Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 91
Filtrer
1.
Front Immunol ; 12: 690821, 2021.
Article de Anglais | MEDLINE | ID: mdl-34177949

RÉSUMÉ

Complement factor B (FB) mutant variants are associated with excessive complement activation in kidney diseases such as atypical hemolytic uremic syndrome (aHUS), C3 glomerulopathy and membranoproliferative glomerulonephritis (MPGN). Patients with aHUS are currently treated with eculizumab while there is no specific treatment for other complement-mediated renal diseases. In this study the phenotype of three FB missense variants, detected in patients with aHUS (D371G and E601K) and MPGN (I242L), was investigated. Patient sera with the D371G and I242L mutations induced hemolysis of sheep erythrocytes. Mutagenesis was performed to study the effect of factor D (FD) inhibition on C3 convertase-induced FB cleavage, complement-mediated hemolysis, and the release of soluble C5b-9 from glomerular endothelial cells. The FD inhibitor danicopan abrogated C3 convertase-associated FB cleavage to the Bb fragment in patient serum, and of the FB constructs, D371G, E601K, I242L, the gain-of-function mutation D279G, and the wild-type construct, in FB-depleted serum. Furthermore, the FD-inhibitor blocked hemolysis induced by the D371G and D279G gain-of-function mutants. In FB-depleted serum the D371G and D279G mutants induced release of C5b-9 from glomerular endothelial cells that was reduced by the FD-inhibitor. These results suggest that FD inhibition can effectively block complement overactivation induced by FB gain-of-function mutations.


Sujet(s)
Syndrome hémolytique et urémique atypique/immunologie , Activation du complément , Facteur B du complément/génétique , Facteur D du complément/antagonistes et inhibiteurs , Glomérulonéphrite membranoproliférative/immunologie , Animaux , Syndrome hémolytique et urémique atypique/génétique , Enfant , Complement C3-C5 Convertases/immunologie , Complément C3b/immunologie , Facteur B du complément/immunologie , Facteur D du complément/immunologie , Cellules endothéliales/immunologie , Érythrocytes , Femelle , Glomérulonéphrite membranoproliférative/génétique , Hémolyse , Humains , Nourrisson , Glomérule rénal/cytologie , Mâle , Adulte d'âge moyen , Mutation , Phénotype , Lapins , Ovis
2.
Methods Mol Biol ; 2227: 141-145, 2021.
Article de Anglais | MEDLINE | ID: mdl-33847939

RÉSUMÉ

Antibodies to autoantigens are implicated in a large number of diseases. Such autoantibodies may cause pathological activation of complement, an ancient humoral recognition and effector system of innate immunity; in addition, complement components or regulators may be target of autoantibodies and cause abnormal complement activation or function. Autoantibodies to complement proteins are in particular involved in kidney diseases. Those binding to complement convertase enzymes can cause enhanced stability of convertases and their increased resistance to regulation, thus promoting complement turnover. Here, we describe an ELISA method to detect factor B autoantibodies that bind to and stabilize the alternative complement pathway C3 convertase enzyme, C3bBb.


Sujet(s)
Autoanticorps/analyse , Facteur B du complément/immunologie , Autoanticorps/sang , Alternative pathway complement C3 convertase/immunologie , Facteur néphritique C3/immunologie , Complement C3-C5 Convertases/immunologie , Test ELISA/méthodes , Glomérulonéphrite/sang , Glomérulonéphrite/diagnostic , Glomérulonéphrite/immunologie , Humains
3.
J Biol Chem ; 295(26): 8746-8758, 2020 06 26.
Article de Anglais | MEDLINE | ID: mdl-32376685

RÉSUMÉ

The complement system is a tightly controlled proteolytic cascade in the innate immune system, which tags intruding pathogens and dying host cells for clearance. An essential protein in this process is complement component C3. Uncontrolled complement activation has been implicated in several human diseases and disorders and has spurred the development of therapeutic approaches that modulate the complement system. Here, using purified proteins and several biochemical assays and surface plasmon resonance, we report that our nanobody, hC3Nb2, inhibits C3 deposition by all complement pathways. We observe that the hC3Nb2 nanobody binds human native C3 and its degradation products with low nanomolar affinity and does not interfere with the endogenous regulation of C3b deposition mediated by Factors H and I. Using negative stain EM analysis and functional assays, we demonstrate that hC3Nb2 inhibits the substrate-convertase interaction by binding to the MG3 and MG4 domains of C3 and C3b. Furthermore, we notice that hC3Nb2 is cross-reactive and inhibits the lectin and alternative pathway in murine serum. We conclude that hC3Nb2 is a potent, general, and versatile inhibitor of the human and murine complement cascades. Its cross-reactivity suggests that this nanobody may be valuable for analysis of complement activation within animal models of both acute and chronic diseases.


Sujet(s)
Activation du complément/effets des médicaments et des substances chimiques , Complément C3/antagonistes et inhibiteurs , Anticorps à domaine unique/pharmacologie , Animaux , Complément C3/immunologie , Complement C3-C5 Convertases/antagonistes et inhibiteurs , Complement C3-C5 Convertases/immunologie , Hémolyse/effets des médicaments et des substances chimiques , Humains , Souris , Modèles moléculaires , Ovis
4.
Front Immunol ; 10: 886, 2019.
Article de Anglais | MEDLINE | ID: mdl-31068950

RÉSUMÉ

Nephritic factors comprise a heterogeneous group of autoantibodies against neoepitopes generated in the C3 and C5 convertases of the complement system, causing its dysregulation. Classification of these autoantibodies can be clustered according to their stabilization of different convertases either from the classical or alternative pathway. The first nephritic factor described with the capacity to stabilize C3 convertase of the alternative pathway was C3 nephritic factor (C3NeF). Another nephritic factor has been characterized by the ability to stabilize C5 convertase of the alternative pathway (C5NeF). In addition, there are autoantibodies against assembled C3/C5 convertase of the classical and lectin pathways (C4NeF). These autoantibodies have been mainly associated with kidney diseases, like C3 glomerulopathy and immune complex-associated-membranoproliferative glomerulonephritis. Other clinical situations where these autoantibodies have been observed include infections and autoimmune disorders such as systemic lupus erythematosus and acquired partial lipodystrophy. C3 hypocomplementemia is a common finding in all patients with nephritic factors. The methods to measure nephritic factors are not standardized, technically complex, and lack of an appropriate quality control. This review will be focused in the description of the mechanism of action of the three known nephritic factors (C3NeF, C4NeF, and C5NeF), and their association with human diseases. Moreover, we present an overview regarding the diagnostic tools for its detection, and the main therapeutic approach for the patients with nephritic factors.


Sujet(s)
Autoanticorps/immunologie , Protéines du système du complément/immunologie , Prédisposition aux maladies , Épitopes/immunologie , Animaux , Activation du complément/immunologie , Facteur néphritique C3/immunologie , Complement C3-C5 Convertases/immunologie , Complément C3a/immunologie , Complément C3a/métabolisme , Complément C5a/immunologie , Complément C5a/métabolisme , Prise en charge de la maladie , Glomérulonéphrite/diagnostic , Glomérulonéphrite/étiologie , Glomérulonéphrite/métabolisme , Glomérulonéphrite/thérapie , Humains , Techniques de diagnostic moléculaire
5.
Cancer Immunol Immunother ; 68(4): 587-598, 2019 Apr.
Article de Anglais | MEDLINE | ID: mdl-30725204

RÉSUMÉ

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.


Sujet(s)
Antigènes CD20/métabolisme , Antinéoplasiques immunologiques/pharmacologie , Complement C3-C5 Convertases/génétique , Complement C3-C5 Convertases/immunologie , Mutation , Rituximab/pharmacologie , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux humanisés , Marqueurs biologiques , Lignée cellulaire tumorale , Voie alterne d'activation du complément/immunologie , Protéines du système du complément/génétique , Protéines du système du complément/immunologie , Cytotoxicité immunologique , Mutation gain de fonction , Humains , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Leucémie chronique lymphocytaire à cellules B/génétique , Leucémie chronique lymphocytaire à cellules B/métabolisme , Leucémie chronique lymphocytaire à cellules B/anatomopathologie
6.
Methods Mol Biol ; 1911: 337-347, 2019.
Article de Anglais | MEDLINE | ID: mdl-30593637

RÉSUMÉ

A prominent role for complement has been identified in the linkage of innate and adaptive immunity. The liver is the main source of complement and hepatocytes are the primary sites for synthesis of complement components in vivo. We have discovered that hepatitis C virus (HCV) impairs C4 and C3 synthesis. Liver damage may diminish capacity of complement synthesis in patients. However, we observed that the changes in measured complement components in chronically HCV infected patients do not correlate with liver fibrosis or rheumatoid factor present in the blood, serum albumin, or alkaline phosphatase levels. Complement component C3 is of critical importance in B cell activation and T cell-dependent antibody responses. C3 activity is required for optimal expansion of CD8+T cells during a systemic viral infection. Deficiencies in complement may predispose patients to infections via ineffective opsonization, and defects in lytic activity via membrane attack complex. Interestingly, C9 is significantly reduced at the mRNA level in chronically HCV infected liver biopsy specimens, while many hepatocyte derived complement components (C6, C8, Factor B, MASP1, and MBL) and unrelated genes remain mostly unaffected. This implies an HCV specific effect, not a global effect from liver disease.


Sujet(s)
Protéines du système du complément/analyse , Hepacivirus/immunologie , Hépatite C chronique/immunologie , Échappement immunitaire , Dosage immunologique/méthodes , Lignée cellulaire tumorale , Complement C3-C5 Convertases/analyse , Complement C3-C5 Convertases/immunologie , Complement C3-C5 Convertases/métabolisme , Voie alterne d'activation du complément/immunologie , Voie classique d'activation du complément/immunologie , Protéines du système du complément/génétique , Protéines du système du complément/immunologie , Protéines du système du complément/métabolisme , Hépatite C chronique/sang , Humains , Foie/immunologie , Foie/virologie , Régions promotrices (génétique)
7.
Front Immunol ; 9: 1191, 2018.
Article de Anglais | MEDLINE | ID: mdl-29892304

RÉSUMÉ

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.


Sujet(s)
Voie alterne d'activation du complément/immunologie , Voie des lectines/immunologie , Glomérulonéphrite lupique/immunologie , Mannose-Binding Protein-Associated Serine Proteases/immunologie , Animaux , Complement C3-C5 Convertases/génétique , Complement C3-C5 Convertases/immunologie , Voie alterne d'activation du complément/génétique , Voie des lectines/génétique , Glomérulonéphrite lupique/génétique , Glomérulonéphrite lupique/anatomopathologie , Mannose-Binding Protein-Associated Serine Proteases/génétique , Souris , Souris de lignée MRL lpr , Souris knockout
8.
J Immunol ; 200(12): 4125-4133, 2018 06 15.
Article de Anglais | MEDLINE | ID: mdl-29752310

RÉSUMÉ

Complement activation leads to membrane attack complex formation, which can lyse not only pathogens but also host cells. Histones can be released from the lysed or damaged cells and serve as a major type of damage-associated molecular pattern, but their effects on the complement system are not clear. In this study, we pulled down two major proteins from human serum using histone-conjugated beads: one was C-reactive protein and the other was C4, as identified by mass spectrometry. In surface plasmon resonance analysis, histone H3 and H4 showed stronger binding to C4 than other histones, with KD around 1 nM. The interaction did not affect C4 cleavage to C4a and C4b. Because histones bind to C4b, a component of C3 and C5 convertases, their activities were significantly inhibited in the presence of histones. Although it is not clear whether the inhibition was achieved through blocking C3 and C5 convertase assembly or just through reducing their activity, the outcome was that both classical and mannose-binding lectin pathways were dramatically inhibited. Using a high concentration of C4 protein, histone-suppressed complement activity could not be fully restored, indicating C4 is not the only target of histones in those pathways. In contrast, the alternative pathway was almost spared, but the overall complement activity activated by zymosan was inhibited by histones. Therefore, we believe that histones inhibiting complement activation is a natural feedback mechanism to prevent the excessive injury of host cells.


Sujet(s)
Activation du complément/immunologie , Complément C4/immunologie , Histone/immunologie , Protéine C-réactive/immunologie , Complement C3-C5 Convertases/immunologie , Humains , Lectine liant le mannose/immunologie , Plasma sanguin/immunologie , Liaison aux protéines/immunologie
10.
Semin Immunopathol ; 40(1): 15-35, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-29167939

RÉSUMÉ

This review is not intended to cover in detail all aspects of the discovery and evolution of our understanding of the "alternative pathway" of complement activation, there are many excellent reviews that do this (see Fearon (CRC Crit Rev Immunol 1:1-32, 1979), Pangburn and Müller-Eberhard (Springer Semin Immunopathol 7:163-192, 1984)), but instead to give sufficient background for current concepts to be put in context. The prevailing textbook view, of components having a primary role as an alternative "pathway" for C3 activation, is challenged, with an argument developed for the primary role of the system being that of providing a surface-dependent amplification loop for both C3 and C5 activation. Whatever the mechanism by which the initial C3b molecule is generated, deposition onto a surface has the potential to target that surface for elimination. Elimination or escape from initial targeting is determined by a sophisticated and highly regulated amplification loop for C3 activation. This viewpoint of the system is then briefly developed to provide a context for therapeutic treatment of disease caused, at least in part, by dysregulated amplification of C3 activation, and to highlight some of the challenges that such therapies will face and need to address.


Sujet(s)
Voie alterne d'activation du complément , Properdine/métabolisme , Transduction du signal , Animaux , Protéines de transport/métabolisme , Membrane cellulaire/immunologie , Membrane cellulaire/métabolisme , Activation du complément/immunologie , Facteur néphritique C3/immunologie , Facteur néphritique C3/métabolisme , Complement C3-C5 Convertases/composition chimique , Complement C3-C5 Convertases/immunologie , Complement C3-C5 Convertases/métabolisme , Protéines inhibitrices de la fraction C3b du complément/immunologie , Protéines inhibitrices de la fraction C3b du complément/métabolisme , Venins des élapidés/immunologie , Venins des élapidés/métabolisme , Interactions hôte-pathogène/immunologie , Humains , Properdine/immunologie , Liaison aux protéines
11.
Semin Immunopathol ; 40(1): 3-14, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-28808775

RÉSUMÉ

During the last decade, the complement field has experienced outstanding advancements in the mechanistic understanding of how complement activators are recognized, what C3 activation means, how protein complexes like the C3 convertases and the membrane attack complex are assembled, and how positive and negative complement regulators perform their function. All of this has been made possible mostly because of the contributions of structural biology to the study of the complement components. The wealth of novel structural data has frequently provided support to previously held knowledge, but often has added alternative and unexpected insights into complement function. Here, we will review some of these findings focusing in the alternative and terminal complement pathways.


Sujet(s)
Protéines du système du complément/composition chimique , Protéines du système du complément/physiologie , Animaux , Protéines de transport , Activation du complément/immunologie , Complement C3-C5 Convertases/composition chimique , Complement C3-C5 Convertases/immunologie , Complement C3-C5 Convertases/métabolisme , Interactions hôte-pathogène/immunologie , Humains , Liaison aux protéines , Conformation des protéines , Transduction du signal , Relation structure-activité , Spécificité du substrat
12.
Kidney Int ; 92(5): 1232-1241, 2017 11.
Article de Anglais | MEDLINE | ID: mdl-28712854

RÉSUMÉ

C3 Glomerulopathies, which include Dense Deposit Disease and C3 Glomerulonephritis, are associated with genetic and acquired dysregulation of the C3 convertase alternative pathway of complement. The potential role of the activation of the C5 convertase has not been studied extensively. Here we analyzed IgG samples from patients with C3 Glomerulopathies to identify circulating autoantibodies that stabilize the C3 alternative pathway (C3 Nephritic Factors) as well as C5 convertases (C5 Nephritic Factors), thus preventing decay of these enzyme complexes. Rare variants in alternative pathway genes were found in 28 of 120 tested patients. C3 and C5 Nephritic Factors were found in 76 of 101 (75%) and 29 of 59 (49%) of the patients, respectively. Therefore, we compared the results of the assays for the C3 and C5 nephritic factors functional activity: 29% were positive for C3 Nephritic Factors alone, 39% were positive for both C3 and C5 Nephritic Factors, and 10% were positive for C5 Nephritic Factors alone. We found that the addition of properdin-enhanced stabilization of C3 convertase in the presence of IgG doubly positive for both Nephritic Factors, while it did not modify the stabilization mediated by IgG solely positive for C3 Nephritic Factors. Both C3 and C5 Nephritic Factors correlated with C3 consumption, while only C5 Nephritic Factors correlated with sC5b9 levels. C5 Nephritic Factors-positive patients were more likely to have C3 Glomerulonephritis than Dense Deposit Disease. Thus, dysregulation of the C5 convertase contributes to C3 Glomerulopathies inter-disease differences and may have direct therapeutic implications.


Sujet(s)
Alternative pathway complement C3 convertase/immunologie , Facteur néphritique C3/immunologie , Complement C3-C5 Convertases/immunologie , Voie alterne d'activation du complément/immunologie , Glomérulonéphrite membranoproliférative/immunologie , Adolescent , Adulte , Enfant , Facteur néphritique C3/analyse , Facteur néphritique C3/génétique , Complement C3-C5 Convertases/métabolisme , Complexe d'attaque membranaire du complément/analyse , Femelle , Études de suivi , Glomérulonéphrite membranoproliférative/sang , Glomérulonéphrite membranoproliférative/génétique , Glomérulonéphrite membranoproliférative/mortalité , Humains , Estimation de Kaplan-Meier , Mâle , Adulte d'âge moyen , Phénotype , Properdine/métabolisme , Tests sérologiques , Jeune adulte
13.
Clin Immunol ; 180: 25-32, 2017 07.
Article de Anglais | MEDLINE | ID: mdl-28366510

RÉSUMÉ

The terminal complement split product C5a has been described as an important mediator in inflammatory diseases. C5a is generated upon cleavage of C5 and earlier research suggests that, besides the known C5 convertases formed upon activation of the complement pathways, various enzymes could activate C5 directly. We demonstrate that eculizumab effectively blocks C5 activation when mediated by C5-convertase formation, but fails to block C5a generation resulting from direct enzymatic cleavage by trypsin and thrombin. C5a generated by these enzymes is shown to be fully biologically functional and can be blocked by IFX-1, a specific monoclonal anti-human C5a antibody. We further report clinical cases of atypical hemolytic uremic syndrome (aHUS) and C3 Glomerulonephritis (C3GN) patients under treatment with eculizumab presenting substantially elevated C5a levels. Thus, blocking the C5 convertase mediated activation of C5 may not be efficient to control C5a-mediated effects in human disease and that a targeted approach is warranted.


Sujet(s)
Anticorps monoclonaux humanisés/pharmacologie , Syndrome hémolytique et urémique atypique/immunologie , Complément C5a/immunologie , Glomérulonéphrite/immunologie , Anticorps monoclonaux humanisés/usage thérapeutique , Syndrome hémolytique et urémique atypique/traitement médicamenteux , Complement C3-C5 Convertases/immunologie , Glomérulonéphrite/traitement médicamenteux , Humains , Thrombine/immunologie , Trypsine/immunologie , Zymosan/pharmacologie
14.
Klin Lab Diagn ; 62(3): 177-81, 2017 Mar.
Article de Russe | MEDLINE | ID: mdl-30620534

RÉSUMÉ

The technique of detection of stabilization of C3-convertase classical way of activation of system of complement inhuman blood serum. The technique comprises two stages and is based on applying a reaction of lysis of erythrocytes of sheep sensitized by antibodies using 0.8% human blood serum. Preliminary an incubation of two samples (experimental and control) is applied during 10 min. and then reaction of activation of complement is stopped by adding a buffer containing 10 mM of EDTA. In control sample degree of lysis of erythrocytes is established and experimental sample is additionally incubated during 30 min at 37oC and then degree of lysis is determined. The activity of C3-convertase is calculated as a difference between degree of lysis and in experimental and control samples. The difference more than 10% is considered as a pathological state conditioned by stabilization of C3-convertase of classical way of activation of system of complement. The studies were carried out concerning stabilization of C3-convertase of classical way of activation of compliment in 31 patients with abdominal obesity. It is demonstrated that in 87% of patients with abdominal obesity stabilization of C3-convertase was established.


Sujet(s)
Activation du complément/immunologie , Complement C3-C5 Convertases/immunologie , Érythrocytes/immunologie , Obésité abdominale/immunologie , Adulte , Animaux , Femelle , Humains , Mâle , Adulte d'âge moyen , Obésité abdominale/enzymologie , Sérum/enzymologie , Sérum/immunologie , Ovis/immunologie
15.
J Biol Chem ; 291(15): 8214-30, 2016 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-26903516

RÉSUMÉ

The activated fragment of C3 (C3b) and factor B form the C3 proconvertase (C3bB), which is cleaved by factor D to C3 convertase (C3bBb). Older studies (Conrad, D. H., Carlo, J. R., and Ruddy, S. (1978)J. Exp. Med.147, 1792-1805; Pangburn, M. K., and Müller-Eberhard, H. J. (1978)Proc. Natl. Acad. Sci. U.S.A.75, 2416-2420; Kazatchkine, M. D., Fearon, D. T., and Austen, K. F. (1979)J. Immunol.122, 75-81) indicated that the complement alternative pathway regulator factor H (FH) competes with factor B for C3b binding; however, the capability of FH to prevent C3bB assembly has not been formally investigated. Moreover, in the few published studies FH did not favor C3bB dissociation. Whether FH may affect C3bBb formation from C3bB is unknown. We set up user-friendly assays based on combined microplate/Western blotting techniques that specifically detect either C3bB or C3bBb, with the aim of investigating the effect of FH on C3bB assembly and decay and C3bBb formation and decay. We document that FH does not affect C3bB assembly, indicating that FH does not efficiently compete with factor B for C3b binding. We also found that FH does not dissociate C3bB. FH showed a strong C3bBb decay-accelerating activity, as reported previously, and also exerted an apparent inhibitory effect on C3bBb formation. The latter effect was not fully attributable to a rapid FH-mediated dissociation of C3bBb complexes, because blocking decay with properdin and C3 nephritic factor did not restore C3bBb formation. FH almost completely prevented release of the smaller cleavage subunit of FB (Ba), without modifying the amount of C3bB complexes, suggesting that FH inhibits the conversion of C3bB to C3bBb. Thus, the inhibitory effect of FH on C3bBb formation is likely the sum of inhibition of C3bB conversion to C3bBb and of C3bBb decay acceleration. Further studies are required to confirm these findings in physiological cell-based settings.


Sujet(s)
Alternative pathway complement C3 convertase/immunologie , Complement C3-C5 Convertases/immunologie , Facteur H du complément/immunologie , Complément C3/immunologie , Alternative pathway complement C3 convertase/analyse , Complement C3-C5 Convertases/analyse , Complément C3b/immunologie , Facteur B du complément/immunologie , Facteur H du complément/analyse , Test ELISA , Humains , Manganèse/analyse , Manganèse/immunologie , Properdine/immunologie
16.
Clin Exp Immunol ; 184(1): 118-25, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-26660535

RÉSUMÉ

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.


Sujet(s)
Complement C3-C5 Convertases/immunologie , Voie alterne d'activation du complément , Glomérulonéphrite/immunologie , Properdine/immunologie , Protéinurie/immunologie , Adolescent , Adulte , Marqueurs biologiques/sang , Enfant , Complément C3/génétique , Complément C3/immunologie , Facteur néphritique C3/génétique , Facteur néphritique C3/immunologie , Complement C3-C5 Convertases/génétique , Complément C5/génétique , Complément C5/immunologie , Facteur B du complément/génétique , Facteur B du complément/immunologie , Inhibiteurs du complément/sang , Complexe d'attaque membranaire du complément/génétique , Complexe d'attaque membranaire du complément/immunologie , Femelle , Régulation de l'expression des gènes , Glomérulonéphrite/sang , Glomérulonéphrite/génétique , Glomérulonéphrite/anatomopathologie , Humains , Mâle , Adulte d'âge moyen , Properdine/génétique , Protéinurie/sang , Protéinurie/génétique , Protéinurie/anatomopathologie , Études rétrospectives , Indice de gravité de la maladie , Transduction du signal
17.
Nat Commun ; 6: 8483, 2015 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-26404464

RÉSUMÉ

The complement system is important for the host defence against infection as well as for the development of inflammatory diseases. Here we show that C1q/TNF-related protein 6 (CTRP6; gene symbol C1qtnf6) expression is elevated in mouse rheumatoid arthritis (RA) models. C1qtnf6(-/-) mice are highly susceptible to induced arthritis due to enhanced complement activation, whereas C1qtnf6-transgenic mice are refractory. The Arthus reaction and the development of experimental autoimmune encephalomyelitis are also enhanced in C1qtnf6(-/-) mice and C1qtnf6(-/-) embryos are semi-lethal. We find that CTRP6 specifically suppresses the alternative pathway of the complement system by competing with factor B for C3(H2O) binding. Furthermore, treatment of arthritis-induced mice with intra-articular injection of recombinant human CTRP6 cures the arthritis. CTRP6 is expressed in human synoviocytes, and CTRP6 levels are increased in RA patients. These results indicate that CTRP6 is an endogenous complement regulator and could be used for the treatment of complement-mediated diseases.


Sujet(s)
Adipokines/immunologie , Arthrite expérimentale/immunologie , Polyarthrite rhumatoïde/immunologie , Voie alterne d'activation du complément/immunologie , Adipokines/génétique , Adulte , Animaux , Arthrite expérimentale/génétique , Arthrite expérimentale/anatomopathologie , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/anatomopathologie , Phénomène d'Arthus/génétique , Phénomène d'Arthus/immunologie , Phénomène d'Arthus/métabolisme , Technique de Western , Collagène/immunologie , Collagène/métabolisme , Complement C3-C5 Convertases/immunologie , Complément C3a/immunologie , Complément C5a/immunologie , Voie alterne d'activation du complément/génétique , Modèles animaux de maladie humaine , Encéphalomyélite auto-immune expérimentale/génétique , Encéphalomyélite auto-immune expérimentale/immunologie , Encéphalomyélite auto-immune expérimentale/métabolisme , Femelle , Cytométrie en flux , Humains , Immunoprécipitation , Macrophages/immunologie , Mâle , Souris , Souris knockout , Souris transgéniques , Adulte d'âge moyen , Réaction de polymérisation en chaine en temps réel , RT-PCR , Membrane synoviale/cytologie , Membrane synoviale/métabolisme
18.
PLoS Negl Trop Dis ; 9(2): e0003513, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25679788

RÉSUMÉ

Hematophagous vectors strictly require ingesting blood from their hosts to complete their life cycles. Exposure of the alimentary canal of these vectors to the host immune effectors necessitates efficient counteractive measures by hematophagous vectors. The Anopheles mosquito transmitting the malaria parasite is an example of hematophagous vectors that within seconds can ingest human blood double its weight. The innate immune defense mechanisms, like the complement system, in the human blood should thereby immediately react against foreign cells in the mosquito midgut. A prerequisite for complement activation is that the target cells lack complement regulators on their surfaces. In this work, we analyzed whether human complement is active in the mosquito midgut, and how the mosquito midgut cells protect themselves against complement attack. We found that complement remained active for a considerable time and was able to kill microbes within the mosquito midgut. However, the Anopheles mosquito midgut cells were not injured. These cells were found to protect themselves by capturing factor H, the main soluble inhibitor of the alternative complement pathway. Factor H inhibited complement on the midgut cells by promoting inactivation of C3b to iC3b and preventing the activity of the alternative pathway amplification C3 convertase enzyme. An interference of the FH regulatory activity by monoclonal antibodies, carried to the midgut via blood, resulted in increased mosquito mortality and reduced fecundity. By using a ligand blotting assay, a putative mosquito midgut FH receptor could be detected. Thereby, we have identified a novel mechanism whereby mosquitoes can tolerate human blood.


Sujet(s)
Anopheles/immunologie , Activation du complément/immunologie , Facteur H du complément/immunologie , Échappement immunitaire/immunologie , Muqueuse intestinale/immunologie , Animaux , Anticorps monoclonaux/immunologie , Complement C3-C5 Convertases/biosynthèse , Complement C3-C5 Convertases/immunologie , Complément C3b/immunologie , Système digestif/immunologie , Humains , Vecteurs insectes/immunologie , Muqueuse intestinale/parasitologie , Étapes du cycle de vie , Récepteurs au complément/immunologie
19.
Clin Exp Immunol ; 178(1): 142-53, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-24853370

RÉSUMÉ

Complement convertases are enzymatic complexes that play a central role in sustaining and amplification of the complement cascade. Impairment of complement function leads directly or indirectly to pathological conditions, including higher infection rate, kidney diseases, autoimmune- or neurodegenerative diseases and ischaemia-reperfusion injury. An assay for direct measurement of activity of the convertases in patient sera is not available. Existing assays testing convertase function are based on purified complement components and, thus, convertase formation occurs under non-physiological conditions. We designed a new assay, in which C5 blocking compounds enabled separation of the complement cascade into two phases: the first ending at the stage of C5 convertases and the second ending with membrane attack complex formation. The use of rabbit erythrocytes or antibody-sensitized sheep erythrocytes as the platforms for convertase formation enabled easy readout based on measurement of haemolysis. Thus, properties of patient sera could be studied directly regarding convertase activity and membrane attack complex formation. Another advantage of this assay was the possibility to screen for host factors such as C3 nephritic factor and other anti-complement autoantibodies, or gain-of-function mutations, which prolong the half-life of complement convertases. Herein, we present proof of concept, detailed description and validation of this novel assay.


Sujet(s)
Complement C3-C5 Convertases/analyse , Érythrocytes/enzymologie , Dosage immunologique/méthodes , Animaux , Autoanticorps/immunologie , Facteur néphritique C3/immunologie , Complement C3-C5 Convertases/immunologie , Voie alterne d'activation du complément/immunologie , Protéines du système du complément/immunologie , Érythrocytes/immunologie , Cochons d'Inde , Période , Humains , Lapins , Ovis
20.
J Immunol ; 192(10): 4844-51, 2014 May 15.
Article de Anglais | MEDLINE | ID: mdl-24729617

RÉSUMÉ

Dysregulation of the complement alternative pathway can cause disease in various organs that may be life-threatening. Severe alternative pathway dysregulation can be triggered by autoantibodies to the C3 convertase, termed nephritic factors, which cause pathological stabilization of the convertase enzyme and confer resistance to innate control mechanisms; unregulated complement consumption followed by deposition of C3 fragments in tissues ensues. The mAb, 3E7, and its humanized derivative, H17, have been shown previously to specifically bind activated C3 and prevent binding of both the activating protein, factor B, and the inhibitor, factor H, which are opposite effects that complicate its potential for therapy. Using ligand binding assays, functional assays, and electron microscopy, we show that these Abs bind C3b via a site that overlaps the binding site on C3 for the Ba domain within factor B, thereby blocking an interaction essential for convertase formation. Both Abs also bind the preformed convertase, C3bBb, and provide powerful inhibition of complement activation by preventing cleavage of C3. Critically, the Abs also bound and inhibited C3 cleavage by the nephritic factor-stabilized convertase. We suggest that by preventing enzyme formation and/or cleavage of C3 to its active downstream fragments, H17 may be an effective therapy for conditions caused by severe dysregulation of the C3 convertase and, in particular, those that involve nephritic factors, such as dense deposit disease.


Sujet(s)
Anticorps monoclonaux humanisés/immunologie , Alternative pathway complement C3 convertase/immunologie , Complement C3-C5 Convertases/immunologie , Facteur B du complément/immunologie , Voie alterne d'activation du complément/immunologie , Maladies du rein/immunologie , Anticorps monoclonaux humanisés/usage thérapeutique , Alternative pathway complement C3 convertase/antagonistes et inhibiteurs , Voie alterne d'activation du complément/effets des médicaments et des substances chimiques , Humains , Maladies du rein/traitement médicamenteux , Maladies du rein/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE