Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 233
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hum Mol Genet ; 30(13): 1188-1199, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33783477

RESUMO

Age-related macular degeneration (AMD) is a complex neurodegenerative eye disease with behavioral and genetic etiology and is the leading cause of irreversible vision loss among elderly Caucasians. Functionally significant genetic variants in the alternative pathway of complement have been strongly linked to disease. More recently, a rare variant in the terminal pathway of complement has been associated with increased risk, Complement component 9 (C9) P167S. To assess the functional consequence of this variant, C9 levels were measured in two independent cohorts of AMD patients. In both cohorts, it was demonstrated that the P167S variant was associated with low C9 plasma levels. Further analysis showed that patients with advanced AMD had elevated sC5b-9 compared to those with non-advanced AMD, although this was not associated with the P167S polymorphism. Electron microscopy of membrane attack complexes (MACs) generated using recombinantly produced wild type or P167S C9 demonstrated identical MAC ring structures. In functional assays, the P167S variant displayed a higher propensity to polymerize and a small increase in its ability to induce hemolysis of sheep erythrocytes when added to C9-depleted serum. The demonstration that this C9 P167S AMD risk polymorphism displays increased polymerization and functional activity provides a rationale for the gene therapy trials of sCD59 to inhibit the terminal pathway of complement in AMD that are underway.


Assuntos
Complemento C9/genética , Predisposição Genética para Doença/genética , Degeneração Macular/genética , Mutação , Idoso , Animais , Células CHO , Estudos de Casos e Controles , Estudos de Coortes , Complemento C9/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/metabolismo , Cricetinae , Cricetulus , Feminino , Cobaias , Hemólise , Humanos , Degeneração Macular/sangue , Degeneração Macular/metabolismo , Masculino , Polimerização , Fatores de Risco , Ovinos
2.
BJOG ; 128(8): 1282-1291, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33539617

RESUMO

OBJECTIVE: To study genetic variants and their function within genes coding for complement receptors in pre-eclampsia. DESIGN: A case-control study. SETTING: Pre-eclampsia is a common vascular disease of pregnancy. The clearance of placenta-derived material is one of the functions of the complement system in pregnancy. POPULATION: We genotyped 500 women with pre-eclamptic pregnancies and 190 pregnant women without pre-eclampsia, as controls, from the FINNPEC cohort, and 122 women with pre-eclamptic pregnancies and 1905 controls from the national FINRISK cohort. METHODS: The functional consequences of genotypes discovered by targeted exomic sequencing were explored by analysing the binding of the main ligand iC3b to mutated CR3 or CR4, which were transiently expressed on the surface of COS-1 cells. MAIN OUTCOME MEASURES: Allele frequencies were compared between pre-eclamptic pregnancies and controls in genetic studies. The functional consequences of selected variants were measured by binding assays. RESULTS: The most significantly pre-eclampsia-linked CR3 variant M441K (P = 4.27E-4, OR = 1.401, 95% CI = 1.167-1.682) displayed a trend of increased adhesion to iC3b (P = 0.051). The CR4 variant A251T was found to enhance the adhesion of CR4 to iC3b, whereas W48R resulted in a decrease of the binding of CR4 to iC3b. CONCLUSIONS: Results suggest that changes in complement-facilitated phagocytosis are associated with pre-eclampsia. Further studies are needed to ascertain whether aberrant CR3 and CR4 activity leads to altered pro- and anti-inflammatory cytokine responses in individuals carrying the associated variants, and the role of these receptors in pre-eclampsia pathogenesis. TWEETABLE ABSTRACT: Genetic variants of complement receptors CR3 and CR4 have functional consequences that are associated with pre-eclampsia.


Assuntos
Antígeno CD11b/genética , Integrina alfaXbeta2/genética , Antígeno de Macrófago 1/genética , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/imunologia , Antígenos CD18/metabolismo , Citocinas/biossíntese , Feminino , Genótipo , Humanos , Integrina alfaXbeta2/metabolismo , Antígeno de Macrófago 1/metabolismo , Mutação , Fagocitose , Gravidez
3.
J Intern Med ; 277(3): 294-305, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25495259

RESUMO

First identified in human serum in the late 19th century as a 'complement' to antibodies in mediating bacterial lysis, the complement system emerged more than a billion years ago probably as the first humoral immune system. The contemporary complement system consists of nearly 60 proteins in three activation pathways (classical, alternative and lectin) and a terminal cytolytic pathway common to all. Modern molecular biology and genetics have not only led to further elucidation of the structure of complement system components, but have also revealed function-altering rare variants and common polymorphisms, particularly in regulators of the alternative pathway, that predispose to human disease by creating 'hyperinflammatory complement phenotypes'. To treat these 'complementopathies', a monoclonal antibody against the initiator of the membrane attack complex, C5, has received approval for use. Additional therapeutic reagents are on the horizon.


Assuntos
Transtornos das Proteínas Sanguíneas/genética , Proteínas do Sistema Complemento/genética , Anticorpos Monoclonais Humanizados/uso terapêutico , Transtornos das Proteínas Sanguíneas/imunologia , Transtornos das Proteínas Sanguíneas/terapia , Ativação do Complemento/genética , Ativação do Complemento/imunologia , Ativação do Complemento/fisiologia , Fator H do Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/fisiologia , Síndrome Hemolítico-Urêmica/imunologia , Humanos , Degeneração Macular/imunologia , Mutação/genética , Polimorfismo Genético/genética
4.
Genes Immun ; 12(4): 270-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21270825

RESUMO

Systemic lupus erythematosus (SLE) is a prototypic autoimmune disorder with a complex pathogenesis in which genetic, hormonal and environmental factors have a role. Rare mutations in the TREX1 gene, the major mammalian 3'-5' exonuclease, have been reported in sporadic SLE cases. Some of these mutations have also been identified in a rare pediatric neurological condition featuring an inflammatory encephalopathy known as Aicardi-Goutières syndrome (AGS). We sought to investigate the frequency of these mutations in a large multi-ancestral cohort of SLE cases and controls. A total of 40 single-nucleotide polymorphisms (SNPs), including both common and rare variants, across the TREX1 gene, were evaluated in ∼8370 patients with SLE and ∼7490 control subjects. Stringent quality control procedures were applied, and principal components and admixture proportions were calculated to identify outliers for removal from analysis. Population-based case-control association analyses were performed. P-values, false-discovery rate q values, and odds ratios (OR) with 95% confidence intervals (CI) were calculated. The estimated frequency of TREX1 mutations in our lupus cohort was 0.5%. Five heterozygous mutations were detected at the Y305C polymorphism in European lupus cases but none were observed in European controls. Five African cases incurred heterozygous mutations at the E266G polymorphism and, again, none were observed in the African controls. A rare homozygous R114H mutation was identified in one Asian SLE patient, whereas all genotypes at this mutation in previous reports for SLE were heterozygous. Analysis of common TREX1 SNPs (minor allele frequency (MAF)>10%) revealed a relatively common risk haplotype in European SLE patients with neurological manifestations, especially seizures, with a frequency of 58% in lupus cases compared with 45% in normal controls (P=0.0008, OR=1.73, 95% CI=1.25-2.39). Finally, the presence or absence of specific autoantibodies in certain populations produced significant genetic associations. For example, a strong association with anti-nRNP was observed in the European cohort at a coding synonymous variant rs56203834 (P=2.99E-13, OR=5.2, 95% CI=3.18-8.56). Our data confirm and expand previous reports and provide additional support for the involvement of TREX1 in lupus pathogenesis.


Assuntos
Exodesoxirribonucleases/genética , Lúpus Eritematoso Sistêmico/genética , Fosfoproteínas/genética , Estudos de Coortes , Feminino , Haplótipos , Humanos , Lúpus Eritematoso Sistêmico/epidemiologia , Masculino , Mutação , Fenótipo , Polimorfismo de Nucleotídeo Único
6.
Curr Top Microbiol Immunol ; 329: 31-57, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19198561

RESUMO

Measles virus (MV) was isolated in 1954 (Enders and Peeble 1954). It is among the most contagious of viruses and a leading cause of mortality in children in developing countries (Murray and Lopez 1997; Griffin 2001; Bryce et al. 2005). Despite intense research over decades on the biology and pathogenesis of the virus and the successful development in 1963 of an effective MV vaccine (Cutts and Markowitz 1994), cell entry receptor(s) for MV remained unidentified until 1993. Two independent studies showed that transfection of nonsusceptible rodent cells with human CD46 renders these cells permissive to infection with the Edmonston and Halle vaccine strains of measles virus (Dorig et al. 1993; Naniche et al. 1993). A key finding in these investigations was that MV binding and infection was inhibited by monoclonal and polyclonal antibodies to CD46. These reports established CD46 as a MV cell entry receptor. This chapter summarizes the role of CD46 in measles virus infection.


Assuntos
Vírus do Sarampo/fisiologia , Sarampo/imunologia , Proteína Cofatora de Membrana/imunologia , Receptores Virais/imunologia , Animais , Sítios de Ligação , Linhagem Celular , Expressão Gênica , Hemaglutininas Virais/química , Hemaglutininas Virais/genética , Hemaglutininas Virais/imunologia , Humanos , Sarampo/virologia , Vírus do Sarampo/imunologia , Vírus do Sarampo/patogenicidade , Proteína Cofatora de Membrana/química , Proteína Cofatora de Membrana/genética , Camundongos , Camundongos Transgênicos , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Receptores Virais/química , Receptores Virais/genética
7.
Mol Immunol ; 44(12): 3162-7, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17368771

RESUMO

Hemolytic uremic syndrome is the clinical triad of thrombocytopenia, microangiopathic hemolytic anaemia and acute renal failure. Cases not associated with a preceding Shiga-like toxin producing Escherichia coli are described as atypical HUS (aHUS). Approximately 50% of patients with aHUS have mutations in one of three complement regulatory proteins, Factor H (CFH), membrane cofactor protein (MCP;CD46) or factor I (IF). A common feature of these three proteins is that they regulate complement by cofactor activity. Decay accelerating factor (DAF; CD55) regulates the complement system by disassociating the alternative and classical pathway convertases. Like CFH and MCP, the gene for DAF lies within the regulators of complement activation (RCA) gene cluster at 1q32. In 1998, we described linkage to this region in families with aHUS which led to the discovery of mutations in CFH and MCP. We therefore genotyped DAF in a panel of 46 aHUS patients including families with linkage to the RCA cluster. A mutation, I197V, was identified in one patient with familial HUS which was not found in 100 healthy controls. Molecular modelling of this mutation shows that the I197V mutation does not reside in an area which would be predicted to be important in decay accelerating activity. The expression of I197V on EBV-transformed B lymphocytes was equivalent to that of wild type controls. There was no significant decrease in decay acceleration activity of the recombinantly produced I197V mutant compared with wild type, as measured by a complement-mediated lytic assay. In conclusion, this study, identifies only one mutation in DAF in 46 patients with aHUS. This mutation, I197V, does not impair complement regulation and cannot be implicated in the pathogenesis of aHUS in this patient. This suggests that the complement regulatory abnormality in aHUS is principally one of deficient cofactor activity rather than of decay acceleration activity.


Assuntos
Antígenos CD55/genética , Proteínas do Sistema Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Mutação de Sentido Incorreto , Fator H do Complemento/genética , Análise Mutacional de DNA , Saúde da Família , Fibrinogênio , Síndrome Hemolítico-Urêmica/etiologia , Humanos , Proteína Cofatora de Membrana/genética , Modelos Moleculares , Mutação
8.
Biochem Soc Trans ; 30(Pt 6): 990-6, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12440959

RESUMO

Members of the regulator of complement activation (RCA) protein family perform a vital role in health and disease. In this report we describe our efforts to solve the structures of human membrane cofactor protein (CD46), the vaccinia virus complement control protein, which mimics mammalian RCA proteins, and human complement receptor type 1 (CD35). These examples illustrate that, despite good progress over the last decade, the regulators of complement, as extracellular multiple domain glycoproteins, still pose formidable problems to structural biologists. Many important questions remain unanswered, in particular with regard to the flexibility of these proteins and the extent to which they undergo conformational rearrangements on engaging their binding partners.


Assuntos
Antígenos CD/química , Ativação do Complemento , Glicoproteínas de Membrana/química , Receptores de Complemento 3b/química , Proteínas Virais/química , Animais , Antígenos CD/metabolismo , Sítios de Ligação , Células CHO , Cricetinae , Humanos , Espectroscopia de Ressonância Magnética , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Virais/metabolismo
9.
Transfusion ; 42(2): 251-6, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11896343

RESUMO

BACKGROUND: Complement receptor type 1 (CR1), which bears the Knops (Kn [KN]) blood group antigens, is involved in the rosetting of Plasmodium falciparum- infected RBCs with uninfected cells. As a first step in understanding this interaction, the molecular basis for the blood group antigens encoded by CR1 was investigated. STUDY DESIGN AND METHODS: An antibody from a white donor who exhibited an apparent anti-Sl(a) was used for population studies of several racial groups. The donor's genomic DNA was sequenced to identify the Sl(a) mutation and other mutations. RESULTS: The donor with anti-Sl(a) typed as Sl(a+) with some sera and had the CR1 genotype AA at bp 4828 (R1601). However, she was homozygous for a new mutation (GG) at bp 4855 changing amino acid 1610 from S1610 to T1610 (S1610T). This mutation occurred in heterozygous form in eight white and one Asian donor. The site is only nine amino acids from the previously described Sl(a) polymorphism and appears to produce a new conformational epitope. CONCLUSION: The antigen formerly known as Sl(a) can now be subdivided. A new terminology is proposed that recognizes both linear and conformational epitopes on the CR1 protein. At amino acid 1601, Sl 1 (Sl(a)) is represented by R, Sl 2 (Vil) is represented by glycine, and Sl 3 requires both R1601 and S1610. Sl 4 and Sl 5 are hypothetical epitopes represented by S1610 and T1610, respectively.


Assuntos
Antígenos de Superfície/genética , Antígenos de Grupos Sanguíneos/genética , Receptores de Complemento/genética , Sequência de Aminoácidos , Antígenos de Superfície/química , Antígenos de Superfície/imunologia , Povo Asiático , População Negra , Antígenos de Grupos Sanguíneos/imunologia , Clonagem Molecular , Sequência Consenso , Epitopos/química , Heterozigoto , Homozigoto , Humanos , Isoanticorpos/sangue , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Hibridização de Ácido Nucleico , Sondas de Oligonucleotídeos , Reação em Cadeia da Polimerase , Receptores de Complemento/química , Receptores de Complemento/imunologia , Sequências Repetitivas de Aminoácidos , Análise de Sequência de DNA , População Branca
10.
Am J Hum Genet ; 69(2): 447-53, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11438888

RESUMO

We performed a genomewide search for linkage in an extended Dutch family with hereditary vascular retinopathy associated with migraine and Raynaud phenomenon. Patients with vascular retinopathy are characterized by microangiopathy of the retina, accompanied by microaneurysms and telangiectatic capillaries. The genome search, using a high throughput capillary sequencer, revealed significant evidence of linkage to chromosome 3p21.1-p21.3 (maximum pairwise LOD score 5.25, with D3S1578). Testing of two additional families that had a similar phenotype, cerebroretinal vasculopathy, and hereditary endotheliopathy with retinopathy, nephropathy, and stroke, revealed linkage to the same chromosomal region (combined maximum LOD score 6.30, with D3S1588). Haplotype analysis of all three families defined a 3-cM candidate region between D3S1578 and D3S3564. Our study shows that three autosomal dominant vasculopathy syndromes with prominent cerebroretinal manifestations map to the same 3-cM interval on 3p21, suggesting a common locus.


Assuntos
Mapeamento Cromossômico , Cromossomos Humanos Par 3/genética , Nefropatias/genética , Doenças Retinianas/genética , Acidente Vascular Cerebral/genética , Doenças Vasculares/genética , Feminino , Haplótipos/genética , Humanos , Nefropatias/patologia , Escore Lod , Masculino , Repetições de Microssatélites/genética , Pessoa de Meia-Idade , Transtornos de Enxaqueca/complicações , Transtornos de Enxaqueca/genética , Dados de Sequência Molecular , Países Baixos , Linhagem , Penetrância , Doença de Raynaud/complicações , Doença de Raynaud/genética , Doenças Retinianas/complicações , Doenças Retinianas/patologia , Doenças Vasculares/complicações , Doenças Vasculares/patologia
11.
Immunol Rev ; 180: 112-22, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11414353

RESUMO

Human complement receptor type 1 (CR1) is a large, multifunctional glycoprotein which is a member of the regulators of complement activation family. Like other members of this family, it is composed mainly of tandemly arranged modules, each about 60-70 amino acids long, known as complement control protein repeats (CCPs). Each domain folds independently and contains a hydrophobic core wrapped in beta sheets. These domains mediate interactions with C3/C4-derived fragments. CR1 is the most versatile inhibitor of both classical and alternative pathway C3 and C5 convertases due to its decay-accelerating activity and co-factor activity for C3b/C4b cleavage. Moreover, CR1 plays a major role in immune complex clearance due to its high affinity for C3b and C4b. CR1 is an excellent model to study structure-function relationships because its functions are mediated by two distinct but highly homologous sites, each composed of three CCPs. CR1 derivatives carrying just one active site were used to define critical sequences/amino acids. This was achieved by testing functional profiles of the proteins carrying a mutated active site produced by substituting peptides/amino acids with their counterparts from the other site. These mutated proteins, of which we analyzed over 100, permitted the fine mapping of the functional sites. CR1 on primate erythrocytes varies in size. In most cases it is smaller and has fewer active sites than does human CR1. This variation was used to determine that increased copy number (3,000 to 20,000 versus 300 for human CR1) compensates for a smaller size. Moreover, studies of primate CR1 led to the finding that subtle differences in the critical areas, as compared to human sites, produce active sites with a broader functional repertoire. These alterations ensure that short CR1 forms possess similar biologic activities to the large CR1 forms. There is much interest in producing therapeutic agents to inhibit unwanted complement activation. Based on these structure-function analyses, smaller and more potent complement inhibitors derived from CR1 can be produced.


Assuntos
Receptores de Complemento 3b/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células Sanguíneas/metabolismo , Ativação do Complemento , Convertases de Complemento C3-C5/antagonistas & inibidores , Complemento C3b/metabolismo , Complemento C4b/metabolismo , Desenho de Fármacos , Humanos , Mamíferos/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Polimorfismo Genético , Primatas/sangue , Primatas/imunologia , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Receptores de Complemento 3b/imunologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Relação Estrutura-Atividade
12.
Clin Exp Immunol ; 124(2): 180-9, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11422193

RESUMO

Human membrane cofactor protein (MCP; CD46) is a widely distributed complement regulator. In the mouse, expression of MCP is largely restricted to the testis while a related, widely expressed protein (Crry) appears to perform MCP's (CD46) regulatory activity. We have developed two mouse strains transgenic for human MCP (CD46) utilizing an approximately 400 kb YAC clone carrying the complete gene. A third mouse strain was generated using an overlapping YAC clone isolated from a second library. The expression of human MCP (CD46) in these mouse strains was characterized by immunohistochemistry, FACS, Western blotting and RT-PCR. No differences were detected in the isoform pattern or distribution among the three strains, although the expression level varied according to how many copies of the gene were integrated. The expression profile closely mimicked that observed in humans, including the same pattern of isoform expression as the donor. In addition, tissue-specific isoform expression in the kidney, salivary gland and brain paralleled that observed in man. The transgenic mice expressed low levels of MCP (CD46) on their E, in contrast to humans but in line with most other primates. These mice should be a useful tool to analyse tissue-specific expression, to establish animal models of infections and to characterize the role of MCP (CD46) in reproduction.


Assuntos
Antígenos CD/biossíntese , Glicoproteínas de Membrana/biossíntese , Receptores Virais/biossíntese , Animais , Antígenos CD/genética , Células Sanguíneas/química , Eritrócitos/química , Feminino , Humanos , Masculino , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Isoformas de Proteínas , Splicing de RNA , Receptores Virais/genética , Especificidade da Espécie , Distribuição Tecidual
15.
Blood ; 97(9): 2879-85, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11313284

RESUMO

Complement receptor 1 (CR1) has been implicated in rosetting of uninfected red blood cells to Plasmodium falciparum-infected cells, and rosette formation is associated with severe malaria. The Knops blood group (KN) is located on CR1 and some of these antigens, ie, McCoy (McC) and Swain-Langley (Sl(a)), show marked frequency differences between Caucasians and Africans. Thus, defining the molecular basis of these antigens may provide new insight into the mechanisms of P falciparum malaria. Monoclonal antibody epitope mapping and serologic inhibition studies using CR1 deletion constructs localized McC and Sl(a) to long homologous repeat D of CR1. Direct DNA sequencing of selected donors identified several single nucleotide polymorphisms in exon 29 coding for complement control protein modules 24 and 25. Two of these appeared to be blood group specific: McC associated with K1590E and Sl(a) with R1601G. These associations were confirmed by inhibition studies using allele-specific mutants. A sequence-specific oligonucleotide probe hybridization assay was developed to genotype several African populations and perform family inheritance studies. Concordance between the 1590 mutation and McC was 94%; that between Sl(a) and 1601 was 88%. All but 2 samples exhibiting discrepancies between the genotype and phenotype were found to be due to low red cell CR1 copy numbers, low or absent expression of some alleles, or heterozygosity combined with low normal levels of CR1. These data further explain the variability observed in previous serologic studies of CR1 and show that DNA and protein-based genetic studies will be needed to clarify the role of the KN antigens in malaria.


Assuntos
Antígenos de Grupos Sanguíneos/genética , Receptores de Complemento 3b/genética , Animais , Antígenos de Grupos Sanguíneos/imunologia , Tipagem e Reações Cruzadas Sanguíneas , Eritrócitos/imunologia , Humanos , Plasmodium falciparum , Polimorfismo Genético , Receptores de Complemento 3b/imunologia
16.
Cell Microbiol ; 3(3): 133-43, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11260136

RESUMO

Pili of Neisseria gonorrhoeae mediate binding of the bacteria to human host cells. Membrane cofactor protein (MCP or CD46), a human cell-surface protein involved in regulation of complement activation, acts as a cellular pilus receptor. In this work, we examined which domains of CD46 mediate bacterial adherence. The CD46 expression was quantified and characterized in human epithelial cell lines. N. gonorrhoeae showed the highest adherence to ME180 cells, which have BC1 as the dominant phenotype. The BC isoforms of CD46 were expressed in all cell lines tested. The adherence was not enhanced by high expression of other isoforms, showing that the BC domain of CD46 is important in adherence of N. gonorrhoeae to human cells. To characterize the pilus-binding site within the CD46 molecule, a set of CD46-BC1 deletion constructs were transfected into COS-7 cells. Piliated N. gonorrhoeae attached well to CD46-BC1-expressing COS-7 cells. We show that the complement control protein repeat 3 (CCP-3) and the serine-threonine-proline (STP)-rich domain of CD46 are important for efficient adherence to host cells. Further, partial deletion of the cytoplasmic tail of CD46 results in low bacterial binding, indicating that the cytoplasmic tail takes part in the process of establishing a stable interaction between N. gonorrhoeae and host cells.


Assuntos
Antígenos CD/química , Antígenos CD/metabolismo , Aderência Bacteriana/fisiologia , Fímbrias Bacterianas/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Neisseria gonorrhoeae/patogenicidade , Animais , Antígenos CD/genética , Western Blotting , Células COS , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Células Epiteliais/microbiologia , Gonorreia/microbiologia , Humanos , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/genética , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/metabolismo , Estrutura Terciária de Proteína , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
17.
J Immunol ; 165(11): 6341-6, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11086071

RESUMO

The malaria parasite Plasmodium falciparum induces a number of novel adhesion properties in the erythrocytes that it infects. One of these properties, the ability of infected erythrocytes to bind uninfected erythrocytes to form rosettes, is associated with severe malaria and may play a direct role in the pathogenesis of disease. Previous work has shown that erythrocytes deficient in complement receptor (CR) 1 (CR1, CD35; C3b/C4b receptor) have greatly reduced rosetting capacity, indicating an essential role for CR1 in rosette formation. Using deletion mutants and mAbs, we have localized the region of CR1 required for the formation of P. falciparum rosettes to the area of long homologous repeat regions B and C that also acts as the binding site for the activated complement component C3b. This result raises the possibility that C3b could be an intermediary in rosetting, bridging between the infected erythrocyte and CR1. We were able to exclude this hypothesis, however, as parasites grown in C3-deficient human serum formed rosettes normally. We have also shown in this report that rosettes can be reversed by mAb J3B11 that recognizes the C3b binding site of CR1. This rosette-reversing activity was demonstrated in a range of laboratory-adapted parasite strains and field isolates from Kenya and Malawi. Thus, we have mapped the region of CR1 required for rosetting and demonstrated that the CR1-dependent rosetting mechanism occurs commonly in P. falciparum isolates, and could therefore be a potential target for future therapeutic interventions to treat severe malaria.


Assuntos
Plasmodium falciparum/imunologia , Plasmodium falciparum/isolamento & purificação , Receptores de Complemento 3b/fisiologia , Formação de Roseta , Animais , Anticorpos Monoclonais/farmacologia , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Sequência Consenso/genética , Sequência Consenso/imunologia , Dimerização , Mapeamento de Epitopos/métodos , Eritrócitos/imunologia , Eritrócitos/parasitologia , Humanos , Plasmodium falciparum/crescimento & desenvolvimento , Receptores de Complemento 3b/sangue , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/imunologia , Sequências Repetitivas de Ácido Nucleico , Deleção de Sequência/imunologia , Homologia de Sequência do Ácido Nucleico
18.
J Immunol ; 165(7): 3999-4006, 2000 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11034410

RESUMO

Decay-accelerating factor (DAF or CD55) and membrane cofactor protein (MCP or CD46) function intrinsically in the membranes of self cells to prevent activation of autologous complement on their surfaces. How these two regulatory proteins cooperate on self-cell surfaces to inhibit autologous complement attack is unknown. In this study, a GPI-anchored form of MCP was generated. The ability of this recombinant protein and that of naturally GPI-anchored DAF to incorporate into cell membranes then was exploited to examine the combined functions of DAF and MCP in regulating complement intermediates assembled from purified alternative pathway components on rabbit erythrocytes. Quantitative studies with complement-coated rabbit erythrocyte intermediates constituted with each protein individually or the two proteins together demonstrated that DAF and MCP synergize the actions of each other in preventing C3b deposition on the cell surface. Further analyses showed that MCP's ability to catalyze the factor I-mediated cleavage of cell-bound C3b is inhibited in the presence of factors B and D and is restored when DAF is incorporated into the cells. Thus, the activities of DAF and MCP, when present together, are greater than the sum of the two proteins individually, and DAF is required for MCP to catalyze the cleavage of cell-bound C3b in the presence of excess factors B and D. These data are relevant to xenotransplantation, pharmacological inhibition of complement in inflammatory diseases, and evasion of tumor cells from humoral immune responses.


Assuntos
Adjuvantes Imunológicos/fisiologia , Antígenos CD/fisiologia , Antígenos CD55/fisiologia , Proteínas Inativadoras do Complemento C3b/fisiologia , Via Alternativa do Complemento/imunologia , Glicoproteínas de Membrana/fisiologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Células CHO , C3 Convertase da Via Alternativa do Complemento , Convertases de Complemento C3-C5 , Complemento C3b/imunologia , Complemento C3b/metabolismo , Complemento C3b/fisiologia , Fator B do Complemento/fisiologia , Cricetinae , Sinergismo Farmacológico , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/metabolismo , Humanos , Células K562 , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Fragmentos de Peptídeos/fisiologia , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo
19.
J Biol Chem ; 275(48): 37692-701, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10960475

RESUMO

Membrane cofactor protein (MCP; CD46), a widely distributed regulator of complement activation, is a cofactor for the factor I-mediated degradation of C3b and C4b deposited on host cells. MCP possesses four extracellular, contiguous complement control protein modules (CCPs) important for this inhibitory activity. The goal of the present study was to delineate functional sites within these modules. We employed multiple approaches including mutagenesis, epitope mapping, and comparisons to primate MCP to make the following observations. First, functional sites were located to each of the four CCPs. Second, some residues were important for both C3b and C4b interactions while others were specific for one or the other. Third, while a reduction in ligand binding was invariably accompanied by a parallel reduction in cofactor activity (CA), other mutants lost or had reduced CA but retained ligand binding. Fourth, two C4b-regulatory domains overlapped measles virus interactive regions, indicating that the hemagglutinin docks to a site important for complement inhibition. Fifth, several MCP regulatory areas corresponded to functionally critical, homologous positions in other CCP-bearing C3b/C4b-binding proteins. Based on these data and the recently derived crystal structure of repeats one and two, computer modeling was employed to predict MCP structure and examine active sites.


Assuntos
Antígenos CD/metabolismo , Glicoproteínas de Membrana/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/imunologia , Sítios de Ligação , Células CHO , Cricetinae , Mapeamento de Epitopos , Humanos , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Conformação Proteica
20.
Dev Comp Immunol ; 24(8): 815-27, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10906393

RESUMO

The decay accelerating factor (DAF, CD55) protects self cells from activation of autologous complement on their surfaces. It functions to disable the C3 convertases, the central amplification enzymes of the cascade. Its active site(s) are contained within four approximately 60 amino acid long units, termed complement control protein repeats (CCPs), which are suspended above the cell surface on a 68 amino acid long serine/threonine (S/T)-rich cushion that derives from three exons. We previously proposed a molecular model of human DAF's four CCPs in which certain amino acids were postulated to be recognition sites for the interaction between DAF and the C3 convertases. In the current study, we characterized DAF in five non-human primates: the great apes, gorilla and common chimpanzee, and the Old World monkeys: hamadryas baboon, Rhesus macaque, and patas monkey. Amino acid homology to human DAF was approximately 98% for the two great apes and 83% for the three Old World monkeys. The above cited putative ligand interactive residues were found to be fully conserved in all of the non-human primates, although there were amino acid changes outside of these areas. In the chimpanzee, alternative splicing of the S/T region was found potentially to be the source of multiple protein isoforms in erythrocytes, whereas in the patas monkey, similar alternative splicing was observed but only one protein band was seen. Interestingly, a Rhesus macaque was found to exhibit a phenomenon paralleling the human Cromer Dr(a-) blood group, in which a 44-base pair deletion in CCP3 leads to a frameshift and early STOP codon.


Assuntos
Antígenos CD55/genética , Primatas/imunologia , Processamento Alternativo , Sequência de Aminoácidos , Animais , Antígenos CD55/química , Linhagem Celular , Complemento C3 , Erythrocebus patas/imunologia , Eritrócitos/imunologia , Deleção de Genes , Variação Genética , Gorilla gorilla/imunologia , Leucócitos Mononucleares/imunologia , Macaca mulatta/imunologia , Masculino , Dados de Sequência Molecular , Pan troglodytes/imunologia , Papio/imunologia , Primatas/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...