Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Am Soc Nephrol ; 29(6): 1649-1661, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29588430

RESUMEN

Background C3 glomerulopathy (C3G) is associated with dysregulation of the alternative pathway of complement activation, and treatment options for C3G remain limited. Complement factor H (FH) is a potent regulator of the alternative pathway and might offer a solution, but the mass and complexity of FH makes generation of full-length FH far from trivial. We previously generated a mini-FH construct, with FH short consensus repeats 1-5 linked to repeats 18-20 (FH1-5^18-20), that was effective in experimental C3G. However, the serum t1/2 of FH1-5^18-20 was significantly shorter than that of serum-purified FH.Methods We introduced the oligomerization domain of human FH-related protein 1 (denoted by R1-2) at the carboxy or amino terminus of human FH1-5^18-20 to generate two homodimeric mini-FH constructs (FHR1-2^1-5^18-20 and FH1-5^18-20^R1-2, respectively) in Chinese hamster ovary cells and tested these constructs using binding, fluid-phase, and erythrocyte lysis assays, followed by experiments in FH-deficient Cfh-/- mice.Results FHR1-2^1-5^18-20 and FH1-5^18-20^R1-2 homodimerized in solution and displayed avid binding profiles on clustered C3b surfaces, particularly FHR1-2^1-5^18-20 Each construct was >10-fold more effective than FH at inhibiting cell surface complement activity in vitro and restricted glomerular basement membrane C3 deposition in vivo significantly better than FH or FH1-5^18-20 FH1-5^18-20^R1-2 had a C3 breakdown fragment binding profile similar to that of FH, a >5-fold increase in serum t1/2 compared with that of FH1-5^18-20, and significantly better retention in the kidney than FH or FH1-5^18-20Conclusions FH1-5^18-20^R1-2 may have utility as a treatment option for C3G or other complement-mediated diseases.


Asunto(s)
Complemento C3/metabolismo , Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Factor H de Complemento/farmacocinética , Glomerulonefritis Membranoproliferativa/metabolismo , Animales , Factor H de Complemento/síntesis química , Factor H de Complemento/genética , Vía Alternativa del Complemento , Cricetinae , Membrana Basal Glomerular/metabolismo , Glomerulonefritis Membranoproliferativa/tratamiento farmacológico , Semivida , Ratones , Unión Proteica , Ingeniería de Proteínas
2.
J Biol Chem ; 292(32): 13345-13360, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28637873

RESUMEN

Spontaneous activation enables the complement system to respond very rapidly to diverse threats. This activation is efficiently suppressed by complement factor H (CFH) on self-surfaces but not on foreign surfaces. The surface selectivity of CFH, a soluble protein containing 20 complement-control protein modules (CCPs 1-20), may be compromised by disease-linked mutations. However, which of the several functions of CFH drives this self-surface selectivity remains unknown. To address this, we expressed human CFH mutants in Pichia pastoris We found that recombinant I62-CFH (protective against age-related macular degeneration) and V62-CFH functioned equivalently, matching or outperforming plasma-derived CFH, whereas R53H-CFH, linked to atypical hemolytic uremic syndrome (aHUS), was defective in C3bBb decay-accelerating activity (DAA) and factor I cofactor activity (CA). The aHUS-linked CCP 19 mutant D1119G-CFH had virtually no CA on (self-like) sheep erythrocytes (ES) but retained DAA. The aHUS-linked CCP 20 mutant S1191L/V1197A-CFH (LA-CFH) had dramatically reduced CA on ES but was less compromised in DAA. D1119G-CFH and LA-CFH both performed poorly at preventing complement-mediated hemolysis of ES PspCN, a CFH-binding Streptococcus pneumoniae protein domain, binds CFH tightly and increases accessibility of CCPs 19 and 20. PspCN did not improve the DAA of any CFH variant on ES Conversely, PspCN boosted the CA, on ES, of I62-CFH, R53H-CFH, and LA-CFH and also enhanced hemolysis protection by I62-CFH and LA-CFH. We conclude that CCPs 19 and 20 are critical for efficient CA on self-surfaces but less important for DAA. Exposing CCPs 19 and 20 with PspCN and thus enhancing CA on self-surfaces may reverse deficiencies of some CFH variants.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Activación de Complemento , Degeneración Macular/genética , Mutación , Sustitución de Aminoácidos , Animales , Síndrome Hemolítico Urémico Atípico/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , C3 Convertasa de la Vía Alternativa del Complemento/química , C3 Convertasa de la Vía Alternativa del Complemento/genética , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , Complemento C3d/química , Complemento C3d/genética , Complemento C3d/metabolismo , Factor H de Complemento/química , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Factor I de Complemento/química , Factor I de Complemento/genética , Factor I de Complemento/metabolismo , Eritrocitos/química , Hemólisis , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/genética , Proteínas Inmovilizadas/metabolismo , Degeneración Macular/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Oveja Doméstica , Solubilidad , Streptococcus pneumoniae/metabolismo , Propiedades de Superficie
3.
Nat Chem Biol ; 11(1): 77-82, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25402769

RESUMEN

The serum protein complement factor H (FH) ensures downregulation of the complement alternative pathway, a branch of innate immunity, upon interaction with specific glycans on host cell surfaces. Using ligand-based NMR, we screened a comprehensive set of sialylated glycans for binding to FH and solved the crystal structure of a ternary complex formed by the two C-terminal domains of FH, a sialylated trisaccharide and the complement C3b thioester-containing domain. Key residues in the sialic acid binding site are conserved from mice to men, and residues linked to atypical hemolytic uremic syndrome cluster within this binding site, suggesting a possible role for sialic acid as a host marker also in other mammals and a critical role in human renal complement homeostasis. Unexpectedly, the FH sialic acid binding site is structurally homologous to the binding sites of two evolutionarily unrelated proteins. The crystal structure also advances our understanding of bacterial immune evasion strategies.


Asunto(s)
Factor H de Complemento/química , Ácido N-Acetilneuramínico/química , Animales , Sitios de Unión , Secuencia de Carbohidratos , Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Vía Alternativa del Complemento/efectos de los fármacos , Secuencia Conservada , Hemólisis/efectos de los fármacos , Síndrome Hemolítico-Urémico/genética , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Ácido N-Acetilneuramínico/metabolismo , Polisacáridos/farmacología , Ovinos
4.
J Immunol ; 195(10): 4986-98, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26459349

RESUMEN

In an attempt to evade annihilation by the vertebrate complement system, many microbes capture factor H (FH), the key soluble complement-regulating protein in human plasma. However, FH is normally an active complement suppressor exclusively on self-surfaces and this selective action of FH is pivotal to self versus non-self discrimination by the complement system. We investigated whether the bacterially captured FH becomes functionally enhanced and, if so, how this is achieved at a structural level. We found, using site-directed and truncation mutagenesis, surface plasmon resonance, nuclear magnetic resonance spectroscopy, and cross-linking and mass spectrometry, that the N-terminal domain of Streptococcus pneumoniae protein PspC (PspCN) not only binds FH extraordinarily tightly but also holds it in a previously uncharacterized conformation. Functional enhancement arises from exposure of a C-terminal cryptic second binding site in FH for C3b, the activation-specific fragment of the pivotal complement component, C3. This conformational change of FH doubles its affinity for C3b and increases 5-fold its ability to accelerate decay of the binary enzyme (C3bBb) responsible for converting C3 to C3b in an amplification loop. Despite not sharing critical FH-binding residues, PspCNs from D39 and Tigr4 S. pneumoniae exhibit similar FH-anchoring and enhancing properties. We propose that these bacterial proteins mimic molecular markers of self-surfaces, providing a compelling hypothesis for how FH prevents complement-mediated injury to host tissue while lacking efficacy on virtually all other surfaces. In hemolysis assays with 2-aminoethylisothiouronium bromide-treated erythrocytes that recapitulate paroxysmal nocturnal hemoglobinuria, PspCN enhanced protection of cells by FH, suggesting a new paradigm for therapeutic complement suppression.


Asunto(s)
Proteínas Bacterianas/química , Complemento C3b/química , Factor H de Complemento/química , Streptococcus pneumoniae/química , Proteínas Bacterianas/inmunología , Complemento C3b/inmunología , Factor H de Complemento/inmunología , Hemoglobinuria Paroxística/inmunología , Humanos , Estructura Terciaria de Proteína , Streptococcus pneumoniae/inmunología
5.
Biochem Soc Trans ; 43(5): 812-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26517887

RESUMEN

Complement control protein modules (CCPs) occur in numerous functionally diverse extracellular proteins. Also known as short consensus repeats (SCRs) or sushi domains each CCP contains approximately 60 amino acid residues, including four consensus cysteines participating in two disulfide bonds. Varying in length and sequence, CCPs adopt a ß-sandwich type fold and have an overall prolate spheroidal shape with N- and C-termini lying close to opposite poles of the long axis. CCP-containing proteins are important as cytokine receptors and in neurotransmission, cell adhesion, blood clotting, extracellular matrix formation, haemoglobin metabolism and development, but CCPs are particularly well represented in the vertebrate complement system. For example, factor H (FH), a key soluble regulator of the alternative pathway of complement activation, is made up entirely from a chain of 20 CCPs joined by short linkers. Collectively, therefore, the 20 CCPs of FH must mediate all its functional capabilities. This is achieved via collaboration and division of labour among these modules. Structural studies have illuminated the dynamic architectures that allow FH and other CCP-rich proteins to perform their biological functions. These are largely the products of a highly varied set of intramolecular interactions between CCPs. The CCP can act as building block, spacer, highly versatile recognition site or dimerization mediator. Tandem CCPs may form composite binding sites or contribute to flexible, rigid or conformationally 'switchable' segments of the parent proteins.


Asunto(s)
Enzimas Activadoras de Complemento/química , Activación de Complemento , Proteínas Inactivadoras de Complemento/química , Diseño de Fármacos , Modelos Moleculares , Ingeniería de Proteínas , Animales , Sitios de Unión , Enzimas Activadoras de Complemento/genética , Enzimas Activadoras de Complemento/metabolismo , Factor H de Complemento/química , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Inactivadores del Complemento/química , Inactivadores del Complemento/metabolismo , Inactivadores del Complemento/farmacología , Proteínas Inactivadoras de Complemento/genética , Proteínas Inactivadoras de Complemento/metabolismo , Humanos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología
6.
J Immunol ; 190(5): 2049-57, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23365078

RESUMEN

Complement factor H (CFH) regulates complement activation in host tissues through its recognition of polyanions, which mediate CFH binding to host cell surfaces and extracellular matrix, promoting the deactivation of deposited C3b. These polyanions include heparan sulfate (HS), a glycosaminoglycan with a highly diverse range of structures, for which two regions of CFH (CCP6-8 and CCP19-20) have been implicated in HS binding. Mutations/polymorphisms within these glycosaminoglycan-binding sites have been associated with age-related macular degeneration (AMD) and atypical hemolytic uremic syndrome. In this study, we demonstrate that CFH has tissue-specific binding properties mediated through its two HS-binding regions. Our data show that the CCP6-8 region of CFH binds more strongly to heparin (a highly sulfated form of HS) than CCP19-20, and that their sulfate specificities are different. Furthermore, the HS binding site in CCP6-8, which is affected by the AMD-associated Y402H polymorphism, plays the principal role in host tissue recognition in the human eye, whereas the CCP19-20 region makes the major contribution to the binding of CFH in the human kidney. This helps provide a biochemical explanation for the genetic basis of tissue-specific diseases such as AMD and atypical hemolytic uremic syndrome, and leads to a better understanding of the pathogenic mechanisms for these diseases of complement dysregulation.


Asunto(s)
Factor H de Complemento/genética , Ojo/metabolismo , Síndrome Hemolítico-Urémico/genética , Heparitina Sulfato/metabolismo , Riñón/metabolismo , Degeneración Macular/genética , Adulto , Anciano , Anciano de 80 o más Años , Sustitución de Aminoácidos , Síndrome Hemolítico Urémico Atípico , Autopsia , Sitios de Unión , Activación de Complemento/genética , Factor H de Complemento/química , Factor H de Complemento/metabolismo , Escherichia coli/genética , Ojo/patología , Femenino , Síndrome Hemolítico-Urémico/metabolismo , Síndrome Hemolítico-Urémico/patología , Humanos , Riñón/patología , Degeneración Macular/metabolismo , Degeneración Macular/patología , Masculino , Persona de Mediana Edad , Mutación , Especificidad de Órganos , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
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
8.
J Am Soc Nephrol ; 24(1): 53-65, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23204401

RESUMEN

Factor H (fH) and properdin both modulate complement; however, fH inhibits activation, and properdin promotes activation of the alternative pathway of complement. Mutations in fH associate with several human kidney diseases, but whether inhibiting properdin would be beneficial in these diseases is unknown. Here, we found that either genetic or pharmacological blockade of properdin, which we expected to be therapeutic, converted the mild C3 GN of an fH-mutant mouse to a lethal C3 GN with features of human dense deposit disease. We attributed this phenotypic change to a differential effect of properdin on the dynamics of alternative pathway complement activation in the fluid phase and the cell surface in the fH-mutant mice. Thus, in fH mutation-related C3 glomerulopathy, additional factors that impact the activation of the alternative pathway of complement critically determine the nature and severity of kidney pathology. These results show that therapeutic manipulation of the complement system requires rigorous disease-specific target validation.


Asunto(s)
Glomerulonefritis Membranoproliferativa/genética , Enfermedades Renales/genética , Properdina/deficiencia , Animales , Complemento C3/metabolismo , Factor H de Complemento/deficiencia , Factor H de Complemento/genética , Vía Alternativa del Complemento , Modelos Animales de Enfermedad , Glomerulonefritis Membranoproliferativa/metabolismo , Glomerulonefritis Membranoproliferativa/patología , Enfermedades por Deficiencia de Complemento Hereditario , Humanos , Glomérulos Renales/ultraestructura , Ratones , Ratones Endogámicos C57BL , Mutación
9.
Biochemistry ; 52(23): 3949-62, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23701234

RESUMEN

Factor H (FH) is a soluble regulator of the proteolytic cascade at the core of the evolutionarily ancient vertebrate complement system. Although FH consists of a single chain of similar protein modules, it has a demanding job description. Its chief role is to prevent complement-mediated injury to healthy host cells and tissues. This entails recognition of molecular patterns on host surfaces combined with control of one of nature's most dangerous examples of a positive-feedback loop. In this way, FH modulates, where and when needed, an amplification process that otherwise exponentially escalates the production of the pro-inflammatory, pro-phagocytic, and pro-cytolytic cleavage products of complement proteins C3 and C5. Mutations and single-nucleotide polymorphisms in the FH gene and autoantibodies against FH predispose individuals to diseases, including age-related macular degeneration, dense-deposit disease, and atypical hemolytic uremic syndrome. Moreover, deletions or variations of genes for FH-related proteins also influence the risk of disease. Numerous pathogens hijack FH and use it for self-defense. As reviewed herein, a molecular understanding of FH function is emerging. While its functional oligomeric status remains uncertain, progress has been achieved in characterizing its three-dimensional architecture and, to a lesser extent, its intermodular flexibility. Models are proposed, based on the reconciliation of older data with a wealth of recent evidence, in which a latent circulating form of FH is activated by its principal target, C3b tethered to a self-surface. Such models suggest hypotheses linking sequence variations to pathophysiology, but improved, more quantitative, functional assays and rigorous data analysis are required to test these ideas.


Asunto(s)
Activación de Complemento , Factor H de Complemento/fisiología , Secuencias de Aminoácidos , Sitios de Unión , Complemento C3b/química , Complemento C3b/fisiología , Factor H de Complemento/química , Humanos , Modelos Moleculares , Unión Proteica , Dominios y Motivos de Interacción de Proteínas
10.
J Biol Chem ; 287(45): 38231-43, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22989873

RESUMEN

Calcium-binding protein 7 (CaBP7) is a member of the calmodulin (CaM) superfamily that harbors two high affinity EF-hand motifs and a C-terminal transmembrane domain. CaBP7 has been previously shown to interact with and modulate phosphatidylinositol 4-kinase III-ß (PI4KIIIß) activity in in vitro assays and affects vesicle transport in neurons when overexpressed. Here we show that the N-terminal domain (NTD) of CaBP7 is sufficient to mediate the interaction of CaBP7 with PI4KIIIß. CaBP7 NTD encompasses the two high affinity Ca(2+) binding sites, and structural characterization through multiangle light scattering, circular dichroism, and NMR reveals unique properties for this domain. CaBP7 NTD binds specifically to Ca(2+) but not Mg(2+) and undergoes significant conformational changes in both secondary and tertiary structure upon Ca(2+) binding. The Ca(2+)-bound form of CaBP7 NTD is monomeric and exhibits an open conformation similar to that of CaM. Ca(2+)-bound CaBP7 NTD has a solvent-exposed hydrophobic surface that is more expansive than observed in CaM or CaBP1. Within this hydrophobic pocket, there is a significant reduction in the number of methionine residues that are conserved in CaM and CaBP1 and shown to be important for target recognition. In CaBP7 NTD, these residues are replaced with isoleucine and leucine residues with branched side chains that are intrinsically more rigid than the flexible methionine side chain. We propose that these differences in surface hydrophobicity, charge, and methionine content may be important in determining highly specific interactions of CaBP7 with target proteins, such as PI4KIIIß.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Calcio/metabolismo , Aparato de Golgi/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Western Blotting , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/genética , Bovinos , Dicroismo Circular , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Antígenos de Histocompatibilidad Menor , Modelos Moleculares , Datos de Secuencia Molecular , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Homología de Secuencia de Aminoácido , Soluciones , Propiedades de Superficie
11.
J Exp Med ; 204(10): 2277-83, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17893204

RESUMEN

Nearly 50 million people worldwide suffer from age-related macular degeneration (AMD), which causes severe loss of central vision. A single-nucleotide polymorphism in the gene for the complement regulator factor H (FH), which causes a Tyr-to-His substitution at position 402, is linked to approximately 50% of attributable risks for AMD. We present the crystal structure of the region of FH containing the polymorphic amino acid His402 in complex with an analogue of the glycosaminoglycans (GAGs) that localize the complement regulator on the cell surface. The structure demonstrates direct coordination of ligand by the disease-associated polymorphic residue, providing a molecular explanation of the genetic observation. This glycan-binding site occupies the center of an extended interaction groove on the regulator's surface, implying multivalent binding of sulfated GAGs. This finding is confirmed by structure-based site-directed mutagenesis, nuclear magnetic resonance-monitored binding experiments performed for both H402 and Y402 variants with this and another model GAG, and analysis of an extended GAG-FH complex.


Asunto(s)
Envejecimiento/fisiología , Factor H de Complemento/química , Factor H de Complemento/metabolismo , Sitios de Unión , Factor H de Complemento/genética , Cristalografía por Rayos X , Productos del Gen gag/química , Productos del Gen gag/genética , Productos del Gen gag/metabolismo , Ligandos , Modelos Moleculares , Mutación/genética , Polisacáridos/química , Polisacáridos/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Sacarosa/análogos & derivados , Sacarosa/química , Sacarosa/metabolismo , Propiedades de Superficie
12.
Biochemistry ; 51(9): 1874-84, 2012 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-22320225

RESUMEN

Numerous complement factor H (FH) mutations predispose patients to atypical hemolytic uremic syndrome (aHUS) and other disorders arising from inadequately regulated complement activation. No unifying structural or mechanistic consequences have been ascribed to these mutants beyond impaired self-cell protection. The S1191L and V1197A mutations toward the C-terminus of FH, which occur in patients singly or together, arose from gene conversion between CFH encoding FH and CFHR1 encoding FH-related 1. We show that neither single nor double mutations structurally perturbed recombinant proteins consisting of the FH C-terminal modules, 19 and 20 (FH19-20), although all three FH19-20 mutants were poor, compared to wild-type FH19-20, at promoting hemolysis of C3b-coated erythrocytes through competition with full-length FH. Indeed, our new crystal structure of the S1191L mutant of FH19-20 complexed with an activation-specific complement fragment, C3d, was nearly identical to that of the wild-type FH19-20:C3d complex, consistent with mutants binding to C3b with wild-type-like affinity. The S1191L mutation enhanced thermal stability of module 20, whereas the V1197A mutation dramatically decreased it. Thus, although mutant proteins were folded at 37 °C, they differ in conformational rigidity. Neither single substitutions nor double substitutions increased measurably the extent of FH19-20 self-association, nor did these mutations significantly affect the affinity of FH19-20 for three glycosaminoglycans, despite critical roles of module 20 in recognizing polyanionic self-surface markers. Unexpectedly, FH19-20 mutants containing Leu1191 self-associated on a heparin-coated surface to a higher degree than on surfaces coated with dermatan or chondroitin sulfates. Thus, potentially disease-related functional distinctions between mutants, and between FH and FH-related 1, may manifest in the presence of specific glycosaminoglycans.


Asunto(s)
Factor H de Complemento/química , Factor H de Complemento/genética , Conversión Génica , Complemento C3b/química , Complemento C3d/química , Factor H de Complemento/metabolismo , Cristalografía por Rayos X , Humanos , Mutación , Pichia/genética , Pichia/metabolismo , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Temperatura
13.
Blood ; 115(2): 379-87, 2010 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-19861685

RESUMEN

Factor H autoantibodies have been reported in approximately 10% of patients with atypical hemolytic uremic syndrome (aHUS) and are associated with deficiency of factor H-related proteins 1 and 3. In this study we examined the prevalence of factor H autoantibodies in the Newcastle cohort of aHUS patients, determined whether the presence of such autoantibodies is always associated with deficiency of factor H-related proteins 1 and 3, and examined whether such patients have additional susceptibility factors and/or mutations in the genes encoding complement regulator/activators. We screened 142 patients with aHUS and found factor H autoantibodies in 13 individuals (age 1-11 years). The presence of the autoantibodies was confirmed by Western blotting. By using multiplex ligation-dependent probe amplification we measured complement factor H-related (CFHR)1 and CFHR3 copy number. In 10 of the 13 patients there were 0 copies of CFHR1, and in 3 patients there were 2. In 3 of the patients with 0 copies of CFHR1 there was 1 copy of CFHR3, and these individuals exhibited a novel deletion incorporating CFHR1 and CFHR4. In 5 patients mutations were identified: 1 in CFH, 1 in CFI, 1 in CD46, and 2 in C3. The latter observation emphasizes that multiple concurrent factors may be necessary in individual patients for disease manifestation.


Asunto(s)
Apolipoproteínas/genética , Autoanticuerpos/sangre , Proteínas Sanguíneas/genética , Complemento C3/genética , Proteínas Inactivadoras del Complemento C3b/genética , Factor H de Complemento/genética , Factor I de Complemento/genética , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/genética , Proteína Cofactora de Membrana/genética , Apolipoproteínas/inmunología , Apolipoproteínas/metabolismo , Autoanticuerpos/inmunología , Proteínas Sanguíneas/inmunología , Proteínas Sanguíneas/metabolismo , Niño , Preescolar , Estudios de Cohortes , Complemento C3/inmunología , Complemento C3/metabolismo , Proteínas Inactivadoras del Complemento C3b/inmunología , Proteínas Inactivadoras del Complemento C3b/metabolismo , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Factor I de Complemento/inmunología , Factor I de Complemento/metabolismo , Femenino , Dosificación de Gen , Síndrome Hemolítico-Urémico/inmunología , Humanos , Lactante , Masculino , Proteína Cofactora de Membrana/inmunología , Proteína Cofactora de Membrana/metabolismo , Eliminación de Secuencia
14.
Nucleic Acids Res ; 38(5): 1723-37, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20007596

RESUMEN

Plasmids, conjugative transposons and phage frequently encode anti-restriction proteins to enhance their chances of entering a new bacterial host that is highly likely to contain a Type I DNA restriction and modification (RM) system. The RM system usually destroys the invading DNA. Some of the anti-restriction proteins are DNA mimics and bind to the RM enzyme to prevent it binding to DNA. In this article, we characterize ArdB anti-restriction proteins and their close homologues, the KlcA proteins from a range of mobile genetic elements; including an ArdB encoded on a pathogenicity island from uropathogenic Escherichia coli and a KlcA from an IncP-1b plasmid, pBP136 isolated from Bordetella pertussis. We show that all the ArdB and KlcA act as anti-restriction proteins and inhibit the four main families of Type I RM systems in vivo, but fail to block the restriction endonuclease activity of the archetypal Type I RM enzyme, EcoKI, in vitro indicating that the action of ArdB is indirect and very different from that of the DNA mimics. We also present the structure determined by NMR spectroscopy of the pBP136 KlcA protein. The structure shows a novel protein fold and it is clearly not a DNA structural mimic.


Asunto(s)
Proteínas Bacterianas/química , Desoxirribonucleasas de Localización Especificada Tipo I/antagonistas & inhibidores , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bordetella pertussis/química , Enzimas de Restricción del ADN/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo I/metabolismo , Endopeptidasa Clp/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Pliegue de Proteína , Homología de Secuencia de Aminoácido , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/metabolismo
15.
Curr Eye Res ; 47(7): 1087-1093, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35282732

RESUMEN

PURPOSE: GEM103 is a recombinantly produced full-length version of the human complement factor H (CFH) under clinical investigation for treatment of age-related macular degeneration (AMD) in individuals carrying an AMD risk-associated genetic variant of CFH. This study aimed to investigate the complement pathway-related functions of GEM103 in comparison with those of native human CFH. METHODS: Key biological activities of GEM103 and human serum-derived CFH (sdCFH) were compared using four independent functional assays. Assays of C3b binding and C3 convertase decay-accelerating activity (DAA) were performed by surface plasmon resonance (SPR). Cofactor activity (CA) was measured using 8-anilinonaphthalene-1-sulfonic acid as a fluorescent probe of C3b integrity. The abilities of GEM103 and sdCFH to protect sheep erythrocytes from hemolysis by CFH-depleted normal human serum were assessed colorimetrically. RESULTS: In multiple SPR-based assays of C3b binding and DAA, the performance of GEM103 was consistently comparable to that of sdCFH across a range of matching concentrations. The EC50 ± SD in the fluorescence-based fluid-phase CA assay was 0.21 ± 0.06 µM for GEM103 compared to 0.20 ± 0.09 µM for sdCFH. In hemolysis assays, the EC50 value of 0.33 ± 0.16 µM for GEM103 versus 0.46 ± 0.06 µM for sdCFH were not significantly different (p = 0.81). CONCLUSIONS: GEM103, a recombinant CFH developed by Gemini Therapeutics, shows activity profiles comparable to sdCFH in all complement-related assays employed in this study, suggesting that GEM103 is equivalent to the native glycoprotein in terms of its in vitro functional activity. These results support further study of GEM103 as a potential therapy for AMD.


Asunto(s)
Factor H de Complemento , Degeneración Macular , Animales , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Hemólisis , Humanos , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/genética , Degeneración Macular/metabolismo , Polimorfismo de Nucleótido Simple , Ovinos
16.
Invest Ophthalmol Vis Sci ; 63(12): 30, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36445700

RESUMEN

Purpose: Factor H (FH, encoded by CFH) prevents activation of the complement system's alternative pathway (AP) on host tissues. FH impedes C3 convertase (C3bBb) formation, accelerates C3bBb decay, and is a cofactor for factor I (FI)-catalyzed C3b cleavage. Numerous CFH variants are associated with age-related macular degeneration (AMD), but their functional consequences frequently remain undetermined. Here, we conduct functional comparisons between a control version of FH (not AMD linked) and 21 AMD-linked FH variants. Methods: Recombinantly produced, untagged, full-length FH versions were assayed for binding to C3b and decay acceleration of C3bBb using surface-plasmon resonance, FI-cofactor activity using a fluorescent probe of C3b integrity, suppression of C5b-9 assembly on an AP-activating surface, and inhibition of human AP-mediated lysis of sheep erythrocytes. Results: All versions were successfully purified despite below-average yields for Arg2Thr, Arg53Cys, Arg175Pro, Arg175Gln, Ile221Val, Tyr402His, Pro503Ala, Arg567Gly, Gly1194Asp, and Arg1210Cys. Compared to control FH, Arg2Thr, Leu3Val, Ser58Ala, Asp90Gly, Asp130Asn, Gln400Lys, Tyr402His, Gly650Val, Ser890Ile, and Thr965Met showed minimal functional differences. Arg1210C, Arg53His, Arg175Gln, Gly1194Asp, Pro503Ala, Arg53Cys, Arg576Gly, and Arg175Pro (in order of decreasing efficacy) underperformed, while Ile221Val, Arg303Gln, and Arg303Trp were "marginal." We newly identified variants toward the center of the molecule, Pro503Ala and Arg567Gly, as potentially pathogenic. Conclusions: Our approach could be extended to other variants of uncertain significance and to assays for noncanonical FH activities, aiming to facilitate selection of cohorts most likely to benefit from therapeutic FH. This is timely as recombinant therapeutic FH is in development for intravitreal treatment of AMD in patients with reduced FH functionality.


Asunto(s)
Factor H de Complemento , Degeneración Macular , Animales , Humanos , Aceleración , Factor H de Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento , Proteínas del Sistema Complemento , Degeneración Macular/genética , Ovinos
17.
J Biol Chem ; 285(6): 3766-3776, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19951950

RESUMEN

Apoptotic cells are opsonized by complement components such as C1q and C3b, which increases their susceptibility to phagocytosis. Soluble complement inhibitors such as factor H (fH) also recognize apoptotic cells to minimize the pro-inflammatory effects of downstream complement activation. We used four radiolabeled protein constructs that span different regions of the 20 complement control protein (CCP) modules that make up fH and found that fragments comprising CCPs 6-8, CCPs 8-15, and CCPs 19-20 but not CCPs 1-4, bound to apoptotic Jurkat T cells. There are four possible ligand types on apoptotic cells that could recruit fH: proteins, carbohydrates, lipids, and DNA. We found that CCPs 6-8 of fH bind to annexin-II, a trypsin-insensitive protein that becomes exposed on surfaces of apoptotic cells. The second ligand of fH, which interacts with CCPs 6-8 and 19-20, is DNA. Confocal microscopy showed co-localization of fH with antibodies specific for DNA. fH also binds to histones devoid of DNA, and CCPs 1-4, 6-8, and 8-15 mediate this interaction. Treatment of apoptotic cells with neuraminidase, chondroitinase, heparitinase, and heparinase did not change fH binding. Treatment of apoptotic cells with phospholipase A(2) dramatically increased both binding of fH and cell-surface DNA. We also excluded the possibility that fH interacts with lysophospholipids using surface plasmon resonance and flow cytometry with lipid-coated beads. Identification of annexin-II as one of the fH ligands on apoptotic cells together with the fact that autoantibodies against annexin-II are found in systemic lupus erythematosus provides further insight into understanding the pathogenesis of this disease.


Asunto(s)
Anexina A2/metabolismo , Apoptosis , Membrana Celular/metabolismo , Factor H de Complemento/metabolismo , ADN de Neoplasias/metabolismo , Histonas/metabolismo , Sitios de Unión , Proteína de Unión al Complemento C4b/metabolismo , Citometría de Flujo , Glicosaminoglicanos/metabolismo , Humanos , Inmunoprecipitación , Células Jurkat , Ligandos , Microscopía Confocal , Ácido N-Acetilneuramínico/metabolismo , Necrosis , Fosfolipasas A2/metabolismo , Fosfolipasas A2/farmacología , Unión Proteica/efectos de los fármacos , Resonancia por Plasmón de Superficie
18.
Acta Crystallogr D Biol Crystallogr ; 67(Pt 7): 593-600, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21697597

RESUMEN

The soluble 155 kDa glycoprotein factor H (FH) protects host tissue from damage by the human complement system. It accelerates decay of the alternative-pathway C3 convertase, C3bBb, and is a cofactor for factor I-mediated cleavage of the opsonin C3b. Numerous mutations and single-nucleotide polymorphisms (SNPs) occur in the gene encoding FH and the resulting missense mutations and truncation products result in altered functionality that predisposes to the development of the serious renal condition atypical haemolytic uraemic syndrome (aHUS). Other polymorphisms are linked to membranoproliferative glomerulonephritis and macular degeneration. The two C-terminal modules of FH (FH19-20) harbour numerous aHUS-associated mutations that disrupt the ability of factor H to protect host cells from complement-mediated damage. In this work, the crystal structure of an aHUS-associated T1184R variant of FH19-20 at a resolution of 1.52 Šis described. It is shown that this mutation has negligible structural effects but causes a significant change in the electrostatic surface of these two domains. Mechanisms are discussed by which this mutation may alter FH-ligand interactions, particularly with regard to the extension of a region of this molecule within module 20 that has been associated with the binding of glycosaminoglycans (GAGs) or sialic acid residues.


Asunto(s)
Factor H de Complemento/química , Síndrome Hemolítico-Urémico/genética , Mutación , Síndrome Hemolítico Urémico Atípico , Factor H de Complemento/genética , Cristalografía por Rayos X , Modelos Moleculares , Estructura Terciaria de Proteína
19.
J Immunol ; 182(11): 7009-18, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19454698

RESUMEN

Factor H (fH) is essential for complement homeostasis in fluid-phase and on surfaces. Its two C-terminal domains (CCP 19-20) anchor fH to self-surfaces where it prevents C3b amplification in a process requiring its N-terminal four domains. In atypical hemolytic uremic syndrome (aHUS), mutations clustering toward the C terminus of fH may disrupt interactions with surface-associated C3b or polyanions and thereby diminish the ability of fH to regulate complement. To test this, we compared a recombinant protein encompassing CCP 19-20 with 16 mutants. The mutations had only very limited and localized effects on protein structure. Although we found four aHUS-linked fH mutations that decreased binding to C3b and/or to heparin (a model compound for cell surface polyanionic carbohydrates), we identified five aHUS-associated mutants with increased affinity for either or both ligands. Strikingly, these variable affinities for the individual ligands did not correlate with the extent to which all the aHUS-associated mutants were found to be impaired in a more physiological assay that measured their ability to inhibit cell surface complement functions of full-length fH. Taken together, our data suggest that disruption of a complex fH-self-surface recognition process, involving a balance of affinities for protein and physiological carbohydrate ligands, predisposes to aHUS.


Asunto(s)
Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Eritrocitos/inmunología , Síndrome Hemolítico-Urémico/genética , Heparina/metabolismo , Mutación , Animales , Células Cultivadas , Factor H de Complemento/genética , Eritrocitos/patología , Predisposición Genética a la Enfermedad , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/patología , Humanos , Polielectrolitos , Polímeros/metabolismo , Unión Proteica/genética , Ovinos
20.
J Immunol ; 183(4): 2565-74, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19625656

RESUMEN

Staphylococcus aureus possesses an impressive arsenal of complement evasion proteins that help the bacterium escape attack of the immune system. The staphylococcal complement inhibitor (SCIN) protein exhibits a particularly high potency and was previously shown to block complement by acting at the level of the C3 convertases. However, many details about the exact binding and inhibitory mechanism remained unclear. In this study, we demonstrate that SCIN directly binds with nanomolar affinity to a functionally important area of C3b that lies near the C terminus of its beta-chain. Direct competition of SCIN with factor B for C3b slightly decreased the formation of surface-bound convertase. However, the main inhibitory effect can be attributed to an entrapment of the assembled convertase in an inactive state. Whereas native C3 is still able to bind to the blocked convertase, no generation and deposition of C3b could be detected in the presence of SCIN. Furthermore, SCIN strongly competes with the binding of factor H to C3b and influences its regulatory activities: the SCIN-stabilized convertase was essentially insensitive to decay acceleration by factor H and the factor I- and H-mediated conversion of surface-bound C3b to iC3b was significantly reduced. By targeting a key area on C3b, SCIN is able to block several essential functions within the alternative pathway, which explains the high potency of the inhibitor. Our findings provide an important insight into complement evasion strategies by S. aureus and may act as a base for further functional studies.


Asunto(s)
Complemento C3b/metabolismo , Proteínas Inactivadoras de Complemento/fisiología , Familia de Multigenes/inmunología , Staphylococcus aureus/inmunología , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , C3 Convertasa de la Vía Alternativa del Complemento/fisiología , Complemento C3b/química , Factor B del Complemento/metabolismo , Factor H de Complemento/metabolismo , Proteínas Inactivadoras de Complemento/metabolismo , Humanos , Unión Proteica/inmunología , Staphylococcus aureus/patogenicidad , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA