Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 9(1): 5316, 2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30552328

RESUMO

The membrane attack complex (MAC) is one of the immune system's first responders. Complement proteins assemble on target membranes to form pores that lyse pathogens and impact tissue homeostasis of self-cells. How MAC disrupts the membrane barrier remains unclear. Here we use electron cryo-microscopy and flicker spectroscopy to show that MAC interacts with lipid bilayers in two distinct ways. Whereas C6 and C7 associate with the outer leaflet and reduce the energy for membrane bending, C8 and C9 traverse the bilayer increasing membrane rigidity. CryoEM reconstructions reveal plasticity of the MAC pore and demonstrate how C5b6 acts as a platform, directing assembly of a giant ß-barrel whose structure is supported by a glycan scaffold. Our work provides a structural basis for understanding how ß-pore forming proteins breach the membrane and reveals a mechanism for how MAC kills pathogens and regulates cell functions.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/química , Complexo de Ataque à Membrana do Sistema Complemento/ultraestrutura , Microscopia Crioeletrônica/métodos , Bicamadas Lipídicas/química , Complemento C6/química , Complemento C6/metabolismo , Complemento C6/ultraestrutura , Complemento C7/química , Complemento C7/metabolismo , Complemento C7/ultraestrutura , Complemento C8/química , Complemento C8/metabolismo , Complemento C8/ultraestrutura , Complemento C9/química , Complemento C9/metabolismo , Complemento C9/ultraestrutura , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Humanos , Processamento de Imagem Assistida por Computador , Bicamadas Lipídicas/metabolismo , Lipossomos , Modelos Moleculares , Polissacarídeos/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Análise Espectral/métodos
2.
J Neuroinflammation ; 15(1): 154, 2018 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-29789012

RESUMO

BACKGROUND: Traumatic brain injury (TBI) begins with the application of mechanical force to the head or brain, which initiates systemic and cellular processes that are hallmarks of the disease. The pathological cascade of secondary injury processes, including inflammation, can exacerbate brain injury-induced morbidities and thus represents a plausible target for pharmaceutical therapies. We have pioneered research on post-traumatic sleep, identifying that injury-induced sleep lasting for 6 h in brain-injured mice coincides with increased cortical levels of inflammatory cytokines, including tumor necrosis factor (TNF). Here, we apply post-traumatic sleep as a physiological bio-indicator of inflammation. We hypothesized the efficacy of novel TNF receptor (TNF-R) inhibitors could be screened using post-traumatic sleep and that these novel compounds would improve functional recovery following diffuse TBI in the mouse. METHODS: Three inhibitors of TNF-R activation were synthesized based on the structure of previously reported TNF CIAM inhibitor F002, which lodges into a defined TNFR1 cavity at the TNF-binding interface, and screened for in vitro efficacy of TNF pathway inhibition (IκB phosphorylation). Compounds were screened for in vivo efficacy in modulating post-traumatic sleep. Compounds were then tested for efficacy in improving functional recovery and verification of cellular mechanism. RESULTS: Brain-injured mice treated with Compound 7 (C7) or SGT11 slept significantly less than those treated with vehicle, suggesting a therapeutic potential to target neuroinflammation. SGT11 restored cognitive, sensorimotor, and neurological function. C7 and SGT11 significantly decreased cortical inflammatory cytokines 3 h post-TBI. CONCLUSIONS: Using sleep as a bio-indicator of TNF-R-dependent neuroinflammation, we identified C7 and SGT11 as potential therapeutic candidates for TBI.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Complemento C7/uso terapêutico , Fatores Imunológicos/uso terapêutico , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Animais , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Complemento C7/química , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fatores Imunológicos/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Microglia/efeitos dos fármacos , Microglia/patologia , Atividade Motora/efeitos dos fármacos , Exame Neurológico , Reconhecimento Psicológico/efeitos dos fármacos , Teste de Desempenho do Rota-Rod , Transtornos do Sono-Vigília/tratamento farmacológico , Transtornos do Sono-Vigília/etiologia
3.
Mol Immunol ; 55(3-4): 400-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23582305

RESUMO

Complement-dependent cytotoxicity (CDC) is a potent promoter of tumor clearance during monoclonal antibody therapy. Complement activation on antibody-bearing tumor cells results in formation of the membrane attack complex (MAC), which activates cell death. The complement activation cascade that bridges between antibody binding and MAC formation is regulated by complement inhibitors that are over-expressed on tumor cells. In order to bypass those complement regulators, we have designed an immunoconjugate composed of a humanized single chain Fv of an anti-Tac (CD25) monoclonal antibody fused at its C terminus either to complement protein C9 (scFv-C9) or to complement C7 (scFv-C7) and tagged with six histidines at the C terminal end. Recombinant scFv-C9 and scFv-C7 were expressed in 293T cells and purified. Both are shown to efficiently bind to CD25-positive tumor cells. In addition, scFv-C9, but not scFv-C7, increases MAC deposition on the cells and enhances complement-mediated cell death of target CD25-positive cells. Thus, scFv-C9 fusion protein is potentially a novel reagent for application in cancer immunotherapy.


Assuntos
Complemento C7/química , Complemento C9/química , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Anticorpos de Cadeia Única/química , Linhagem Celular Tumoral , Complemento C7/genética , Complemento C7/metabolismo , Complemento C9/genética , Complemento C9/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/química , Complexo de Ataque à Membrana do Sistema Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Citotoxicidade Imunológica , Células HEK293 , Humanos , Imunoconjugados/química , Imunoconjugados/genética , Imunoterapia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/metabolismo
4.
PLoS One ; 8(3): e58657, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23536807

RESUMO

Glaucoma and age-related macular degeneration (AMD) are the two leading causes of visual loss in the United States. We utilized a novel study design to perform a genome-wide association for both primary open angle glaucoma (POAG) and AMD. This study design utilized a two-stage process for hypothesis generation and validation, in which each disease cohort was utilized as a control for the other. A total of 400 POAG patients and 400 AMD patients were ascertained and genotyped at 500,000 loci. This study identified a novel association of complement component 7 (C7) to POAG. Additionally, an association of central corneal thickness, a known risk factor for POAG, was found to be associated with ribophorin II (RPN2). Linked monogenic loci for POAG and AMD were also evaluated for evidence of association, none of which were found to be significantly associated. However, several yielded putative associations requiring validation. Our data suggest that POAG is more genetically complex than AMD, with no common risk alleles of large effect.


Assuntos
Estudo de Associação Genômica Ampla , Glaucoma de Ângulo Aberto/genética , Degeneração Macular/genética , Locos de Características Quantitativas , Idoso , Alelos , Sequência de Aminoácidos , Complemento C7/química , Complemento C7/genética , Córnea/patologia , Feminino , Predisposição Genética para Doença , Genótipo , Glaucoma de Ângulo Aberto/patologia , Humanos , Degeneração Macular/patologia , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Característica Quantitativa Herdável , Alinhamento de Sequência
5.
Biomol NMR Assign ; 7(2): 285-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23054935

RESUMO

Human C7 is one of four homologous complement proteins that self-assemble on the nascent activation-specific fragment, C5b, thus forming the cytolytic membrane attack complex (MAC). In addition to the conserved modular core of the MAC/perforin protein family, C7 has four C-terminal domains comprising a pair of complement control protein modules (CCPs) preceding two Factor-I like modules (FIMs). It is proposed that the C7-CCPs might serve as a molecular arm for delivery of C7-FIMs to their binding site on C5b. Here we present the NMR chemical shift assignments for the C7-CCPs produced as a 14-kDa recombinant protein. Based upon triple-resonance experiments, 98 and 94 % of the backbone and side-chain ((1)H, (13)C and (15)N) assignments, respectively, have been completed. The chemical shifts and assignments have been deposited in the BioMagResBank database under accession number 18530.


Assuntos
Complemento C7/química , Ressonância Magnética Nuclear Biomolecular , Prótons , Sequência de Aminoácidos , Isótopos de Carbono , Humanos , Dados de Sequência Molecular , Isótopos de Nitrogênio , Estrutura Secundária de Proteína , Alinhamento de Sequência
6.
Mol Immunol ; 47(4): 671-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19931914

RESUMO

Meningococcal disease is caused by Neisseria meningitidis which is associated with high morbidity and mortality. Recurrences of meningococcal infection have been observed in patients with terminal complement component defects, because of the inefficient formation of the lytic membrane attack complex (MAC), C5b-9. Complement component C7 is one of the five plasma proteins to form the MAC. The gene C7 may carry mutations that cause functional abnormalities or the mere absence of the C7 protein. More than 200 patients were screened for aberrant C7 protein by isoelectric focusing (C7 IEF). These were compared with patients in whom recurrent meningococcal infection had resulted in the diagnosis of complete C7 absence (C7Q0). A higher proportion of C7 IEF variants were found in meningitis cases compared to controls (p=0.03). In contrast to C7Q0 patients, recurrent meningococcal infection was never observed in C7 IEF cases. Whereas C7Q0 sera were defective in meningococcal serogroup B and W135 killing assays, the sera of patients with C7 IEF variants were only defective in complement-mediated killing when classical pathway activation by (endogenous) anti-meningococcal antibodies was blocked. Upon sequence analysis we characterized the genetic background of the C7*6 and C7*8 IEF pattern and identified three novel C7 gene mutations in 13 C7Q0 patients. In conclusion, C7 IEF variants can determine meningococcal killing in the early stage of infection when antibody-independent killing prevails. The results endorse the lack of clinical recurrences once antibodies are present, whereas in C7Q0 patients the anti-meningococcal antibodies may not suffice to protect from recurrent meningococcal infection.


Assuntos
Complemento C7/genética , Complemento C7/imunologia , Infecções Meningocócicas/genética , Infecções Meningocócicas/imunologia , Mutação/genética , Adolescente , Adulto , Estudos de Casos e Controles , Criança , Pré-Escolar , Complemento C7/química , Citotoxicidade Imunológica , Feminino , Humanos , Immunoblotting , Focalização Isoelétrica , Masculino , Infecções Meningocócicas/prevenção & controle , Neisseria meningitidis/classificação , Neisseria meningitidis/imunologia , Linhagem , Fenótipo , Recidiva , Sorotipagem , Adulto Jovem
7.
Biomol NMR Assign ; 3(1): 49-52, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19636945

RESUMO

The carboxy terminus of human complement component C7 comprises two Factor I-like Modules (FIMs) which are essential for formation of the Membrane Attack Complex, the terminal pathway of the innate immune system. C7-FIMs is a 16.9 kDa, recombinant, disulphide-rich, protein encompassing this C-terminal domain. Using conventional triple resonance experiments 93% of the (1)H, (15)N and (13)C assignment has been achieved, accounting for all assignment apart from a flexible N-terminus cloning artefact and an undefined loop. The chemical shifts have been deposited in the BioMagResBank; Accession No. 15996.


Assuntos
Complemento C7/química , Complemento C7/ultraestrutura , Fibrinogênio/química , Fibrinogênio/ultraestrutura , Espectroscopia de Ressonância Magnética/métodos , Sequência de Aminoácidos , Isótopos de Carbono/química , Dados de Sequência Molecular , Isótopos de Nitrogênio/química , Conformação Proteica , Estrutura Terciária de Proteína , Prótons
8.
J Biol Chem ; 284(29): 19637-49, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19419965

RESUMO

Factor I-like modules (FIMs) of complement proteins C6, C7, and factor I participate in protein-protein interactions critical to the progress of a complement-mediated immune response to infections and other trauma. For instance, the carboxyl-terminal FIM pair of C7 (C7-FIMs) binds to the C345C domain of C5 and its activated product, C5b, during self-assembly of the cytolytic membrane-attack complex. FIMs share sequence similarity with follistatin domains (FDs) of known three-dimensional structure, suggesting that FIM structures could be reliably modeled. However, conflicting disulfide maps, inconsistent orientations of subdomains within FDs, and the presence of binding partners in all FD structures led us to determine the three-dimensional structure of C7-FIMs by NMR spectroscopy. The solution structure reveals that each FIM within C7 contains a small amino-terminal FOLN subdomain connected to a larger carboxyl-terminal KAZAL domain. The open arrangement of the subdomains within FIMs resembles that of first FDs within structures of tandem FDs but differs from the more compact subdomain arrangement of second or third FDs. Unexpectedly, the two C7-FIMs pack closely together with an approximate 2-fold rotational symmetry that is rarely seen in module pairs and has not been observed in FD-containing proteins. Interfaces between subdomains and between modules include numerous hydrophobic and electrostatic contributions, suggesting that this is a physiologically relevant conformation that persists in the context of the parent protein. Similar interfaces were predicted in a homology-based model of the C6-FIM pair. The C7-FIM structures also facilitated construction of a model of the single FIM of factor I.


Assuntos
Complemento C7/química , Modelos Moleculares , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Sítios de Ligação/genética , Complemento C7/genética , Complemento C7/metabolismo , Folistatina/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Conformação Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Soluções , Eletricidade Estática
9.
J Biol Chem ; 280(11): 10636-45, 2005 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-15598652

RESUMO

The complement protein C5 initiates assembly of the membrane attack complex. This remarkable process results in lysis of target cells and is fundamental to mammalian defense against infection. The 150-amino acid residue domain at the C terminus of C5 (C5-C345C) is pivotal to C5 function. It interacts with enzymes that convert C5 to C5b, the first step in the assembly of the membrane attack complex; it also binds to the membrane attack complex components C6 and C7 with high affinity. Here a recombinant version of this C5-C345C domain is shown to adopt the oligosaccharide/oligonucleotide binding fold, with two helices packed against a five-stranded beta-barrel. The structure is compared with those from the netrin-like module family that have a similar fold. Residues critical to the interaction with C5-convertase cluster on a mobile, hydrophobic inter-strand loop that protrudes from the open face of the beta-barrel. The opposite, helix-dominated face of C5-C345C carries a pair of exposed hydrophobic side chains adjacent to a striking negatively charged patch, consistent with affinity for positively charged factor I modules in C6 and C7. Modeling of homologous domains from complement proteins C3 and C4, which do not participate in membrane attack complex assembly, suggests that this provisionally identified C6/C7-interacting face is indeed specific to C5.


Assuntos
Complemento C5/química , Sequência de Aminoácidos , Membrana Celular/metabolismo , Complemento C6/química , Complemento C7/química , Escherichia coli/metabolismo , Vetores Genéticos , Humanos , Cinética , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Oligonucleotídeos/química , Oligossacarídeos/química , Mutação Puntual , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Estereoisomerismo , Ressonância de Plasmônio de Superfície , Fatores de Tempo
10.
J Immunol ; 173(7): 4547-52, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15383587

RESUMO

Studies reported over 30 years ago revealed that latent, nonactivated C5 binds specifically and reversibly to C6 and C7. These reversible reactions are distinct from the essentially nonreversible associations with activated C5b that occur during assembly of the membrane attack complex, but they likely involve some, perhaps many, of the same molecular contacts. We recently reported that these reversible reactions are mediated by the C345C (NTR) domain at the C terminus of the C5 alpha-chain. Earlier work by others localized the complementary binding sites to a tryptic fragment of C6 composed entirely of two adjacent factor I modules (FIMs), and to a larger fragment of C7 composed of its homologous FIMs as well as two adjoining short consensus repeat modules. In this work, we expressed the tandem FIMs from C7 in bacteria. The mobility on SDS-polyacrylamide gels, lack of free sulfhydryl groups, and atypical circular dichroism spectrum of the recombinant product rC7-FIMs were all consistent with a native structure. Using surface plasmon resonance, we found that rC7-FIMs binds specifically to both C5 and the rC5-C345C domain with K(D) approximately 50 nM, and competes with C7 for binding to C5, as expected for an active domain. These results indicate that, like C6, the FIMs alone in C7 mediate reversible binding to C5. Based on available evidence, we suggest a model for an irreversible membrane attack complex assembly in which the C7 FIMs, but not those in C6, are bound to the C345C domain of C5 within the fully assembled complex.


Assuntos
Complemento C5/metabolismo , Complemento C7/metabolismo , Fator I do Complemento/metabolismo , Proteínas Recombinantes/metabolismo , Motivos de Aminoácidos/genética , Dicroísmo Circular , Complemento C3/química , Complemento C6/química , Complemento C6/genética , Complemento C6/metabolismo , Complemento C7/química , Complemento C7/genética , Fator I do Complemento/química , Fator I do Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/química , Complexo de Ataque à Membrana do Sistema Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Dissulfetos/química , Escherichia coli/genética , Humanos , Receptores de Netrina , Oxirredução , Ligação Proteica/genética , Ligação Proteica/imunologia , Estrutura Terciária de Proteína/genética , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Ressonância de Plasmônio de Superfície
11.
Genes Immun ; 4(1): 54-9, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12595902

RESUMO

To study the transcriptional regulation of the human complement component C7, a 1 kb promoter fragment was cloned and the transcription start site was determined. C7 is expressed by the hepatoma-derived cell line Hep-3B, but not by Hep-G2. Transfection of these cell lines with different C7 promoter-luciferase constructs demonstrated that 1 kb of the 5'-flanking region contains the necessary elements for driving C7 transcription in a tissue-specific manner and showed that the sequence between -29/+102 retained the majority of C7 promoter activity in Hep-3B. Electrophoretic mobility shift assays suggested that the binding of the C/EBPalpha transcription factor to a C/EBP sequence located at +42 is essential for C7 expression. To investigate whether the absence of C/EBPalpha expression in Hep-G2 cells is responsible for the lack of C7 transcription, Hep-G2 cells were transfected with a C/EBPalpha expression vector. C/EBPalpha transactivated the C7 luciferase reported gene and restored the C7 expression in Hep-G2 cells.


Assuntos
Clonagem Molecular/métodos , Complemento C7/genética , Regiões Promotoras Genéticas/genética , Sequência de Bases , Linhagem Celular , Complemento C7/química , Regulação da Expressão Gênica/imunologia , Humanos , Dados de Sequência Molecular , Células Tumorais Cultivadas
12.
Biochemistry ; 40(50): 15127-34, 2001 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-11735395

RESUMO

We have determined the X-ray crystal structure to 1.8 A resolution of the Ca(2+) complex of complement-like repeat 7 (CR7) from the low-density lipoprotein receptor-related protein (LRP) and characterized its calcium binding properties at pH 7.4 and 5. CR7 occurs in a region of the LRP that binds to the receptor-associated protein, RAP, and other protein ligands in a Ca(2+)-dependent manner. The calcium coordination is identical to that found in LB5 and consists of carboxyls from three conserved aspartates and one conserved glutamate, and the backbone carbonyls of a tryptophan and another aspartate. The overall fold of CR7 is similar to those of CR3 and CR8 from the LRP and LB5 from the LDL receptor, though the low degree of sequence homology of residues not involved in calcium coordination or in disulfide formation results in a distinct pattern of surface residues for each domain, including CR7. The thermodynamic parameters for Ca(2+) binding at both extracellular and endosomal pHs were determined by isothermal titration calorimetry for CR7 and for related complement-like repeats CR3, CR8, and LB5. Although the drop in pH resulted in a reduction in calcium affinity in each case, the changes were very variable in magnitude, being as low as a 2-fold reduction for CR3. This suggests that a pH-dependent change in calcium affinity alone cannot be responsible for the release of bound protein ligands from the LRP at the pH prevailing in the endosome, which in turn requires one or more other pH-dependent effects for regulating protein ligand release.


Assuntos
Proteínas Relacionadas a Receptor de LDL/química , Proteínas Relacionadas a Receptor de LDL/metabolismo , Receptores de LDL/química , Receptores de LDL/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cálcio/metabolismo , Complemento C7/química , Complemento C7/genética , Cristalografia por Raios X , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Proteínas Relacionadas a Receptor de LDL/genética , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Receptores de LDL/genética , Sequências Repetitivas de Aminoácidos , Eletricidade Estática
13.
J Immunol ; 165(2): 1059-65, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10878384

RESUMO

Activation of the complement system through the classical, alternative, or lectin pathway results in the formation of the terminal complement complex. C7 plays an integral role in the assembly of this complex with target cell membranes. To date, only human C7 has been cloned and characterized; thus, in this study, we characterized the porcine complement component C7. Porcine C7 was isolated by affinity chromatography as a single glycoprotein with an approximate molecular mass of 90 kDa and 100 kDa under reducing and nonreducing conditions, respectively. The full-length porcine C7 cDNA was isolated, and the predicted amino acid sequence exhibited 80% identity with human C7 with conservation of the cysteine backbone and two putative N-linked glycosylation sites. Porcine C7 mRNA expression was detected in all tissues investigated, except polymorphonuclear and mononuclear leukocytes. Addition of purified porcine C7 restored the hemolytic activity of C7-depleted human sera in a dose-dependent manner. A functionally inhibitory mAb against porcine C7 attenuated the hemolytic activity of human, rabbit, or rat sera, suggesting an important conserved C7 epitope among species. These data demonstrate that porcine and human C7 are highly conserved, sharing structural and functional characteristics.


Assuntos
Complemento C7/química , Complemento C7/isolamento & purificação , Suínos/imunologia , Sequência de Aminoácidos , Animais , Clonagem Molecular , Complemento C7/genética , Complemento C7/metabolismo , Ensaio de Atividade Hemolítica de Complemento , DNA Complementar/isolamento & purificação , Humanos , Dados de Sequência Molecular , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Testes de Precipitina , Coelhos , Ratos , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Suínos/genética
14.
J Mol Evol ; 49(2): 282-9, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10441679

RESUMO

The plasma complement system comprises several activation pathways that share a common terminal route involving the assembly of the terminal complement complex (TCC), formed by C5b-C9. The order of emergence of the homologous components of TCC (C6, C7, C8alpha, C8beta, and C9) has been determined by phylogenetic analyses of their amino acid sequences. Using all the sequence data available for C6-C9 proteins, as well as for perforins, the results suggested that these TCC components originated from a single ancestral gene and that C6 and C7 were the earliest to emerge. Our evidence supports the notion that the ancestral gene had a complex modular composition. A series of gene duplications in combination with a tendency to lose modules resulted in successive complement proteins with decreasing modular complexity. C9 and perforin apparently are the result of different selective conditions to acquire pore-forming function. Thus C9 and perforin are examples of evolutionary parallelism.


Assuntos
Complemento C6/genética , Complemento C7/genética , Complemento C8/genética , Complemento C9/genética , Evolução Molecular , Filogenia , Sequência de Aminoácidos , Animais , Complemento C5/genética , Complemento C5b , Complemento C6/química , Complemento C7/química , Complemento C8/química , Complemento C9/química , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Perforina , Proteínas Citotóxicas Formadoras de Poros , Coelhos , Ratos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...