Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
J Biol Chem ; 298(11): 102557, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36183830

RESUMEN

Proteolytic cascades comprise several important physiological systems, including a primary arm of innate immunity called the complement cascade. To safeguard against complement-mediated attack, the etiologic agent of Lyme disease, Borreliella burgdorferi, produces numerous outer surface-localized lipoproteins that contribute to successful complement evasion. Recently, we discovered a pair of B. burgdorferi surface lipoproteins of the OspEF-related protein family-termed ElpB and ElpQ-that inhibit antibody-mediated complement activation. In this study, we investigate the molecular mechanism of ElpB and ElpQ complement inhibition using an array of biochemical and biophysical approaches. In vitro assays of complement activation show that an independently folded homologous C-terminal domain of each Elp protein maintains full complement inhibitory activity and selectively inhibits the classical pathway. Using binding assays and complement component C1s enzyme assays, we show that binding of Elp proteins to activated C1s blocks complement component C4 cleavage by competing with C1s-C4 binding without occluding the active site. C1s-mediated C4 cleavage is dependent on activation-induced binding sites, termed exosites. To test whether these exosites are involved in Elp-C1s binding, we performed site-directed mutagenesis, which showed that ElpB and ElpQ binding require C1s residues in the anion-binding exosite located on the serine protease domain of C1s. Based on these results, we propose a model whereby ElpB and ElpQ exploit activation-induced conformational changes that are normally important for C1s-mediated C4 cleavage. Our study expands the known complement evasion mechanisms of microbial pathogens and reveals a novel molecular mechanism for selective C1s inhibition by Lyme disease spirochetes.


Asunto(s)
Borrelia burgdorferi , Enfermedad de Lyme , Humanos , Complemento C1s/química , Complemento C1s/metabolismo , Borrelia burgdorferi/genética , Complemento C4/química , Proteínas del Sistema Complemento/metabolismo , Serina Proteasas , Lipoproteínas/genética
2.
Methods Mol Biol ; 2227: 249-264, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33847947

RESUMEN

Activated complement component C4 (C4b) is the nonenzymatic component of the classical pathway (CP) convertases of the complement system. Preparation of C4 and C4b samples suitable for structural biology studies is challenging due to low yields and complexity of recombinant C4 production protocols reported so far and heterogeneity of C4 in native sources. Here we present a purification protocol for human C4 and describe sample preparation methods for structural investigation of C4 and its complexes by crystallography, small angle X-ray scattering, and electron microscopy.


Asunto(s)
Complemento C4/química , Complemento C4/aislamiento & purificación , Proteómica/métodos , Biología/métodos , Cromatografía en Gel/métodos , Cristalografía por Rayos X/métodos , Humanos , Intercambio Iónico , Microscopía Electrónica/métodos , Coloración Negativa/métodos , Conformación Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X/métodos
3.
Viruses ; 13(1)2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33467558

RESUMEN

Adenovirus (AdV) infection elicits a strong immune response with the production of neutralizing antibodies and opsonization by complement and coagulation factors. One anti-hexon neutralizing antibody, called 9C12, is known to activate the complement cascade, resulting in the deposition of complement component C4b on the capsid, and the neutralization of the virus. The mechanism of AdV neutralization by C4b is independent of downstream complement proteins and involves the blockage of the release of protein VI, which is required for viral escape from the endosome. To investigate the structural basis underlying how C4b blocks the uncoating of AdV, we built a model for the complex of human adenovirus type-5 (HAdV5) with 9C12, together with complement components C1 and C4b. This model positions C4b near the Arg-Gly-Asp (RGD) loops of the penton base. There are multiple amino acids in the RGD loop that might serve as covalent binding sites for the reactive thioester of C4b. Molecular dynamics simulations with a multimeric penton base and C4b indicated that stabilizing interactions may form between C4b and multiple RGD loops. We propose that C4b deposition on one RGD loop leads to the entanglement of C4b with additional RGD loops on the same penton base multimer and that this entanglement blocks AdV uncoating.


Asunto(s)
Adenoviridae/inmunología , Complemento C4/química , Complemento C4/inmunología , Modelos Moleculares , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/química , Anticuerpos Antivirales/inmunología , Sitios de Unión , Cápside/química , Cápside/metabolismo , Cápside/ultraestructura , Proteínas de la Cápside/química , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/ultraestructura , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
4.
Int J Mol Med ; 47(1): 276-288, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33236147

RESUMEN

Preeclampsia (PE) is a pregnancy­specific complication characterized by hypertension and proteinuria, and it is one of the primary global causes of maternal and perinatal mortality. Poor remodeling of placental arteries and endothelial dysfunction serve important roles in the pathogenesis of PE. Peptide derived from complement C4 A chain (PDCC4) was identified in our previous peptidome analysis of serum from patients with PE. The present study aimed to investigate the effect of PDCC4 on endothelial dysfunction in PE. TNF­α stimulated HUVECs were employed to mimic endothelial dysfunction in PE, and Cell Counting Kit 8 assay, wound healing assay, tube formation assay, RNA­sequencing (seq) and western blot analysis were performed using HUVECs. Moreover, an in vivo model of PE was established using pregnant rats treated with lipopolysaccharide (LPS), and blood pressure monitoring, histopathological examination, ELISA and immunohistochemistry were performed on rats. It was found that TNF­α impaired proliferation, migration and tube formation of HUVECs, but pretreatment with PDCC4 moderated these effects. RNA­seq and western blotting demonstrated that the PI3K/mTOR/HIF1α signaling pathway was activated by PDCC4, and a selective PI3K inhibitor reversed the protective function of PDCC4 on TNF­α stimulated HUVECs. Additionally, PDCC4 alleviated hypertension, histopathological changes of placenta and kidney and the expression levels of endothelial injury markers and inflammatory cytokines induced by LPS in rats. These results suggested that PDCC4 relieved endothelial dysfunction in PE via PI3K/mTOR/HIF1α signaling pathway and may be a potential therapy for PE.


Asunto(s)
Complemento C4/química , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Preeclampsia/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Péptidos/química , Preeclampsia/patología , Embarazo
5.
Fish Shellfish Immunol ; 104: 527-536, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32599058

RESUMEN

Complement component 4 (C4) has critical immunological functions in vertebrates. In the current study, a C4 homolog (gcC4) was identified in grass carp (Ctenopharyngodon idella). The full-length 5458 bp gcC4 cDNA contained a 5148 bp open reading frame (ORF) encoding a protein of 1715 amino acids with a signal peptide and eight conservative domains. The gcC4 protein has a high level of identity with other fish C4 counterparts and is phylogenetically clustered with cyprinid fish C4. The gcC4 transcript shows wide tissue distribution and is inducible by Aeromonas hydrophila in vivo and in vitro. Furthermore, its expression also fluctuates upon lipopolysaccharide or flagellin stimulation in vitro. During infection, the gcC4 protein level decreases or increases to varying degrees, and the intrahepatic C4 expression location changes. With gcC4 overexpression, interleukin 1 beta, tumor necrosis factor alpha, and interferon transcripts are all upregulated by A. hydrophila infection. Meanwhile, overexpression of gcC4 reduces bacterial invasion or proliferation. Moreover, gcC4 may activate the NF-κB signaling pathway. These findings demonstrate the vital role of gcC4 in the innate immunity of grass carp.


Asunto(s)
Carpas/genética , Carpas/inmunología , Complemento C4/genética , Complemento C4/inmunología , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Aeromonas hydrophila/fisiología , Secuencia de Aminoácidos , Animales , Complemento C4/química , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/veterinaria , FN-kappa B/fisiología , Filogenia , Alineación de Secuencia/veterinaria , Transducción de Señal/inmunología
6.
PLoS One ; 14(4): e0214514, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30958843

RESUMEN

The kidney is especially sensitive to diseases associated with overactivation of the complement system. While most of these diseases affect kidney glomeruli and the microvasculature, there is also evidence for tubulointerstitial deposition of complement factors. Complement inactivating factors on cell membranes comprise CD55, CD59 and CD46, which is also termed membrane cofactor protein (MCP). CD46 has been described as localized to glomeruli, but especially also to proximal tubular epithelial cells (RPTECs). However, human cell culture models to assess CD46 function on RPTECs are still missing. Therefore, we here performed gene editing of RPTEC/TERT1 cells generating a monoclonal CD46-/- cell line that did not show changes of the primary cell like characteristics. In addition, factor I and CD46-mediated cleavage of C4b into soluble C4c and membrane deposited C4d was clearly reduced in the knock-out cell line as compared to the maternal cells. Thus, human CD46-/- proximal tubular epithelial cells will be of interest to dissect the roles of the epithelium and the kidney in various complement activation mediated tubulointerstitial pathologies or in studying CD46 mediated uropathogenic internalization of bacteria. In addition, this gives proof-of-principle, that telomerized cells can be used in the generation of knock-out, knock-in or any kind of reporter cell lines without losing the primary cell characteristics of the maternal cells.


Asunto(s)
Sistemas CRISPR-Cas , Activación de Complemento , Células Epiteliales/citología , Técnicas de Inactivación de Genes , Proteína Cofactora de Membrana/genética , Telomerasa/metabolismo , Línea Celular , Complemento C4/química , Complemento C4b/química , Edición Génica , Humanos , Túbulos Renales/citología , Telómero/ultraestructura , gamma-Glutamiltransferasa/metabolismo
7.
Kidney Int ; 94(4): 689-700, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29884545

RESUMEN

Certain kidney diseases are associated with complement activation although a renal triggering factor has not been identified. Here we demonstrated that renin, a kidney-specific enzyme, cleaves C3 into C3b and C3a, in a manner identical to the C3 convertase. Cleavage was specifically blocked by the renin inhibitor aliskiren. Renin-mediated C3 cleavage and its inhibition by aliskiren also occurred in serum. Generation of C3 cleavage products was demonstrated by immunoblotting, detecting the cleavage product C3b, by N-terminal sequencing of the cleavage product, and by ELISA for C3a release. Functional assays showed mast cell chemotaxis towards the cleavage product C3a and release of factor Ba when the cleavage product C3b was combined with factor B and factor D. The renin-mediated C3 cleavage product bound to factor B. In the presence of aliskiren this did not occur, and less C3 deposited on renin-producing cells. The effect of aliskiren was studied in three patients with dense deposit disease and this demonstrated decreased systemic and renal complement activation (increased C3, decreased C3a and C5a, decreased renal C3 and C5b-9 deposition and/or decreased glomerular basement membrane thickness) over a follow-up period of four to seven years. Thus, renin can trigger complement activation, an effect inhibited by aliskiren. Since renin concentrations are higher in renal tissue than systemically, this may explain the renal propensity of complement-mediated disease in the presence of complement mutations or auto-antibodies.


Asunto(s)
Amidas/farmacología , Activación de Complemento/efectos de los fármacos , Complemento C3/química , Fumaratos/farmacología , Glomerulonefritis Membranoproliferativa/metabolismo , Glomerulonefritis Membranoproliferativa/terapia , Renina/química , Amidas/uso terapéutico , Quimiotaxis/efectos de los fármacos , Niño , Complemento C3/metabolismo , Complemento C3a/química , Complemento C3a/metabolismo , Complemento C3b/química , Complemento C3b/metabolismo , Complemento C4/química , Complemento C5a/química , Complemento C5a/metabolismo , Complemento C5b/química , Complemento C5b/metabolismo , Factor B del Complemento/química , Factor D del Complemento/química , Femenino , Fumaratos/uso terapéutico , Membrana Basal Glomerular/patología , Glomerulonefritis Membranoproliferativa/patología , Humanos , Mastocitos/fisiología , Renina/antagonistas & inhibidores , Renina/metabolismo
8.
J Korean Med Sci ; 33(18): e136, 2018 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-29713256

RESUMEN

C4 glomerulopathy is a recently introduced entity that presents with bright C4d staining and minimal or absent immunoglobulin and C3 staining. We report a case of a 62-year-old man with C4 glomerulonephritis (GN) and uveitis. He presented to the nephrology department with proteinuria and hematuria. The patient also had intermediate uveitis along with proteinuria and hematuria. A kidney biopsy that was performed in light of continuing proteinuria and hematuria showed a focal proliferative, focal sclerotic glomerulopathy pattern on light microscopy, absent staining for immunoglobulin or C3 by immunofluorescence microscopy, with bright staining for C4d on immunohistochemistry, and electron-dense deposits on electron microscopy. Consequently, C4 GN was suggested as the pathologic diagnosis. Although laser microdissection and mass spectrometry for glomerular deposit and pathologic evaluation of the retinal tissue were not performed, this is the first report of C4 GN in Korea and the first case of coexisting C4 GN and uveitis in the English literature.


Asunto(s)
Complemento C4/metabolismo , Glomerulonefritis/diagnóstico , Uveítis Intermedia/diagnóstico , Complemento C4/química , Glomerulonefritis/complicaciones , Glomerulonefritis/patología , Humanos , Riñón/patología , Masculino , Microscopía Electrónica , Persona de Mediana Edad , Proteinuria/etiología , Uveítis Intermedia/complicaciones
9.
J Biomol Struct Dyn ; 36(3): 575-589, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28132614

RESUMEN

Serine protease cleaved-complement component 4 (C4) at sessile loop, which is significant for completion of lectin and classical complement pathways at the time of infections. The co-crystalized structure of C4 with Mannose-binding protein-associated serine protease 2 (MASP2) provided the structural and functional aspects of its interaction and underlined the C4 activation by MASP2. The same study also revealed the significance of complement control protein (CCP) domain through mutational study, where mutated CCP domain led to the inhibition of C4 activation. However, the interaction of trypsin serine domain with C4α sessile loop revealed another aspect of C4 activation. The human C4 cleavage by Trypsin (Tryp) in a control manner was explored but not yet revealed the identification of cleaved fragments. Hence, the present study investigated the Tryp mediated C4 activation using computational approach (protein-protein docking and molecular dynamics simulation) by comparing with the co-crystalized structure of C4-MASP2. Docking result identified the crucial interacting residues Gly219, Gln178, and Asn102 of Tryp catalytic pocket which were interacting with Arg756 and Glu759 (sessile loop) of α-Chain (C4) in a similar manner to C4-MASP2 co-crystallized complex. Moreover, MD simulation results and mutational study underlined the conformational rearrangements in the C4 due to the Tryp interaction. Comparative analysis of C4 alone, C4-Tryp, and C4-MASP2 revealed the impact of Tryp on C4 was similar as MASP2. These studies designate the role of sessile loop in the interaction with serine domain, which could be useful to understand the various interactions of C4 with other complement components.


Asunto(s)
Complemento C4/química , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/química , Conformación Proteica , Serina Proteasas/química , Activación de Complemento/genética , Cristalografía por Rayos X , Humanos , Lectinas/química , Simulación del Acoplamiento Molecular , Unión Proteica , Mapas de Interacción de Proteínas , Tripsina/química
10.
Acta Crystallogr D Struct Biol ; 72(Pt 9): 1006-16, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27599733

RESUMEN

While the rapid proliferation of high-resolution structures in the Protein Data Bank provides a rich set of templates for starting models, it remains the case that a great many structures both past and present are built at least in part by hand-threading through low-resolution and/or weak electron density. With current model-building tools this task can be challenging, and the de facto standard for acceptable error rates (in the form of atomic clashes and unfavourable backbone and side-chain conformations) in structures based on data with dmax not exceeding 3.5 Šreflects this. When combined with other factors such as model bias, these residual errors can conspire to make more serious errors in the protein fold difficult or impossible to detect. The three recently published 3.6-4.2 Šresolution structures of complement C4 (PDB entries 4fxg, 4fxk and 4xam) rank in the top quartile of structures of comparable resolution both in terms of Rfree and MolProbity score, yet, as shown here, contain register errors in six ß-strands. By applying a molecular-dynamics force field that explicitly models interatomic forces and hence excludes most physically impossible conformations, the recently developed interactive molecular-dynamics flexible fitting (iMDFF) approach significantly reduces the complexity of the conformational space to be searched during manual rebuilding. This substantially improves the rate of detection and correction of register errors, and allows user-guided model building in maps with a resolution lower than 3.5 Što converge to solutions with a stereochemical quality comparable to atomic resolution structures. Here, iMDFF has been used to individually correct and re-refine these three structures to MolProbity scores of <1.7, and strategies for working with such challenging data sets are suggested. Notably, the improved model allowed the resolution for complement C4b to be extended from 4.2 to 3.5 Šas demonstrated by paired refinement.


Asunto(s)
Complemento C4/química , Simulación de Dinámica Molecular , Cristalografía por Rayos X , Humanos , Conformación Proteica , Dominios Proteicos
11.
J Biol Chem ; 291(32): 16494-507, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27252379

RESUMEN

The lectin (LP) and classical (CP) pathways are two of the three main activation cascades of the complement system. These pathways start with recognition of different pathogen- or danger-associated molecular patterns and include identical steps of proteolytic activation of complement component C4, formation of the C3 proconvertase C4b2, followed by cleavage of complement component C2 within C4b2 resulting in the C3 convertase C4b2a. Here, we describe the solution structures of the two central complexes of the pathways, C3 proconvertase and C3 convertase, as well as the unbound zymogen C2 obtained by small angle x-ray scattering analysis. We analyzed both native and enzymatically deglycosylated C4b2 and C2 and showed that the resulting structural models were independent of the glycans. The small angle x-ray scattering-derived models suggest a different activation mode for the CP/LP C3 proconvertase as compared with that established for the alternative pathway proconvertase C3bB. This is likely due to the rather different structural and functional properties of the proteases activating the proconvertases. The solution structure of a stabilized form of the active CP/LP C3 convertase C4b2a is strikingly similar to the crystal structure of the alternative pathway C3 convertase C3bBb, which is in accordance with their identical functions in cleaving the complement proteins C3 and C5.


Asunto(s)
Complemento C2/química , Convertasas de Complemento C3-C5/química , Complemento C4/química , Humanos , Difracción de Rayos X
12.
Nature ; 530(7589): 177-83, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26814963

RESUMEN

Schizophrenia is a heritable brain illness with unknown pathogenic mechanisms. Schizophrenia's strongest genetic association at a population level involves variation in the major histocompatibility complex (MHC) locus, but the genes and molecular mechanisms accounting for this have been challenging to identify. Here we show that this association arises in part from many structurally diverse alleles of the complement component 4 (C4) genes. We found that these alleles generated widely varying levels of C4A and C4B expression in the brain, with each common C4 allele associating with schizophrenia in proportion to its tendency to generate greater expression of C4A. Human C4 protein localized to neuronal synapses, dendrites, axons, and cell bodies. In mice, C4 mediated synapse elimination during postnatal development. These results implicate excessive complement activity in the development of schizophrenia and may help explain the reduced numbers of synapses in the brains of individuals with schizophrenia.


Asunto(s)
Complemento C4/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Esquizofrenia/genética , Alelos , Secuencia de Aminoácidos , Animales , Axones/metabolismo , Secuencia de Bases , Encéfalo/metabolismo , Encéfalo/patología , Complemento C4/química , Vía Clásica del Complemento , Dendritas/metabolismo , Dosificación de Gen/genética , Regulación de la Expresión Génica/genética , Haplotipos/genética , Humanos , Complejo Mayor de Histocompatibilidad/genética , Ratones , Modelos Animales , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Polimorfismo de Nucleótido Simple/genética , ARN Mensajero/análisis , ARN Mensajero/genética , Factores de Riesgo , Esquizofrenia/patología , Sinapsis/metabolismo
13.
MAbs ; 7(6): 1094-103, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26338058

RESUMEN

Many therapeutic monoclonal antibodies (mAbs) are clinically administered through intravenous infusion after mixing with a diluent, e.g., saline, 5% dextrose. Such a clinical setting increases the likelihood of interactions among mAb molecules, diluent, and plasma components, which may adversely affect product safety and efficacy. Avastin® (bevacizumab) and Herceptin® (trastuzumab), but not Remicade® (infliximab), were shown to undergo rapid aggregation upon dilution into 5% dextrose when mixed with human plasma in vitro; however, the biochemical pathways leading to the aggregation were not clearly defined. Here, we show that dextrose-mediated aggregation of Avastin or Herceptin in plasma involves isoelectric precipitation of complement proteins. Using mass spectrometry, we found that dextrose-induced insoluble aggregates were composed of mAb itself and multiple abundant plasma proteins, namely complement proteins C3, C4, factor H, fibronectin, and apolipoprotein. These plasma proteins, which are characterized by an isoelectronic point of 5.5-6.7, lost solubility at the resulting pH in the mixture with formulated Avastin (pH 6.2) and Herceptin (pH 6.0). Notably, switching formulation buffers for Avastin (pH 6.2) and Remicade (pH 7.2) reversed their aggregation profiles. Avastin formed little, if any, insoluble aggregates in dextrose-plasma upon raising the buffer pH to 7.2 or above. Furthermore, dextrose induced pH-dependent precipitation of plasma proteins, with massive insoluble aggregates being detected at pH 6.5-6.8. These data show that isoelectric precipitation of complement proteins is a prerequisite of dextrose-induced aggregation of mAb in human plasma. This finding highlights the importance of assessing the compatibility of a therapeutic mAb with diluent and human plasma during product development.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Proteínas Sanguíneas/metabolismo , Proteínas del Sistema Complemento/metabolismo , Glucosa/metabolismo , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/química , Apolipoproteínas/química , Apolipoproteínas/metabolismo , Bevacizumab/sangre , Bevacizumab/química , Bevacizumab/metabolismo , Proteínas Sanguíneas/química , Cromatografía Liquida , Complemento C3/química , Complemento C3/metabolismo , Complemento C4/química , Complemento C4/metabolismo , Factor H de Complemento/química , Factor H de Complemento/metabolismo , Proteínas del Sistema Complemento/química , Electroforesis en Gel de Poliacrilamida , Fibronectinas/química , Fibronectinas/metabolismo , Glucosa/química , Humanos , Concentración de Iones de Hidrógeno , Infliximab/sangre , Infliximab/química , Infliximab/metabolismo , Punto Isoeléctrico , Espectrometría de Masas , Agregación Patológica de Proteínas , Unión Proteica , Trastuzumab/sangre , Trastuzumab/química , Trastuzumab/metabolismo
14.
Eur J Immunol ; 45(2): 544-50, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25359215

RESUMEN

The lectin pathway (LP) of complement has a protective function against invading pathogens. Recent studies have also shown that the LP plays an important role in ischemia/reperfusion (I/R)-injury. MBL-associated serine protease (MASP)-2 appears to be crucial in this process. The serpin C1-inhibitor is the major inhibitor of MASP-2. In addition, aprotinin, a Kunitz-type inhibitor, was shown to inhibit MASP-2 activity in vitro. In this study we investigated whether the Kunitz-type inhibitor tissue factor pathway inhibitor (TFPI) is also able to inhibit MASP-2. Ex vivo LP was induced and detected by C4-deposition on mannan-coated plates. The MASP-2 activity was measured in a fluid-phase chromogenic assay. rTFPI in the absence or presence of specific monoclonal antibodies was used to investigate which TFPI-domains contribute to MASP-2 inhibition. Here, we identify TFPI as a novel selective inhibitor of MASP-2, without affecting MASP-1 or the classical pathway proteases C1s and C1r. Kunitz-2 domain of TFPI is required for the inhibition of MASP-2. Considering the role of MASP-2 in complement-mediated I/R-injury, the inhibition of this protease by TFPI could be an interesting therapeutic approach to limit the tissue damage in conditions such as cerebral stroke, myocardial infarction or solid organ transplantation.


Asunto(s)
Complemento C4/inmunología , Lectina de Unión a Manosa de la Vía del Complemento , Lipoproteínas/inmunología , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/antagonistas & inhibidores , Proteínas Recombinantes/inmunología , Inhibidores de Serina Proteinasa/inmunología , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Compuestos Cromogénicos , Complemento C1r/química , Complemento C1r/inmunología , Complemento C1s/química , Complemento C1s/inmunología , Complemento C4/química , Humanos , Inmunoensayo , Lipoproteínas/química , Lipoproteínas/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/química , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/inmunología , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/genética , Soluciones
15.
PLoS One ; 8(12): e82094, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24349192

RESUMEN

BACKGROUND: It has been hypothesized that persistent hepatitis C virus (HCV) infection is mediated in part by viral proteins that abrogate the host immune response, including the complement system, but the precise mechanisms are not well understood. We investigated whether HCV proteins are involved in the fragmentation of complement component 4 (C4), composed of subunits C4α, C4ß, and C4γ, and the role of HCV proteins in complement activation. METHODS: Human C4 was incubated with HCV nonstructural (NS) 3/4A protease, core, or NS5. Samples were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and then subjected to peptide sequencing. The activity of the classical complement pathway was examined using an erythrocyte hemolysis assay. The cleavage pattern of C4 in NS3/4A-expressing and HCV-infected cells, respectively, was also examined. RESULTS: HCV NS3/4A protease cleaved C4γ in a concentration-dependent manner, but viral core and NS5 did not. A specific inhibitor of NS3/4A protease reduced C4γ cleavage. NS3/4A protease-mediated cleavage of C4 inhibited classical pathway activation, which was abrogated by a NS3/4A protease inhibitor. In addition, co-transfection of cells with C4 and wild-type NS3/4A, but not a catalytic-site mutant of NS3/4A, produced cleaved C4γ fragments. Such C4 processing, with a concomitant reduction in levels of full-length C4γ, was also observed in HCV-infected cells expressing C4. CONCLUSIONS: C4 is a novel cellular substrate of the HCV NS3/4A protease. Understanding disturbances in the complement system mediated by NS3/4A protease may provide new insights into the mechanisms underlying persistent HCV infection.


Asunto(s)
Proteínas Portadoras/metabolismo , Activación de Complemento , Complemento C4/metabolismo , Proteínas no Estructurales Virales/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Activación de Complemento/efectos de los fármacos , Complemento C4/química , Hemólisis/efectos de los fármacos , Hepatitis C/virología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Datos de Secuencia Molecular , Oligopéptidos/farmacología , Inhibidores de Proteasas/farmacología , Ovinos
16.
Mol Immunol ; 56(3): 222-31, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23810291

RESUMEN

To initiate the lectin pathway of complement pattern recognition molecules bind to surface-linked carbohydrates or acetyl groups on pathogens or damaged self-tissue. This leads to activation of the serine proteases MASP-1 and MASP-2 resulting in deposition of C4 on the activator and assembly of the C3 convertase. In addition MASP-3 and the non-catalytic MAp19 and MAp44 presumably play regulatory functions, but the exact function of the MASP-3 protease remains to be established. Recent functional studies have significantly advanced our understanding of the molecular events occurring as activation progresses from pattern recognition to convertase assembly. Furthermore, atomic structures derived by crystallography or solution scattering of most proteins acting in the lectin pathway and two key complexes have become available. Here we integrate the current functional and structural knowledge concerning the lectin pathway proteins and derive overall models for their glycan bound complexes. These models are used to discuss cis- versus trans-activation of MASP proteases and the geometry of C4 deposition occurring on glycans in the lectin pathway.


Asunto(s)
Activación de Complemento/fisiología , Lectina de Unión a Manosa de la Vía del Complemento/fisiología , Proteínas del Sistema Complemento/inmunología , Animales , Complemento C4/química , Complemento C4/inmunología , Complemento C4/metabolismo , Proteínas del Sistema Complemento/química , Proteínas del Sistema Complemento/metabolismo , Humanos , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/química , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo
17.
J Biol Chem ; 288(22): 15821-9, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23592783

RESUMEN

The complement system is an ancient innate immune defense pathway that plays a front line role in eliminating microbial pathogens. Recognition of foreign targets by antibodies drives sequential activation of two serine proteases, C1r and C1s, which reside within the complement Component 1 (C1) complex. Active C1s propagates the immune response through its ability to bind and cleave the effector molecule complement Component 4 (C4). Currently, the precise structural and biochemical basis for the control of the interaction between C1s and C4 is unclear. Here, using surface plasmon resonance, we show that the transition of the C1s zymogen to the active form is essential for C1s binding to C4. To understand this, we determined the crystal structure of a zymogen C1s construct (comprising two complement control protein (CCP) domains and the serine protease (SP) domain). These data reveal that two loops (492-499 and 573-580) in the zymogen serine protease domain adopt a conformation that would be predicted to sterically abrogate C4 binding. The transition from zymogen to active C1s repositions both loops such that they would be able to interact with sulfotyrosine residues on C4. The structure also shows the junction of the CCP1 and CCP2 domains of C1s for the first time, yielding valuable information about the exosite for C4 binding located at this position. Together, these data provide a structural explanation for the control of the interaction with C1s and C4 and, furthermore, point to alternative strategies for developing therapeutic approaches for controlling activation of the complement cascade.


Asunto(s)
Complemento C1s/química , Complemento C4/química , Precursores Enzimáticos/química , Complemento C1s/genética , Complemento C1s/metabolismo , Complemento C4/genética , Complemento C4/metabolismo , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Humanos , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad
18.
Eur J Pharmacol ; 709(1-3): 72-9, 2013 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23567070

RESUMEN

Secondary hyperparathyroidism is a common complication in patients with chronic kidney disease and frequently persists after kidney transplantation. Paricalcitol, a selective vitamin D receptor activator, is indicated in the management of this disorder and recent evidences have suggested that this drug has other beneficial effects. Aiming to elucidate these effects, our study included 52 stable kidney transplant recipients randomized 2:1 to treatment with paricalcitol or to no treatment. Bone mineral parameters, kidney function and inflammatory status were assessed at baseline, at 3 and at 12 months. Moreover, a proteomic approach, based on magnetic beads technology coupled to MALDI-TOF mass spectrometry readout, was used to determine changes in patients' plasma peptidome. Patients treated with paricalcitol showed a significant decrease in parathyroid hormone and alkaline phosphatase levels, and an increase of bone mineral density and glomerular filtration rate. The proteomic analysis revealed a decrease in bradykinin after paricalcitol treatment, whereas 2 peptides identified as fragments of the complement factor C4 decreased only in those patients not treated with paricalcitol. These findings suggest that paricalcitol may offer additional benefits due to immunomodulatory effects via the kallikrein-kinin and complement systems.


Asunto(s)
Conservadores de la Densidad Ósea/uso terapéutico , Resorción Ósea/prevención & control , Ergocalciferoles/uso terapéutico , Hiperparatiroidismo Secundario/tratamiento farmacológico , Fallo Renal Crónico/fisiopatología , Riñón/efectos de los fármacos , Receptores de Calcitriol/agonistas , Anciano , Biomarcadores/sangre , Biomarcadores/química , Densidad Ósea/efectos de los fármacos , Resorción Ósea/etiología , Bradiquinina/sangre , Bradiquinina/química , Complemento C4/análisis , Complemento C4/química , Femenino , Humanos , Hiperparatiroidismo Secundario/sangre , Hiperparatiroidismo Secundario/etiología , Hiperparatiroidismo Secundario/fisiopatología , Riñón/inmunología , Riñón/fisiopatología , Fallo Renal Crónico/inmunología , Fallo Renal Crónico/terapia , Trasplante de Riñón/inmunología , Fenómenos Magnéticos , Masculino , Microesferas , Persona de Mediana Edad , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/química , Proteómica/métodos , Receptores de Calcitriol/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
19.
Proc Natl Acad Sci U S A ; 109(38): 15425-30, 2012 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-22949645

RESUMEN

An essential aspect of innate immunity is recognition of molecular patterns on the surface of pathogens or altered self through the lectin and classical pathways, two of the three well-established activation pathways of the complement system. This recognition causes activation of the MASP-2 or the C1s serine proteases followed by cleavage of the protein C4. Here we present the crystal structures of the 203-kDa human C4 and the 245-kDa C4·MASP-2 substrate·enzyme complex. When C4 binds to MASP-2, substantial conformational changes in C4 are induced, and its scissile bond region becomes ordered and inserted into the protease catalytic site in a manner canonical to serine proteases. In MASP-2, an exosite located within the CCP domains recognizes the C4 C345C domain 60 Å from the scissile bond. Mutations in C4 and MASP-2 residues at the C345C-CCP interface inhibit the intermolecular interaction and C4 cleavage. The possible assembly of the huge in vivo enzyme-substrate complex consisting of glycan-bound mannan-binding lectin, MASP-2, and C4 is discussed. Our own and prior functional data suggest that C1s in the classical pathway of complement activated by, e.g., antigen-antibody complexes, also recognizes the C4 C345C domain through a CCP exosite. Our results provide a unified structural framework for understanding the early and essential step of C4 cleavage in the elimination of pathogens and altered self through two major pathways of complement activation.


Asunto(s)
Complemento C4/química , Sitios de Unión , Cristalografía/métodos , Células HEK293 , Humanos , Inmunidad Innata , Mananos/química , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/química , Conformación Molecular , Mutación , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas/química , Proteolisis , Proteínas Recombinantes/química , Electricidad Estática , Especificidad por Sustrato
20.
Immunol Lett ; 143(1): 101-5, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22306105

RESUMEN

Antibody effector functions other than neutralization depend on interactions with soluble and cellular components of the immune system. Antigen recognition is usually oligoclonal, with the different clones of antibodies belonging to different classes, subclasses, glycoforms and having different affinities and epitope specificities. Thus, composition of immune complexes determines biological effects mainly via interactions with FcR and complement proteins. Antibodies are capable of triggering any of the three pathways of complement activation and antigen recognition of complex antigens often results in the activation of more than one pathway. These events can be tracked in a multiplex format using antigen microarrays, where complement products bind to elements of the microarray. By controlling cation concentrations and detecting various complement components (C1q, C4, C3) contribution of the different pathways can be identified. Parallel measurement of antibodies and complement proteins provides a novel way of looking at interactions between antigen and antibodies. We propose the use of immune complex signatures, composite depictions of antibody and complement content of immune complexes characterizing healthy and diseased populations. Normalized interquartile ranges of antibody binding (IgM, IgG) and complement deposition (C4, C3) are projected onto radar charts to produce patterns that can distinguish normal and altered immune responses. We propose that comprehensive interaction studies of serum antibodies and complement with arrays of antigens can generate functional antibody profiles and help better understand immunological disease mechanism.


Asunto(s)
Antígenos/inmunología , Complemento C1q/inmunología , Complemento C3/inmunología , Complemento C4/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/inmunología , Antígenos/química , Complemento C1q/química , Complemento C3/química , Complemento C4/química , Humanos , Inmunoglobulina G/química , Inmunoglobulina M/química , Modelos Moleculares , Análisis por Matrices de Proteínas , Dominios y Motivos de Interacción de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA