate this result with disease severity. We studied and characterized cases of invasive NTHi disease in detail. All patients with invasive NTHi isolates were adults, and 35% had a clinical presentation of severe sepsis according to the ACCP/SCCM classification of sepsis grading. Moreover, 41% of the patients had evidence of immune deficiency. The different isolates were analyzed for survival in human serum and for binding of 125I-labeled, purified human complement inhibitors C4b-binding protein (C4BP), factor H, and vitronectin, in addition to binding of regulators directly from serum. No significant differences were found when blood-derived and nasopharyngeal isolates were compared, suggesting that interactions with the complement system are equally important for NTHi strains, irrespective of isolation site. Interestingly, a correlation between serum resistance and invasive disease severity was found. The ability to resist the attack of the complement system seems to be important for NTHi strains infecting the respiratory tract as well as the bloodstream.


Assuntos
Atividade Bactericida do Sangue , Portador Sadio/microbiologia , Proteínas do Sistema Complemento/imunologia , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/imunologia , Haemophilus influenzae/patogenicidade , Sepse/microbiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas do Sistema Complemento/metabolismo , Feminino , Humanos , Masculino , Viabilidade Microbiana , Pessoa de Meia-Idade , Ligação Proteica
16.
Microbes Infect ; 10(4): 374-81, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18400547

RESUMO

The immunoglobulin D (IgD)-binding protein MID/Hag of the human respiratory pathogen Moraxella catarrhalis is an outer membrane protein of approximately 200kDa belonging to the autotransporter family. MID also functions as an adhesin and hemagglutinin. In the present paper, the ultrastructure of MID was mapped. Using a series of Escherichia coli transformants, the last 210 aa of the C-terminal region were shown to translocate protein MID through the outer membrane suggesting that MID has a beta-barrel structure comprising of 10 transmembrane beta-sheets. Electron microscopy mapping with gold-labelled specific antibodies, and partial unravelling using guanidine hydrochloride showed that the rest of the MID protein forms an approximately 120nm long, fibrillar structure in which the individual monomers fold back on themselves to expose a globular distal domain at their tips comprising both the IgD-binding (MID962-1200) and adhesive (MID764-913) regions. This positions their N-termini close to the C-terminal membrane spanning domains. Mass measurements by scanning transmission electron microscopy (STEM) verified that the MID molecule is an oligomer.


Assuntos
Adesinas Bacterianas/química , Adesinas Bacterianas/ultraestrutura , Moraxella catarrhalis/química , Moraxella catarrhalis/metabolismo , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Animais , Análise Mutacional de DNA , Escherichia coli/genética , Humanos , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , Modelos Moleculares , Peso Molecular , Moraxella catarrhalis/genética , Desnaturação Proteica , Estrutura Quaternária de Proteína , Transporte Proteico , Coelhos
17.
J Innate Immun ; 9(1): 33-51, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27798934

RESUMO

Complement resistance is an important virulence trait of Yersinia enterocolitica (Ye). The predominant virulence factor expressed by Ye is Yersinia adhesin A (YadA), which enables bacterial attachment to host cells and extracellular matrix and additionally allows the acquisition of soluble serum factors. The serum glycoprotein vitronectin (Vn) acts as an inhibitory regulator of the terminal complement complex by inhibiting the lytic pore formation. Here, we show YadA-mediated direct interaction of Ye with Vn and investigated the role of this Vn binding during mouse infection in vivo. Using different Yersinia strains, we identified a short stretch in the YadA head domain of Ye O:9 E40, similar to the 'uptake region' of Y. pseudotuberculosis YPIII YadA, as crucial for efficient Vn binding. Using recombinant fragments of Vn, we found the C-terminal part of Vn, including heparin-binding domain 3, to be responsible for binding to YadA. Moreover, we found that Vn bound to the bacterial surface is still functionally active and thus inhibits C5b-9 formation. In a mouse infection model, we demonstrate that Vn reduces complement-mediated killing of Ye O:9 E40 and, thus, improved bacterial survival. Taken together, these findings show that YadA-mediated Vn binding influences Ye pathogenesis.


Assuntos
Adesinas Bacterianas/metabolismo , Vitronectina/metabolismo , Yersiniose/imunologia , Yersinia enterocolitica/fisiologia , Animais , Bacteriólise , Proteínas do Sistema Complemento/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Imunomodulação , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Domínios Proteicos/genética , Especificidade da Espécie , Virulência , Vitronectina/genética , Yersinia enterocolitica/patogenicidade
18.
PLoS One ; 11(1): e0147709, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26808444

RESUMO

Pathogenicity of many microbes relies on their capacity to resist innate immunity, and to survive and persist in an immunocompetent human host microbes have developed highly efficient and sophisticated complement evasion strategies. Here we show that different human pathogens including Gram-negative and Gram-positive bacteria, as well as the fungal pathogen Candida albicans, acquire the human terminal complement regulator vitronectin to their surface. By using truncated vitronectin fragments we found that all analyzed microbial pathogens (n = 13) bound human vitronectin via the same C-terminal heparin-binding domain (amino acids 352-374). This specific interaction leaves the terminal complement complex (TCC) regulatory region of vitronectin accessible, allowing inhibition of C5b-7 membrane insertion and C9 polymerization. Vitronectin complexed with the various microbes and corresponding proteins was thus functionally active and inhibited complement-mediated C5b-9 deposition. Taken together, diverse microbial pathogens expressing different structurally unrelated vitronectin-binding molecules interact with host vitronectin via the same conserved region to allow versatile control of the host innate immune response.


Assuntos
Candida albicans/imunologia , Inativadores do Complemento/metabolismo , Bactérias Gram-Negativas/imunologia , Bactérias Gram-Positivas/imunologia , Evasão da Resposta Imune , Vitronectina/metabolismo , Humanos
19.
PLoS One ; 10(9): e0137630, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26368530

RESUMO

The opportunistic human pathogen Pseudomonas aeruginosa controls host innate immune and complement attack. Here we identify Dihydrolipoamide dehydrogenase (Lpd), a 57 kDa moonlighting protein, as the first P. aeruginosa protein that binds the two human terminal pathway inhibitors vitronectin and clusterin. Both human regulators when bound to the bacterium inhibited effector function of the terminal complement, blocked C5b-9 deposition and protected the bacterium from complement damage. P. aeruginosa when challenged with complement active human serum depleted from vitronectin was severely damaged and bacterial survival was reduced by over 50%. Similarly, when in human serum clusterin was blocked by a mAb, bacterial survival was reduced by 44%. Thus, demonstrating that Pseudomonas benefits from attachment of each human regulator and controls complement attack. The Lpd binding site in vitronectin was localized to the C-terminal region, i.e. to residues 354-363. Thus, Lpd of P. aeruginosa is a surface exposed moonlighting protein that binds two human terminal pathway inhibitors, vitronectin and clusterin and each human inhibitor when attached protected the bacterial pathogen from the action of the terminal complement pathway. Our results showed insights into the important function of Lpd as a complement regulator binding protein that might play an important role in virulence of P. aeruginosa.


Assuntos
Proteínas de Bactérias/metabolismo , Clusterina/metabolismo , Ativação do Complemento , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Di-Hidrolipoamida Desidrogenase/metabolismo , Pseudomonas aeruginosa/enzimologia , Vitronectina/metabolismo , Proteínas de Bactérias/genética , Clusterina/genética , Complexo de Ataque à Membrana do Sistema Complemento/genética , Di-Hidrolipoamida Desidrogenase/genética , Humanos , Viabilidade Microbiana , Ligação Proteica , Estrutura Terciária de Proteína , Pseudomonas aeruginosa/genética , Vitronectina/genética
20.
Thromb Haemost ; 113(1): 125-42, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25181963

RESUMO

Streptococcus pneumoniae serotype 3 strains are highly resistant to opsonophagocytosis due to recruitment of the complement inhibitor Factor H via Hic, a member of the pneumococcal surface protein C (PspC) family. In this study, we demonstrated that Hic also interacts with vitronectin, a fluid-phase regulator involved in haemostasis, angiogenesis, and the terminal complement cascade as well as a component of the extracellular matrix. Blocking of Hic by specific antiserum or genetic deletion significantly reduced pneumococcal binding to soluble and immobilised vitronectin and to Factor H, respectively. In parallel, ectopic expression of Hic on the surface of Lactococcus lactis conferred binding to soluble and immobilised vitronectin as well as Factor H. Molecular analyses with truncated Hic fragments narrowed down the vitronectin-binding site to the central core of Hic (aa 151-201). This vitronectin-binding region is separate from that of Factor H, which binds to the N-terminus of Hic (aa 38-92). Binding of pneumococcal Hic was localised to the C-terminal heparin-binding domain (HBD3) of vitronectin. However, an N-terminal region to HBD3 was further involved in Hic-binding to immobilised vitronectin. Finally, vitronectin bound to Hic was functionally active and inhibited formation of the terminal complement complex. In conclusion, Hic interacts with vitronectin and simultaneously with Factor H, and both human proteins may contribute to colonisation and invasive disease caused by serotype 3 pneumococci.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Evasão da Resposta Imune , Sorogrupo , Streptococcus pneumoniae/metabolismo , Vitronectina/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Fator H do Complemento/imunologia , Fator H do Complemento/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Lactococcus lactis/imunologia , Lactococcus lactis/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Transdução de Sinais , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/imunologia , Vitronectina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA