Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Immunobiology ; 228(5): 152410, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37478687

RESUMO

Complement factor I (FI) is the nexus for classical, lectin and alternative pathway complement regulation. FI is an 88 kDa plasma protein that circulates in an inactive configuration until it forms a trimolecular complex with its cofactor and substrate whereupon a structural reorganization allows the catalytic triad to cleave its substrates, C3b and C4b. In keeping with its role as the master complement regulatory enzyme, deficiency has been linked to immunopathology. In the setting of complete FI deficiency, a consumptive C3 deficiency results in recurrent infections with encapsulated microorganisms. Aseptic cerebral inflammation and vasculitic presentations are also less commonly observed. Heterozygous mutations in the factor I gene (CFI) have been demonstrated to be enriched in atypical haemolytic uraemic syndrome, albeit with a very low penetrance. Haploinsufficiency of CFI has also been associated with decreased retinal thickness and is a strong risk factor for the development of age-related macular degeneration. Supplementation of FI using plasma purified or recombinant protein has long been postulated, however, technical difficulties prevented progression into clinical trials. It is only using gene therapy that CFI supplementation has reached the clinic with GT005 in phase I/II clinical trials for geographic atrophy.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Fator I do Complemento , Humanos , Fator I do Complemento/química , Complemento C3/genética , Mutação , Inflamação , Complemento C3b
2.
Blood ; 142(6): 607-610, 2023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37363824

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a rare thrombotic microangiopathy. Genetic variants in complement proteins are found in ≈60% of patients. Of these patients, ≈15% carry mutations in complement factor I (CFI). Factor I (FI) is a multidomain serine protease that cleaves and thereby inactivates C3b and C4b in the presence of cofactor proteins. Crystal structures have shown that FI possesses 2 calcium-binding domains, low-density lipoprotein receptor class A (LDLRA) 1 and LDLRA2. Yet, the role of calcium in FI is unknown. We determined that 9 genetic variants identified in aHUS (N151S, G162D, G188A, V230E, A240G, G243R, C247G, A258T, and Q260D) cluster around the calcium-binding site of LDLRA1. Using site-directed mutagenesis, we established that the synthesis of all, except A258T, was impaired, implying defective protein folding, perhaps due to loss of calcium binding. To further explore this possibility, we generated 12 alanine mutants that coordinate with the calcium in LDLRA1 and LDLRA2 (K239A, D242A, I244A, D246A, D252A, E253A, Y276A, N279A, E281A, D283A, D289A, and D290A) and are expected to perturb calcium binding. Except for K239A and Y276A, none of the mutants was secreted. These observations suggest that calcium ions play key structural and functional roles in FI.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Humanos , Síndrome Hemolítico-Urêmica Atípica/genética , Cálcio , Fator I do Complemento/genética , Fator I do Complemento/química , Fator I do Complemento/metabolismo , Proteínas do Sistema Complemento , Mutação
3.
Fish Shellfish Immunol ; 98: 988-994, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31712129

RESUMO

Complement factor I (CFI) is a serine protease which plays a key role in the modulation of complement system and the induced-fit factor responsible for controlling the complement-mediated processes. In this study, a CFI gene was cloned and characterized from Lampetra morii (designated as L-CFI) at molecular and cellular levels. The L-CFI protein included a factor I membrane attack complex domain (FIMAC), a scavenger receptor cysteine-rich domain (SRCR), a trypsin-like serine protease domain (Tryp_SPc) and 2 low-density lipoprotein receptor class A domains (LDLa) which would exhibit functional similarities to CFI superfamily proteins. Tissue expression profile analysis showed that L-CFI mRNA constitutively expressed in all tested tissues except erythrocytes, with the predominant expression in liver. The mRNA expression level of L-CFI increased significantly after Vibrio anguillarum and Staphylocccus aureus stimulation. It is demonstrated that L-CFI interacted with L-C3 protein and affected the deposition of L-C3 on the cell surface. Furthermore, lamprey serum after deplete L-CFI and L-C3 reduced the cytotoxic activity against HeLa cells. These findings suggest that L-CFI plays an important role in lamprey immunity and involved in the lamprey complement system.


Assuntos
Ativação do Complemento/imunologia , Fator I do Complemento/genética , Proteínas de Peixes/genética , Imunidade Inata/genética , Lampreias/genética , Lampreias/imunologia , Sequência de Aminoácidos , Animais , Fator I do Complemento/química , Fator I do Complemento/metabolismo , Proteínas de Peixes/química , Proteínas de Peixes/metabolismo , Alinhamento de Sequência
4.
Immunobiology ; 224(4): 511-517, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31109748

RESUMO

Factor I was first discovered in 1966. Its importance became apparent with the description of the original Factor I deficient patient in Boston in 1967. This patient presented with a hyperactive alternative complement pathway resulting in secondary complement deficiency due to continuous complement consumption. On the basis of these findings, the mechanism of the alternative pathway was worked out. In 1975, the surprise finding was made that elevating levels of Factor I in plasma down-regulated the alternative pathway. Attempts to exploit this finding for clinical use had a long and frustrating history and it was not until 2019 that the first patient was treated with the gene therapy vector for age related macular degeneration by Professor Sir Robert MacLaren in Oxford. This review follows the long and contorted course from initial observations to clinical use of complement Factor I.


Assuntos
Fator I do Complemento/fisiologia , Animais , Ativação do Complemento/genética , Ativação do Complemento/imunologia , Fator I do Complemento/química , Fator I do Complemento/uso terapêutico , Via Alternativa do Complemento/genética , Via Alternativa do Complemento/imunologia , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Suscetibilidade a Doenças , Humanos , Imunoconglutininas/imunologia , Transdução de Sinais , Relação Estrutura-Atividade
5.
Nat Struct Mol Biol ; 24(8): 643-651, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28671664

RESUMO

The complement system labels microbes and host debris for clearance. Degradation of surface-bound C3b is pivotal to direct immune responses and protect host cells. How the serine protease factor I (FI), assisted by regulators, cleaves either two or three distant peptide bonds in the CUB domain of C3b remains unclear. We present a crystal structure of C3b in complex with FI and regulator factor H (FH; domains 1-4 with 19-20). FI binds C3b-FH between FH domains 2 and 3 and a reoriented C3b C-terminal domain and docks onto the first scissile bond, while stabilizing its catalytic domain for proteolytic activity. One cleavage in C3b does not affect its overall structure, whereas two cleavages unfold CUB and dislodge the thioester-containing domain (TED), affecting binding of regulators and thereby determining the number of cleavages. These data explain how FI generates late-stage opsonins iC3b or C3dg in a context-dependent manner, to react to foreign, danger or healthy self signals.


Assuntos
Complemento C3b/química , Complemento C3b/metabolismo , Fator H do Complemento/química , Fator H do Complemento/metabolismo , Fator I do Complemento/química , Fator I do Complemento/metabolismo , Cristalografia por Raios X , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Proteólise
6.
J Biol Chem ; 292(32): 13345-13360, 2017 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-28637873

RESUMO

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.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Ativação do Complemento , Degeneração Macular/genética , Mutação , Substituição de Aminoácidos , Animais , Síndrome Hemolítico-Urêmica Atípica/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , C3 Convertase da Via Alternativa do Complemento/química , C3 Convertase da Via Alternativa do Complemento/genética , C3 Convertase da Via Alternativa do Complemento/metabolismo , Complemento C3d/química , Complemento C3d/genética , Complemento C3d/metabolismo , Fator H do Complemento/química , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Fator I do Complemento/química , Fator I do Complemento/genética , Fator I do Complemento/metabolismo , Eritrócitos/química , Hemólise , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/genética , Proteínas Imobilizadas/metabolismo , Degeneração Macular/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Carneiro Doméstico , Solubilidade , Streptococcus pneumoniae/metabolismo , Propriedades de Superfície
7.
Proc Natl Acad Sci U S A ; 112(41): 12794-9, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26420870

RESUMO

The complement system has evolved to annul pathogens, but its improper regulation is linked with diseases. Efficient regulation of the system is primarily provided by a family of proteins termed regulators of complement activation (RCA). The knowledge of precise structural determinants of RCA proteins critical for imparting the regulatory activities and the molecular events underlying the regulatory processes, nonetheless, is still limited. Here, we have dissected the structural requirements of RCA proteins that are crucial for one of their two regulatory activities, the cofactor activity (CFA), by using the Kaposi's sarcoma-associated herpesvirus RCA homolog Kaposica as a model protein. We have scanned the entire Kaposica molecule by sequential mutagenesis using swapping and site-directed mutagenesis, which identified residues critical for its interaction with C3b and factor I. Mapping of these residues onto the modeled structure of C3b-Kaposica-factor I complex supported the mutagenesis data. Furthermore, the model suggested that the C3b-interacting residues bridge the CUB (complement C1r-C1s, Uegf, Bmp1) and MG2 (macroglobulin-2) domains of C3b. Thus, it seems that stabilization of the CUB domain with respect to the core of the C3b molecule is central for its CFA. Identification of CFA-critical regions in Kaposica guided experiments in which the equivalent regions of membrane cofactor protein were swapped into decay-accelerating factor. This strategy allowed CFA to be introduced into decay-accelerating factor, suggesting that viral and human regulators use a common mechanism for CFA.


Assuntos
Complemento C3b/química , Fator I do Complemento/química , Herpesvirus Humano 8/química , Modelos Moleculares , Complexos Multiproteicos/química , Proteínas Virais/química , Complemento C3b/genética , Fator I do Complemento/genética , Herpesvirus Humano 8/genética , Humanos , Complexos Multiproteicos/genética , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas Virais/genética
8.
Biosci Rep ; 34(5)2014 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-25188723

RESUMO

aHUS (atypical haemolytic uraemic syndrome), AMD (age-related macular degeneration) and other diseases are associated with defective AP (alternative pathway) regulation. CFH (complement factor H), CFI (complement factor I), MCP (membrane cofactor protein) and C3 exhibited the most disease-associated genetic alterations in the AP. Our interactive structural database for these was updated with a total of 324 genetic alterations. A consensus structure for the SCR (short complement regulator) domain showed that the majority (37%) of SCR mutations occurred at its hypervariable loop and its four conserved Cys residues. Mapping 113 missense mutations onto the CFH structure showed that over half occurred in the C-terminal domains SCR-15 to -20. In particular, SCR-20 with the highest total of affected residues is associated with binding to C3d and heparin-like oligosaccharides. No clustering of 49 missense mutations in CFI was seen. In MCP, SCR-3 was the most affected by 23 missense mutations. In C3, the neighbouring thioester and MG (macroglobulin) domains exhibited most of 47 missense mutations. The mutations in the regulators CFH, CFI and MCP involve loss-of-function, whereas those for C3 involve gain-of-function. This combined update emphasizes the importance of the complement AP in inflammatory disease, clarifies the functionally important regions in these proteins, and will facilitate diagnosis and therapy.


Assuntos
Complemento C3 , Fator H do Complemento , Fator I do Complemento , Bases de Dados de Ácidos Nucleicos , Bases de Dados de Proteínas , Proteína Cofatora de Membrana , Mutação de Sentido Incorreto , Complemento C3/química , Complemento C3/genética , Fator H do Complemento/química , Fator H do Complemento/genética , Fator I do Complemento/química , Fator I do Complemento/genética , Via Alternativa do Complemento/genética , Humanos , Inflamação/diagnóstico , Inflamação/genética , Inflamação/terapia , Proteína Cofatora de Membrana/química , Proteína Cofatora de Membrana/genética , Estrutura Terciária de Proteína
9.
Fish Shellfish Immunol ; 40(2): 414-23, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25108086

RESUMO

Complement factor I (FI) is a plasma serine proteinase that plays an essential role in the modulation of the complement cascade. In the presence of substrate modulating cofactors (Factor H, C4bp, CR1, etc), FI cleaves the activation products of C3 (i.e. C3b) and C4 (i.e. C4b) to limit complement activity. In this study, the full length cDNA of factor I (CpFI) is isolated from the liver of the whitespotted bamboo shark (Chiloscyllium plagiosum). The CpFI cDNA is 2326 bp in length, encoding a protein of 671 amino acids, which shares 72-80% identity with FI molecules of other sharks, higher than the teleosts (37-40%) and mammals (44-47%). The sequence alignment and comparative analysis indicates the FI proteins are well conserved, with the typical modular architecture and identical active sites throughout vertebrate evolution, suggesting the conserved function. However, the additional sequence present between the leader peptide (LP) and the factor I membrane attack complex (FIMAC) domain in other fishes is also found in CpFI, which consists of two kind of tandem repeats. Phylogenetic analysis suggests that CpFI belongs to the elasmobranch clade, in parallel with the higher vertebrates, to form a sister taxa to teleosts. Expression analysis revealed that CpFI is ubiquitously distributed in a variety of tissues, with the constitutive expression in liver, which might reflect the species-specific distribution patterns of FI. Together with earlier reports, the presence of FI in various sharks might suggest the existence of a well-developed complement regulation mechanism in cartilaginous fish.


Assuntos
Fator I do Complemento/genética , Proteínas de Peixes/genética , Regulação da Expressão Gênica , Tubarões/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Fator I do Complemento/química , Fator I do Complemento/metabolismo , DNA Complementar/genética , Proteínas de Peixes/química , Proteínas de Peixes/metabolismo , Perfilação da Expressão Gênica , Masculino , Dados de Sequência Molecular , Especificidade de Órgãos , Filogenia , Reação em Cadeia da Polimerase/veterinária , Alinhamento de Sequência/veterinária , Tubarões/metabolismo
10.
Proc Natl Acad Sci U S A ; 111(1): 427-32, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24367091

RESUMO

The complement system is a crucial component of the innate immune response against invading bacterial pathogens. The human pathogen Neisseria meningitidis (Nm) is known to possess several mechanisms to evade the complement system, including binding to complement inhibitors. In this study, we describe an additional mechanism used by Nm to evade the complement system and survive in human blood. Using an isogenic NalP deletion mutant and NalP complementing strains, we show that the autotransporter protease NalP cleaves C3, the central component of the complement cascade. The cleavage occurs 4 aa upstream from the natural C3 cleavage site and produces shorter C3a-like and longer C3b-like fragments. The C3b-like fragment is degraded in the presence of the complement regulators (factors H and I), and this degradation results in lower deposition of C3b on the bacterial surface. We conclude that NalP is an important factor to increase the survival of Nm in human serum.


Assuntos
Complemento C3/química , Complemento C3b/química , Proteínas de Membrana Transportadoras/metabolismo , Neisseria meningitidis/metabolismo , Serina Endopeptidases/metabolismo , Soro/microbiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Complemento C3/imunologia , Complemento C3b/imunologia , Fator H do Complemento/química , Fator I do Complemento/química , DNA/genética , Escherichia coli/metabolismo , Deleção de Genes , Humanos , Camundongos , Dados de Sequência Molecular , Fenótipo , Ligação Proteica , Coelhos , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Frações Subcelulares/metabolismo , Fatores de Tempo
11.
PLoS One ; 8(11): e78617, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24260121

RESUMO

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.


Assuntos
Proteínas Inativadoras do Complemento C3b/metabolismo , Via Alternativa do Complemento/fisiologia , Proteólise , Complemento C3/química , Complemento C3/genética , Complemento C3/metabolismo , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Proteínas Inativadoras do Complemento C3b/química , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/química , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Fator I do Complemento/química , Fator I do Complemento/genética , Fator I do Complemento/metabolismo , Humanos , Multimerização Proteica/fisiologia , Estrutura Terciária de Proteína , Sequências Repetitivas de Aminoácidos
12.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 4): 418-24, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22505261

RESUMO

Tetartohedral crystal twinning is discussed as a particular case of (pseudo)merohedral twinning when the number of twinned domains is four. Tetartohedrally twinned crystals often possess pseudosymmetry, with the rotational part of the pseudosymmetry operators coinciding with the twinning operators. Tetartohedrally twinned structures from the literature are reviewed and the recent structure determination of tetartohedrally twinned triclinic crystals of human complement factor I is discussed.


Assuntos
Cristalografia por Raios X/métodos , Fator I do Complemento/análise , Fator I do Complemento/química , Bases de Dados de Proteínas , Humanos
13.
J Biol Chem ; 287(17): 13572-83, 2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-22393059

RESUMO

Factor I (FI) is a serine protease that inhibits all complement pathways by degrading activated complement components C3b and C4b. FI functions only in the presence of several cofactors, such as factor H, C4b-binding protein, complement receptor 1, and membrane cofactor protein. FI is composed of two chains linked by a disulfide bridge; the light chain comprises only the serine protease (SP) domain, whereas the heavy chain contains the FI membrane attack complex domain (FIMAC), CD5 domain, and low density lipoprotein receptor 1 (LDLr1) and LDLr2 domains. To better understand how FI inhibits complement, we used homology-based three-dimensional models of FI domains in an attempt to identify potential protein-protein interaction sites. Specific amino acids were then mutated to yield 20 recombinant mutants of FI carrying additional surface-exposed N-glycosylation sites that were expected to sterically hinder interactions. The Michaelis constant (K(m)) of all FI mutants toward a small substrate was not increased. We found that many mutations in the FIMAC and SP domains nearly abolished the ability of FI to degrade C4b and C3b in the fluid phase and on the surface, irrespective of the cofactor used. On the other hand, only a few alterations in the CD5 and LDLr1/2 domains impaired this activity. In conclusion, all analyzed cofactors form similar trimolecular complexes with FI and C3b/C4b, and the accessibility of FIMAC and SP domains is crucial for the function of FI.


Assuntos
Bioquímica/métodos , Fator I do Complemento/química , Sítios de Ligação , Complemento C3b/química , Complemento C4b/química , Glicosilação , Células HEK293 , Humanos , Imageamento Tridimensional , Cinética , Mutagênese Sítio-Dirigida , Mutação , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Recombinantes/química
14.
Proc Natl Acad Sci U S A ; 108(31): 12839-44, 2011 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-21768352

RESUMO

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.


Assuntos
Fator I do Complemento/química , Fator I do Complemento/genética , Polimorfismo Genético , Estrutura Terciária de Proteína , Regulação Alostérica , Sítios de Ligação/genética , Domínio Catalítico , Complemento C3b/química , Complemento C3b/metabolismo , Complemento C4b/química , Complemento C4b/metabolismo , Fator I do Complemento/metabolismo , Cristalização , Cristalografia por Raios X , Precursores Enzimáticos/química , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Predisposição Genética para Doença/genética , Glicosilação , Humanos , Modelos Moleculares , Mutação , Ligação Proteica
15.
Mol Immunol ; 48(14): 1611-20, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21529951

RESUMO

Factor I (FI) is a crucial inhibitor controlling all complement pathways due to its ability to degrade activated complement proteins C3b and C4b in the presence of cofactors such as factor H, C4b-binding protein, complement receptor 1 or CD46. Complete deficiency of FI, which is synthesized mainly in the liver is rare and leads to complement consumption resulting in recurrent severe infections, glomerulonephritis or autoimmune diseases. Incomplete FI deficiency is in turn associated with atypical haemolytic uremic syndrome, a severe disease characterized by thrombocytopenia, microangiopathic haemolytic anaemia and acute renal failure. Structurally, FI is a 88kDa heterodimer of a heavy chain consisting of one FI-membrane attack complex (FIMAC) domain, one CD5 domain and two low-density lipoprotein receptor domains (LDLr), and a light chain which is a serine protease domain (SP), linked to the heavy chain by a disulfide bond. FI cleaves its in vivo substrates C3b and C4b only in the presence of cofactors, it shows poor enzymatic activity towards synthetic substrates tested so far and it has no natural inhibitor.


Assuntos
Fator I do Complemento/metabolismo , Síndrome Hemolítico-Urêmica Atípica , Fator I do Complemento/química , Fator I do Complemento/deficiência , Fator I do Complemento/genética , Dimerização , Síndrome Hemolítico-Urêmica/imunologia , Humanos , Modelos Imunológicos , Mutação , Estrutura Terciária de Proteína , Especificidade por Substrato
16.
J Biol Chem ; 285(9): 6235-45, 2010 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-20044478

RESUMO

The central complement inhibitor factor I (FI) degrades activated complement factors C4b and C3b in the presence of cofactors such as C4b-binding protein, factor H, complement receptor 1, and membrane cofactor protein. FI is a serine protease composed of two chains. The light chain comprises the serine protease domain, whereas the heavy chain contains several domains; that is, the FI and membrane attack complex domain (FIMAC), CD5, low density lipoprotein receptor 1 (LDLr1) and LDLr2 domains. To understand better how FI acts as a complement inhibitor, we used homology-based models of FI domains to predict potential binding sites. Specific amino acids were then mutated to yield 16 well expressed mutants, which were then purified from media of eukaryotic cells for functional analyses. The Michaelis constant (K(m)) of all FI mutants toward a small substrate was not altered, whereas some mutants showed increased maximum initial velocity (V(max)). All the mutations in the FIMAC domain affected the ability of FI to degrade C4b and C3b irrespective of the cofactor used, whereas only some mutations in the CD5 and LDLr1/2 domains had a similar effect. These same mutants also showed impaired binding to C3met. In conclusion, the FIMAC domain appears to harbor the main binding sites important for the ability of FI to degrade C4b and C3b.


Assuntos
Fator I do Complemento/metabolismo , Sítios de Ligação , Catálise , Complemento C3b/metabolismo , Complemento C4b/metabolismo , Fator I do Complemento/química , Fator I do Complemento/genética , Humanos , Cinética , Mutagênese Sítio-Dirigida , Homologia Estrutural de Proteína
17.
Eur J Immunol ; 40(1): 172-85, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19877009

RESUMO

The complement system is regulated by inhibitors such as factor I (FI), a serine protease that degrades activated complement factors C4b and C3b in the presence of specific cofactors. Mutations and polymorphisms in FI and its cofactors are associated with atypical hemolytic uremic syndrome (aHUS). All 14 complement factor I mutations associated with aHUS analyzed in this study were heterozygous and generated premature stop codons (six) or amino acid substitutions (eight). Almost all of the mutants were expressed by human embryonic kidney 293 cells but only six mutants were secreted into the medium, three of which were at lower levels than WT. The remaining eight mutants were not secreted but sensitive to deglycosylation with endoglycosidase H, indicating that they were retained early in the secretory pathway. Six secreted mutants were purified and five of them were functionally altered in degradation of C4b/C3b in the fluid-phase in the presence of various cofactors and on endothelial cells. Three mutants cleaved surface-bound C3b less efficiently than WT. The D501N mutant was severely impaired both in solution and on surface irrespective of the cofactor used. In conclusion, mutations in complement factor I affect both secretion and function of FI, which leads to impaired regulation of the complement system in aHUS.


Assuntos
Fator I do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/metabolismo , Mutação , Animais , Células Cultivadas , Complemento C3b/metabolismo , Complemento C4b/metabolismo , Fator I do Complemento/química , Fator I do Complemento/metabolismo , Células Endoteliais/metabolismo , Eritrócitos/metabolismo , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína , Ovinos
18.
Eur J Immunol ; 39(1): 310-23, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19065647

RESUMO

Complete deficiency of complement inhibitor factor I (FI) results in secondary complement deficiency due to uncontrolled spontaneous alternative pathway activation leading to susceptibility to infections. Current genetic examination of two patients with near complete FI deficiency and three patients with no detectable serum FI and also close family members revealed homozygous or compound heterozygous mutations in several domains of FI. These mutations were introduced into recombinant FI and the resulting proteins were purified for functional studies, while transient transfection was used to analyze expression and secretion. The G170V mutation resulted in a protein that was not expressed, whereas the mutations Q232K, C237Y, S250L, I339M and H400L affected secretion. Furthermore, the C237Y and the S250L mutants did not degrade C4b and C3b as efficiently as the WT. The truncated Q336x mutant could be expressed, in vitro, but was not functional because it lacks the serine protease domain. Furthermore, this truncated FI was not detected in serum of the patient. Structural investigations using molecular modeling were performed to predict the potential impact the mutations have on FI structure. This is the first study that investigates, at the functional level, the consequences of molecular defects identified in patients with full FI deficiency.


Assuntos
Fator I do Complemento/deficiência , Fator I do Complemento/genética , Mutação de Sentido Incorreto , Adulto , Pré-Escolar , Complemento C3b/metabolismo , Complemento C4b/metabolismo , Fator I do Complemento/química , Feminino , Humanos , Lactente , Polimorfismo Genético , Conformação Proteica , Dobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Adulto Jovem
19.
Mol Immunol ; 45(10): 2764-71, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18374984

RESUMO

Complement Factor I (CFI) is a regulator of the classical and alternative pathways. CFI has enzymatic activity and is able to cleave C3b and C4b. Homozygous Factor I deficiency is associated with infectious and/or autoimmune diseases. Here we describe the biochemical and genetic characterization in two Spanish families with complete Factor I deficiency. In Family 1, the propositus suffered from several episodes of meningitis for more than a year. Biochemical complement studies showed undetectable Factor I levels in the propositus and in her sister, while their parents and a brother had partial Factor I deficiency and were healthy. In Family 2, three out of five children were homozygous for Factor I deficiency, two of whom suffered from meningitis and the third one from several infections. The parents and the other two siblings were healthy and heterozygous for Factor I deficiency. Molecular studies showed that the two families had different mutations at exon 5 of the Factor I gene, which codifies for module LDLr1. One mutation corresponds to a 772G>A change at the donor splice site that was originally found in a family from Northern England. The second is a new missense mutation 739T>G, that generates a Cys to Gly change.


Assuntos
Fator I do Complemento/deficiência , Fator I do Complemento/genética , População Branca/genética , Adolescente , Adulto , Sequência de Bases , Western Blotting , Criança , Pré-Escolar , Fator I do Complemento/química , Mapeamento de Sequências Contíguas , Análise Mutacional de DNA , DNA Complementar , Família , Feminino , Genoma Humano/genética , Humanos , Masculino , Dados de Sequência Molecular , Mutação/genética , Linhagem , Estrutura Terciária de Proteína , Espanha
20.
Transpl Immunol ; 19(1): 30-6, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18346635

RESUMO

The use of a bioartificial liver with pig liver cells in the treatment of fulminant hepatic failure has already begun as a clinical trial in several countries. Therefore, studies on plasma complement regulatory proteins of the pig are necessary, because the liver produces them. Complement factor I is a serine protease that cleaves C3b and C4b. The DNA sequences of factor I have been reported in many species, with the noted exception of pigs. In this study, porcine factor I was cloned and an open reading frame of 1794 base pairs were identified. The predicted amino acid sequence maintained a relatively high homology compared to those of other mammals, especially in the serine protease (SP) region. The cell membrane-bound forms of the porcine and the human SP domain of factor I were constructed. Amelioration of complement-mediated cell lysis by these molecules was then tested, using several kinds of sera and Chinese hamster ovary (CHO) cell transfectants. Both molecules had a suppressing effect on pig, human and dog complements, indicating little species-specificity. Further studies of other plasma complement regulatory proteins produced from pig liver cells will need to be considered as the primary feature of the pig bioartificial liver.


Assuntos
Fator I do Complemento/imunologia , Fator I do Complemento/metabolismo , Hepatócitos/imunologia , Suínos/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Clonagem Molecular , Fator I do Complemento/química , Fator I do Complemento/genética , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Cricetinae , Cricetulus , Cães , Hepatócitos/metabolismo , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência , Serina Endopeptidases/imunologia , Serina Endopeptidases/metabolismo , Especificidade da Espécie , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA