Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell Proteomics ; 22(7): 100580, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37211046

RESUMEN

Current proteomic technologies focus on the quantification of protein levels, while little effort is dedicated to the development of system approaches to simultaneously monitor proteome variability and abundance. Protein variants may display different immunogenic epitopes detectable by monoclonal antibodies. Epitope variability results from alternative splicing, posttranslational modifications, processing, degradation, and complex formation and possesses dynamically changing availability of interacting surface structures that frequently serve as reachable epitopes and often carry different functions. Thus, it is highly likely that the presence of some of the accessible epitopes correlates with function under physiological and pathological conditions. To enable the exploration of the impact of protein variation on the immunogenic epitome first, here, we present a robust and analytically validated PEP technology for characterizing immunogenic epitopes of the plasma. To this end, we prepared mAb libraries directed against the normalized human plasma proteome as a complex natural immunogen. Antibody producing hybridomas were selected and cloned. Monoclonal antibodies react with single epitopes, thus profiling with the libraries is expected to profile many epitopes which we define by the mimotopes, as we present here. Screening blood plasma samples from control subjects (n = 558) and cancer patients (n = 598) for merely 69 native epitopes displayed by 20 abundant plasma proteins resulted in distinct cancer-specific epitope panels that showed high accuracy (AUC 0.826-0.966) and specificity for lung, breast, and colon cancer. Deeper profiling (≈290 epitopes of approximately 100 proteins) showed unexpected granularity of the epitope-level expression data and detected neutral and lung cancer-associated epitopes of individual proteins. Biomarker epitope panels selected from a pool of 21 epitopes of 12 proteins were validated in independent clinical cohorts. The results demonstrate the value of PEP as a rich and thus far unexplored source of protein biomarkers with diagnostic potential.


Asunto(s)
Biomarcadores de Tumor , Neoplasias , Humanos , Proteoma , Proteómica/métodos , Epítopos , Anticuerpos Monoclonales/química
2.
Int J Mol Sci ; 25(13)2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-39000451

RESUMEN

The nucleocapsid (N) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a viral structural protein that is abundant in the circulation of infected individuals. Previous published studies reported controversial data about the role of the N protein in the activation of the complement system. It was suggested that the N protein directly interacts with mannose-binding lectin-associated serine protease-2 (MASP-2) and stimulates lectin pathway overactivation/activity. In order to check these data and to reveal the mechanism of activation, we examined the effect of the N protein on lectin pathway activation. We found that the N protein does not bind to MASP-2 and MASP-1 and it does not stimulate lectin pathway activity in normal human serum. Furthermore, the N protein does not facilitate the activation of zymogen MASP-2, which is MASP-1 dependent. Moreover, the N protein does not boost the enzymatic activity of MASP-2 either on synthetic or on protein substrates. In some of our experiments, we observed that MASP-2 digests the N protein. However, it is questionable, whether this activity is biologically relevant. Although surface-bound N protein did not activate the lectin pathway, it did trigger the alternative pathway in 10% human serum. Additionally, we detected some classical pathway activation by the N protein. Nevertheless, we demonstrated that this activation was induced by the bound nucleic acid, rather than by the N protein itself.


Asunto(s)
Lectina de Unión a Manosa de la Vía del Complemento , Proteínas de la Nucleocápside de Coronavirus , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa , SARS-CoV-2 , Humanos , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , SARS-CoV-2/metabolismo , Proteínas de la Nucleocápside de Coronavirus/metabolismo , COVID-19/virología , COVID-19/metabolismo , COVID-19/inmunología , Fosfoproteínas/metabolismo , Unión Proteica , Activación de Complemento
3.
BMC Ophthalmol ; 23(1): 14, 2023 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-36627583

RESUMEN

PURPOSE: To detect immunoglobulins in aqueous humour of AMD patients after repeated administration of intravitreal aflibercept. PATIENTS AND METHODS: Twenty-one patients (age: 77.85 ± 9.21 years) previously treated with intravitreal aflibercept due to wet type age-related macular degeneration (AMD group) and 18 age-matched control subjects (age: 69.75 ± 12.67 years) were included in this study. Patients in the AMD group received a mean of 5 intravitreal injections (min: 1 max: 17) prior to the cataract surgery. Samples of aqueous humour (50 µl) were obtained by anterior chamber paracentesis as the first step of routine cataract surgery. The IgG content of the samples was analysed by an in-house developed ELISA system. RESULTS: A significant increase in nonspecific IgG levels in the AMD group was detected compared to the control group (13.37 ± 6.65 vs. 9.44 ± 6.55 µg/ml; p = 0.03). In 11 patients, intraocular anti-aflibercept immunoglobulins could be detected (0.05 ± 0.01 µg/ml) which was significantly higher than the limit of detection for anti-aflibercept (0.04 µg/ml; p = 0.001). No correlation was found between the number of injections or the type of CNV and the aqueous level of anti-aflibercept (r = 0.02; p = 0.95). CONCLUSION: According to our results, penetration of non-specific systemic antibodies through the impaired blood-retinal barrier is higher in patients with neovascular AMD than in subjects with an intact structural barrier. Evaluation of neutralizing antibodies to anti-VEGF agents in the aqueous humour can lead us to understanding tachyphylaxis and changes in intraocular immune mechanisms due to AMD.


Asunto(s)
Catarata , Degeneración Macular Húmeda , Humanos , Anciano , Anciano de 80 o más Años , Persona de Mediana Edad , Inhibidores de la Angiogénesis/uso terapéutico , Ranibizumab/uso terapéutico , Degeneración Macular Húmeda/tratamiento farmacológico , Degeneración Macular Húmeda/diagnóstico , Factor A de Crecimiento Endotelial Vascular , Anticuerpos Neutralizantes/uso terapéutico , Agudeza Visual , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Inyecciones Intravítreas , Catarata/tratamiento farmacológico , Inmunoglobulina G , Proteínas Recombinantes de Fusión/uso terapéutico
4.
Semin Immunol ; 45: 101341, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31757608

RESUMEN

The complement system, while being an essential and very efficient effector component of innate immunity, may cause damage to the host and result in various inflammatory, autoimmune and infectious diseases or cancer, when it is improperly activated or regulated. Factor H is a serum glycoprotein and the main regulator of the activity of the alternative complement pathway. Factor H, together with its splice variant factor H-like protein 1 (FHL-1), inhibits complement activation at the level of the central complement component C3 and beyond. In humans, there are also five factor H-related (FHR) proteins, whose function is poorly characterized. While data indicate complement inhibiting activity for some of the FHRs, there is increasing evidence that FHRs have an opposite role compared with factor H and FHL-1, namely, they enhance complement activation directly and also by competing with the regulators FH and FHL-1. This review summarizes the current stand and recent data on the roles of factor H family proteins in health and disease, with focus on the function of FHR proteins.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Proteínas Portadoras/metabolismo , Activación de Complemento/inmunología , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Animales , Proteínas Sanguíneas/química , Proteínas Sanguíneas/genética , Proteínas Portadoras/química , Proteínas Portadoras/genética , Factor H de Complemento/química , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Susceptibilidad a Enfermedades , Humanos , Inmunomodulación , Ligandos , Familia de Multigenes , Unión Proteica , Relación Estructura-Actividad
5.
Semin Cell Dev Biol ; 85: 122-131, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29305301

RESUMEN

Complement factor H is a major regulator of the alternative pathway of the complement system. The factor H-related proteins are less characterized, but recent data indicate that they rather promote complement activation. These proteins have some common ligands with factor H and have both overlapping and distinct functions depending on domain composition and the degree of conservation of amino acid sequence. Factor H and some of the factor H-related proteins also appear in a non-canonical function that is beyond their role in the modulation of complement activation. This review covers our current understanding on this emerging role of factor H family proteins in modulating the activation and function of various cells by binding to receptors or receptor ligands.


Asunto(s)
Linfocitos B/metabolismo , Células Dendríticas/metabolismo , Granulocitos/metabolismo , Neutrófilos/metabolismo , Fagocitos/metabolismo , Activación de Complemento , Factor H de Complemento/metabolismo , Humanos
6.
PLoS Pathog ; 15(12): e1008232, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31860690

RESUMEN

Ecotin is a serine protease inhibitor produced by hundreds of microbial species, including pathogens. Here we show, that ecotin orthologs from Escherichia coli, Yersinia pestis, Pseudomonas aeruginosa and Leishmania major are potent inhibitors of MASP-1 and MASP-2, the two key activator proteases of the complement lectin pathway. Factor D is the key activator protease of another complement activation route, the alternative pathway. We show that ecotin inhibits MASP-3, which is the sole factor D activator in resting human blood. In pathway-specific ELISA tests, we found that all ecotin orthologs are potent lectin pathway inhibitors, and at high concentration, they block the alternative pathway as well. In flow cytometry experiments, we compared the extent of complement-mediated opsonization and lysis of wild-type and ecotin-knockout variants of two E. coli strains carrying different surface lipopolysaccharides. We show, that endogenous ecotin provides significant protections against these microbicidal activities for both bacteria. By using pathway specific complement inhibitors, we detected classical-, lectin- and alternative pathway-driven complement attack from normal serum, with the relative contributions of the activation routes depending on the lipopolysaccharide type. Moreover, in cell proliferation experiments we observed an additional, complement-unrelated antimicrobial activity exerted by heat-inactivated serum. While ecotin-knockout cells are highly vulnerable to these activities, endogenous ecotin of wild-type bacteria provides complete protection against the lectin pathway-related and the complement-unrelated attack, and partial protection against the alternative pathway-related damage. In all, ecotin emerges as a potent, versatile self-defense tool that blocks multiple antimicrobial activities of the serum. These findings suggest that ecotin might be a relevant antimicrobial drug target.


Asunto(s)
Lectina de Unión a Manosa de la Vía del Complemento/fisiología , Proteínas del Sistema Complemento/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Periplasmáticas/metabolismo , Serina Proteasas/sangre , Activación de Complemento/fisiología , Escherichia coli/metabolismo , Humanos , Pseudomonas aeruginosa/metabolismo , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/metabolismo , Yersinia pestis/metabolismo
7.
J Immunol ; 199(1): 292-303, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28533443

RESUMEN

Factor H-related protein (FHR) 1 is one of the five human FHRs that share sequence and structural homology with the alternative pathway complement inhibitor FH. Genetic studies on disease associations and functional analyses indicate that FHR-1 enhances complement activation by competitive inhibition of FH binding to some surfaces and immune proteins. We have recently shown that FHR-1 binds to pentraxin 3. In this study, our aim was to investigate whether FHR-1 binds to another pentraxin, C-reactive protein (CRP), analyze the functional relevance of this interaction, and study the role of FHR-1 in complement activation and regulation. FHR-1 did not bind to native, pentameric CRP, but it bound strongly to monomeric CRP via its C-terminal domains. FHR-1 at high concentration competed with FH for CRP binding, indicating possible complement deregulation also on this ligand. FHR-1 did not inhibit regulation of solid-phase C3 convertase by FH and did not inhibit terminal complement complex formation induced by zymosan. On the contrary, by binding C3b, FHR-1 allowed C3 convertase formation and thereby enhanced complement activation. FHR-1/CRP interactions increased complement activation via the classical and alternative pathways on surfaces such as the extracellular matrix and necrotic cells. Altogether, these results identify CRP as a ligand for FHR-1 and suggest that FHR-1 enhances, rather than inhibits, complement activation, which may explain the protective effect of FHR-1 deficiency in age-related macular degeneration.


Asunto(s)
Proteína C-Reactiva/inmunología , Proteína C-Reactiva/metabolismo , Activación de Complemento , Proteínas Inactivadoras del Complemento C3b/inmunología , Proteínas Inactivadoras del Complemento C3b/metabolismo , Sitios de Unión , Proteína C-Reactiva/química , Proteína C-Reactiva/farmacología , Convertasas de Complemento C3-C5 , Complemento C3b/inmunología , Complemento C3b/farmacología , Proteínas Inactivadoras del Complemento C3b/farmacología , Factor H de Complemento , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/inmunología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Ligandos , Degeneración Macular/inmunología , Unión Proteica , Componente Amiloide P Sérico/inmunología , Componente Amiloide P Sérico/metabolismo
8.
Trends Immunol ; 36(6): 374-84, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25979655

RESUMEN

Complement factor H-related proteins (FHRs) are strongly associated with different diseases involving complement dysregulation, which suggests a major role for these proteins regulating complement activation. Because FHRs are evolutionarily and structurally related to complement inhibitor factor H (FH), the initial assumption was that the FHRs are also negative complement regulators. Whereas weak complement inhibiting activities were originally reported for these molecules, recent developments indicate that FHRs may enhance complement activation, with important implications for the role of these proteins in health and disease. We review these findings here, and propose that FHRs represent a complex set of surface recognition molecules that, by competing with FH, provide improved discrimination of self and non-self surfaces and play a central role in determining appropriate activation of the complement pathway.


Asunto(s)
Apolipoproteínas/inmunología , Proteínas Sanguíneas/inmunología , Activación de Complemento/inmunología , Proteínas Inactivadoras del Complemento C3b/inmunología , Proteínas del Sistema Complemento/inmunología , Apolipoproteínas/genética , Proteínas Sanguíneas/genética , Proteínas Inactivadoras del Complemento C3b/genética , Factor H de Complemento/genética , Factor H de Complemento/inmunología , Proteínas del Sistema Complemento/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Modelos Inmunológicos
9.
J Immunol ; 194(10): 4963-73, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25855355

RESUMEN

The physiological roles of the factor H (FH)-related proteins are controversial and poorly understood. Based on genetic studies, FH-related protein 5 (CFHR5) is implicated in glomerular diseases, such as atypical hemolytic uremic syndrome, dense deposit disease, and CFHR5 nephropathy. CFHR5 was also identified in glomerular immune deposits at the protein level. For CFHR5, weak complement regulatory activity and competition for C3b binding with the plasma complement inhibitor FH have been reported, but its function remains elusive. In this study, we identify pentraxin 3 (PTX3) as a novel ligand of CFHR5. Binding of native CFHR5 to PTX3 was detected in human plasma and the interaction was characterized using recombinant proteins. The binding of PTX3 to CFHR5 is of ∼2-fold higher affinity compared with that of FH. CFHR5 dose-dependently inhibited FH binding to PTX3 and also to the monomeric, denatured form of the short pentraxin C-reactive protein. Binding of PTX3 to CFHR5 resulted in increased C1q binding. Additionally, CFHR5 bound to extracellular matrix in vitro in a dose-dependent manner and competed with FH for binding. Altogether, CFHR5 reduced FH binding and its cofactor activity on pentraxins and the extracellular matrix, while at the same time allowed for enhanced C1q binding. Furthermore, CFHR5 allowed formation of the alternative pathway C3 convertase and supported complement activation. Thus, CFHR5 may locally enhance complement activation via interference with the complement-inhibiting function of FH, by enhancement of C1q binding, and by activating complement, thereby contributing to glomerular disease.


Asunto(s)
Proteína C-Reactiva/metabolismo , Activación de Complemento/fisiología , Proteínas del Sistema Complemento/metabolismo , Matriz Extracelular/metabolismo , Componente Amiloide P Sérico/metabolismo , Humanos , Ligandos , Unión Proteica , Proteínas Recombinantes
10.
J Biol Chem ; 290(15): 9500-10, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25659429

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is characterized by complement attack against host cells due to mutations in complement proteins or autoantibodies against complement factor H (CFH). It is unknown why nearly all patients with autoimmune aHUS lack CFHR1 (CFH-related protein-1). These patients have autoantibodies against CFH domains 19 and 20 (CFH19-20), which are nearly identical to CFHR1 domains 4 and 5 (CFHR14-5). Here, binding site mapping of autoantibodies from 17 patients using mutant CFH19-20 constructs revealed an autoantibody epitope cluster within a loop on domain 20, next to the two buried residues that are different in CFH19-20 and CFHR14-5. The crystal structure of CFHR14-5 revealed a difference in conformation of the autoantigenic loop in the C-terminal domains of CFH and CFHR1, explaining the variation in binding of autoantibodies from some aHUS patients to CFH19-20 and CFHR14-5. The autoantigenic loop on CFH seems to be generally flexible, as its conformation in previously published structures of CFH19-20 bound to the microbial protein OspE and a sialic acid glycan is somewhat altered. Cumulatively, our data suggest that association of CFHR1 deficiency with autoimmune aHUS could be due to the structural difference between CFHR1 and the autoantigenic CFH epitope, suggesting a novel explanation for CFHR1 deficiency in the pathogenesis of autoimmune aHUS.


Asunto(s)
Autoanticuerpos/química , Proteínas Inactivadoras del Complemento C3b/química , Factor H de Complemento/química , Epítopos/química , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/inmunología , Síndrome Hemolítico Urémico Atípico/metabolismo , Autoanticuerpos/inmunología , Autoinmunidad/genética , Autoinmunidad/inmunología , Sitios de Unión/genética , Sitios de Unión/inmunología , Proteínas Inactivadoras del Complemento C3b/genética , Proteínas Inactivadoras del Complemento C3b/inmunología , Factor H de Complemento/genética , Factor H de Complemento/inmunología , Cristalografía por Rayos X , Mapeo Epitopo , Epítopos/inmunología , Humanos , Modelos Moleculares , Mutación , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Unión Proteica/inmunología , Estructura Terciaria de Proteína
11.
Nanomedicine ; 12(4): 1023-1031, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26733258

RESUMEN

Hypersensitivity reactions to particulate drugs can partly be caused by complement activation and represent a major complication during intravenous application of nanomedicines. Several liposomal and micellar drugs and carriers, and therapeutic antibodies, were shown to activate complement and induce complement activation-related pseudoallergy (CARPA) in model animals. To explore the possible use of the natural complement inhibitor factor H (FH) against CARPA, we examined the effect of FH on complement activation induced by CARPAgenic drugs. Exogenous FH inhibited complement activation induced by the antifungal liposomal Amphotericin-B (AmBisome), the widely used solvent of anticancer drugs Cremophor EL, and the anticancer monoclonal antibody rituximab in vitro. An engineered form of FH (mini-FH) was more potent inhibitor of Ambisome-, Cremophor EL- and rituximab-induced complement activation than FH. The FH-related protein CFHR1 had no inhibitory effect. Our data suggest that FH or its derivatives may be considered in the pharmacological prevention of CARPA. FROM THE CLINICAL EDITOR: Although liposomes and micelles are already in use in the clinical setting as drug carriers, there remains the potential problem of hypersensitivity due to complement activation. In this article, the authors investigated the use of complement inhibitor factor H (FH) on complement activation and showed good efficacy. The results would therefore suggest the potential application of complement inhibitor in the future.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Factor H de Complemento/administración & dosificación , Hipersensibilidad a las Drogas/tratamiento farmacológico , Liposomas/efectos adversos , Activación de Complemento/inmunología , Factor H de Complemento/inmunología , Portadores de Fármacos/efectos adversos , Hipersensibilidad a las Drogas/inmunología , Hipersensibilidad a las Drogas/patología , Voluntarios Sanos , Humanos , Micelas , Nanomedicina , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Rituximab/efectos adversos
12.
Infect Immun ; 83(3): 888-97, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534939

RESUMEN

Upon infection, pathogenic Leptospira species bind several complement regulators in order to overcome host innate immunity. We previously characterized a 20-kDa leptospiral surface protein which interacts with C4b binding protein (C4BP): leptospiral complement regulator-acquiring protein A (LcpA). Here we show that LcpA also interacts with human factor H (FH), which remains functionally active once bound to the protein. Antibodies directed against short consensus repeat 20 (SCR20) inhibited binding of FH to LcpA by approximately 90%, thus confirming that this particular domain is involved in the interaction. We have also shown for the first time that leptospires bind human vitronectin and that the interaction is mediated by LcpA. Coincubation with heparin blocked LcpA-vitronectin interaction in a dose-dependent manner, strongly suggesting that binding may occur through the heparin binding domains of vitronectin. LcpA also bound to the terminal pathway component C9 and inhibited Zn(2+)-induced polymerization and membrane attack complex (MAC) formation. Competitive binding assays indicated that LcpA interacts with C4BP, FH, and vitronectin through distinct sites. Taken together, our findings indicate that LcpA may play a role in leptospiral immune evasion.


Asunto(s)
Proteínas Bacterianas/química , Leptospira interrogans/química , Leptospira/química , Fragmentos de Péptidos/química , Vitronectina/química , Anticuerpos Monoclonales/química , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/inmunología , Sitios de Unión , Unión Competitiva , Activación de Complemento , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/inmunología , Complemento C9/química , Complemento C9/inmunología , Factor H de Complemento/química , Factor H de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/química , Heparina/química , Humanos , Evasión Inmune , Leptospira/inmunología , Leptospira/patogenicidad , Leptospira interrogans/inmunología , Leptospira interrogans/patogenicidad , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/inmunología , Unión Proteica , Vitronectina/inmunología , Zinc/química
13.
J Immunol ; 191(2): 912-21, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23772024

RESUMEN

Complement is an essential humoral component of innate immunity; however, its inappropriate activation leads to pathology. Polymorphisms, mutations, and autoantibodies affecting factor H (FH), a major regulator of the alternative complement pathway, are associated with various diseases, including age-related macular degeneration, atypical hemolytic uremic syndrome, and C3 glomerulopathies. Restoring FH function could be a treatment option for such pathologies. In this article, we report on an engineered FH construct that directly combines the two major functional regions of FH: the N-terminal complement regulatory domains and the C-terminal surface-recognition domains. This minimal-size FH (mini-FH) binds C3b and has complement regulatory functions similar to those of the full-length protein. In addition, we demonstrate that mini-FH binds to the FH ligands C-reactive protein, pentraxin 3, and malondialdehyde epitopes. Mini-FH was functionally active when bound to the extracellular matrix and endothelial cells in vitro, and it inhibited C3 deposition on the cells. Furthermore, mini-FH efficiently inhibited complement-mediated lysis of host-like cells caused by a disease-associated FH mutation or by anti-FH autoantibodies. Therefore, mini-FH could potentially be used as a complement inhibitor targeting host surfaces, as well as to replace compromised FH in diseases associated with FH dysfunction.


Asunto(s)
Complemento C3b/inmunología , Factor H de Complemento/genética , Factor H de Complemento/inmunología , Secuencia de Aminoácidos , Secuencia de Bases , Proteína C-Reactiva/metabolismo , Activación de Complemento , Complemento C3b/antagonistas & inhibidores , Complemento C3b/metabolismo , Vía Alternativa del Complemento , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Humanos , Malondialdehído/metabolismo , Unión Proteica , Ingeniería de Proteínas , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Análisis de Secuencia de ADN , Componente Amiloide P Sérico/metabolismo
14.
Semin Thromb Hemost ; 40(4): 431-43, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24799303

RESUMEN

The presence of circulating autoantibodies, primarily to complement factor H antibodies (CFH-Abs) in plasma characterizes the autoimmune form of atypical hemolytic uremic syndrome (aHUS). This acquired form of aHUS defines a distinct subgroup of aHUS patients, which requires diagnostic and treatment approaches in part different from those of the genetically defined forms. The mechanisms leading to CFH-Ab production and disease onset are not completely understood, but CFH-Ab HUS seems to be secondary to a combination of genetic predisposition and environmental factors. Early diagnosis of this specific aHUS entity is important, as prompt induction of plasma exchange and concomitant immunosuppression leads to a favorable outcome. Nevertheless, information on clinical features and outcome in children is limited. Here, we review the literature on the biological and clinical features of CFH-Ab HUS and discuss therapeutic options.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/terapia , Factor H de Complemento/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Síndrome Hemolítico Urémico Atípico/inmunología , Autoanticuerpos/sangre , Sitios de Unión , Ensayos Clínicos como Asunto , Proteínas Inactivadoras del Complemento C3b/inmunología , Predisposición Genética a la Enfermedad , Humanos , Trasplante de Riñón , Estructura Terciaria de Proteína , Resultado del Tratamiento
15.
J Immunol ; 189(5): 2502-11, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22851712

RESUMEN

Candida albicans, the most common facultative human pathogenic fungus is of major medical importance, whereas the closely related species Candida dubliniensis is less virulent and rarely causes life-threatening, systemic infections. Little is known, however, about the reasons for this difference in pathogenicity, and especially on the interactions of C. dubliniensis with the human immune system. Because innate immunity and, in particular, neutrophil granulocytes play a major role in host antifungal defense, we studied the responses of human neutrophils to clinical isolates of both C. albicans and C. dubliniensis. C. dubliniensis was found to support neutrophil migration and fungal cell uptake to a greater extent in comparison with C. albicans, whereas inducing less neutrophil damage and extracellular trap formation. The production of antimicrobial reactive oxygen species, myeloperoxidase, and lactoferrin, as well as the inflammatory chemokine IL-8 by neutrophils was increased when stimulated with C. dubliniensis as compared with C. albicans. However, most of the analyzed macrophage-derived inflammatory and regulatory cytokines and chemokines, such as IL-1α, IL-1ß, IL-1ra, TNF-α, IL-10, G-CSF, and GM-CSF, were less induced by C. dubliniensis. Similarly, the amounts of the antifungal immunity-related IL-17A produced by PBMCs was significantly lower when challenged with C. dubliniensis than with C. albicans. These data indicate that C. dubliniensis triggers stronger early neutrophil responses than C. albicans, thus providing insight into the differential virulence of these two closely related fungal species, and suggest that this is, in part, due to their differential capacity to form hyphae.


Asunto(s)
Candida albicans/inmunología , Comunicación Celular/inmunología , Neutrófilos/inmunología , Neutrófilos/microbiología , Candida albicans/metabolismo , Candida albicans/patogenicidad , Candidiasis/inmunología , Candidiasis/metabolismo , Candidiasis/patología , Movimiento Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Humanos , Neutrófilos/parasitología , Fagocitosis/inmunología , Especificidad de la Especie , Virulencia/inmunología
16.
J Immunol ; 189(4): 1858-67, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22786770

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a renal disease associated with complement alternative pathway dysregulation and is characterized by endothelial injury. Pentraxin 3 (PTX3) is a soluble pattern recognition molecule expressed by endothelial cells and upregulated under inflammatory conditions. PTX3 activates complement, but it also binds the complement inhibitor factor H. In this study, we show that native factor H, factor H-like protein 1, and factor H-related protein 1 (CFHR1) bind to PTX3 and that PTX3-bound factor H and factor H-like protein 1 maintain their complement regulatory activities. PTX3, when bound to extracellular matrix, recruited functionally active factor H. Residues within short consensus repeat 20 of factor H that are relevant for PTX3 binding were identified using a peptide array. aHUS-associated factor H mutations within this binding site caused a reduced factor H binding to PTX3. Similarly, seven of nine analyzed anti-factor H autoantibodies isolated from aHUS patients inhibited the interaction between factor H and PTX3, and five autoantibodies also inhibited PTX3 binding to CFHR1. Moreover, the aHUS-associated CFHR1*B variant showed reduced binding to PTX3 in comparison with CFHR1*A. Thus, the interactions of PTX3 with complement regulators are impaired by certain mutations and autoantibodies affecting factor H and CFHR1, which could result in an enhanced local complement-mediated inflammation, endothelial cell activation, and damage in aHUS.


Asunto(s)
Autoanticuerpos/inmunología , Proteína C-Reactiva/metabolismo , Proteínas Inactivadoras del Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Síndrome Hemolítico-Urémico/metabolismo , Componente Amiloide P Sérico/metabolismo , Síndrome Hemolítico Urémico Atípico , Autoantígenos/inmunología , Proteína C-Reactiva/inmunología , Proteínas Inactivadoras del Complemento C3b/inmunología , Factor H de Complemento/inmunología , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Síndrome Hemolítico-Urémico/inmunología , Humanos , Componente Amiloide P Sérico/inmunología
17.
Front Immunol ; 15: 1135490, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38410512

RESUMEN

Complement is an ancient and complex network of the immune system and, as such, it plays vital physiological roles, but it is also involved in numerous pathological processes. The proper regulation of the complement system is important to allow its sufficient and targeted activity without deleterious side-effects. Factor H is a major complement regulator, and together with its splice variant factor H-like protein 1 and the five human factor H-related (FHR) proteins, they have been linked to various diseases. The role of factor H in inhibiting complement activation is well studied, but the function of the FHRs is less characterized. Current evidence supports the main role of the FHRs as enhancers of complement activation and opsonization, i.e., counter-balancing the inhibitory effect of factor H. FHRs emerge as soluble pattern recognition molecules and positive regulators of the complement system. In addition, factor H and some of the FHR proteins were shown to modulate the activity of immune cells, a non-canonical function outside the complement cascade. Recent efforts have intensified to study factor H and the FHRs and develop new tools for the distinction, quantification and functional characterization of members of this protein family. Here, we provide an update and overview on the versatile roles of factor H family proteins, what we know about their biological functions in healthy conditions and in diseases.


Asunto(s)
Factor H de Complemento , Proteínas del Sistema Complemento , Humanos , Factor H de Complemento/metabolismo , Activación de Complemento
18.
J Biol Chem ; 287(23): 19528-36, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22518841

RESUMEN

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.


Asunto(s)
Apolipoproteínas/metabolismo , Convertasas de Complemento C3-C5/metabolismo , Complemento C3b/metabolismo , Vía Alternativa del Complemento/fisiología , Animales , Apolipoproteínas/genética , Sitios de Unión , Línea Celular , Convertasas de Complemento C3-C5/genética , Complemento C3b/genética , Factor B del Complemento/genética , Factor B del Complemento/metabolismo , Humanos , Mutación , Properdina/genética , Properdina/metabolismo , Spodoptera
19.
J Infect Dis ; 205(6): 995-1004, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22291192

RESUMEN

Leptospira, the causative agent of leptospirosis, interacts with several host molecules, including extracellular matrix components, coagulation cascade proteins, and human complement regulators. Here we demonstrate that acquisition of factor H (FH) on the Leptospira surface is crucial for bacterial survival in the serum and that these spirochetes, besides interacting with FH, FH related-1, and C4b binding protein (C4BP), also acquire FH like-1 from human serum. We also demonstrate that binding to these complement regulators is mediated by leptospiral immunoglobulin-like (Lig) proteins, previously shown to interact with fibronectin, laminin, collagen, elastin, tropoelastin, and fibrinogen. Factor H binds to Lig proteins via short consensus repeat domains 5 and 20. Competition assays suggest that FH and C4BP have distinct binding sites on Lig proteins. Moreover, FH and C4BP bound to immobilized Ligs display cofactor activity, mediating C3b and C4b degradation by factor I. In conclusion, Lig proteins are multifunctional molecules, contributing to leptospiral adhesion and immune evasion.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Sanguíneas/metabolismo , Proteínas Inactivadoras del Complemento C3b/metabolismo , Antígenos de Histocompatibilidad/metabolismo , Leptospira/patogenicidad , Leptospirosis/inmunología , Adhesión Bacteriana , Proteínas Bacterianas/genética , Sitios de Unión , Clonación Molecular , Complemento C3b/metabolismo , Proteína de Unión al Complemento C4b/metabolismo , Factor H de Complemento/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Evasión Inmune , Inmunoglobulinas/química , Leptospira/genética , Leptospira/metabolismo , Leptospirosis/microbiología , Plásmidos
20.
Front Immunol ; 14: 1113015, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36891314

RESUMEN

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder affecting the joints. Many patients carry anti-citrullinated protein autoantibodies (ACPA). Overactivation of the complement system seems to be part of the pathogenesis of RA, and autoantibodies against the pathway initiators C1q and MBL, and the regulator of the complement alternative pathway, factor H (FH), were previously reported. Our aim was to analyze the presence and role of autoantibodies against complement proteins in a Hungarian RA cohort. To this end, serum samples of 97 ACPA-positive RA patients and 117 healthy controls were analyzed for autoantibodies against FH, factor B (FB), C3b, C3-convertase (C3bBbP), C1q, MBL and factor I. In this cohort, we did not detect any patient with FH autoantibodies but detected C1q autoantibodies in four patients, MBL autoantibodies in two patients and FB autoantibodies in five patients. Since the latter autoantibodies were previously reported in patients with kidney diseases but not in RA, we set out to further characterize such FB autoantibodies. The isotypes of the analyzed autoantibodies were IgG2, IgG3, IgGκ, IgGλ and their binding site was localized in the Bb part of FB. We detected in vivo formed FB-autoanti-FB complexes by Western blot. The effect of the autoantibodies on the formation, activity and FH-mediated decay of the C3 convertase in solid phase convertase assays was determined. In order to investigate the effect of the autoantibodies on complement functions, hemolysis assays and fluid phase complement activation assays were performed. The autoantibodies partially inhibited the complement-mediated hemolysis of rabbit red blood cells, inhibited the activity of the solid phase C3-convertase and C3 and C5b-9 deposition on complement activating surfaces. In summary, in ACPA-positive RA patients we identified FB autoantibodies. The characterized FB autoantibodies did not enhance complement activation, rather, they had inhibitory effect on complement. These results support the involvement of the complement system in the pathomechanism of RA and raise the possibility that protective autoantibodies may be generated in some patients against the alternative pathway C3 convertase. However, further analyses are needed to assess the exact role of such autoantibodies.


Asunto(s)
Artritis Reumatoide , Factor B del Complemento , Animales , Conejos , Autoanticuerpos , Hemólisis , Complemento C1q , Convertasas de Complemento C3-C5/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA