Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Immunol ; 67(1): 21-30, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25929733

RESUMEN

C3 glomerulopathy describes a spectrum of disorders with glomerular pathology associated with C3 cleavage product deposition and with defective complement action and regulation (Fakhouri et al., 2010; Sethi et al., 2012b). Kidney biopsies from these patients show glomerular accumulation or deposition of C3 cleavage fragments, but no or minor deposition of immunoglobulins (Appel et al., 2005; D'Agati and Bomback, 2012; Servais et al., 2007; Sethi and Fervenza, 2011). At present the current situation asks for a better definition of the underlining disease mechanisms, for precise biomarkers, and for a treatment for this disease. The complement system is a self activating and propelling enzymatic cascade type system in which inactive, soluble plasma components are activated spontaneously and lead into an amplification loop (Zipfel and Skerka, 2009). Activation of the alternative pathway is spontaneous, occurs by default, and cascade progression leads to amplification by complement activators. The system however is self-controlled by multiple regulators and inhibitors, like Factor H that control cascade progression in fluid phase and on surfaces. The activated complement system generates a series of potent effector components and activation products, which damage foreign-, as well as modified self cells, recruit innate immune cells to the site of action, coordinate inflammation and the response of the adaptive immune system in form of B cells and T lymphocytes (Kohl, 2006; Medzhitov and Janeway, 2002; Ogden and Elkon, 2006; Carroll, 2004; Kemper and Atkinson, 2007; Morgan, 1999; Muller-Eberhard, 1986; Ricklin et al., 2010). Complement controls homeostasis and multiple reactions in the vertebrate organism including defense against microbial infections (Diaz-Guillen et al., 1999; Mastellos and Lambris, 2002; Nordahl et al., 2004; Ricklin et al., 2010). In consequence defective control of the spontaneous self amplifying cascade or regulation is associated with numerous human disorders (Ricklin and Lambris, 2007; Skerka and Zipfel, 2008; Zipfel et al., 2006). Understanding the exact action and regulation of this sophisticated homeotic cascade system is relevant to understand disease pathology of various complement associated human disorders. Furthermore this knowledge is relevant for a better diagnosis and appropriate therapy. At present diagnosis of C3 glomerulopathy is primarily based on the kidney biopsy, and histological, immmunohistological and electron microscopical evaluation (D'Agati and Bomback, 2012; Fakhouri et al., 2010; Medjeral-Thomas et al., 2014a,b; Sethi et al., 2012b). The challenge is to define the actual cause of the diverse glomerular changes or damages, to define how C3 deposition results in the reported glomerular changes, the location of the cell damage and the formation of deposits.


Asunto(s)
Factor Nefrítico del Complemento 3/química , Complemento C3/química , Proteínas Inactivadoras del Complemento C3b/química , Glomerulonefritis Membranoproliferativa/patología , Glomérulos Renales/patología , Agregación Patológica de Proteínas/patología , Inmunidad Adaptativa , Linfocitos B/inmunología , Linfocitos B/patología , Biomarcadores/sangre , Activación de Complemento , Complemento C3/genética , Factor Nefrítico del Complemento 3/genética , Proteínas Inactivadoras del Complemento C3b/genética , Expresión Génica , Glomerulonefritis Membranoproliferativa/diagnóstico , Glomerulonefritis Membranoproliferativa/genética , Glomerulonefritis Membranoproliferativa/inmunología , Humanos , Glomérulos Renales/química , Glomérulos Renales/inmunología , Agregación Patológica de Proteínas/diagnóstico , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/inmunología , Linfocitos T/inmunología , Linfocitos T/patología
2.
J Biol Chem ; 290(15): 9500-10, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25659429

RESUMEN

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


Asunto(s)
Autoanticuerpos/química , Proteínas Inactivadoras del Complemento C3b/química , Factor H de Complemento/química , Epítopos/química , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/inmunología , Síndrome Hemolítico Urémico Atípico/metabolismo , Autoanticuerpos/inmunología , Autoinmunidad/genética , Autoinmunidad/inmunología , Sitios de Unión/genética , Sitios de Unión/inmunología , Proteínas Inactivadoras del Complemento C3b/genética , Proteínas Inactivadoras del Complemento C3b/inmunología , Factor H de Complemento/genética , Factor H de Complemento/inmunología , Cristalografía por Rayos X , Mapeo Epitopo , Epítopos/inmunología , Humanos , Modelos Moleculares , Mutación , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Unión Proteica/inmunología , Estructura Terciaria de Proteína
3.
PLoS One ; 8(11): e78617, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260121

RESUMEN

Mutations and deletions within the human CFHR gene cluster on chromosome 1 are associated with diseases, such as dense deposit disease, CFHR nephropathy or age-related macular degeneration. Resulting mutant CFHR proteins can affect complement regulation. Here we identify human CFHR2 as a novel alternative pathway complement regulator that inhibits the C3 alternative pathway convertase and terminal pathway assembly. CFHR2 is composed of four short consensus repeat domains (SCRs). Two CFHR2 molecules form a dimer through their N-terminal SCRs, and each of the two C-terminal ends can bind C3b. C3b bound CFHR2 still allows C3 convertase formation but the CFHR2 bound convertases do not cleave the substrate C3. Interestingly CFHR2 hardly competes off factor H from C3b. Thus CFHR2 likely acts in concert with factor H, as CFHR2 inhibits convertases while simultaneously allowing factor H assisted degradation by factor I.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/metabolismo , Vía Alternativa del Complemento/fisiología , Proteolisis , Complemento C3/química , Complemento C3/genética , Complemento C3/metabolismo , Convertasas de Complemento C3-C5/química , Convertasas de Complemento C3-C5/genética , Convertasas de Complemento C3-C5/metabolismo , Proteínas Inactivadoras del Complemento C3b/química , Proteínas Inactivadoras del Complemento C3b/genética , 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 , Humanos , Multimerización de Proteína/fisiología , Estructura Terciaria de Proteína , Secuencias Repetitivas de Aminoácido
4.
J Clin Invest ; 123(6): 2434-46, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23728178

RESUMEN

C3 glomerulopathies (C3G) are a group of severe renal diseases with distinct patterns of glomerular inflammation and C3 deposition caused by complement dysregulation. Here we report the identification of a familial C3G-associated genomic mutation in the gene complement factor H­related 1 (CFHR1), which encodes FHR1. The mutation resulted in the duplication of the N-terminal short consensus repeats (SCRs) that are conserved in FHR2 and FHR5. We determined that native FHR1, FHR2, and FHR5 circulate in plasma as homo- and hetero-oligomeric complexes, the formation of which is likely mediated by the conserved N-terminal domain. In mutant FHR1, duplication of the N-terminal domain resulted in the formation of unusually large multimeric FHR complexes that exhibited increased avidity for the FHR1 ligands C3b, iC3b, and C3dg and enhanced competition with complement factor H (FH) in surface plasmon resonance (SPR) studies and hemolytic assays. These data revealed that FHR1, FHR2, and FHR5 organize a combinatorial repertoire of oligomeric complexes and demonstrated that changes in FHR oligomerization influence the regulation of complement activation. In summary, our identification and characterization of a unique CFHR1 mutation provides insights into the biology of the FHRs and contributes to our understanding of the pathogenic mechanisms underlying C3G.


Asunto(s)
Complemento C3/metabolismo , Proteínas Inactivadoras del Complemento C3b/genética , Enfermedades Renales/genética , Niño , Complemento C3/química , Proteínas Inactivadoras del Complemento C3b/química , Proteínas Inactivadoras del Complemento C3b/metabolismo , Proteínas del Sistema Complemento/aislamiento & purificación , Proteínas del Sistema Complemento/metabolismo , Femenino , Duplicación de Gen , Hemólisis , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Riñón/patología , Masculino , Persona de Mediana Edad , Mutagénesis Insercional , Linaje , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Análisis de Secuencia de ADN
5.
Adv Exp Med Biol ; 946: 113-33, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21948365

RESUMEN

Our understanding of both the nature and diversity of Staphylococcal immune evasion proteins has increased tremendously throughout the last several years. Among this group of molecules, members of the SCIN and Efb families of complement inhibitors have been the subject of particularly intense study. This work has demonstrated that both types of proteins exert their primary function by inhibiting C3 convertases, which lie at the heart of the complement-mediated immune response. Despite this similarity, however, significant differences in structure/function relationships and mechanisms of action exist between these bacterial proteins. Furthermore, divergent secondary effects on host immune responses have also been described for these two protein families. This chapter summarizes recent advances toward understanding the structure, function, and mechanism of the SCIN and Efb families, and suggests potential directions for the field over the coming years.


Asunto(s)
Proteínas Bacterianas/inmunología , Proteínas Inactivadoras del Complemento C3b/inmunología , Proteínas Inactivadoras de Complemento/inmunología , Inmunidad Innata/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Proteínas Bacterianas/química , Proteínas Inactivadoras del Complemento C3b/química , Proteínas Inactivadoras de Complemento/química , Humanos , Infecciones Estafilocócicas/microbiología , Relación Estructura-Actividad
6.
Artículo en Inglés | MEDLINE | ID: mdl-19407382

RESUMEN

Staphylococcus aureus secretes a number of small proteins that effectively attenuate the human innate immune response. Among these, the staphylococcal complement-inhibitor protein (SCIN) disrupts the function of the complement component 3 (C3) convertase that is initiated through either the classical or the alternative pathway and thereby prevents amplification of the complement response on the bacterial surface. Recent studies have shown that SCIN may affect the activities of the C3 convertase by binding in an equimolar fashion to C3b, which is itself an integral although non-enzymatic component of the convertase. In order to better understand the nature of the C3b-SCIN interaction, the hanging-drop vapor-diffusion technique was used to crystallize human C3b in the presence of a recombinant form of SCIN. These crystals diffracted synchrotron X-rays to approximately 6 A Bragg spacing and grew in a primitive tetragonal space group (P4(1)2(1)2 or P4(3)2(1)2; unit-cell parameters a = b = 128.03, c = 468.59 A). Cell-content analysis of these crystals was consistent with the presence of either two 1:1 complexes or a single 2:2 assembly in the asymmetric unit, both of which correspond to a solvent content of 51.9%. By making use of these crystals, solution of the C3b-SCIN structure should further our understanding of complement inhibition and immune evasion by this pathogen.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/química , Proteínas Inactivadoras del Complemento C3b/metabolismo , Complemento C3b/química , Complemento C3b/metabolismo , Staphylococcus aureus/metabolismo , Complemento C3b/genética , Proteínas Inactivadoras del Complemento C3b/genética , Cristalización , Cristalografía por Rayos X , Humanos , Staphylococcus aureus/genética
7.
J Immunol ; 174(4): 2143-51, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15699145

RESUMEN

Regulation of complement activation by pathogens and the host are critical for survival. Using two highly related orthopoxvirus proteins, the vaccinia and variola (smallpox) virus complement control proteins, which differ by only 11 aa, but differ 1000-fold in their ability to regulate complement activation, we investigated the role of electrostatic potential in predicting functional activity. Electrostatic modeling of the two proteins predicted that altering the vaccinia virus protein to contain the amino acids present in the second short consensus repeat domain of the smallpox protein would result in a vaccinia virus protein with increased complement regulatory activity. Mutagenesis of the vaccinia virus protein confirmed that changing the electrostatic potential of specific regions of the molecule influences its activity and identifies critical residues that result in enhanced function as measured by binding to C3b, inhibition of the alternative pathway of complement activation, and cofactor activity. In addition, we also demonstrate that despite the enhanced activity of the variola virus protein, its cofactor activity in the factor I-mediated degradation of C3b does not result in the cleavage of the alpha' chain of C3b between residues 954-955. Our data have important implications in our understanding of how regulators of complement activation interact with complement, the regulation of the innate immune system, and the rational design of potent complement inhibitors that might be used as therapeutic agents.


Asunto(s)
Proteínas Inactivadoras de Complemento/química , Proteínas Inactivadoras de Complemento/metabolismo , Modelos Moleculares , Orthopoxvirus/inmunología , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Inactivadoras del Complemento C3b/química , Proteínas Inactivadoras del Complemento C3b/genética , Proteínas Inactivadoras del Complemento C3b/metabolismo , Proteínas Inactivadoras de Complemento/genética , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/inmunología , Secuencia de Consenso , Ácido Glutámico/genética , Humanos , Lisina/genética , Orthopoxvirus/genética , Mapeo Peptídico , Mutación Puntual , Valor Predictivo de las Pruebas , Unión Proteica/inmunología , Secuencias Repetitivas de Aminoácido , Electricidad Estática , Virus de la Viruela/inmunología , Proteínas Virales/biosíntesis , Proteínas Virales/genética
8.
Mol Immunol ; 35(9): 537-44, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9809581

RESUMEN

We designed a cDNA construct encoding an artificial membrane molecule consisting of all 8 short consensus repeats (SCRs) of human monomeric C4b-binding protein (C4bp) followed by DAF's GPI anchor, named mC4bp, and expressed the protein on swine endothelial cells (SEC). At the same level of expression, mC4bp protected host cells as effectively as DAF, the most potent complement (C) regulator on the membrane. This result was unexpected from the reported functional properties of natural multimeric C4bp. Here, we investigated the mechanism whereby mC4bp has potent cell-protective activity. Our results were as follows: (1) mC4bp serves more efficiently as a methylamine-treated C3 (C3ma)-inactivating factor I-cofactor than natural C4bp and as efficiently as MCP as a methylamine-treated (C4ma)-inactivating cofactor by fluid-phase cofactor assay: (2) the potency of C3ma inactivation by mC4bp and factor I is quite high compared to those of other cofactors: (3)blocking studies using mAbs against C4bp suggested that both the 48 kDa N-terminal fragment and the C-terminal domain near the portion responsible for bundle formation participate in the high C3ma-inactivating capacity of mC4bp. Thus, acquiring high C3ma-inactivating capacity secondary to monomeric alteration leads to high C regulatory activity of mC4bp. These results infer that mC4bp differs from C4bp in its potent factor I-cofactor activity and is a good candidate as a safeguard against hyperacute rejection of xenografts.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/metabolismo , Proteínas Inactivadoras de Complemento , Glicoproteínas , Receptores de Complemento/metabolismo , Proteínas Inactivadoras del Complemento C3b/química , Proteínas Inactivadoras del Complemento C3b/genética , Factor I de Complemento/metabolismo , Glicosilfosfatidilinositoles , Humanos , Fragmentos de Péptidos , Conformación Proteica , Receptores de Complemento/química , Receptores de Complemento/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Secuencias Repetitivas de Aminoácido
9.
Biochemistry ; 31(14): 3626-34, 1992 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-1533152

RESUMEN

Modules which share the same consensus sequence are assumed to have common structural features, at the secondary and tertiary level. In order to test the extent of such similarities, it is necessary to examine the structures of several examples from each module family. Recently, the first three-dimensional structure of a complement control protein (CCP) module (the 16th repeat of human factor H, H16) was determined using a combination of two-dimensional NMR and simulated annealing [Norman, D.G., Barlow, P.N., Baron, M., Day, A.J., Sim, R.B., & Campbell, I.D. (1991) J. Mol. Biol. 219, 717-725]. Using the same techniques, the three-dimensional structure of a second CCP module (the 5th repeat of human factor H, H5) has now been determined. The primary sequence of H5 contains 17 residues which are identical and in equivalent position to those in H16. Thirteen of these 17 are part of the consensus sequence. The similarities between the secondary structure of H5 and that of H16 are extensive. This implies that the consensus sequence dictates a particular secondary structure. The tertiary structure of H5, a compact hydrophobic core wrapped in beta-strand and sheet, bears much overall resemblance to that of H16. However, there is a deletion in the first strand of H5, and an insertion in a loop, resulting in slightly shorter overall length. This is associated with a rearrangement of residues within the hydrophobic core. The side chain of the highly conserved Tyr29, which occupies a central position within the core of H16, lies on the periphery of the core of H5.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/química , Secuencia de Aminoácidos , Secuencia de Bases , Clonación Molecular , Proteínas Inactivadoras del Complemento C3b/genética , Proteínas Inactivadoras del Complemento C3b/aislamiento & purificación , Factor H de Complemento , ADN/genética , Expresión Génica , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Plásmidos , Reacción en Cadena de la Polimerasa , Conformación Proteica , Secuencias Repetitivas de Ácidos Nucleicos
10.
Biochem J ; 283 ( Pt 2): 317-9, 1992 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-1533511

RESUMEN

The potency of complement factor H (H) in accelerating the decay of the alternative pathway C3 convertase, C3b,Bb (decay-accelerating activity), was used as a measure of the affinity of native versus trypsin-treated H for the complement protein C3b bound to surfaces. When about 99% of H was cleaved at the primary tryptic cleavage site 34 kDa from the N-terminus, its decay-accelerating activity on C3b,Bb on sheep erythrocytes fell about 60-fold, whereas the trypsin-treated H was only 3-4 times less potent than native H in dissociating C3b,Bb on Sepharose 4B. The residual decay-accelerating activity, remaining after the primary cleavage, was not affected by secondary cleavage at a site 120 kDa from the N-terminus, as shown with H preparations cleaved to different degrees. Because cell surface sialic acid is known to be responsible for the high affinity of H for C3b bound on sheep erythrocytes, the results strongly suggest that the integrity of the primary tryptic cleavage site of H is essential for the recognition of sialic acid-containing surfaces by the C3b-H complex.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/metabolismo , Ácidos Siálicos/análisis , Tripsina/metabolismo , Animales , Complemento C3b/metabolismo , Proteínas Inactivadoras del Complemento C3b/química , Factor H de Complemento , Electroforesis en Gel de Poliacrilamida , Eritrocitos/metabolismo , Cinética , Peso Molecular , Ovinos
11.
Eur J Immunogenet ; 18(5-6): 399-403, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1837734

RESUMEN

The authors report the functional analysis of the purified mouse factor H allotypes H.1 and H.2, which were clearly distinguished from each other by an immunodiffusion test. Both allotypes acted as a co-factor for factor I in cleaving mouse C3b and we found no significant difference between their activities. The results strongly suggest that the function of mouse factor H for the co-factor activity has been well conserved between two allotypes.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/fisiología , Serina Endopeptidasas/fisiología , Alelos , Animales , Proteínas Inactivadoras del Complemento C3b/química , Proteínas Inactivadoras del Complemento C3b/genética , Factor H de Complemento , Factor I de Complemento , Ratones , Peso Molecular , Transfección
12.
J Biol Chem ; 266(25): 16847-53, 1991 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-1832158

RESUMEN

Factor H is a regulator of complement activation and, in this capacity, it prevents activation of the alternative pathway on host cells and tissues when it recognizes markers on these surfaces. This report describes the binding characteristics and location of the site on factor H that is responsible for host recognition. Factor H was found to bind a variety of polyanions, including heparin, heparan sulfate, dextran sulfate, and clusters of sialic acid. In heparin-agarose binding assays it exhibited an affinity for heparin only 2-fold weaker than that of antithrombin III. Factor H exhibited little or no affinity for polyaspartic acid or bacterial colominic acid (polysialic acid). Factor H (Mr 150,000 with approximate dimensions of 30 x 600 A) is composed of 20 highly homologous domains (SCRs) that are arranged as beads on a string. Polyanions were found to block a tryptic cleavage site in domain 15, and a photoaffinity-tagged heparin probe labeled the region between domains 12 and 15. Affinity chromatography of tryptic fragments on heparin-Sepharose confirmed that this region contained the heparin-binding site. CNBr cleavage at Met787 located between SCRs 13 and 14 split the photoaffinity-tagged region. Sequence analysis strongly suggests that domain 13 contains the primary site of polyanion binding. Factor H expresses its complement regulatory function through a site located in domains 4-6 where C3b binds. Thus, the polyanion-binding site that regulates the affinity of factor H for C3b appears to reside more than 200 A away from the C3b-binding site.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/metabolismo , Heparina/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Unión Competitiva , Complemento C3b/metabolismo , Proteínas Inactivadoras del Complemento C3b/química , Factor H de Complemento , Datos de Secuencia Molecular , Fotoquímica , Sefarosa/metabolismo , Tripsina/metabolismo , Inhibidores de Tripsina/metabolismo
13.
J Mol Biol ; 219(4): 717-25, 1991 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-1829116

RESUMEN

The complement control protein (CCP) modules (also known as short consensus repeats) are defined by a consensus sequence within a stretch of about 60 amino acid residues. These modules have been identified more than 140 times in over 20 proteins, including 12 proteins of the complement system. The solution structure of the 16th CCP module from human complement factor H has been determined by a combination of 2-dimensional nuclear magnetic resonance spectroscopy and restrained simulated annealing. In all, 548 structurally important nuclear Overhauser enhancement cross-peaks were quantified as distance restraints and, together with 41 experimentally measured angle restraints, were incorporated into a simulated annealing protocol to determine a family of closely related structures that satisfied the experimental observations. The CCP structure is shown to be based on a beta-sandwich arrangement; one face made up of three beta-strands hydrogen-bonded to form a triple-stranded region at its centre and the other face formed from two separate beta-strands. Both faces of the molecule contribute highly conserved hydrophobic side-chains to a compact core. The regions between the beta-strands are composed of both well-defined turns and less well-defined loops. Analysis of CCP sequence alignments, in light of the determined structure, reveals a high degree of conservation amongst residues of obvious structural importance, while almost all insertions, deletions or replacements observed in the known sequences are found in the less well-defined loop regions. On the basis of these observations it is postulated that models of other CCP modules that are based on the structure presented here will be accurate. Certain families of CCP modules differ from the consensus in that they contain extra cysteine residues. As a test of structural consensus, the extra disulphide bridges are shown to be easily accommodated within the determined CCP model.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/química , Secuencia de Aminoácidos , Factor H de Complemento , Secuencia de Consenso , Humanos , Enlace de Hidrógeno , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Conformación Proteica , Secuencias Repetitivas de Ácidos Nucleicos , Alineación de Secuencia
14.
Mol Immunol ; 28(4-5): 375-82, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1829503

RESUMEN

Two proteins that are involved in cleavage of methylamine-treated C3 of guinea-pig origin (C3(MA)gp) have been isolated from guinea-pig serum. One of them functioned as a cofactor of human factor I (Ihu) for cleavage of C3(MA)gp and its molecular size was 150 kDa. The other was functionally pure and able to cleave C3(MA)gp together with human factor H (Hhu). They appear to be analogous to human factors H and I in the guinea-pig and will be referred to as Hgp and Igp. Methylamine-treated human C3 [C3(MA)hu] was not a compatible substrate for Hgp or Igp: little cleavage of C3(MA)hu was observed if human factor H (Hhu) or I was substituted with the guinea-pig counterpart. C3(MA)gp, on the other hand, served as a substrate, though less efficiently, for Hhu and Ihu. Human C4b-binding protein (C4bp) and membrane cofactor protein (MCP) as well as Hhu could participate in cleavage of C3(MA)gp by Igp or Ihu. In these assays, C3(MA)gp was degraded again less efficiently than C3(MA)hu. Interestingly, human C3b/C4b receptor (CR1) mediated factor I-dependent cleavage of C3(MA)hu and C3(MA)gp to a similar extent regardless the sources of factor I. These results suggest that factor I-dependent C3b regulatory system is species-specific except in the case of CR1, which may function as a cofactor irrespective of species.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/fisiología , Complemento C3b/fisiología , Cobayas/fisiología , Serina Endopeptidasas/fisiología , Animales , Proteínas Inactivadoras del Complemento C3b/química , Proteínas Inactivadoras del Complemento C3b/aislamiento & purificación , Factor H de Complemento , Factor I de Complemento , Humanos , Técnicas In Vitro , Peso Molecular , Receptores de Complemento/fisiología , Receptores de Complemento 3b , Serina Endopeptidasas/química , Serina Endopeptidasas/aislamiento & purificación , Especificidad de la Especie
15.
Eur J Immunol ; 21(4): 967-72, 1991 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1826890

RESUMEN

We studied the capacity of human factor H to promote the secretion of a lymphocyte-activating factor (LAF) by human monocytes cultured under serum-free conditions. Presence of LAF in the culture supernatants was assessed with the mouse thymocyte assay. Highly purified factor H alone had no effect on thymocyte proliferation. When monocytes were cultured with factor H for 24 h, a significant secretion of LAF was observed. The effect was dose dependent over a range of factor H concentrations from 1 to 15 micrograms/ml. Polymyxin B did not abrogate the capacity of factor H to induce LAF secretion. Adsorption of factor H preparations onto anti-factor H-Sepharose completely suppressed the phenomenon. Conversely, the activity was recovered in the acidic eluate. Furthermore, factor H subpopulation phi 2, that was able to bind to phenyl-Sepharose, was a stronger inducer of LAF secretion by monocytes than the subpopulation phi 1 (which did not bind to phenyl-Sepharose). Using a specific radioimmunoassay for interleukin 1-beta (IL 1 beta), we observed a good correlation between the LAF activity and the amount of IL 1 beta secreted by human monocytes stimulated with factor H. We have shown previously that factor H (phi 2) bound specifically on Raji cells whereas factor H (phi 1) did not. These results argue for the participation of the interaction of factor H with its receptor to stimulate the secretion of IL 1 by monocytes and that the phi 2 form of factor H is a ligand for the human factor H receptor.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/farmacología , Interleucina-1/metabolismo , Monocitos/efectos de los fármacos , Células Cultivadas , Proteínas Inactivadoras del Complemento C3b/química , Factor H de Complemento , Humanos , Monocitos/metabolismo
16.
Biochemistry ; 30(11): 2847-57, 1991 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-1826087

RESUMEN

Factor H is a regulatory component of the complement system. It has a monomer Mr of 150,000. Primary structure analysis shows that the polypeptide is divided into 20 homologous regions, each 60 amino acid residues long. These are independently folding domains and are termed "short consensus repeats" (SCRs) or "complement control protein" (CCP) repeats. High-flux synchrotron X-ray and neutron scattering studies were performed in order to define its solution structure in conditions close to physiological. The Mr of factor H was determined as 250,000-320,000 to show that factor H is dimeric. This structure is maintained at concentrations between 1 and 11 mg/mL in the pH range 5-9. Zn2+ ions are an inhibitor of C3b cleavage by factor I, a reaction in which factor H acts as a cofactor. Additions of Zn2+ to factor H caused it to form oligomers containing 4-10 monomers. The radius of gyration RG of native factor H by X-rays or by neutrons in 0% or 100% 2H2O buffers is not measurable but is greater than 12.5 nm. Two cross-sectional radii of gyration RXS-1 and RXS-2 were determined as 3.0-3.1 and 1.8 nm, respectively. Analyses of the cross-sectional intensities show that factor H is composed of two distinct subunits. The RXS-1 corresponds to the cross-sectional properties of both subunits and exhibits an unusual radiation dependence on the X-ray flux. Since RXS-2 is close to the corresponding RXS of C4b binding protein (91% of which is formed from SCR/CCP domains), it is inferred that the SCR/CCP domains of factor H and C4b binding protein have similar solution structures. The use of hydrodynamic spheres to reproduce literature sedimentation coefficients of 5.5-5.6 S showed that these were compatible with a V-shaped arrangement of two rods (36 spheres each, length 87 +/- 5 nm) joined at an angle of 5 degrees. The use of a similar arrangement of 244 spheres arranged in two rods (length 77 nm) to fit the experimental X-ray and neutron scattering curves showed that the two rods are joined at an angle of 5 degrees. This model corresponds to an actual RG of 21-23 nm. The separation between each SCR/CCP in factor H is close to 4 nm. In the solution structure of factor H, the SCR/CCP domains are in a highly extended conformation.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/química , Complemento C3b/metabolismo , Proteínas Inactivadoras del Complemento C3b/metabolismo , Factor H de Complemento , Humanos , Concentración de Iones de Hidrógeno , Sustancias Macromoleculares , Matemática , Modelos Moleculares , Neutrones , Aceleradores de Partículas , Conformación Proteica , Dispersión de Radiación , Difracción de Rayos X/métodos , Zinc/farmacología
17.
Biochemistry ; 30(4): 997-1004, 1991 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-1824927

RESUMEN

The complement control protein (CCP) module (also known as the short consensus repeat) is a consensus sequence of about 60 amino acid residues which is thought to fold independently. It occurs over 140 times in more than 20 extracellular mosaic proteins including 12 proteins of the complement cascade. An isolated CCP module, the 16th repeat from human complement factor H, has been expressed in a yeast vector and shown to fold with the same pattern of disulfide bond formation as is seen in the native protein. Two-dimensional 600-MHz 1H NMR spectra of this module have been recorded at pH 3.3 and 6.0 and analyzed to permit determination of secondary structure in solution. The CCP module comprises two predominantly extended segments (Glu1-His13 and Ala17-Glu27), two segments of double-stranded antiparallel beta-sheet (Gly14-Val16 paired with Tyr31-Cys33 and Gly38-Asp40 paired with Ser57-Ile59), and a short piece of triple-stranded beta-sheet (Glu27-Thr30, Ile44-Leu48, and Lys51-Ser53). Turns occur at Asp22, Gly36, and Glu50, while Gly41-Ala43 appear to form a looped-out segment or bulge. This structure is compared with a secondary structure prediction made on the basis of an alignment scheme of 101 sequences for CCP modules [Perkins, S. J., Haris, P. I., Sim, R. B., & Chapman, D. (1988) Biochemistry 27, 4004-4012]--the experimentally determined secondary structure bears an overall resemblance to the predicted one but differs in the number and position of turns. Some of those amino acid residues which are highly conserved throughout the range of CCP modules appear to play a role in stabilizing the global fold.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/química , Proteínas del Sistema Complemento/genética , Secuencia de Consenso , Proteínas/genética , Secuencia de Aminoácidos , Secuencia de Bases , Clonación Molecular , Proteínas Inactivadoras del Complemento C3b/genética , Factor H de Complemento , Humanos , Hidrógeno , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Conformación Proteica , Levaduras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...