Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunol Rev ; 313(1): 402-419, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36369963

RESUMEN

The complement alternative pathway (AP) is implicated in numerous diseases affecting many organs, ranging from the rare hematological disease paroxysmal nocturnal hemoglobinuria (PNH), to the common blinding disease age-related macular degeneration (AMD). Critically, the AP amplifies any activating trigger driving a downstream inflammatory response; thus, components of the pathway have become targets for drugs of varying modality. Recent validation from clinical trials using drug modalities such as inhibitory antibodies has paved the path for gene targeting of the AP or downstream effectors. Gene targeting in the complement field currently focuses on supplementation or suppression of complement regulators in AMD and PNH, largely because the eye and liver are highly amenable to drug delivery through local (eye) or systemic (liver) routes. Targeting the liver could facilitate treatment of numerous diseases as this organ generates most of the systemic complement pool. This review explains key concepts of RNA and DNA targeting and discusses assets in clinical development for the treatment of diseases driven by the alternative pathway, including the RNA-targeting therapeutics ALN-CC5, ARO-C3, and IONIS-FB-LRX, and the gene therapies GT005 and HMR59. These therapies are but the spearhead of potential drug candidates that might revolutionize the field in coming years.


Asunto(s)
Proteínas del Sistema Complemento , Hemoglobinuria Paroxística , Humanos , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/metabolismo , Hemoglobinuria Paroxística/tratamiento farmacológico , Hemoglobinuria Paroxística/genética , Marcación de Gen , Vía Alternativa del Complemento
2.
J Biol Chem ; : 107452, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38852887

RESUMEN

Rare variants (RVs) in the gene encoding the regulatory enzyme complement factor I (CFI; FI) that reduce protein function or levels increase age-related macular degeneration (AMD) risk. A total of 3357 subjects underwent screening in the SCOPE natural history study for Geographic Atrophy (GA) secondary to AMD, including CFI sequencing followed by serum FI measurement. Eleven CFI RV genotypes that were challenging to categorise as Type I (low serum level) or Type II (normal serum level but reduced enzymatic function) were characterized in the context of pure FI protein in C3b and C4b fluid phase cleavage assays and a novel bead-based functional assay (BBFA) of surface-bound C3b cleavage. A further 4 variants predicted or previously characterized as benign, were analysed using the BBFA to add confidence to their classification. In all, 3 variants [W51S, C67R, I370T] resulted in low expression. A further 4 variants [P64L, R339Q, G527V and P528T] were identified as being highly deleterious with IC50s for C3b breakdown >1 log increased vs the WT protein, while 2 variants [K476E and R474Q] were ∼1 log reduced in function. Meanwhile, 6 variants [P50A, T203I, K441R, E548Q, P553S, S570T] had IC50s similar to wild-type (WT). Odds ratios (ORs) and BBFA IC50s were positively correlated (r=0.76, P<0.01), whilst ORs vs combined annotation dependent depletion (CADD) scores were not (r=0.43, P=0.16). Overall, 15 CFI RVs were functionally characterized which may aid future patient stratification approaches for complement-targeted therapies. Pure protein in vitro analysis remains the gold standard for determining the functional consequence of CFI RVs.

3.
Immunology ; 168(3): 473-492, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36175370

RESUMEN

Complement, a critical defence against pathogens, has been implicated as a driver of pathology in COVID-19. Complement activation products are detected in plasma and tissues and complement blockade is considered for therapy. To delineate roles of complement in immunopathogenesis, we undertook the largest comprehensive study of complement in COVID-19 to date, comprehensive profiling of 16 complement biomarkers, including key components, regulators and activation products, in 966 plasma samples from 682 hospitalized COVID-19 patients collected across the hospitalization period as part of the UK ISARIC4C (International Acute Respiratory and Emerging Infection Consortium) study. Unsupervised clustering of complement biomarkers mapped to disease severity and supervised machine learning identified marker sets in early samples that predicted peak severity. Compared to healthy controls, complement proteins and activation products (Ba, iC3b, terminal complement complex) were significantly altered in COVID-19 admission samples in all severity groups. Elevated alternative pathway activation markers (Ba and iC3b) and decreased alternative pathway regulator (properdin) in admission samples were associated with more severe disease and risk of death. Levels of most complement biomarkers were reduced in severe disease, consistent with consumption and tissue deposition. Latent class mixed modelling and cumulative incidence analysis identified the trajectory of increase of Ba to be a strong predictor of peak COVID-19 disease severity and death. The data demonstrate that early-onset, uncontrolled activation of complement, driven by sustained and progressive amplification through the alternative pathway amplification loop is a ubiquitous feature of COVID-19, further exacerbated in severe disease. These findings provide novel insights into COVID-19 immunopathogenesis and inform strategies for therapeutic intervention.


Asunto(s)
COVID-19 , Humanos , Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Complemento C3b , Biomarcadores , Progresión de la Enfermedad , Vía Alternativa del Complemento
5.
Kidney Int ; 97(6): 1260-1274, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32386968

RESUMEN

Recessive mutations in diacylglycerol kinase epsilon (DGKE) display genetic pleiotropy, with pathological features reported as either thrombotic microangiopathy or membranoproliferative glomerulonephritis (MPGN), and clinical features of atypical hemolytic uremic syndrome (aHUS), nephrotic syndrome or both. Pathophysiological mechanisms and optimal management strategies have not yet been defined. In prospective and retrospective studies of aHUS referred to the United Kingdom National aHUS service and prospective studies of MPGN referred to the National Registry of Rare Kidney Diseases for MPGN we defined the incidence of DGKE aHUS as 0.009/million/year and so-called DGKE MPGN as 0.006/million/year, giving a combined incidence of 0.015/million/year. Here, we describe a cohort of sixteen individuals with DGKE nephropathy. One presented with isolated nephrotic syndrome. Analysis of pathological features reveals that DGKE mutations give an MPGN-like appearance to different extents, with but more often without changes in arterioles or arteries. In 15 patients presenting with aHUS, ten had concurrent substantial proteinuria. Identified triggering events were rare but coexistent developmental disorders were seen in six. Nine with aHUS experienced at least one relapse, although in only one did a relapse of aHUS occur after age five years. Persistent proteinuria was seen in the majority of cases. Only two individuals have reached end stage renal disease, 20 years after the initial presentation, and in one, renal transplantation was successfully undertaken without relapse. Six individuals received eculizumab. Relapses on treatment occurred in one individual. In four individuals eculizumab was withdrawn, with one spontaneously resolving aHUS relapse occurring. Thus we suggest that DGKE-mediated aHUS is eculizumab non-responsive and that in individuals who currently receive eculizumab therapy it can be safely withdrawn. This has important patient safety and economic implications.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Diacilglicerol Quinasa , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/epidemiología , Síndrome Hemolítico Urémico Atípico/genética , Preescolar , Diacilglicerol Quinasa/genética , Humanos , Estudios Prospectivos , Estudios Retrospectivos , Reino Unido
6.
Immunology ; 155(3): 396-403, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29981529

RESUMEN

C5 plays a major role in complement activation; C5 convertase cleaves C5 into the pro-inflammatory C5a, and C5b, the nidus for the formation of the lytic membrane attack complex. C5 is a major target for anti-complement drugs, necessitating better methods for the study of C5 function. Purification of C5 is complicated; classical methods involve precipitation or pH shifts that result in functional loss and low yield. We here present a method for C5 purification using a novel anti-C5 monoclonal antibody (mAb); RO7112689 (C5i mAb, SKY59), pH-switch engineered to induce antibody-antigen dissociation in the acidic endosome (~ pH 5·5). RO7112689 was bound on an affinity column; applied serum was completely depleted of C5. Elution at pH 5 produced fully active C5 at 98% yield. The mAb also bound C5b in the C5b6 complex, preventing C5b6 binding to target membranes and enabling purification of C5b6 from activated serum. RO7112689 inhibited C5 in mouse serum and efficiently purified mouse C5. Used as capture, RO7112689 produced sensitive and specific assays for human and mouse C5. This novel antibody enables efficient production of intact, fully active, pure human and mouse C5, and quantification of C5 in these species. The demonstration that RO7112689 binds C5b6 adds an additional mechanism of membrane attack complex inhibition by this mAb.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/química , Complemento C5 , Proteínas del Sistema Complemento , Animales , Cromatografía de Afinidad/métodos , Complemento C5/química , Complemento C5/inmunología , Complemento C5/aislamiento & purificación , Proteínas del Sistema Complemento/química , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/aislamiento & purificación , Humanos , Concentración de Iones de Hidrógeno , Ratones , Especificidad de la Especie
7.
Proc Natl Acad Sci U S A ; 112(46): 14319-24, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578778

RESUMEN

Activation of complement is a key determinant of neuropathology and disability after traumatic brain injury (TBI), and inhibition is neuroprotective. However, systemic complement is essential to fight infections, a critical complication of TBI. We describe a targeted complement inhibitor, comprising complement receptor of the Ig superfamily (CRIg) fused with complement regulator CD59a, designed to inhibit membrane attack complex (MAC) assembly at sites of C3b/iC3b deposition. CRIg and CD59a were linked via the IgG2a hinge, yielding CD59-2a-CRIg dimer with increased iC3b/C3b binding avidity and MAC inhibitory activity. CD59-2a-CRIg inhibited MAC formation and prevented complement-mediated lysis in vitro. CD59-2a-CRIg dimer bound C3b-coated surfaces with submicromolar affinity (KD). In experimental TBI, CD59-2a-CRIg administered posttrauma homed to sites of injury and significantly reduced MAC deposition, microglial accumulation, mitochondrial stress, and axonal damage and enhanced neurologic recovery compared with placebo controls. CD59-2a-CRIg inhibited MAC-induced inflammasome activation and IL-1ß production in microglia. Given the important anti-infection roles of complement opsonization, site-targeted inhibition of MAC should be considered to promote recovery postneurotrauma.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Antígenos CD59/farmacología , Receptores de Complemento , Animales , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Antígenos CD59/genética , Complemento C3b/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Ratones , Microglía/metabolismo , Microglía/patología , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
8.
J Immunol ; 194(7): 3029-34, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25725109

RESUMEN

Complement is implicated in the pathogenesis of rheumatoid arthritis (RA); elevated levels of complement activation products have been measured in plasma, synovial fluid, and synovial tissues of patients. Complement polymorphisms are associated with RA in genome-wide association studies. Coding-region polymorphisms may directly impact protein activity; indeed, we have shown that complement polymorphisms affecting a single amino acid change cause subtle changes in individual component function that in combination have dramatic effects on complement activity and disease risk. In this study, we explore the functional consequences of a single nucleotide polymorphism (SNP) (rs17611) encoding a V802I polymorphism in C5 and propose a mechanism for its link to RA pathology. Plasma levels of C5, C5a, and terminal complement complex were measured in healthy and RA donors and correlated to rs17611 polymorphic status. Impact of the SNP on C5 functionality was assessed. Plasma C5a levels were significantly increased and C5 levels significantly lower with higher copy number of the RA risk allele for rs17611, suggesting increased turnover of C5 V802. Functional assays using purified C5 variants revealed no significant differences in lytic activity, suggesting that increased C5 V802 turnover was not mediated by complement convertase enzymes. C5 is also cleaved in vivo by proteases; the C5 V802 variant was more sensitive to cleavage with elastase and the "C5a" generated was biologically active. We hypothesize that this SNP in C5 alters the rate at which elastase generates active C5a in rheumatoid joints, hence recruiting neutrophils to the site thus maintaining a state of inflammation in arthritic joints.


Asunto(s)
Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Complemento C5/genética , Polimorfismo Genético , Alelos , Activación de Complemento/genética , Activación de Complemento/inmunología , Complemento C5/inmunología , Complemento C5/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/inmunología , Citotoxicidad Inmunológica , Frecuencia de los Genes , Estudios de Asociación Genética , Genotipo , Humanos , Hidrólisis , Elastasa de Leucocito/metabolismo , Polimorfismo de Nucleótido Simple
9.
J Am Soc Nephrol ; 27(6): 1617-24, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26490391

RESUMEN

The regulators of complement activation cluster at chromosome 1q32 contains the complement factor H (CFH) and five complement factor H-related (CFHR) genes. This area of the genome arose from several large genomic duplications, and these low-copy repeats can cause genome instability in this region. Genomic disorders affecting these genes have been described in atypical hemolytic uremic syndrome, arising commonly through nonallelic homologous recombination. We describe a novel CFH/CFHR3 hybrid gene secondary to a de novo 6.3-kb deletion that arose through microhomology-mediated end joining rather than nonallelic homologous recombination. We confirmed a transcript from this hybrid gene and showed a secreted protein product that lacks the recognition domain of factor H and exhibits impaired cell surface complement regulation. The fact that the formation of this hybrid gene arose as a de novo event suggests that this cluster is a dynamic area of the genome in which additional genomic disorders may arise.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Proteínas Sanguíneas/genética , Activación de Complemento/genética , Eliminación de Gen , Animales , Células Cultivadas , Factor H de Complemento/genética , Humanos , Ovinos
10.
J Immunol ; 192(10): 4844-51, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24729617

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , C3 Convertasa de la Vía Alternativa del Complemento/inmunología , Convertasas de Complemento C3-C5/inmunología , Factor B del Complemento/inmunología , Vía Alternativa del Complemento/inmunología , Enfermedades Renales/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , C3 Convertasa de la Vía Alternativa del Complemento/antagonistas & inhibidores , Vía Alternativa del Complemento/efectos de los fármacos , Humanos , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología
11.
Proc Natl Acad Sci U S A ; 110(12): 4685-90, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23487775

RESUMEN

The complement system is a key component regulation influences susceptibility to age-related macular degeneration, meningitis, and kidney disease. Variation includes genomic rearrangements within the complement factor H-related (CFHR) locus. Elucidating the mechanism underlying these associations has been hindered by the lack of understanding of the biological role of CFHR proteins. Here we present unique structural data demonstrating that three of the CFHR proteins contain a shared dimerization motif and that this hitherto unrecognized structural property enables formation of both homodimers and heterodimers. Dimerization confers avidity for tissue-bound complement fragments and enables these proteins to efficiently compete with the physiological complement inhibitor, complement factor H (CFH), for ligand binding. Our data demonstrate that these CFHR proteins function as competitive antagonists of CFH to modulate complement activation in vivo and explain why variation in the CFHRs predisposes to disease.


Asunto(s)
Activación de Complemento/fisiología , Proteínas del Sistema Complemento , Dimerización , Sitios Genéticos , Secuencias de Aminoácidos , Proteínas del Sistema Complemento/química , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/metabolismo , Humanos , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Colapso de la Estructura
12.
PLoS Pathog ; 9(4): e1003323, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23637608

RESUMEN

Many pathogens express a surface protein that binds the human complement regulator factor H (FH), as first described for Streptococcus pyogenes and the antiphagocytic M6 protein. It is commonly assumed that FH recruited to an M protein enhances virulence by protecting the bacteria against complement deposition and phagocytosis, but the role of FH-binding in S. pyogenes pathogenesis has remained unclear and controversial. Here, we studied seven purified M proteins for ability to bind FH and found that FH binds to the M5, M6 and M18 proteins but not the M1, M3, M4 and M22 proteins. Extensive immunochemical analysis indicated that FH binds solely to the hypervariable region (HVR) of an M protein, suggesting that selection has favored the ability of certain HVRs to bind FH. These FH-binding HVRs could be studied as isolated polypeptides that retain ability to bind FH, implying that an FH-binding HVR represents a distinct ligand-binding domain. The isolated HVRs specifically interacted with FH among all human serum proteins, interacted with the same region in FH and showed species specificity, but exhibited little or no antigenic cross-reactivity. Although these findings suggested that FH recruited to an M protein promotes virulence, studies in transgenic mice did not demonstrate a role for bound FH during acute infection. Moreover, phagocytosis tests indicated that ability to bind FH is neither sufficient nor necessary for S. pyogenes to resist killing in whole human blood. While these data shed new light on the HVR of M proteins, they suggest that FH-binding may affect S. pyogenes virulence by mechanisms not assessed in currently used model systems.


Asunto(s)
Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/patogenicidad , Animales , Sitios de Unión , Proteína de Unión al Complemento C4b/metabolismo , Factor H de Complemento/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagocitosis , Unión Proteica , Estructura Terciaria de Proteína , Especificidad de la Especie , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo
13.
Trends Immunol ; 33(10): 513-21, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22749446

RESUMEN

Complement is a key component of immune defence against infection; it potently drives inflammation at sites of pathology and is essential for killing of pathogens. Genetic linkage of common complement polymorphisms to disease has advanced the concept that subtle changes in complement activity significantly affect disease risk. Functional analyses of disease-linked polymorphic variants demonstrate that, although individual polymorphisms cause only small changes in activity, when combined, the aggregate effects are large. The inherited set of common variants, the complotype, thus has a major impact on susceptibility to inflammatory and infectious diseases. Assessing the complotype of an individual will aid prediction of disease risk and inform intervention to reduce or eliminate risk.


Asunto(s)
Infecciones/inmunología , Inflamación/inmunología , Animales , Genotipo , Humanos , Infecciones/genética , Inflamación/genética , Polimorfismo Genético , Factores de Riesgo
14.
J Am Soc Nephrol ; 25(11): 2425-33, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24722444

RESUMEN

Complement C3 activation is a characteristic finding in membranoproliferative GN (MPGN). This activation can be caused by immune complex deposition or an acquired or inherited defect in complement regulation. Deficiency of complement factor H has long been associated with MPGN. More recently, heterozygous genetic variants have been reported in sporadic cases of MPGN, although their functional significance has not been assessed. We describe a family with MPGN and acquired partial lipodystrophy. Although C3 nephritic factor was shown in family members with acquired partial lipodystrophy, it did not segregate with the renal phenotype. Genetic analysis revealed a novel heterozygous mutation in complement factor H (R83S) in addition to known risk polymorphisms carried by individuals with MPGN. Patients with MPGN had normal levels of factor H, and structural analysis of the mutant revealed only subtle alterations. However, functional analysis revealed profoundly reduced C3b binding, cofactor activity, and decay accelerating activity leading to loss of regulation of the alternative pathway. In summary, this family showed a confluence of common and rare functionally significant genetic risk factors causing disease. Data from our analysis of these factors highlight the role of the alternative pathway of complement in MPGN.


Asunto(s)
Factor H de Complemento/deficiencia , Factor H de Complemento/genética , Vía Alternativa del Complemento/genética , Eritrocitos/inmunología , Glomerulonefritis Membranoproliferativa/genética , Glomerulonefritis Membranoproliferativa/inmunología , Enfermedades Renales/genética , Animales , Factor H de Complemento/química , Factor H de Complemento/inmunología , Vía Alternativa del Complemento/inmunología , Cristalografía por Rayos X , Eritrocitos/citología , Salud de la Familia , Femenino , Haplotipos , Enfermedades por Deficiencia de Complemento Hereditario , Heterocigoto , Humanos , Enfermedades Renales/inmunología , Masculino , Linaje , Polimorfismo Genético , Estructura Terciaria de Proteína , Ovinos , Relación Estructura-Actividad
15.
Blood ; 119(2): 591-601, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-22058112

RESUMEN

Genomic disorders affecting the genes encoding factor H (fH) and the 5 factor H related proteins have been described in association with atypical hemolytic uremic syndrome. These include deletions of CFHR3, CFHR1, and CFHR4 in association with fH autoantibodies and the formation of a hybrid CFH/CFHR1 gene. These occur through nonallelic homologous recombination secondary to the presence of large segmental duplications (macrohomology) in this region. Using multiplex ligation-dependent probe amplification to screen for such genomic disorders, we have identified a large atypical hemolytic uremic syndrome family where a deletion has occurred through microhomology-mediated end joining rather than nonallelic homologous recombination. In the 3 affected persons of this family, we have shown that the deletion results in formation of a CFH/CFHR3 gene. We have shown that the protein product of this is a 24 SCR protein that is secreted with normal fluid-phase activity but marked loss of complement regulation at cell surfaces despite increased heparin binding. In this study, we have therefore shown that microhomology in this area of chromosome 1 predisposes to disease associated genomic disorders and that the complement regulatory function of fH at the cell surface is critically dependent on the structural integrity of the whole molecule.


Asunto(s)
Apolipoproteínas/genética , Proteínas Sanguíneas/genética , Proteínas Inactivadoras del Complemento C3b/genética , Factor H de Complemento/genética , Eliminación de Gen , Predisposición Genética a la Enfermedad , Síndrome Hemolítico-Urémico/genética , Animales , Apolipoproteínas/metabolismo , Síndrome Hemolítico Urémico Atípico , Autoanticuerpos , Secuencia de Bases , Proteínas Sanguíneas/metabolismo , Western Blotting , Activación de Complemento , Proteínas Inactivadoras del Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Eritrocitos/metabolismo , Hemólisis , Síndrome Hemolítico-Urémico/metabolismo , Síndrome Hemolítico-Urémico/patología , Recombinación Homóloga , Humanos , Datos de Secuencia Molecular , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/metabolismo , Mutación/genética , Linaje , Homología de Secuencia de Ácido Nucleico , Ovinos , Resonancia por Plasmón de Superficie
16.
Proc Natl Acad Sci U S A ; 108(32): 13236-40, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21788512

RESUMEN

Activation of C3, deposition of C3b on the target surface, and subsequent amplification by formation of a C3-cleaving enzyme (C3-convertase; C3bBb) triggers the effector functions of complement that result in inflammation and cell lysis. Concurrently, surface-bound C3b is proteolyzed to iC3b by factor I and appropriate cofactors. iC3b then interacts with the complement receptors (CR) of the Ig superfamily, CR2 (CD21), CR3 (CD11b/CD18), and CR4 (CD11c/CD18) on leukocytes, down-modulating inflammation, enhancing B cell-mediated immunity, and targeting pathogens for clearance by phagocytosis. Using EM and small-angle X-ray scattering, we now present a medium-resolution structure of iC3b (24 Å). iC3b displays a unique conformation with structural features distinct from any other C3 fragment. The macroglobulin ring in iC3b is similar to that in C3b, whereas the TED (thioester-containing domain) domain and the remnants of the CUB (complement protein subcomponents C1r/C1s, urchin embryonic growth factor and bone morphogenetic protein 1) domain have moved to locations more similar to where they were in native C3. A consequence of this large conformational change is the disruption of the factor B binding site, which renders iC3b unable to assemble a C3-convertase. This structural model also justifies the decreased interaction between iC3b and complement regulators and the recognition of iC3b by the CR of the Ig superfamily, CR2, CR3, and CR4. These data further illustrate the extraordinary conformational versatility of C3 to accommodate a great diversity of functional activities.


Asunto(s)
Complemento C3b/química , Complemento C3b/ultraestructura , Microscopía Electrónica , Complemento C3b/aislamiento & purificación , Humanos , Modelos Moleculares , Estructura Terciaria de Proteína , Receptores de Complemento 3b/química , Dispersión del Ángulo Pequeño , Soluciones , Difracción de Rayos X
17.
Proc Natl Acad Sci U S A ; 108(21): 8761-6, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21555552

RESUMEN

Common polymorphisms in complement alternative pathway (AP) proteins C3 (C3(R102G)), factor B (fB(R32Q)), and factor H (fH(V62I)) are associated with age-related macular degeneration (AMD) and other pathologies. Our published work showed that fB(R32Q) influences C3 convertase formation, whereas fH(V62I) affects factor I cofactor activity. Here we show how C3(R102G) (C3S/F) influences AP activity. In hemolysis assays, C3(102G) activated AP more efficiently (EC(50) C3(102G): 157 nM; C3(102R): 191 nM; P < 0.0001). fB binding kinetics and convertase stability were identical, but native and recombinant fH bound more strongly to C3b(102R) (K(D) C3b(102R): 1.0 µM; C3b(102G): 1.4 µM; P < 0.0001). Accelerated decay was unaltered, but fH cofactor activity was reduced for C3b(102G), favoring AP amplification. Combining disease "risk" variants (C3(102G), fB(32R), and fH(62V)) in add-back assays yielded sixfold higher hemolytic activity compared with "protective" variants (C3(102R), fB(32Q), and fH(62I); P < 0.0001). These data introduce the concept of a functional complotype (combination of polymorphisms) defining complement activity in an individual, thereby influencing susceptibility to AP-driven disease.


Asunto(s)
Activación de Complemento/genética , Complemento C3/genética , Factor B del Complemento/genética , Factor H de Complemento/genética , Susceptibilidad a Enfermedades/inmunología , Polimorfismo Genético/inmunología , Convertasas de Complemento C3-C5/metabolismo , Fibrinógeno/metabolismo , Humanos , Degeneración Macular , Unión Proteica , Factores de Riesgo
18.
Proc Natl Acad Sci U S A ; 108(31): 12839-44, 2011 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-21768352

RESUMEN

The complement system is a key component of innate and adaptive immune responses. Complement regulation is critical for prevention and control of disease. We have determined the crystal structure of the complement regulatory enzyme human factor I (fI). FI is in a proteolytically inactive form, demonstrating that it circulates in a zymogen-like state despite being fully processed to the mature sequence. Mapping of functional data from mutants of fI onto the structure suggests that this inactive form is maintained by the noncatalytic heavy-chain allosterically modulating activity of the light chain. Once the ternary complex of fI, a cofactor and a substrate is formed, the allosteric inhibition is released, and fI is oriented for cleavage. In addition to explaining how circulating fI is limited to cleaving only C3b/C4b, our model explains the molecular basis of disease-associated polymorphisms in fI and its cofactors.


Asunto(s)
Factor I de Complemento/química , Factor I de Complemento/genética , Polimorfismo Genético , Estructura Terciaria de Proteína , Regulación Alostérica , Sitios de Unión/genética , Dominio Catalítico , Complemento C3b/química , Complemento C3b/metabolismo , Complemento C4b/química , Complemento C4b/metabolismo , Factor I de Complemento/metabolismo , Cristalización , Cristalografía por Rayos X , Precursores Enzimáticos/química , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Predisposición Genética a la Enfermedad/genética , Glicosilación , Humanos , Modelos Moleculares , Mutación , Unión Proteica
19.
Kidney Int ; 84(6): 1079-89, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24172683

RESUMEN

C3 glomerulopathy is a recently introduced pathological entity whose original definition was glomerular pathology characterized by C3 accumulation with absent or scanty immunoglobulin deposition. In August 2012, an invited group of experts (comprising the authors of this document) in renal pathology, nephrology, complement biology, and complement therapeutics met to discuss C3 glomerulopathy in the first C3 Glomerulopathy Meeting. The objectives were to reach a consensus on: the definition of C3 glomerulopathy, appropriate complement investigations that should be performed in these patients, and how complement therapeutics should be explored in the condition. This meeting report represents the current consensus view of the group.


Asunto(s)
Complemento C3/análisis , Glomerulonefritis/inmunología , Glomérulos Renales/inmunología , Investigación Biomédica , Biopsia , Conducta Cooperativa , Glomerulonefritis/diagnóstico , Glomerulonefritis/terapia , Humanos , Cooperación Internacional , Glomérulos Renales/patología , Valor Predictivo de las Pruebas , Pronóstico
20.
Am J Kidney Dis ; 62(5): 978-83, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23870792

RESUMEN

We report a male infant who presented at 8 months of age with atypical hemolytic uremic syndrome (aHUS) responsive to plasma therapy. Investigation showed him to have complement factor H (CFH) deficiency associated with a homozygous CFH mutation (c.2880delT [p.Phe960fs]). Mutation screening of the child's parents revealed that the father was heterozygous for this change but that it was not present in his mother. Chromosome 1 uniparental isodisomy of paternal origin was confirmed by genotyping chromosome 1 SNPs. CD46 SNP genotyping was undertaken in this individual and another patient with CFH deficiency associated with chromosome 1 uniparental isodisomy. This showed a homozygous aHUS risk haplotype (CD46GGAAC) in the patient with aHUS and a homozygous glomerulonephritis risk haplotype (CD46AAGGT) in the patient with endocapillary glomerulonephritis. We also showed that FHL-1 (factor H-like protein 1) was present in the patient with aHUS and absent in the patient with glomerulonephritis. This study emphasizes that modifiers such as CD46 and FHL-1 may determine the kidney phenotype of patients who present with homozygous CFH deficiency.


Asunto(s)
Factor H de Complemento/deficiencia , Genotipo , Síndrome Hemolítico-Urémico/genética , Enfermedades Renales/genética , Fenotipo , Disomía Uniparental/genética , Síndrome Hemolítico Urémico Atípico , Factor H de Complemento/genética , Haplotipos/genética , Enfermedades por Deficiencia de Complemento Hereditario , Homocigoto , Humanos , Lactante , Masculino , Proteína Cofactora de Membrana/genética , Mutación/genética , Polimorfismo de Nucleótido Simple/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA