Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Transgenic Res ; 30(1): 11-21, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33387103

RESUMO

Decay accelerating factor (DAF), a key complement activation control protein, is a 70 kDa membrane bound glycoprotein which controls extent of formation of the C3 and C5 convertases by accelerating their decay. Using clustered regularly-interspaced short palindromic repeats, (CRISPR)/associated protein 9 (Cas9) genome editing we generated a novel DAF deficient (Daf-/-) rat model. The present study describes the renal and extrarenal phenotype of this model and assesses renal response to complement-dependent injury induced by administration of a complement-fixing antibody (anti-Fx1A) against the glomerular epithelial cell (podocyte). Rats generated were healthy, viable and able to reproduce normally. Complete absence of DAF was documented in renal as well as extra-renal tissues at both protein and mRNA level compared to Daf+/+ rats. Renal histology in Daf-/- rats showed no differences regarding glomerular or tubulointerstitial pathology compared to Daf+/+ rats. Moreover, there was no difference in urine protein excretion (ratio of urine albumin to creatinine) or in serum creatinine and urea levels. In Daf-/- rats, proteinuria was significantly increased following binding of anti-Fx1A antibody to podocytes while increased C3b deposition was observed. The DAF knock-out rat model developed validates the role of this complement cascade regulator in immune-mediated podocyte injury. Given the increasing role of dysregulated complement activation in various forms of kidney disease and the fact that the rat is the preferred animal for renal pathophysiology studies, the rat DAF deficient model may serve as a useful tool to study the role of this complement activation regulator in complement-dependent forms of kidney injury.


Assuntos
Injúria Renal Aguda/genética , Antígenos CD55/genética , Ativação do Complemento/genética , Podócitos/metabolismo , Injúria Renal Aguda/patologia , Albuminúria , Animais , Anticorpos Anti-Idiotípicos/farmacologia , Antígenos CD55/deficiência , Antígenos CD55/imunologia , Sistemas CRISPR-Cas/genética , Ativação do Complemento/imunologia , Convertases de Complemento C3-C5/genética , Complemento C5/genética , Técnicas de Inativação de Genes , Complexo Antigênico da Nefrite de Heymann/genética , Complexo Antigênico da Nefrite de Heymann/imunologia , Humanos , Podócitos/patologia , Ratos
2.
FASEB J ; 35(1): e21221, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33337564

RESUMO

Alterations in complement component 3 (C3) expression has been reported to be linked to several bowel diseases including Crohn's disease, inflammatory bowel disease, and ulcerative colitis; however, the association with constipation has never been investigated. In this study, we aimed to investigate the correlation between C3 regulation and constipation development using a C3 deficiency model. To achieve these, alterations in stool excretion, transverse colon histological structure, and mucin secretion were analyzed in FVB/N-C3em1Hlee /Korl (C3 knockout, C3 KO) mice with the deletion of 11 nucleotides in exon 2 of the C3 gene. The stool excretion parameters, gastrointestinal transit, and intestine length were remarkably decreased in C3 KO mice compared with wild-type (WT) mice, although there was no specific change in feeding behavior. Furthermore, C3 KO mice showed a decrease in mucosal and muscle layer thickness, alterations in crypt structure, irregular distribution of goblet cells, and an increase of mucin droplets in the transverse colon. Mucin secretion was suppressed, and they accumulated in the crypts of C3 KO mice. In addition, the constipation phenotypes detected during C3 deficiency were confirmed in FVB/N mice treated with C3 convertase inhibitor (rosmarinic acid (RA)). Similar phenotypes were observed with respect to stool excretion parameters, gastrointestinal transit, intestine length, alterations in crypt structure, and mucin secretion in RA-treated FVB/N mice. Therefore, the results of the present study provide the first scientific evidence that C3 deficiency may play an important role in the development of constipation phenotypes in C3 KO mice.


Assuntos
Complemento C3/deficiência , Constipação Intestinal/metabolismo , Éxons , Animais , Cinamatos/farmacologia , Complemento C3/metabolismo , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Constipação Intestinal/genética , Constipação Intestinal/patologia , Depsídeos/farmacologia , Camundongos , Camundongos Knockout , Ácido Rosmarínico
3.
J Biol Chem ; 296: 100083, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33199367

RESUMO

Inhibition of the alternative pathway (AP) of complement by saliva from Anopheles mosquitoes facilitates feeding by blocking production of the anaphylatoxins C3a and C5a, which activate mast cells leading to plasma extravasation, pain, and itching. We have previously shown that albicin, a member of the SG7 protein family from An. Albimanus, blocks the AP by binding to and inhibiting the function of the C3 convertase, C3bBb. Here we show that SG7.AF, the albicin homolog from An. freeborni, has a similar potency to albicin but is more active in the presence of properdin, a plasma protein that acts to stabilize C3bBb. Conversely, albicin is highly active in the absence or presence of properdin. Albicin and SG7.AF stabilize the C3bBb complex in a form that accumulates on surface plasmon resonance (SPR) surfaces coated with properdin, but SG7.AF binds with lower affinity than albicin. Albicin induces oligomerization of the complex in solution, suggesting that it is oligomerization that leads to stabilization on SPR surfaces. Anophensin, the albicin ortholog from An. stephensi, is only weakly active as an inhibitor of the AP, suggesting that the SG7 family may play a different functional role in this species and other species of the subgenus Cellia, containing the major malaria vectors in Africa and Asia. Crystal structures of albicin and SG7.AF reveal a novel four-helix bundle arrangement that is stabilized by an N-terminal hydrogen bonding network. These structures provide insight into the SG7 family and related mosquito salivary proteins including the platelet-inhibitory 30 kDa family.


Assuntos
Inativadores do Complemento/química , Inativadores do Complemento/metabolismo , Properdina/metabolismo , Saliva/química , Animais , Anopheles , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Via Alternativa do Complemento/genética , Via Alternativa do Complemento/fisiologia , Cristalografia por Raios X , Culicidae , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Properdina/genética , Ressonância de Plasmônio de Superfície
4.
Mol Vis ; 26: 48-63, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32165826

RESUMO

Purpose: The use of small non-coding nucleic acids, such as siRNA and miRNA, has allowed for a deeper understanding of gene functions, as well as for development of gene therapies for complex neurodegenerative diseases, including retinal degeneration. For effective delivery into the eye and transfection of the retina, suitable transfection methods are required. We investigated the use of a lipid-based transfection agent, Invivofectamine® 3.0 (Thermo Fisher Scientific), as a potential method for delivery of nucleic acids to the retina. Methods: Rodents were injected intravitreally with formulations of Invivofectamine 3.0 containing scrambled, Gapdh, Il-1ß, and C3 siRNAs, or sterile PBS (control) using a modified protocol for encapsulation of nucleic acids. TdT-mediated dUTP nick-end labeling (TUNEL) and IBA1 immunohistochemistry was used to determine histological cell death and inflammation. qPCR were used to determine the stress and inflammatory profile of the retina. Electroretinography (ERG) and optical coherence tomography (OCT) were employed as clinical indicators of retinal health. Results: We showed that macrophage recruitment, retinal stress, and photoreceptor cell death in animals receiving Invivofectamine 3.0 were comparable to those in negative controls. Following delivery of Invivofectamine 3.0 alone, no statistically significant changes in expression were found in a suite of inflammatory and stress genes, and ERG and OCT analyses revealed no changes in retinal function or morphology. Injections with siRNAs for proinflammatory genes (C3 and Il-1ß) and Gapdh, in combination with Invivofectamine 3.0, resulted in statistically significant targeted gene knockdown in the retina for up to 4 days following injection. Using a fluorescent Block-It siRNA, transfection was visualized throughout the neural retina with evidence of transfection observed in cells of the ganglion cell layer, inner nuclear layer, and outer nuclear layer. Conclusions: This work supports the use of Invivofectamine 3.0 as a transfection agent for effective delivery of nucleic acids to the retina for gene function studies and as potential therapeutics.


Assuntos
Técnicas de Silenciamento de Genes/métodos , Lipoproteínas/farmacologia , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Transfecção/métodos , Animais , Morte Celular/genética , Convertases de Complemento C3-C5/genética , Modelos Animais de Doenças , Portadores de Fármacos/química , Eletrorretinografia , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/genética , Marcação In Situ das Extremidades Cortadas , Interleucina-1beta/genética , Lipídeos/química , Lipídeos/farmacologia , Lipoproteínas/química , Camundongos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ratos , Retina/diagnóstico por imagem , Tomografia de Coerência Óptica
5.
Cancer Immunol Immunother ; 68(4): 587-598, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30725204

RESUMO

Anti-CD20 monoclonal antibodies (mAbs) rituximab and ofatumumab are potent activators of the classical complement pathway, and have been approved for the treatment of B-cell malignancies. However, complement exhaustion and overexpression of complement inhibitors by cancer cells diminish their therapeutic potential. The strategies of targeting membrane complement inhibitors by function-blocking antibodies and the supplementation with fresh frozen plasma have been proposed to overcome tumour cell resistance. We present a novel approach, which utilizes gain-of-function variants of complement factor B (FB), a component of alternative C3/C5 convertases, which augment mAb-activated reactions through a positive feedback mechanism called an amplification loop. If complement concentration is limited, an addition of quadruple gain-of-function FB mutant p.D279G p.F286L p.K323E p.Y363A (or selected single mutants) results in significantly increased complement-mediated lysis of ofatumumab-resistant tumour cells, as well as the complete lysis of moderately sensitive cells. Importantly, this effect cannot be achieved by further increasing ofatumumab concentration. Potentiation of cytotoxic effect towards moderately sensitive cells was less apparent at physiological serum concentration. However, an addition of hyperactive FB could compensate the loss of cytotoxic potential of serum collected from the NHL and CLL patients after infusion of rituximab. Residual levels of rituximab in such sera, in combination with added FB, were able to efficiently lyse tumour cells. We suggest that the administration of gain-of-function variants of FB can restore cytotoxic potential of complement-exhausted serum and maximize the therapeutic effect of circulating anti-CD20 mAbs.


Assuntos
Antígenos CD20/metabolismo , Antineoplásicos Imunológicos/farmacologia , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/imunologia , Mutação , Rituximab/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Biomarcadores , Linhagem Celular Tumoral , Via Alternativa do Complemento/imunologia , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/imunologia , Citotoxicidade Imunológica , Mutação com Ganho de Função , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/metabolismo , Leucemia Linfocítica Crônica de Células B/patologia
6.
Front Immunol ; 9: 1191, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29892304

RESUMO

The complement system, composed of the three activation pathways, has both protective and pathogenic roles in the development of systemic lupus erythematosus (or lupus), a prototypic autoimmune disease. The classical pathway contributes to the clearance of immune complexes (ICs) and apoptotic cells, whereas the alternative pathway (AP) exacerbates renal inflammation. The role of the lectin pathway (LP) in lupus has remained largely unknown. Mannose-binding lectin (MBL)-associated serine proteases (MASPs), which are associated with humoral pattern recognition molecules (MBL or ficolins), are the enzymatic constituents of the LP and AP. MASP-1 encoded by the Masp1 gene significantly contributes to the activation of the LP. After the binding of MBL/ficolins to pathogens or self-altered cells, MASP-1 autoactivates first, then activates MASP-2, and both participate in the formation of the LP C3 convertase C4b2a, whereas, MASP-3, the splice variant of the Masp1 gene, is required for the activation of the zymogen of factor D (FD), and finally participates in the formation of the AP C3 convertase C3bBb. To investigate the roles of MASP-1 and MASP-3 in lupus, we generated Masp1 gene knockout lupus-prone MRL/lpr mice (Masp1/3-/- MRL/lpr mice), lacking both MASP-1 and MASP-3, and analyzed their renal disease. As expected, sera from Masp1/3-/- MRL/lpr mice had no or markedly reduced activation of the LP and AP with zymogen forms of complement FD. Compared to their wild-type littermates, the Masp1/3-/- MRL/lpr mice had maintained serum C3 levels, little-to-no albuminuria, as well as significantly reduced glomerular C3 deposition levels and glomerular pathological score. On the other hand, there were no significant differences in the levels of serum anti-dsDNA antibody, circulating ICs, glomerular IgG and MBL/ficolins deposition, renal interstitial pathological score, urea nitrogen, and mortality between the wild-type and Masp1/3-/- MRL/lpr mice. Our data indicate that MASP-1/3 plays essential roles in the development of lupus-like glomerulonephritis in MRL/lpr mice, most likely via activation of the LP and/or AP.


Assuntos
Via Alternativa do Complemento/imunologia , Lectina de Ligação a Manose da Via do Complemento/imunologia , Nefrite Lúpica/imunologia , Serina Proteases Associadas a Proteína de Ligação a Manose/imunologia , Animais , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/imunologia , Via Alternativa do Complemento/genética , Lectina de Ligação a Manose da Via do Complemento/genética , Nefrite Lúpica/genética , Nefrite Lúpica/patologia , Serina Proteases Associadas a Proteína de Ligação a Manose/genética , Camundongos , Camundongos Endogâmicos MRL lpr , Camundongos Knockout
7.
EMBO J ; 36(8): 1084-1099, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28264884

RESUMO

Properdin (FP) is an essential positive regulator of the complement alternative pathway (AP) providing stabilization of the C3 and C5 convertases, but its oligomeric nature challenges structural analysis. We describe here a novel FP deficiency (E244K) caused by a single point mutation which results in a very low level of AP activity. Recombinant FP E244K is monomeric, fails to support bacteriolysis, and binds weakly to C3 products. We compare this to a monomeric unit excised from oligomeric FP, which is also dysfunctional in bacteriolysis but binds the AP proconvertase, C3 convertase, C3 products and partially stabilizes the convertase. The crystal structure of such a FP-convertase complex suggests that the major contact between FP and the AP convertase is mediated by a single FP thrombospondin repeat and a small region in C3b. Small angle X-ray scattering indicates that FP E244K is trapped in a compact conformation preventing its oligomerization. Our studies demonstrate an essential role of FP oligomerization in vivo while our monomers enable detailed structural insight paving the way for novel modulators of complement.


Assuntos
Convertases de Complemento C3-C5/química , Via Alternativa do Complemento , Properdina/química , Multimerização Proteica , Substituição de Aminoácidos , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/metabolismo , Humanos , Mutação de Sentido Incorreto , Properdina/deficiência , Properdina/genética , Properdina/metabolismo , Domínios Proteicos
8.
Clin Exp Immunol ; 184(1): 118-25, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26660535

RESUMO

Properdin (P) stabilizes the alternative pathway (AP) convertases, being the only known positive regulator of the complement system. In addition, P is a pattern recognition molecule able to initiate directly the AP on non-self surfaces. Although P deficiencies have long been known to be associated with Neisseria infections and P is often found deposited at sites of AP activation and tissue injury, the potential role of P in the pathogenesis of complement dysregulation-associated disorders has not been studied extensively. Serum P levels were measured in 49 patients with histological and clinical evidence of C3 glomerulopathy (C3G). Patients were divided into two groups according to the presence or absence of C3 nephritic factor (C3NeF), an autoantibody that stabilizes the AP C3 convertase. The presence of this autoantibody results in a significant reduction in circulating C3 (P < 0·001) and C5 levels (P < 0·05), but does not alter factor B, P and sC5b-9 levels. Interestingly, in our cohort, serum P levels were low in 17 of the 32 C3NeF-negative patients. This group exhibited significant reduction of C3 (P < 0·001) and C5 (P < 0·001) and increase of sC5b-9 (P < 0·001) plasma levels compared to the control group. Also, P consumption was correlated significantly with C3 (r = 0·798, P = 0·0001), C5 (r = 0·806, P < 0·0001), sC5b-9 (r = -0·683, P = 0·043) and a higher degree of proteinuria (r = -0·862, P = 0·013). These results illustrate further the heterogeneity among C3G patients and suggest that P serum levels could be a reliable clinical biomarker to identify patients with underlying surface AP C5 convertase dysregulation.


Assuntos
Convertases de Complemento C3-C5/imunologia , Via Alternativa do Complemento , Glomerulonefrite/imunologia , Properdina/imunologia , Proteinúria/imunologia , Adolescente , Adulto , Biomarcadores/sangue , Criança , Complemento C3/genética , Complemento C3/imunologia , Fator Nefrítico do Complemento 3/genética , Fator Nefrítico do Complemento 3/imunologia , Convertases de Complemento C3-C5/genética , Complemento C5/genética , Complemento C5/imunologia , Fator B do Complemento/genética , Fator B do Complemento/imunologia , Inativadores do Complemento/sangue , Complexo de Ataque à Membrana do Sistema Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Feminino , Regulação da Expressão Gênica , Glomerulonefrite/sangue , Glomerulonefrite/genética , Glomerulonefrite/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Properdina/genética , Proteinúria/sangue , Proteinúria/genética , Proteinúria/patologia , Estudos Retrospectivos , Índice de Gravidade de Doença , Transdução de Sinais
9.
J Am Soc Nephrol ; 26(12): 2917-29, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26185203

RESUMO

Kidney diseases caused by genetic or acquired dysregulation of the complement alternative pathway (AP) are traditionally classified on the basis of clinical presentation (atypical hemolytic uremic syndrome as thrombotic microangiopathy), biopsy appearance (dense deposit disease and C3 GN), or clinical course (atypical postinfectious GN). Each is characterized by an inappropriate activation of the AP, eventuating in renal damage. The clinical diversity of these disorders highlights important differences in the triggers, the sites and intensity of involvement, and the outcome of the AP dysregulation. Nevertheless, we contend that these diseases should be grouped as disorders of the AP and classified on an etiologic basis. In this review, we define different pathophysiologic categories of AP dysfunction. The precise identification of the underlying abnormality is the key to predict the response to immune suppression, plasma infusion, and complement-inhibitory drugs and the outcome after transplantation. In a patient with presumed dysregulation of the AP, the collaboration of the clinician, the renal pathologist, and the biochemical and genetic laboratory is very much encouraged, because this enables the elucidation of both the underlying pathogenesis and the optimal therapeutic approach.


Assuntos
Fator H do Complemento/deficiência , Via Alternativa do Complemento/fisiologia , Nefropatias/imunologia , Animais , Autoanticorpos , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/imunologia , Fator H do Complemento/metabolismo , Doenças da Deficiência Hereditária de Complemento , Humanos , Nefropatias/classificação , Nefropatias/terapia
10.
J Immunol ; 193(3): 1485-95, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24973443

RESUMO

Binding of C1q to target-bound IgG initiates complement-mediated lysis (CML) of pathogens, as well as of malignant or apoptotic cells, and thus constitutes an integral part of the innate immune system. Despite its prominent molecular flexibility and higher C1q binding affinity compared with human IgG1, IgG3 does not consistently promote superior CML. Hence the aim of this study was to investigate underlying molecular mechanisms of IgG1- and IgG3-driven complement activation using isotype variants of the therapeutic epidermal growth factor receptor (EGFR) Ab cetuximab. Both IgG1 and IgG3 Abs demonstrated similar EGFR binding and similar efficiency in Fab-mediated effector mechanisms. Whereas anti-EGFR-IgG1 did not promote CML of investigated target cells, anti-EGFR-IgG3 triggered significant CML of some, but not all tested cell lines. CML triggered by anti-EGFR-IgG3 negatively correlated with expression levels of the membrane-bound complement regulatory proteins CD55 and CD59, but not CD46. Notably, anti-EGFR-IgG3 promoted strong C1q and C3b, but relatively low C4b and C5b-9 deposition on analyzed cell lines. Furthermore, anti-EGFR-IgG3 triggered C4a release on all cells but failed to induce C3a and C5a release on CD55/CD59 highly expressing cells. RNA interference-induced knockdown or overexpression of membrane-bound complement regulatory proteins revealed CD55 expression to be a pivotal determinant of anti-EGFR-IgG3-triggered CML and to force a switch from classical complement pathway activation to C1q-dependent alternative pathway amplification. Together, these data suggest human anti-EGFR-IgG3, although highly reactive with C1q, to weakly promote assembly of the classical C3 convertase that is further suppressed in the presence of CD55, forcing human IgG3 to act mainly through the alternative pathway.


Assuntos
Antígenos CD55/genética , Via Alternativa do Complemento/genética , Via Alternativa do Complemento/imunologia , Citotoxicidade Imunológica/genética , Receptores ErbB/genética , Imunoglobulina G/genética , Animais , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/uso terapêutico , Antígenos CD55/biossíntese , Antígenos CD55/química , Células CHO , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/genética , Cricetinae , Cricetulus , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/imunologia , Feminino , Marcação de Genes/métodos , Humanos , Imunoglobulina G/uso terapêutico , Técnicas de Amplificação de Ácido Nucleico/métodos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia
11.
Dev Comp Immunol ; 46(2): 430-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24877658

RESUMO

The complement system is a fundamental effector mechanism of the innate immunity in both vertebrates and invertebrates. The comprehension of its roots in the evolution is a useful step to understand how the main complement-related proteins had changed in order to adapt to new environmental conditions and life-cycles or, in the case of vertebrates, to interact with the adaptive immunity. Data on organisms evolutionary close to vertebrates, such as tunicates, are of primary importance for a better understanding of the changes in immune responses associated with the invertebrate-vertebrate transition. Here we report on the characterization of C3 and Bf transcripts from the colonial ascidian Botryllus schlosseri (BsC3 and BsBf, respectively), a reliable model organism for immunobiological research, and present a comparative analysis of amino acid sequences of C3s and Bfs suggesting that, in deuterostomes, the structure of these proteins remained largely unchanged. We also present new data on the cells responsible of the expression of BsC3 and BsBf showing that cytotoxic immunocytes are the sole cells where the relative transcripts can be found. Finally, using the C3 specific inhibitor compstatin, we demonstrate the opsonic role of BsC3 in accordance with the idea that promotion of phagocytosis is one of the main function of C3 in metazoans.


Assuntos
Convertases de Complemento C3-C5/metabolismo , Complemento C3/metabolismo , Peptídeos Cíclicos/farmacologia , Urocordados/imunologia , Animais , Células Cultivadas , Complemento C3/genética , Convertases de Complemento C3-C5/genética , Hemócitos/imunologia , Hemócitos/metabolismo , Imunidade Inata , Proteínas Opsonizantes/genética , Proteínas Opsonizantes/metabolismo , Fagocitose , Filogenia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica , Urocordados/citologia , Urocordados/metabolismo
12.
Immunol Lett ; 160(2): 163-71, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24491679

RESUMO

The alternative pathway of complement is implicated in the pathogenesis of several renal diseases, such as atypical hemolytic uremic syndrome, dense deposit disease and other forms of C3 glomerulopathy. The underlying complement defects include genetic and/or acquired factors, the latter in the form of autoantibodies. Because the autoimmune forms require a specific treatment, in part different from that of the genetic forms, it is important to detect the autoantibodies as soon as possible and understand their characteristics. In this overview, we summarize the types of anti-complement autoantibodies detected in such diseases, i.e. autoantibodies to factor H, factor I, C3b, factor B and those against the C3 convertases (C3 nephritic factor and C4 nephritic factor). We draw attention to newly described autoantibodies and their characteristics, and highlight similarities and differences in the autoimmune forms of these diseases.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/imunologia , Autoanticorpos/biossíntese , Glomerulonefrite por IGA/imunologia , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/patologia , Autoanticorpos/genética , Autoanticorpos/imunologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/imunologia , Complemento C3b/antagonistas & inibidores , Complemento C3b/genética , Complemento C3b/imunologia , Fator B do Complemento/antagonistas & inibidores , Fator B do Complemento/genética , Fator B do Complemento/imunologia , Fator H do Complemento/antagonistas & inibidores , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/imunologia , Fibrinogênio/antagonistas & inibidores , Fibrinogênio/genética , Fibrinogênio/imunologia , Expressão Gênica , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/patologia , Humanos
13.
PLoS One ; 8(11): e78617, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24260121

RESUMO

Mutations and deletions within the human CFHR gene cluster on chromosome 1 are associated with diseases, such as dense deposit disease, CFHR nephropathy or age-related macular degeneration. Resulting mutant CFHR proteins can affect complement regulation. Here we identify human CFHR2 as a novel alternative pathway complement regulator that inhibits the C3 alternative pathway convertase and terminal pathway assembly. CFHR2 is composed of four short consensus repeat domains (SCRs). Two CFHR2 molecules form a dimer through their N-terminal SCRs, and each of the two C-terminal ends can bind C3b. C3b bound CFHR2 still allows C3 convertase formation but the CFHR2 bound convertases do not cleave the substrate C3. Interestingly CFHR2 hardly competes off factor H from C3b. Thus CFHR2 likely acts in concert with factor H, as CFHR2 inhibits convertases while simultaneously allowing factor H assisted degradation by factor I.


Assuntos
Proteínas Inativadoras do Complemento C3b/metabolismo , Via Alternativa do Complemento/fisiologia , Proteólise , Complemento C3/química , Complemento C3/genética , Complemento C3/metabolismo , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Proteínas Inativadoras do Complemento C3b/química , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/química , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Fator I do Complemento/química , Fator I do Complemento/genética , Fator I do Complemento/metabolismo , Humanos , Multimerização Proteica/fisiologia , Estrutura Terciária de Proteína , Sequências Repetitivas de Aminoácidos
14.
Immunobiology ; 217(11): 1034-46, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22964229

RESUMO

During the last decade, numerous studies have associated genetic variations in complement components and regulators with a number of chronic and infectious diseases. The functional characterization of these complement protein variants, in addition to recent structural advances in understanding of the assembly, activation and regulation of the AP C3 convertase, have provided important insights into the pathogenic mechanisms involved in some of these complement related disorders. This knowledge has identified potential targets for complement inhibitory therapies which are demonstrating efficacy and generating considerable expectation in changing the natural history of these diseases. Comprehensive understanding of the genetic and non-genetic risk factors contributing to these disorders will also result in targeting of the right patient groups in a stratified medicine approach through better diagnostics and individually tailored treatments, thereby improving management of patients.


Assuntos
Ativação do Complemento/genética , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/metabolismo , Inflamação/genética , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Proteínas Inativadoras do Complemento/genética , Variação Genética , Glomerulonefrite Membranoproliferativa/genética , Glomerulonefrite Membranoproliferativa/imunologia , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/imunologia , Humanos , Inflamação/imunologia , Degeneração Macular/genética , Degeneração Macular/imunologia
15.
J Biol Chem ; 287(29): 24734-8, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22689574

RESUMO

Cerebral malaria (CM) is the most severe manifestation of clinical malaria syndromes and has a high fatality rate especially in the developing world. Recent studies demonstrated that C5(-/-) mice are resistant to experimental CM (ECM) and that protection was due to the inability to form the membrane attack complex. Unexpectedly, we observed that C4(-/-) and factor B(-/-) mice were fully susceptible to disease, indicating that activation of the classical or alternative pathways is not required for ECM. C3(-/-) mice were also susceptible to ECM, indicating that the canonical C5 convertases are not required for ECM development and progression. Abrogation of ECM by treatment with anti-C9 antibody and detection of C5a in serum of C3(-/-) mice confirmed that C5 activation occurs in ECM independent of C5 convertases. Our data indicate that activation of C5 in ECM likely occurs via coagulation enzymes of the extrinsic protease pathway.


Assuntos
Convertases de Complemento C3-C5/metabolismo , Malária Cerebral/imunologia , Malária Cerebral/metabolismo , Animais , Ativação do Complemento/genética , Ativação do Complemento/fisiologia , Complemento C3/genética , Complemento C3/metabolismo , Convertases de Complemento C3-C5/genética , Complemento C4/genética , Complemento C4/metabolismo , Fator B do Complemento/genética , Fator B do Complemento/metabolismo , Malária Cerebral/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Plasmodium berghei/imunologia , Plasmodium berghei/patogenicidade
16.
J Biol Chem ; 287(23): 19528-36, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22518841

RESUMO

Human complement factor H-related protein (CFHR) 4 belongs to the factor H family of plasma glycoproteins that are composed of short consensus repeat (SCR) domains. Although factor H is a well known inhibitor of the alternative complement pathway, the functions of the CFHR proteins are poorly understood. CFHR4 lacks SCRs homologous to the complement inhibitory domains of factor H and, accordingly, has no significant complement regulatory activities. We have previously shown that CFHR4 binds C-reactive protein via its most N-terminal SCR, which leads to classical complement pathway activation. CFHR4 binds C3b via its C terminus, but the significance of this interaction is unclear. Therefore, we set out to clarify the functional relevance of C3b binding by CFHR4. Here, we report a novel role for CFHR4 in the complement system. CFHR4 serves as a platform for the assembly of an alternative pathway C3 convertase by binding C3b. This is based on the sustained ability of CFHR4-bound C3b to bind factor B and properdin, leading to an active convertase that generates C3a and C3b from C3. The CFHR4-C3bBb convertase is less sensitive to the factor H-mediated decay compared with the C3bBb convertase. CFHR4 mutants containing exchanges of conserved residues within the C-terminal C3b-binding site showed significantly reduced C3b binding and alternative pathway complement activation. In conclusion, our results suggest that, in contrast to the complement inhibitor factor H, CFHR4 acts as an enhancer of opsonization by promoting complement activation.


Assuntos
Apolipoproteínas/metabolismo , Convertases de Complemento C3-C5/metabolismo , Complemento C3b/metabolismo , Via Alternativa do Complemento/fisiologia , Animais , Apolipoproteínas/genética , Sítios de Ligação , Linhagem Celular , Convertases de Complemento C3-C5/genética , Complemento C3b/genética , Fator B do Complemento/genética , Fator B do Complemento/metabolismo , Humanos , Mutação , Properdina/genética , Properdina/metabolismo , Spodoptera
17.
J Immunol ; 188(4): 2030-7, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22250080

RESUMO

Atypical hemolytic uremic syndrome has been associated with dysregulation of the alternative complement pathway. In this study, a novel heterozygous C3 mutation was identified in a factor B-binding region in exon 41, V1636A (4973 T > C). The mutation was found in three family members affected with late-onset atypical hemolytic uremic syndrome and symptoms of glomerulonephritis. All three patients exhibited increased complement activation detected by decreased C3 levels and glomerular C3 deposits. Platelets from two of the patients had C3 and C9 deposits on the cell surface. Patient sera exhibited more C3 cleavage and higher levels of C3a. The C3 mutation resulted in increased C3 binding to factor B and increased net formation of the C3 convertase, even after decay induced by decay-accelerating factor and factor H, as assayed by surface plasmon resonance. Patient sera incubated with washed human platelets induced more C3 and C9 deposition on the cell surface in comparison with normal sera. More C3a was released into serum over time when washed platelets were exposed to patient sera. Results regarding C3 and C9 deposition on washed platelets were confirmed using purified patient C3 in C3-depleted serum. The results indicated enhanced convertase formation leading to increased complement activation on cell surfaces. Previously described C3 mutations showed loss of function with regard to C3 binding to complement regulators. To our knowledge, this study presents the first known C3 mutation inducing increased formation of the C3 convertase, thus explaining enhanced activation of the alternative pathway of complement.


Assuntos
Convertases de Complemento C3-C5/biossíntese , Complemento C3a/genética , Complemento C3a/metabolismo , Complemento C3b/genética , Complemento C3b/metabolismo , Síndrome Hemolítico-Urêmica/imunologia , Animais , Síndrome Hemolítico-Urêmica Atípica , Células COS , Chlorocebus aethiops , Ativação do Complemento , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Complemento C3a/biossíntese , Complemento C3b/biossíntese , Complemento C9/biossíntese , Complemento C9/metabolismo , Fator B do Complemento/metabolismo , Fator H do Complemento/metabolismo , Via Alternativa do Complemento/genética , Glomerulonefrite/genética , Síndrome Hemolítico-Urêmica/genética , Humanos , Masculino , Pessoa de Meia-Idade
18.
J Biol Chem ; 287(11): 8092-100, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22267731

RESUMO

PRELP is a 58-kDa proteoglycan found in a variety of extracellular matrices, including cartilage and at several basement membranes. In rheumatoid arthritis (RA), the cartilage tissue is destroyed and fragmented molecules, including PRELP, are released into the synovial fluid where they may interact with components of the complement system. In a previous study, PRELP was found to interact with the complement inhibitor C4b-binding protein, which was suggested to locally down-regulate complement activation in joints during RA. Here we show that PRELP directly inhibits all pathways of complement by binding C9 and thereby prevents the formation of the membrane attack complex (MAC). PRELP does not interfere with the interaction between C9 and already formed C5b-8, but inhibits C9 polymerization thereby preventing formation of the lytic pore. The alternative pathway is moreover inhibited already at the level of C3-convertase formation due to an interaction between PRELP and C3. This suggests that PRELP may down-regulate complement attack at basement membranes and on damaged cartilage and therefore limit pathological complement activation in inflammatory disease such as RA. The net outcome of PRELP-mediated complement inhibition will highly depend on the local concentration of other complement modulating molecules as well as on the local concentration of available complement proteins.


Assuntos
Artrite Reumatoide/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Membrana Basal/química , Membrana Basal/metabolismo , Membrana Basal/patologia , Ativação do Complemento/genética , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Proteína de Ligação ao Complemento C4b/química , Proteína de Ligação ao Complemento C4b/genética , Proteína de Ligação ao Complemento C4b/metabolismo , Complemento C9/química , Complemento C9/genética , Complemento C9/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/química , Complexo de Ataque à Membrana do Sistema Complemento/genética , Proteínas do Sistema Complemento/química , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/metabolismo , Regulação para Baixo/genética , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/genética , Feminino , Glicoproteínas/química , Glicoproteínas/genética , Células HEK293 , Humanos , Masculino
19.
J Biol Chem ; 285(49): 38473-85, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20855886

RESUMO

Group A streptococci (GAS) utilize soluble human complement regulators to evade host complement attack. Here, we characterized the binding of the terminal complement complex inhibitor complement Factor H-related protein 1 (CFHR1) and of the C3 convertase regulator Factor H to the streptococcal collagen-like proteins (Scl). CFHR1 and Factor H, but no other member of the Factor H protein family (CFHR2, CFHR3, or CFHR4A), bound to the two streptococcal proteins Scl1.6 and Scl1.55, which are expressed by GAS serotypes M6 and M55. The two human regulators bound to the Scl1 proteins via their conserved C-terminal attachment region, i.e. CFHR1 short consensus repeats 3-5 (SCR3-5) and Factor H SCR18-20. Binding was affected by ionic strength and by heparin. CFHR1 and the C-terminal attachment region of Factor H did not bind to Scl1.1 and Scl2.28 proteins but did bind to intact M1-type and M28-type GAS, which express Scl1.1 and Scl2.28, respectively, thus arguing for the presence of an additional binding mechanism to CFHR1 and Factor H. Furthermore mutations within the C-terminal heparin-binding region and Factor H mutations that are associated with the acute renal disease atypical hemolytic uremic syndrome blocked the interaction with the two streptococcal proteins. Binding of CFHR1 affected the complement regulatory functions of Factor H on the level of the C3 convertase. Apparently, streptococci utilize two types of complement regulator-acquiring surface proteins; type A proteins, as represented by Scl1.6 and Scl1.55, bind to CFHR1 and Factor H via their conserved C-terminal region and do not bind the Factor H-like protein 1 (FHL-1). On the contrary, type B proteins, represented by M-, M-like, and the fibronectin-binding protein Fba proteins, bind Factor H and FHL-1 via domain SCR7 and do not bind CFHR1. In conclusion, binding of CFHR1 is at the expense of Factor H-mediated regulatory function at the level of C3 convertase and at the gain of a regulator that controls complement at the level of the C5 convertase and formation of the terminal complement complex.


Assuntos
Proteínas de Bactérias/metabolismo , Colágeno/metabolismo , Ativação do Complemento , Proteínas Inativadoras do Complemento C3b/metabolismo , Fator H do Complemento/metabolismo , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes/metabolismo , Doença Aguda , Síndrome Hemolítico-Urêmica Atípica , Proteínas de Bactérias/genética , Colágeno/genética , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/metabolismo , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Infecções Estreptocócicas/genética , Streptococcus pyogenes/genética
20.
Am J Transplant ; 9(8): 1784-95, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19563342

RESUMO

CD8 T cells primed by transplantation recognize allogeneic class I MHC molecules expressed on graft vascular endothelium and contribute to allograft injury. We previously showed that immune cell-derived complement activation fragments are integral to T cell activation/expansion. Herein we tested the impact of local complement production/activation on T cell/endothelial cell (EC) interactions. We found that proinflammatory cytokines upregulated alternative pathway complement production by ECs, yielding C5a. We further found that ECs deficient in the cell surface C3/C5 convertase regulator decay accelerating factor (DAF, CD55) induced greater CD8 T-cell proliferation and more IFNgamma(+) and perforin(+) effector cells than wild-type (WT) ECs. Allogeneic C3(-/-) EC induced little or no CD8 responses. Abrogation of responses following C5a receptor (C5aR) blockade, or augmentation following addition of recombinant C5a demonstrated that the effects were mediated through T-cell-expressed-C5aR interactions. Analyses of in vivo CD8 cell responses to transplanted heart grafts deficient in EC DAF showed similar augmentation. The findings reveal that EC-derived complement triggers secondary CD8 T-cell differentiation and expansion and argue that targeting complement and/or C5aR could limit T-cell-mediated graft injury.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Comunicação Celular/fisiologia , Proteínas do Sistema Complemento/metabolismo , Endotélio Vascular/metabolismo , Animais , Antígenos CD55/genética , Antígenos CD55/metabolismo , Linfócitos T CD8-Positivos/patologia , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Complemento C3/genética , Complemento C3/metabolismo , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Complemento C5a/genética , Complemento C5a/metabolismo , Proteínas do Sistema Complemento/genética , Citocinas/metabolismo , Endotélio Vascular/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA