Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
J Intern Med ; 282(2): 116-128, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28345259

RESUMO

The complement system is an arm of innate immunity that aids in the removal of pathogens and dying cells. Due to its harmful, pro-inflammatory potential, complement is controlled by several soluble and membrane-bound inhibitors. This family of complement regulators has been recently extended by the discovery of several new members, and it is becoming apparent that these proteins harbour additional functions. In this review, the current state of knowledge of the physiological functions of four complement regulators will be described: cartilage oligomeric matrix protein (COMP), CUB and sushi multiple domains 1 (CSMD1), sushi domain-containing protein 4 (SUSD4) and CD59. Complement activation is involved in both the development of and defence against cancer. COMP expression is pro-oncogenic, whereas CSMD1 and SUSD4 act as tumour suppressors. These effects may be related in part to the complex influence of complement on cancer but also depend on unrelated functions such as the protection of cells from endoplasmic reticulum stress conveyed by intracellular COMP. CD59 is the main inhibitor of the membrane attack complex, and its deficiency leads to complement attack on erythrocytes and severe haemolytic anaemia, which is now amenable to treatment with an inhibitor of C5 cleavage. Unexpectedly, the intracellular pool of CD59 is crucial for insulin secretion from pancreatic ß-cells. This finding is one of several relating to the intracellular functions of complement proteins, which until recently were only considered to be present in the extracellular space. Understanding the alternative functions of complement inhibitors may unravel unexpected links between complement and other physiological systems, but is also important for better design of therapeutic complement inhibition.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Animais , Antígenos CD59/fisiologia , Proteína de Matriz Oligomérica de Cartilagem/fisiologia , Ativação do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Humanos , Infecções/fisiopatologia , Inflamação/fisiopatologia , Artropatias/fisiopatologia , Proteínas de Membrana/fisiologia , Neoplasias/fisiopatologia , Proteínas Supressoras de Tumor
2.
J Immunol ; 191(4): 1775-84, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23863906

RESUMO

Staphylococcus aureus is a major human pathogen causing more than a tenth of all septicemia cases and often superficial and deep infections in various tissues. One of the immune evasion strategies of S. aureus is to secrete proteins that bind to the central complement opsonin C3b. One of these, extracellular complement binding protein (Ecb), is known to interfere directly with functions of C3b. Because C3b is also the target of the physiological plasma complement regulator, factor H (FH), we studied the effect of Ecb on the complement regulatory functions of FH. We show that Ecb enhances acquisition of FH from serum onto staphylococcal surfaces. Ecb and FH enhance mutual binding to C3b and also the function of each other in downregulating complement activation. Both Ecb and the C-terminal domains 19-20 of FH bind to the C3d part of C3b. We show that the mutual enhancing effect of Ecb and FH on binding to C3b depends on binding of the FH domain 19 to the C3d part of C3b next to the binding site of Ecb on C3d. Our results show that Ecb, FH, and C3b form a tripartite complex. Upon exposure of serum-sensitive Haemophilus influenzae to human serum, Ecb protected the bacteria, and this effect was enhanced by the addition of the C-terminal domains 19-20 of FH. This finding indicates that the tripartite complex formation could give additional protection to bacteria and that S. aureus is thereby able to use host FH and bacterial Ecb in a concerted action to eliminate C3b at the site of infection.


Assuntos
Proteínas de Bactérias/fisiologia , Complemento C3b/metabolismo , Fator H do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Via Alternativa do Complemento , Evasão da Resposta Imune/imunologia , Staphylococcus aureus/imunologia , Fatores de Virulência/fisiologia , Proteínas de Bactérias/química , Sítios de Ligação , Complemento C3b/antagonistas & inibidores , Complemento C3b/química , Fator H do Complemento/química , Fator H do Complemento/genética , Proteínas Inativadoras do Complemento/química , Haemophilus influenzae/imunologia , Humanos , Imunidade Inata , Modelos Moleculares , Complexos Multiproteicos , Fragmentos de Peptídeos/metabolismo , Mutação Puntual , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Soro/imunologia , Infecções Estafilocócicas , Fatores de Virulência/química
3.
FASEB J ; 27(6): 2355-66, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23482636

RESUMO

Recently discovered Sushi domain-containing protein 4 (SUSD4) contains several Sushi or complement control protein domains; therefore, we hypothesized that it may act as complement inhibitor. Two isoforms of human SUSD4, fused to the Fc part of human IgG, were recombinantly expressed in Chinese hamster ovary (CHO) cells. The secreted soluble isoform of SUSD4 (SUSD4b) inhibited the classical and lectin complement pathways by 50% at a concentration of 0.5 µM. This effect was due to the fact that 1 µM SUSD4b inhibited the formation of the classical C3 convertase by 90%. The membrane-bound isoform (SUSD4a) inhibited the classical and alternative complement pathways when expressed on the surface of CHO cells but not when expressed as a soluble, truncated protein. In all functional studies, we used known complement inhibitors as positive controls, while Coxsackie adenovirus receptor, which has no effect on complement, expressed with Fc tag, was a negative control. We also studied the mRNA expression of both isoforms of SUSD4 in a panel of human tissues using quantitative PCR and primarily found SUSD4a in esophagus and brain, while SUSD4b was highly expressed in esophagus, ovary, and heart. Overall, our results show that SUSD4 is a novel complement inhibitor with restricted expression.


Assuntos
C3 Convertase da Via Clássica do Complemento/antagonistas & inibidores , C3 Convertase da Via Clássica do Complemento/biossíntese , Proteínas Inativadoras do Complemento/fisiologia , Proteínas de Membrana/fisiologia , Animais , Células CHO , Complemento C3/metabolismo , Proteínas Inativadoras do Complemento/genética , Via Clássica do Complemento , Cricetinae , Cricetulus , Feminino , Humanos , Imunidade Inata/genética , Proteínas de Membrana/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Distribuição Tecidual
4.
Thromb Haemost ; 108(6): 1141-53, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23014597

RESUMO

Coagulation and complement regulators belong to two interactive systems constituting emerging mechanisms of diabetic nephropathy. Thrombomodulin (TM) regulates both coagulation and complement activation, in part through discrete domains. TM's lectin like domain dampens complement activation, while its EGF-like domains independently enhance activation of the anti-coagulant and cytoprotective serine protease protein C (PC). A protective effect of activated PC in diabetic nephropathy is established. We hypothesised that TM controls diabetic nephropathy independent of PC through its lectin-like domain by regulating complement. Diabetic nephropathy was analysed in mice lacking TM's lectin-like domain (TMLeD/LeD) and controls (TMwt/wt). Albuminuria (290 µg/mg vs. 166 µg/mg, p=0.03) and other indices of experimental diabetic nephropathy were aggravated in diabetic TMLeD/LeD mice. Complement deposition (C3 and C5b-9) was markedly increased in glomeruli of diabetic TMLeD/LeD mice. Complement inhibition with enoxaparin ameliorated diabetic nephropathy in TMLeD/LeD mice (e.g. albuminuria 85 µg/mg vs. 290 µg/mg, p<0.001). In vitro TM's lectin-like domain cell-autonomously prevented glucose-induced complement activation on endothelial cells and - notably - on podocytes. Podocyte injury, which was enhanced in diabetic TMLeD/LeD mice, was reduced following complement inhibition with enoxaparin. The current study identifies a novel mechanism regulating complement activation in diabetic nephropathy. TM's lectin-like domain constrains glucose-induced complement activation on endothelial cells and podocytes and ameliorates albuminuria and glomerular damage in mice.


Assuntos
Nefropatias Diabéticas/etiologia , Trombomodulina/química , Trombomodulina/fisiologia , Animais , Linhagem Celular , Ativação do Complemento/fisiologia , Proteínas Inativadoras do Complemento/química , Proteínas Inativadoras do Complemento/deficiência , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/fisiologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/fisiopatologia , Nefropatias Diabéticas/fisiopatologia , Nefropatias Diabéticas/prevenção & controle , Células Endoteliais/imunologia , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Glomérulos Renais/imunologia , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/fisiologia , Podócitos/imunologia , Podócitos/patologia , Podócitos/fisiologia , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombomodulina/deficiência , Trombomodulina/genética
5.
J Immunol ; 188(5): 2338-49, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22287711

RESUMO

Tannerella forsythia is a poorly studied pathogen despite being one of the main causes of periodontitis, which is an inflammatory disease of the supporting structures of the teeth. We found that despite being recognized by all complement pathways, T. forsythia is resistant to killing by human complement, which is present at up to 70% of serum concentration in gingival crevicular fluid. Incubation of human serum with karilysin, a metalloproteinase of T. forsythia, resulted in a decrease in bactericidal activity of the serum. T. forsythia strains expressing karilysin at higher levels were more resistant than low-expressing strains. Furthermore, the low-expressing strain was significantly more opsonized with activated complement factor 3 and membrane attack complex from serum compared with the other strains. The high-expressing strain was more resistant to killing in human blood. The protective effect of karilysin against serum bactericidal activity was attributable to its ability to inhibit complement at several stages. The classical and lectin complement pathways were inhibited because of the efficient degradation of mannose-binding lectin, ficolin-2, ficolin-3, and C4 by karilysin, whereas inhibition of the terminal pathway was caused by degradation of C5. Interestingly, karilysin was able to release biologically active C5a peptide in human plasma and induce migration of neutrophils. Importantly, we detected the karilysin gene in >90% of gingival crevicular fluid samples containing T. forsythia obtained from patients with periodontitis. Taken together, the newly characterized karilysin appears to be an important virulence factor of T. forsythia and might have several important implications for immune evasion.


Assuntos
Proteínas de Bactérias/fisiologia , Bacteroides/enzimologia , Bacteroides/imunologia , Ativação do Complemento/imunologia , Proteínas Inativadoras do Complemento/fisiologia , Metaloproteinases da Matriz/fisiologia , Proteínas de Bactérias/isolamento & purificação , Bacteroides/isolamento & purificação , Humanos , Metaloproteinases da Matriz/sangue , Metaloproteinases da Matriz/isolamento & purificação , Fatores de Virulência/sangue , Fatores de Virulência/fisiologia
6.
J Immunol ; 187(9): 4913-9, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21964028

RESUMO

Experimental evidence suggests that C inhibition and more particularly combined inhibition of C and the TLR coreceptor CD14 may be of therapeutic benefit in sepsis and other inflammatory conditions. A barrier to the testing and further development of many inhibitors is that their activity is species specific. Pig is a relevant species for experimental models of human disease, and this study undertakes a comprehensive comparison of the inhibitory efficacy of the C5 inhibitor Ornithodoros moubata C inhibitor (OmCI) in human and porcine whole blood ex vivo models of Escherichia coli-induced sepsis. The effect of OmCI on complement activity in pigs undergoing E. coli sepsis was also examined. Porcine and human serum, and whole blood anticoagulated with lepirudin, was incubated with E. coli and the effect of OmCI investigated. The ex vivo results were virtually identical in pig and human. OmCI completely ablated the activity of all three C pathways at 0.64 µM. E. coli-induced C activation and expression of CD11b (wCD11R3 in the pig), was abolished ex vivo at 0.32 µM OmCI. Combining anti-CD14 and OmCI reduced the formation of IL-8 and TNF-α more potently than the single inhibitors. OmCI also efficiently bound E. coli-induced leukotriene B(4) in pig and human plasma. In support of our ex vivo findings, in vivo the activity of all C pathways was inhibited at 0.6 mg OmCI/kg pig. In conclusion, OmCI efficiently inhibited pig and human C activation, has accompanying anti-inflammatory effects and is a promising candidate inhibitor for further in vivo studies of sepsis.


Assuntos
Complemento C5a/antagonistas & inibidores , Proteínas Inativadoras do Complemento/fisiologia , Ornithodoros/imunologia , Animais , Complemento C5a/metabolismo , Proteínas Inativadoras do Complemento/uso terapêutico , Via Alternativa do Complemento/imunologia , Via Clássica do Complemento/imunologia , Modelos Animais de Doenças , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/prevenção & controle , Feminino , Humanos , Masculino , Projetos Piloto , Proteínas e Peptídeos Salivares/fisiologia , Proteínas e Peptídeos Salivares/uso terapêutico , Sepse/imunologia , Sepse/prevenção & controle , Suínos
7.
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
8.
J Immunol ; 185(11): 7007-13, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20962256

RESUMO

Ischemia reperfusion injury (IRI) is an unavoidable event during solid organ transplantation and is a major contributor to early graft dysfunction and subsequent graft immunogenicity. In a therapeutic paradigm using targeted complement inhibitors, we investigated the role of complement, and specifically the alternative pathway of complement, in IRI to heart isografts. Mouse heterotopic isograft heart transplants were performed in C57BL/6 mice treated with a single injection of either CR2-Crry (inhibits all complement pathways) or CR2-fH (inhibits alternative complement pathway) immediately posttransplantation. Transplanted hearts were harvested at 12 and 48 h for analysis. Both inhibitors resulted in a significant reduction in myocardial IRI, as measured by histology and serum cardiac troponin I levels. Furthermore, compared with untreated controls, both inhibitors reduced graft complement deposition, neutrophil and macrophage infiltration, adhesion molecule expression (P-selectin, E-selectin, and I-CAM-1), and proinflammatory cytokine expression (TNF-α, IL-1ß, KC, and MCP-1). The reduction in myocardial damage and cellular infiltration was not significantly different between CR2-Crry- and CR2-fH-treated mice, although adhesion molecule and cytokine levels were significantly lower in CR2-Crry-treated mice compared with CR2-fH-treated mice. In conclusion, the alternative complement pathway plays a major contributing role in myocardial IRI after heart transplantation, and local (targeted) complement inhibition has the potential to provide an effective and safe therapeutic strategy to reduce graft injury. Although total complement blockade may be somewhat more efficacious in terms of reducing inflammation, specific blockade of the alternative pathway is likely to be less immunosuppressive in an already immunocompromised recipient.


Assuntos
Fator H do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Via Alternativa do Complemento/imunologia , Transplante de Coração/imunologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Proteínas Recombinantes de Fusão/fisiologia , Animais , Fator H do Complemento/uso terapêutico , Proteínas Inativadoras do Complemento/uso terapêutico , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Sistemas de Liberação de Medicamentos/métodos , Transplante de Coração/patologia , Contagem de Leucócitos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Distribuição Aleatória , Receptores de Complemento 3d/fisiologia , Receptores de Complemento 3d/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Transplante Homólogo
9.
J Immunol ; 185(5): 3086-94, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20675597

RESUMO

Congenital and acquired deficiencies of complement regulatory proteins are associated with pathologic complement activation in several renal diseases. To elucidate the mechanisms by which renal tubular epithelial cells (TECs) control the complement system, we examined the expression of complement regulatory proteins by the cells. We found that Crry is the only membrane-bound complement regulator expressed by murine TECs, and its expression is concentrated on the basolateral surface. Consistent with the polarized localization of Crry, less complement activation was observed when the basolateral surface of TECs was exposed to serum than when the apical surface was exposed. Furthermore, greater complement activation occurred when the basolateral surface of TECs from Crry(-/-)fB(-/-) mice was exposed to normal serum compared with TECs from wild-type mice. Complement activation on the apical and basolateral surfaces was also greater when factor H, an alternative pathway regulatory protein found in serum, was blocked from interacting with the cells. Finally, we injected Crry(-/-)fB(-/-) and Crry(+/+)fB(-/-) mice with purified factor B (an essential protein of the alternative pathway). Spontaneous complement activation was seen on the tubules of Crry(-/-)fB(-/-) mice after injection with factor B, and the mice developed acute tubular injury. These studies indicate that factor H and Crry regulate complement activation on the basolateral surface of TECs and that factor H regulates complement activation on the apical surface. However, congenital deficiency of Crry or reduced expression of the protein on the basolateral surface of injured cells permits spontaneous complement activation and tubular injury.


Assuntos
Fator H do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Células Epiteliais/imunologia , Túbulos Renais/imunologia , Receptores de Complemento/fisiologia , Animais , Células Cultivadas , Fator H do Complemento/biossíntese , Fator H do Complemento/deficiência , Proteínas Inativadoras do Complemento/deficiência , Via Alternativa do Complemento/imunologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Membrana/deficiência , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica/imunologia , Receptores de Complemento/biossíntese , Receptores de Complemento/deficiência , Receptores de Complemento 3b
10.
Pharmacol Rep ; 62(2): 220-32, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20508277

RESUMO

Guillain-Barré syndrome (GBS) is an autoimmune and post-infectious immune disease. The syndrome includes several pathological subtypes, the most common of which is a multifocal demyelinating disorder of the peripheral nerves. In the present review, the main clinical aspects and the basic features of GBS are discussed along with approaches to diagnosis and treatment. Furthermore, the pathophysiology of GBS is reviewed, with an emphasis on the production of symptoms and the course of the disease.


Assuntos
Síndrome de Guillain-Barré/terapia , Corticosteroides/uso terapêutico , Proteínas Inativadoras do Complemento/fisiologia , Síndrome de Guillain-Barré/diagnóstico , Síndrome de Guillain-Barré/epidemiologia , Síndrome de Guillain-Barré/etiologia , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Troca Plasmática
11.
J Immunol ; 184(4): 1956-67, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20089702

RESUMO

Kaposica, the complement regulator of Kaposi's sarcoma-associated herpesvirus, inhibits complement by supporting factor I-mediated inactivation of the proteolytically activated form of C3 (C3b) and C4 (C4b) (cofactor activity [CFA]) and by accelerating the decay of classical and alternative pathway C3-convertases (decay-accelerating activity [DAA]). Previous data suggested that electrostatic interactions play a critical role in the binding of viral complement regulators to their targets, C3b and C4b. We therefore investigated how electrostatic potential on Kaposica influences its activities. We built a homology structure of Kaposica and calculated the electrostatic potential of the molecule, using the Poisson-Boltzmann equation. Mutants were then designed to alter the overall positive potential of the molecule or of each of its domains and linkers by mutating Lys/Arg to Glu/Gln, and the functional activities of the expressed mutants were analyzed. Our data indicate that 1) positive potential at specific sites and not the overall positive potential on the molecule guides the CFAs and classical pathway DAA; 2) positive potential around the linkers between complement control protein domains (CCPs) 1-2 and 2-3 is more important for DAAs than for CFAs; 3) positive potential in CCP1 is crucial for binding to C3b and C4b, and thereby its functional activities; 4) conversion to negative or enhancement of negative potential for CCPs 2-4 has a marked effect on C3b-linked activities as opposed to C4b-linked activities; and 5) reversal of the electrostatic potential of CCP4 to negative has a differential effect on classical and alternative pathway DAAs. Together, our data provide functional relevance to conservation of positive potential in CCPs 1 and 4 and the linkers of viral complement regulators.


Assuntos
Proteínas Inativadoras do Complemento/fisiologia , Herpesvirus Humano 8/imunologia , Eletricidade Estática , Proteínas Virais/fisiologia , Proteínas Inativadoras do Complemento/genética , Via Alternativa do Complemento/genética , Via Alternativa do Complemento/imunologia , Herpesvirus Humano 8/genética , Humanos , Mutagênese Sítio-Dirigida , Proteínas Virais/genética
12.
J Virol ; 84(7): 3210-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20071581

RESUMO

A major obstacle to gene transduction by viral vectors is inactivation by human complement in vivo. One way to overcome this is to incorporate complement regulatory proteins, such as CD55/decay accelerating factor (DAF), into viral particles. Lentivirus vectors pseudotyped with the baculovirus envelope protein GP64 have been shown to acquire more potent resistance to serum inactivation and longer transgene expression than those pseudotyped with the vesicular stomatitis virus (VSV) envelope protein G. However, the molecular mechanisms underlying resistance to serum inactivation in pseudotype particles bearing the GP64 have not been precisely elucidated. In this study, we generated pseudotype and recombinant VSVs bearing the GP64. Recombinant VSVs generated in human cell lines exhibited the incorporation of human DAF in viral particles and were resistant to serum inactivation, whereas those generated in insect cells exhibited no incorporation of human DAF and were sensitive to complement inactivation. The GP64 and human DAF were detected on the detergent-resistant membrane and were coprecipitated by immunoprecipitation analysis. A pseudotype VSV bearing GP64 produced in human DAF knockdown cells reduced resistance to serum inactivation. In contrast, recombinant baculoviruses generated in insect cells expressing human DAF or carrying the human DAF gene exhibited resistance to complement inactivation. These results suggest that the incorporation of human DAF into viral particles by interacting with baculovirus GP64 is involved in the acquisition of resistance to serum inactivation.


Assuntos
Baculoviridae/genética , Antígenos CD55/genética , Proteínas Inativadoras do Complemento/genética , Proteínas Virais de Fusão/genética , Vírion/fisiologia , Animais , Bombyx , Antígenos CD55/fisiologia , Células Cultivadas , Proteínas Inativadoras do Complemento/fisiologia , Terapia Genética , Glicosilação , Humanos , Spodoptera , Vírus da Estomatite Vesicular Indiana/genética
13.
J Immunol ; 183(9): 5928-37, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19828624

RESUMO

The alternative pathway (AP) of complement is required for the induction of collagen Ab-induced arthritis (CAIA) in mice. The objective of this study was to examine the effect of a recombinant AP inhibitor containing complement receptor 2 and factor H (CR2-fH) on CAIA in mice. CR2 binds to tissue-fixed activation fragments of C3, and the linked fH is a potent local inhibitor of the AP. CAIA was induced in C57BL/6 mice by i.p. injections of 4 mAb to type II collagen (CII) on day 0 and LPS on day 3. PBS or CR2-fH (250 or 500 microg) were injected i.p. 15 min after the mAb to CII on day 0 and 15 min after LPS on day 3; the mice were sacrificed on day 10. The disease activity score (DAS) was decreased significantly (p < 0.001) in both groups receiving CR2-fH compared with the PBS. Histology scores for inflammation, pannus, bone damage, and cartilage damage decreased in parallel with the DAS. C3 deposition in the synovium and cartilage was significantly reduced (p < 0.0001) in the mice treated with CR2-fH. In vitro studies with immune complexes containing type II collagen and mAb to CII showed that CR2-fH specifically inhibited the AP with minimal effect on the classical pathway (CP) and no effect on the lectin pathway (LP). The relative potency of CR2-fH in vitro was superior to mAbs to factor B and C5. Thus, CR2-fH specifically targets and inhibits the AP of complement in vitro and is effective in CAIA in vivo.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Artrite Experimental/imunologia , Colágeno Tipo II/imunologia , Fator H do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Via Alternativa do Complemento/imunologia , Receptores de Complemento 3d/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Animais , Artrite Experimental/patologia , Artrite Experimental/terapia , Bovinos , Fator H do Complemento/administração & dosagem , Proteínas Inativadoras do Complemento/administração & dosagem , Combinação de Medicamentos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Complemento 3d/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem
14.
J Immunol ; 183(4): 2565-74, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19625656

RESUMO

Staphylococcus aureus possesses an impressive arsenal of complement evasion proteins that help the bacterium escape attack of the immune system. The staphylococcal complement inhibitor (SCIN) protein exhibits a particularly high potency and was previously shown to block complement by acting at the level of the C3 convertases. However, many details about the exact binding and inhibitory mechanism remained unclear. In this study, we demonstrate that SCIN directly binds with nanomolar affinity to a functionally important area of C3b that lies near the C terminus of its beta-chain. Direct competition of SCIN with factor B for C3b slightly decreased the formation of surface-bound convertase. However, the main inhibitory effect can be attributed to an entrapment of the assembled convertase in an inactive state. Whereas native C3 is still able to bind to the blocked convertase, no generation and deposition of C3b could be detected in the presence of SCIN. Furthermore, SCIN strongly competes with the binding of factor H to C3b and influences its regulatory activities: the SCIN-stabilized convertase was essentially insensitive to decay acceleration by factor H and the factor I- and H-mediated conversion of surface-bound C3b to iC3b was significantly reduced. By targeting a key area on C3b, SCIN is able to block several essential functions within the alternative pathway, which explains the high potency of the inhibitor. Our findings provide an important insight into complement evasion strategies by S. aureus and may act as a base for further functional studies.


Assuntos
Complemento C3b/metabolismo , Proteínas Inativadoras do Complemento/fisiologia , Família Multigênica/imunologia , Staphylococcus aureus/imunologia , C3 Convertase da Via Alternativa do Complemento/metabolismo , C3 Convertase da Via Alternativa do Complemento/fisiologia , Complemento C3b/química , Fator B do Complemento/metabolismo , Fator H do Complemento/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Humanos , Ligação Proteica/imunologia , Staphylococcus aureus/patogenicidade , Virulência
15.
Adv Drug Deliv Rev ; 61(4): 303-9, 2009 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-19385090

RESUMO

Thioredoxin 1 (Trx 1) is a redox-active small protein ubiquitously present in human body. It is one of the defensive proteins induced in response to various stress conditions. In addition to its anti-oxidative effect by dithiol-disulfide exchange in its active site, Trx 1 has anti-apoptotic and anti-inflammatory effects. Trx 1 overexpression has been shown to be effective in a wide variety of animal models for oxidative and inflammatory disorders. An administration of recombinant Trx 1 protein is also effective in animal models especially for severe acute lung diseases where Trx 1 is likely to act with its anti-inflammatory properties. Trx 1 in circulation shows anti-chemotactic effects for neutrophils and inhibitory effects against macrophage migration inhibitory factor (MIF). Neovascularization is also suppressed by Trx 1 via inhibition of the complement activation. Here we discuss precise mechanisms of Trx 1 and potential therapeutic approach of this molecule.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Tiorredoxinas/administração & dosagem , Tiorredoxinas/uso terapêutico , Animais , Fatores Quimiotáticos/antagonistas & inibidores , Proteínas Inativadoras do Complemento/administração & dosagem , Proteínas Inativadoras do Complemento/fisiologia , Proteínas Inativadoras do Complemento/uso terapêutico , Citoproteção/fisiologia , Modelos Animais de Doenças , Humanos , Neutrófilos/patologia , Neutrófilos/fisiologia , Tiorredoxinas/fisiologia
16.
J Immunol ; 181(6): 4199-207, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768877

RESUMO

Despite eradication of smallpox three decades ago, public health concerns remain due to its potential use as a bioterrorist weapon. Smallpox and other orthopoxviruses express virulence factors that inhibit the host's complement system. In this study, our goals were to characterize the ability of the smallpox inhibitor of complement enzymes, SPICE, to regulate human complement on the cell surface. We demonstrate that SPICE binds to a variety of cell types and that the heparan sulfate and chondroitin sulfate glycosaminoglycans serve as attachment sites. A transmembrane-engineered version as well as soluble recombinant SPICE inhibited complement activation at the C3 convertase step with equal or greater efficiency than that of the related host regulators. Moreover, SPICE attached to glycosaminoglycans was more efficient than transmembrane SPICE. We also demonstrate that this virulence activity of SPICE on cells could be blocked by a mAb to SPICE. These results provide insights related to the complement inhibitory activities of poxviral inhibitors of complement and describe a mAb with therapeutic potential.


Assuntos
Membrana Celular/imunologia , Enzimas Ativadoras do Complemento/antagonistas & inibidores , Ativação do Complemento/imunologia , Proteínas Inativadoras do Complemento/fisiologia , Vírus da Varíola/imunologia , Proteínas da Matriz Viral/fisiologia , Proteínas Virais/fisiologia , Fatores de Virulência/fisiologia , Ligação Viral , Animais , Células CHO , Membrana Celular/enzimologia , Membrana Celular/genética , Membrana Celular/virologia , Enzimas Ativadoras do Complemento/metabolismo , Ativação do Complemento/genética , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Convertases de Complemento C3-C5/fisiologia , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/metabolismo , Cricetinae , Cricetulus , Glicosaminoglicanos/metabolismo , Células HeLa , Humanos , Ligação Proteica/genética , Ligação Proteica/imunologia , Estrutura Terciária de Proteína/genética , Vírus da Varíola/patogenicidade , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
17.
J Immunol ; 180(6): 3964-8, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18322205

RESUMO

Ixodes scapularis salivary protein 20 (Salp20) is a member of the Ixodes scapularis anti-complement protein-like family of tick salivary proteins that inhibit the alternative complement pathway. In this study, we demonstrate that the target of Salp20 is properdin. Properdin is a natural, positive regulator of the alternative pathway that binds to the C3 convertase, stabilizing the molecule. Salp20 directly bound to and displaced properdin from the C3 convertase. Displacement of properdin accelerated the decay of the C3 convertase, leading to inhibition of the alternative pathway. S20NS is distinct from known decay accelerating factors, such as decay accelerating factor, complement receptor 1, and factor H, which directly interact with either C3b or cleaved factor B.


Assuntos
Proteínas Inativadoras do Complemento/química , Proteínas Inativadoras do Complemento/metabolismo , Properdina/metabolismo , Proteínas e Peptídeos Salivares/metabolismo , Animais , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Proteínas Inativadoras do Complemento/fisiologia , Via Alternativa do Complemento/imunologia , Humanos , Ixodes , Ligação Proteica/imunologia , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/fisiologia
18.
J Immunol ; 180(5): 3426-35, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18292569

RESUMO

Complement forms a key arm of innate immune defenses against gonococcal infection. Sialylation of gonococcal lipo-oligosaccharide, or expression of porin 1A (Por1A) protein, enables Neisseria gonorrhoeae to bind the alternative pathway complement inhibitor, factor H (fH), and evade killing by human complement. Using recombinant fH fragment-murine Fc fusion proteins, we localized two N. gonorrhoeae Por1A-binding regions in fH: one in complement control protein domain 6, the other in complement control proteins 18-20. The latter is similar to that reported previously for sialylated Por1B gonococci. Upon incubation with human serum, Por1A and sialylated Por1B strains bound full-length human fH (HufH) and fH-related protein 1. In addition, Por1A strains bound fH-like protein 1 weakly. Only HufH, but not fH from other primates, bound directly to gonococci. Consistent with direct HufH binding, unsialylated Por1A gonococci resisted killing only by human complement, but not complement from other primates, rodents or lagomorphs; adding HufH to these heterologous sera restored serum resistance. Lipo-oligosaccharide sialylation of N. gonorrhoeae resulted in classical pathway regulation as evidenced by decreased C4 binding in human, chimpanzee, and rhesus serum but was accompanied by serum resistance only in human and chimpanzee serum. Direct-binding specificity of HufH only to gonococci that prevents serum killing is restricted to humans and may in part explain species-specific restriction of natural gonococcal infection. Our findings may help to improve animal models for gonorrhea while also having implications in the choice of complement sources to evaluate neisserial vaccine candidates.


Assuntos
Atividade Bactericida do Sangue/imunologia , Fator H do Complemento/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Via Clássica do Complemento/imunologia , Neisseria gonorrhoeae/imunologia , Motivos de Aminoácidos/imunologia , Animais , Fator H do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Lipopolissacarídeos/sangue , Macaca mulatta , Camundongos , Neisseria gonorrhoeae/metabolismo , Oligossacarídeos/sangue , Pan troglodytes , Papio , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Porinas/metabolismo , Ligação Proteica/imunologia , Especificidade da Espécie
19.
Mol Immunol ; 45(5): 1485-93, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17915330

RESUMO

Pathogenic fungi represent a major threat particularly to immunocompromised hosts, leading to severe, and often lethal, systemic opportunistic infections. Although the impaired immune status of the host is clearly the most important factor leading to disease, virulence factors of the fungus also play a role. Factor H (FH) and its splice product FHL-1 represent the major fluid phase inhibitors of the alternative pathway of complement, whereas C4b-binding protein (C4bp) is the main fluid phase inhibitor of the classical and lectin pathways. Both proteins can bind to the surface of various human pathogens conveying resistance to complement destruction and thus contribute to their pathogenic potential. We have recently shown that Candida albicans evades complement by binding both Factor H and C4bp. Here we show that moulds such as Aspergillus spp. bind Factor H, the splicing variant FHL-1 and also C4bp. Immunofluorescence and flow cytometry studies show that the binding of Factor H and C4bp to Aspergillus spp. appears to be even stronger than to Candida spp. and that different, albeit possibly nearby, binding moieties mediate this surface attachment.


Assuntos
Proteína de Ligação ao Complemento C4b/metabolismo , Fator H do Complemento/metabolismo , Proteínas Inativadoras do Complemento/fisiologia , Imunidade , Aspergillus/imunologia , Proteínas Inativadoras do Complemento C3b , Humanos , Ligação Proteica
20.
J Immunol ; 179(6): 4187-92, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17785858

RESUMO

Complement activation is a central component of inflammation and sepsis and can lead to significant tissue injury. Complement factors are serum proteins that work through a cascade of proteolytic reactions to amplify proinflammatory signals. Inter-alpha-trypsin inhibitor (IaI) is an abundant serum protease inhibitor that contains potential complement-binding domains, and has been shown to improve survival in animal sepsis models. We hypothesized that IaI can bind complement and inhibit complement activation, thus ameliorating complement-dependent inflammation. We evaluated this hypothesis with in vitro complement activation assays and in vivo in a murine model of complement-dependent lung injury. We found that IaI inhibited complement activation through the classical and alternative pathways, inhibited complement-dependent phagocytosis in vitro, and reduced complement-dependent lung injury in vivo. This novel function of IaI provides a mechanistic explanation for its observed salutary effects in sepsis and opens new possibilities for its use as a treatment agent in inflammatory diseases.


Assuntos
alfa-Globulinas/fisiologia , Ativação do Complemento/imunologia , Proteínas Inativadoras do Complemento/fisiologia , Proteínas do Sistema Complemento/toxicidade , Pulmão/imunologia , Pulmão/patologia , alfa-Globulinas/deficiência , alfa-Globulinas/genética , alfa-Globulinas/metabolismo , Animais , Ativação do Complemento/genética , Proteínas Inativadoras do Complemento/deficiência , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Feminino , Doenças do Complexo Imune/imunologia , Doenças do Complexo Imune/metabolismo , Doenças do Complexo Imune/patologia , Doenças do Complexo Imune/prevenção & controle , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/imunologia , Ligação Proteica/imunologia , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Subunidades Proteicas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA