Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Clin Lab Med ; 39(4): 579-590, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31668271

RESUMO

The complement system is a critical component of both the innate and adaptive immune systems that augments the function of antibodies and phagocytes. Antigen-antibody immune complexes, lectin binding, and accelerated C3 tick-over can activate this well-coordinated and carefully regulated process. The importance of this system is highlighted by the disorders that arise when complement components or regulators are deficient or dysregulated. This article describes the pathways involved in complement activation and function, the regulation of these various pathways, and the interpretation of laboratory testing performed for the diagnosis of diseases of complement deficiency, exuberant complement activation, and complement dysregulation.


Assuntos
Proteínas do Sistema Complemento , Doenças da Deficiência Hereditária de Complemento , Testes Imunológicos , Convertases de Complemento C3-C5/análise , Convertases de Complemento C3-C5/metabolismo , Convertases de Complemento C3-C5/fisiologia , Proteínas do Sistema Complemento/análise , Proteínas do Sistema Complemento/metabolismo , Proteínas do Sistema Complemento/fisiologia , Doenças da Deficiência Hereditária de Complemento/sangue , Doenças da Deficiência Hereditária de Complemento/diagnóstico , Humanos , Modelos Biológicos
2.
Kidney Int ; 83(2): 293-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23235567

RESUMO

Postinfectious glomerulonephritis is a common disorder that develops following an infection. In the majority of cases, there is complete recovery of renal function within a few days to weeks following resolution of the infection. In a small percentage of patients, however, the glomerulonephritis takes longer to resolve, resulting in persistent hematuria and proteinuria, or even progression to end-stage kidney disease. In some cases of persistent hematuria and proteinuria, kidney biopsies show findings of a postinfectious glomerulonephritis even in the absence of any evidence of a preceding infection. The cause of such 'atypical' postinfectious glomerulonephritis, with or without evidence of preceding infection, is unknown. Here we show that most patients diagnosed with this 'atypical' postinfectious glomerulonephritis have an underlying defect in the regulation of the alternative pathway of complement. These defects include mutations in complement-regulating proteins and antibodies to the C3 convertase known as C3 nephritic factors. As a result, the activated alternative pathway is not brought under control even after resolution of the infection. Hence, the sequela is continual glomerular deposition of complement factors with resultant inflammation and development of an 'atypical' postinfectious glomerulonephritis.


Assuntos
Via Alternativa do Complemento , Glomerulonefrite/imunologia , Infecções/complicações , Adolescente , Adulto , Idoso , Biópsia , Convertases de Complemento C3-C5/fisiologia , Feminino , Glomerulonefrite/tratamento farmacológico , Glomerulonefrite/etiologia , Glomerulonefrite/patologia , Humanos , Rim/patologia , Masculino , Pessoa de Meia-Idade
3.
PLoS One ; 7(10): e47245, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23071769

RESUMO

C3 and C5 convertases are central stages of the complement cascade since they converge the different initiation pathways, augment complement activation by an amplification loop and lead to a common terminal pathway resulting in the formation of the membrane attack complex. Several complement inhibitors attenuate convertase formation and/or accelerate dissociation of convertase complexes. Functional assays used to study these processes are often performed using purified complement components, from which enzymatic complexes are reconstituted on the surface of erythrocytes or artificial matrices. This strategy enables identification of individual interactions between convertase components and putative regulators but carries an inherent risk of detecting non-physiological interactions that would not occur in a milieu of whole serum. Here we describe a novel, alternative method based on C3 or C5-depleted sera, which support activation of the complement cascade up to the desired stages of convertases. This approach allows fast and simple assessment of the influence of putative regulators on convertase formation and stability. As an example of practical utility of the assay, we performed studies on thioredoxin-1 in order to clarify the mechanism of its influence on complement convertases.


Assuntos
Ativação do Complemento/fisiologia , Convertases de Complemento C3-C5/fisiologia , Ensaios Enzimáticos/métodos , Cromatografia de Afinidade , Humanos , Soros Imunes/química , Tiorredoxinas/química , Tiorredoxinas/metabolismo
4.
Curr Mol Med ; 12(8): 1083-97, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22812419

RESUMO

As part of the innate immune system, the complement system recognises a wide range of non-self structures present on pathogens or altered self cells. Its activation elicits proteolytic cascades which eventually results in the cleavage of the C5 protein into two fragments, C5a and C5b. The small anaphylatoxin C5a induces a variety of biological responses upon binding to the 7TM receptors C5aR and the C5L2, while the large C5b fragment nucleates formation of the membrane attack complex capable of killing susceptible pathogens by the formation of a pore structure in association with complement components C6, C7, C8, and C9. A number of regulatory molecules help to control C5 mediated immune responses towards host cells, but in several major inflammatory conditions including sepsis and arthritis, C5a is believed to contribute significantly to disease etiology. Inhibition of membrane attack complex assembly is already approved for treatment of paroxysmal nocturnal haemoglobinuria and atypical hemolytic uremic syndrome. A number of recent crystal structures have provided a comprehensive insight into the architecture and properties of intact C5 and its fragments, and how pathogens interfere with their function. Here we review the functional and structural aspects of C5 and its fragments, the pathological conditions associated with them, and strategies employed by pathogens to interfere with the biological function of C5. Structural insight and elucidation of evasion strategies employed by pathogens present a unique opportunity for promoting the development of novel selective C5 inhibitors with therapeutic applications.


Assuntos
Complemento C5/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/fisiologia , Complemento C5/genética , Complemento C5/metabolismo , Complemento C5/fisiologia , Humanos , Inflamação/genética , Cinética , Modelos Moleculares , Mutação , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/fisiologia , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
5.
J Clin Invest ; 120(10): 3702-12, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20852386

RESUMO

Dense deposit disease (DDD) is a severe renal disease characterized by accumulation of electron-dense material in the mesangium and glomerular basement membrane. Previously, DDD has been associated with deficiency of factor H (fH), a plasma regulator of the alternative pathway (AP) of complement activation, and studies in animal models have linked pathogenesis to the massive complement factor 3 (C3) activation caused by this deficiency. Here, we identified a unique DDD pedigree that associates disease with a mutation in the C3 gene. Mutant C(3923ΔDG), which lacks 2 amino acids, could not be cleaved to C3b by the AP C3-convertase and was therefore the predominant circulating C3 protein in the patients. However, upon activation to C3b by proteases, or to C3(H2O) by spontaneous thioester hydrolysis, C(3923ΔDG) generated an active AP C3-convertase that was regulated normally by decay accelerating factor (DAF) but was resistant to decay by fH. Moreover, activated C(3b923ΔDG) and C3(H2O)(923ΔDG) were resistant to proteolysis by factor I (fI) in the presence of fH, but were efficiently inactivated in the presence of membrane cofactor protein (MCP). These characteristics cause a fluid phase-restricted AP dysregulation in the patients that continuously activated and consumed C3 produced by the normal C3 allele. These findings expose structural requirements in C3 that are critical for recognition of the substrate C3 by the AP C3-convertase and for the regulatory activities of fH, DAF, and MCP, all of which have implications for therapeutic developments.


Assuntos
Ativação do Complemento , Complemento C3/genética , Glomerulonefrite Membranoproliferativa/genética , Mutação , Adulto , Complemento C3/análise , Convertases de Complemento C3-C5/fisiologia , Complemento C3b/metabolismo , Fator H do Complemento/metabolismo , Feminino , Glomerulonefrite Membranoproliferativa/etiologia , Glomerulonefrite Membranoproliferativa/imunologia , Humanos , Masculino , Pessoa de Meia-Idade
6.
Blood ; 114(13): 2837-45, 2009 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-19584399

RESUMO

Complement is a major innate immune defense against pathogens, tightly regulated to prevent host tissue damage. Atypical hemolytic uremic syndrome (aHUS) is characterized by endothelial damage leading to renal failure and is highly associated with abnormal alternative pathway regulation. We characterized the functional consequences of 2 aHUS-associated mutations (D(254)G and K(325)N) in factor B, a key participant in the alternative C3 convertase. Mutant proteins formed high-affinity C3-binding site, leading to a hyperfunctional C3 convertase, resistant to decay by factor H. This led to enhanced complement deposition on the surface of alternative pathway activator cells. In contrast to native factor B, the 2 mutants bound to inactivated C3 and induced formation of functional C3-convertase on iC3b-coated surface. We demonstrated for the first time that factor B mutations lead to enhanced C3-fragment deposition on quiescent and adherent human glomerular cells (GEnCs) and human umbilical vein endothelial cells (HUVECs), together with the formation of sC5b-9 complexes. These results could explain the occurrence of the disease, since excessive complement deposition on endothelial cells is a central event in the pathogenesis of aHUS. Therefore, risk factors for aHUS are not only mutations leading to loss of regulation, but also mutations, resulting in hyperactive C3 convertase.


Assuntos
Convertases de Complemento C3-C5/fisiologia , Proteínas do Sistema Complemento/metabolismo , Células Endoteliais/metabolismo , Síndrome Hemolítico-Urêmica/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Criança , Pré-Escolar , Estudos de Coortes , Ativação do Complemento/genética , Convertases de Complemento C3-C5/genética , Proteínas do Sistema Complemento/genética , Família , Feminino , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Proteínas Mutantes/fisiologia , Linhagem , Adulto Jovem
7.
J Immunol ; 181(6): 4199-207, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768877

RESUMO

Despite eradication of smallpox three decades ago, public health concerns remain due to its potential use as a bioterrorist weapon. Smallpox and other orthopoxviruses express virulence factors that inhibit the host's complement system. In this study, our goals were to characterize the ability of the smallpox inhibitor of complement enzymes, SPICE, to regulate human complement on the cell surface. We demonstrate that SPICE binds to a variety of cell types and that the heparan sulfate and chondroitin sulfate glycosaminoglycans serve as attachment sites. A transmembrane-engineered version as well as soluble recombinant SPICE inhibited complement activation at the C3 convertase step with equal or greater efficiency than that of the related host regulators. Moreover, SPICE attached to glycosaminoglycans was more efficient than transmembrane SPICE. We also demonstrate that this virulence activity of SPICE on cells could be blocked by a mAb to SPICE. These results provide insights related to the complement inhibitory activities of poxviral inhibitors of complement and describe a mAb with therapeutic potential.


Assuntos
Membrana Celular/imunologia , Enzimas Ativadoras do Complemento/antagonistas & inibidores , Ativação do Complemento/imunologia , Proteínas Inativadoras do Complemento/fisiologia , Vírus da Varíola/imunologia , Proteínas da Matriz Viral/fisiologia , Proteínas Virais/fisiologia , Fatores de Virulência/fisiologia , Ligação Viral , Animais , Células CHO , Membrana Celular/enzimologia , Membrana Celular/genética , Membrana Celular/virologia , Enzimas Ativadoras do Complemento/metabolismo , Ativação do Complemento/genética , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Convertases de Complemento C3-C5/fisiologia , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/metabolismo , Cricetinae , Cricetulus , Glicosaminoglicanos/metabolismo , Células HeLa , Humanos , Ligação Proteica/genética , Ligação Proteica/imunologia , Estrutura Terciária de Proteína/genética , Vírus da Varíola/patogenicidade , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
8.
Mol Immunol ; 44(1-3): 23-32, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16875737

RESUMO

The human complement system is elemental to recognize bacteria, opsonize them for handling by phagocytes, or kill them by direct lysis. However, successful bacterial pathogens have in turn evolved ingenious strategies to overcome this part of the immune system. In this review we discuss the different stages of complement activation sequentially and illustrate the immune evasion strategies that various bacteria have developed to evade each subsequent step. The focus is on bacterial proteins, either surface-bound or excreted, that block complement activation. The underlying molecular mechanism of action and the possible role in pathophysiology of bacterial infections are discussed.


Assuntos
Infecções Bacterianas/imunologia , Ativação do Complemento/imunologia , Animais , Proteínas de Bactérias/fisiologia , Antígenos CD59/fisiologia , Proteína Inibidora do Complemento C1/metabolismo , Complemento C3/imunologia , Complemento C3/metabolismo , Convertases de Complemento C3-C5/fisiologia , Humanos , Receptores de Complemento/fisiologia
9.
J Immunol ; 175(7): 4528-35, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16177096

RESUMO

The extracellular domain of the complement receptor type 1 (CR1; CD35) consists entirely of 30 complement control protein repeats (CCPs). CR1 has two distinct functional sites, site 1 (CCPs 1-3) and two copies of site 2 (CCPs 8-10 and CCPs 15-17). In this report we further define the structural requirements for decay-accelerating activity (DAA) for the classical pathway (CP) C3 and C5 convertases and, using these results, generate more potent decay accelerators. Previously, we demonstrated that both sites 1 and 2, tandemly arranged, are required for efficient DAA for C5 convertases. We show that site 1 dissociates the CP C5 convertase, whereas the role of site 2 is to bind the C3b subunit. The intervening CCPs between two functional sites are required for optimal DAA, suggesting that a spatial orientation of the two sites is important. DAA for the CP C3 convertase is increased synergistically if two copies of site 1, particularly those carrying DAA-increasing mutations, are contained within one protein. DAA in such constructs may exceed that of long homologous repeat A (CCPs 1-7) by up to 58-fold. To explain this synergy, we propose a dimeric structure for the CP C3 convertase on cell surfaces. We also extended our previous studies of the amino acid requirements for DAA of site 1 and found that the CCP 1/CCP 2 junction is critical and that Phe82 may contact the C3 convertases. These observations increase our understanding of the mechanism of DAA. In addition, a more potent decay-accelerating form of CR1 was generated.


Assuntos
Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/fisiologia , Proteínas Inativadoras do Complemento/metabolismo , Via Clássica do Complemento , Receptores de Complemento 3b/fisiologia , Sítios de Ligação , Antígenos CD55/fisiologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Complemento C3b/metabolismo , Proteínas Inativadoras do Complemento/química , Via Clássica do Complemento/genética , Glicina/genética , Glicina/metabolismo , Humanos , Mutação , Fenilalanina/genética , Fenilalanina/metabolismo , Estrutura Terciária de Proteína , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/metabolismo
10.
Immunol Rev ; 180: 123-35, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11414354

RESUMO

Complement convertases are bimolecular complexes expressing protease activity only against C3 and C5. Their action is necessary for production of the biological activities of the complement system. Formation of these complexes proceeds through sequential protein-protein interactions and proteolytic cleavages of high specificity. Recent structural, mutational and functional data on factors D and B have significantly enhanced our understanding of the assembly, action, and regulation of the alternative pathway convertase. These processes were shown to depend critically on conformational changes, only some of which are reversible. The need for such changes is dictated by the zymogen-like configurations of the active centers of these unique serine proteases. The structural determinants of some of these changes have been defined from structural and mutational analyses of the two enzymes. Transition of factor D from the zymogen-like to the catalytically active conformation is completely reversible, while the active conformation of the catalytic center of the Bb fragment of factor B is irreversibly attenuated to a great extent on dissociation of the convertase complex. Both mechanisms contribute to the regulation of the proteolytic activity of these enzymes. Additional studies are necessary for a complete description of the elegant mechanisms mediating these processes.


Assuntos
Convertases de Complemento C3-C5/biossíntese , Convertases de Complemento C3-C5/química , Fator D do Complemento/química , Via Alternativa do Complemento , Animais , Sítios de Ligação , Domínio Catalítico , C3 Convertase da Via Alternativa do Complemento , Convertases de Complemento C3-C5/fisiologia , Complemento C3b/imunologia , Complemento C3b/metabolismo , Fator B do Complemento/metabolismo , Fator D do Complemento/fisiologia , Ativação Enzimática , Precursores Enzimáticos/química , Camundongos , Modelos Moleculares , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Serina Endopeptidases/química , Especificidade por Substrato , Fator de von Willebrand/química
11.
Immunol Rev ; 180: 16-34, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11414357

RESUMO

The origin of adaptive immunity in the vertebrates can be traced to the appearance of the ancestral RAG genes in the ancestral jawed vertebrate; however, the innate immune system is more ancient. A central subsystem within innate immunity is the complement system, which has been identified throughout and seems to be restricted to the deuterostomes. The evolutionary history of complement can be traced from the sea urchins (members of the echinoderm phylum), which have a simplified system homologous to the alternative pathway, through the agnathans (hagfish and lamprey) and the elasmobranchs (sharks and rays) to the teleosts (bony fish) and tetrapods, with increases in the numbers of complement components and duplications in complement pathways. Increasing complexity in the complement system parallels increasing complexity in the deuterostome animals. This review focuses on the simplest of the complement systems that is present in the sea urchin. Two components have been identified that show significant homology to vertebrate C3 and factor B (Bf), called SpC3 and SpBf, respectively. Sequence analysis from both molecules reveals their ancestral characteristics. Immune challenge of sea urchins indicates that SpC3 is inducible and is present in coelomic fluid (the body fluids) in relatively high concentrations, while SpBf expression is constitutive and is present in much lower concentrations. Opsonization of foreign cells and particles followed by augmented uptake by phagocytic coelomocytes appears to be a central function for this simpler complement system and important for host defense in the sea urchin. These activities are similar to some of the functions of the homologous proteins in the vertebrate complement system. The selective advantage for the ancestral deuterostome may have been the amplification feedback loop that is still of central importance in the alternative pathway of complement in higher vertebrates. Feedback loop functions would quickly coat pathogens with complement leading to phagocytosis and removal of foreign cells, a system that would be significantly more effective than an opsonin that binds upon contact as a result of simple diffusion. An understanding of the immune response of the sea urchin, an animal that is a good estimator of what the ancestral deuterostome immune system was like, will aid us in understanding how adaptive immunity might have been selected for during the early evolution of the vertebrates and how it might have been integrated into the pre-existing innate immune system that was already in place in those animals.


Assuntos
Proteínas do Sistema Complemento/genética , Evolução Molecular , Ouriços-do-Mar/imunologia , Sequência de Aminoácidos , Animais , Ativação do Complemento , Complemento C3/química , Complemento C3/imunologia , Convertases de Complemento C3-C5/fisiologia , Fator B do Complemento/química , Fator B do Complemento/imunologia , Sequência Consenso , Equinodermos/imunologia , Peixes/imunologia , Regulação da Expressão Gênica/imunologia , Rejeição de Enxerto/imunologia , Sistema Imunitário/citologia , Invertebrados/imunologia , Mamíferos/genética , Mamíferos/imunologia , Dados de Sequência Molecular , Proteínas Opsonizantes/imunologia , Fagocitose , Filogenia , Estrutura Terciária de Proteína , Ouriços-do-Mar/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Serina Endopeptidases/química , Fator de von Willebrand/química
12.
Am J Pathol ; 157(3): 905-18, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10980130

RESUMO

This study investigated the capacity of neurons and astrocytes to spontaneously activate the complement system and control activation by expressing complement regulators. Human fetal neurons spontaneously activated complement through the classical pathway in normal and immunoglobulin-deficient serum and C1q binding was noted on neurons but not on astrocytes. A strong staining for C4, C3b, iC3b neoepitope and C9 neoepitope was also found on neurons. More than 40% of human fetal neurons were lysed when exposed to normal human serum in the presence of a CD59-blocking antibody, whereas astrocytes were unaffected. Significant reduction in neuronal cell lysis was observed after the addition of soluble complement receptor 1 at 10 microg/ml. Fetal neurons were stained for CD59 and CD46 and were negative for CD55 and CD35. In contrast, fetal astrocytes were strongly stained for CD59, CD46, CD55, and were negative for CD35. This study demonstrates that human fetal neurons activate spontaneously the classical pathway of complement in an antibody-independent manner to assemble the cytolytic membrane attack complex on their membranes, whereas astrocytes are unaffected. One reason for the susceptibility of neurons to complement-mediated damage in vivo may reside in their poor capacity to control complement activation.


Assuntos
Ativação do Complemento/fisiologia , Proteínas Inativadoras do Complemento/deficiência , Via Clássica do Complemento/fisiologia , Glicoproteínas de Membrana/deficiência , Neurônios/fisiologia , Anticorpos Bloqueadores/farmacologia , Antígenos CD/genética , Antígenos CD/metabolismo , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Encéfalo/citologia , Encéfalo/embriologia , Células Cultivadas , Convertases de Complemento C3-C5/fisiologia , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/metabolismo , Feto , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Processamento de Imagem Assistida por Computador , Hibridização In Situ , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , RNA/análise , RNA Mensageiro/metabolismo , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Immunol ; 164(3): 1379-85, 2000 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-10640753

RESUMO

The C5 convertase is a serine protease that consists of two subunits: a catalytic subunit which is bound in a Mg2+-dependent complex to a noncatalytic subunit. To understand the functional role of the noncatalytic subunit, we have determined the C5-cleaving properties of the cobra venom factor-dependent C5 convertase (CVF, Bb) made with CVF purified from the venom of Naja naja (CVFn) and Naja haje (CVFh) and compared them to those for two C3b-dependent C5 convertases (ZymC3b,Bb and C3b,Bb). A comparison of the kinetic parameters indicated that although the four C5 convertases (CVFn,Bb, ZymC3b,Bb, CVFh,Bb, and C3b,Bb) had similar catalytic rate constants (kcat = 0.004-0.012 s-1) they differed 700-fold in their affinity for the substrate as indicated by the Km values (CVFn,Bb = 0.036 microM, ZymC3b,Bb = 1.24 microM, CVFh,Bb = 14.0 microM, and C3b,Bb = 24 microM). Analysis of binding interactions between C5 and the noncatalytic subunits (CVFh or C3b, or CVFn) using the BIAcore, revealed dissociation binding constants (Kd) that were similar to the Km values of the respective enzymes. The kinetic and binding data demonstrate that the binding site for C5 resides in the noncatalytic subunit of the enzyme, the affinity for the substrate is solely determined by the noncatalytic subunit and the catalytic efficiency of the enzyme appears not to be influenced by the nature of this subunit.


Assuntos
Domínio Catalítico/imunologia , Convertases de Complemento C3-C5/fisiologia , Animais , Sítios de Ligação/imunologia , Técnicas Biossensoriais , Convertases de Complemento C3-C5/sangue , Convertases de Complemento C3-C5/metabolismo , Fator B do Complemento/metabolismo , Via Alternativa do Complemento/imunologia , Relação Dose-Resposta Imunológica , Venenos Elapídicos/metabolismo , Elapidae/imunologia , Humanos , Cinética , Relação Estrutura-Atividade
14.
Mol Immunol ; 33(3): 297-309, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8649451

RESUMO

It has long been known that mouse C4 has unusually low hemolytic activity relative to the C4 of other mammalian species (e.g. human and guinea pig), the measurements being done in most cases using a C4-deficient guinea pig serum reagent in a one-step assay with EA. This low activity for mouse C4 previously had been attributed to "technical" difficulties such as lability of the protein during blood collection and partial species incompatibilities with guinea pig components. Recently, we presented evidence for the involvement of human C4 beta-chain residues 455-469, a putatively exposed hydrophilic segment, in contributing to a C5 binding site in the C4b subunit of the classical pathway C5 convertase, C4b3b2a. Given that there were five sequence differences between the human and mouse protein within this segment, we hypothesized that these substitutions may have compromised the C5 convertase subunit activity of mouse C4, thereby resulting in its low hemolytic activity. Using a multi-step hemolytic assay which was totally dependent upon C5 cleavage by the classical pathway, we found that mouse C4 was completely devoid of classical pathway C5 convertase subunit activity. We have been able to rule out the most obvious potential species incompatibilities (e.g. between C4mo and C5gp) as being responsible for this lack of activity. Moreover, we found that the low level of hemolytic activity of mouse C4 measured in the one-step assay can be ascribed totally to C5 cleavage, and subsequent terminal component assembly, by the alternative pathway C5 convertase, (C3b)2Bb. However, the assembly of the latter enzyme complex is dependent upon the presence of C3b molecules deposited initially via the classical pathway C3 convertase in which mouse C4b is a subunit. Finally, whereas conversion of human residues 458RP to the mouse-like sequence PL was sufficient to abrogate classical pathway C5 convertase subunit activity in human C4, the five substitutions which "humanized" the 452-466 segment of mouse C4 (corresponding to human residues 455-469) were on their own insufficient to impart this activity to mouse C4. This implies that, in addition to the 455-469 beta-chain segment of human C4, there are other regions of the molecule contributing to C5 binding which are also non-conserved between human and mouse C4.


Assuntos
Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/fisiologia , Complemento C4/química , Complemento C4/deficiência , Via Clássica do Complemento , Sequência de Aminoácidos , Animais , Complemento C3b/química , Complemento C3b/fisiologia , Complemento C4/fisiologia , Complemento C4b/química , Complemento C4b/fisiologia , Via Alternativa do Complemento/genética , Via Clássica do Complemento/genética , Cobaias , Hemólise , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida/imunologia , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Relação Estrutura-Atividade
15.
J Immunol ; 148(9): 2803-11, 1992 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-1573269

RESUMO

In general, C4A allotypes of human C4 show one-fourth to one-third the hemolytic activity of C4B allotypes. An exception to this rule is C4A6 which is almost totally deficient in hemolytic activity. Previous studies have localized the defect in C4A6 to the C5 convertase stage. Of the two critical events required for C5 cleavage, namely formation of a covalent adduct between C3b and the C4b subunit of the C3 convertase (C4b2a), and binding of C5 to this C4b-C3b complex, it is a defect in the latter step that accounts for the aberrant activity of C4A6. DNA sequencing studies described in a companion paper have suggested that the sole C4A6-specific difference was a Trp for Arg replacement at beta-chain residue 458. To directly ascertain whether this single substitution was responsible for the hemolytic defect in C4A6, we have used site-directed mutagenesis to introduce this change into both C4A and C4B cDNA expression plasmids. We found that the R to W replacement totally abrogated hemolytic activity. However, irrespective of the amino acid at residue 458, the mutant proteins behaved like their wild-type counterparts with respect to covalent binding to C1-bearing targets, i.e., the C4B recombinants displayed higher binding to sheep and human red cells than did the C4A counterparts. Furthermore, the mutants were able to form covalent C4b-C3b adducts. There was, however, substantially less C5 cleavage produced by cell-bound C4boxy23b complexes made with R458W mutant C4B than with wild-type C4B. These results are consistent with the sole defect in the mutants being at the C5 binding stage and strongly suggest that Arg 458 of the C4 beta-chain contributes to the C5 binding site of the molecule.


Assuntos
Convertases de Complemento C3-C5/fisiologia , Complemento C4a/fisiologia , Complemento C5/metabolismo , Via Clássica do Complemento/genética , Complemento C1/metabolismo , Complemento C4a/genética , Complemento C4b/fisiologia , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Hemólise/genética , Humanos , Mutagênese Sítio-Dirigida , Plasmídeos , Testes de Precipitina , Transfecção
16.
J Immunol ; 147(3): 927-32, 1991 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-1861081

RESUMO

A C intermediate, LAC14, was prepared from TNP-aminocaproyl liposomes sensitized with anti-TNP antibody (Ab) and purified human C1 and C4. LAC14, containing radiolabeled C4, was analyzed by SDS-PAGE followed by autoradiography, and yielded a 210-kDa band and a predominant 400-kDa band. The 210-kDa band consisted of monomeric C4b bound to low molecular mass acceptors. The 400-kDa band was comprised of a 200-kDa moiety, as well as beta- and gamma-chains of C4. The 200-kDa moiety contained neither C1 nor sensitizing Ab, but it was largely decreased by treatment with NH2OH to the 90-kDa moiety with the mobility corresponding to the alpha'-chain of C4b. A covalent dimer of C4b, therefore, is the predominant form of C4b deposited on liposomes sensitized with antibody. The C4b-C4b dimer formed rapidly (within 5 min) followed by slow dissociation into monomers. The LAC14 bearing the C4b dimer but not the monomer was lysed, although with relatively low efficiency, by the addition of oxyC2 and EDTA-supplemented C3-deficient serum (C3DS), and, furthermore, LAC142 possessed the ability to convert C5 into C5a and C5b. Moreover, lysis was inhibited not by anti-C3 Ab but by anti-C4 Ab. In other experiments, the dimer served as an element of C3 convertase, as well. These findings imply that the C4b dimer, when complexed with C2, expresses C3/C5 convertase activity without participation of C3, and may provide a molecular mechanism whereby sera from patients with complete C3 deficiency retain the ability to induce C-mediated cytolysis.


Assuntos
Convertases de Complemento C3-C5/biossíntese , Complemento C4b/metabolismo , Autorradiografia , Complemento C3/metabolismo , Convertases de Complemento C3-C5/fisiologia , Via Alternativa do Complemento , Via Clássica do Complemento , Eletroforese em Gel Bidimensional , Humanos , Lipossomos/imunologia
17.
Curr Top Microbiol Immunol ; 153: 1-21, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2404692

RESUMO

C3, the most abundant complement protein in blood, plays a central role in the activation sequence of the complement system as well as in host defense. Expression of the multiple functions of C3 requires its cleavage by highly specific enzymes termed C3 convertases. C3 in a conformationally altered form, C3H2O, resulting from the slow spontaneous hydrolysis of the internal thioester bond of native C3, initiates the assembly of a C3 convertase which continuously cleaves C3 in the blood at slow rates generating a constant supply of small amounts of C3b. When an activator of the alternative complement pathway is present, C3b becomes covalently attached to its surface via an ester or amide bond. Activator surface-bound C3b initiates the assembly of an "amplification" C3 convertase, C3bBb(P), which can efficiently activate C3 and generate additional convertase complexes on the surface of the activator. C3b generated by an amplification or classical pathway C3 convertase can also bind covalently to the noncatalytic subunit, C3b or C4b, respectively, resulting in the generation of a C5 convertase, an enzyme catalyzing the cleavage/activation of C5. In terms of participation in host defense, several fragments of C3, including C3a, C3b, iC3b, and C3dg, mediate a number of important functions such as increased vascular permeability, enhancement of phagocytosis, elimination of immune complexes, and perhaps also proliferative responses and/or differentiation of B cells.


Assuntos
Ativação do Complemento/fisiologia , Complemento C3/fisiologia , Convertases de Complemento C3-C5/fisiologia , Fator B do Complemento/fisiologia , Fator D do Complemento/fisiologia , Humanos
20.
J Immunol ; 135(4): 2661-7, 1985 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-3161944

RESUMO

Four CR1 variants have been found in the normal population and are designated CR1-A (190,000 daltons), CR1-B (220,000 daltons), CR1-C (160,000 daltons), and CR1-D (250,000 daltons). In the present study, we first developed an improved chromatographic purification scheme for CR1 that does not employ a C3b affinity step. CR1 variants (A, B, and C) were then isolated, and their individual functional activity was assessed. Each possessed similar co-factor activity for I-mediated cleavage of C3(H2O), as well as for the inhibitory activity for fluid phase C3 convertases. These results indicate that, despite relatively large Mr differences, in the purified state these three CR1 variants have similar functional activities. The functional activity of CR1 was also compared with C4bp, H, and decay accelerating factor (DAF) in fluid phase assays designed to assess the inhibition of the C3 convertases and co-factor activity. On a molar basis, CR1 had approximately the same inhibitory activity as C4bp for the classical pathway convertase, and had the same as H for the alternative pathway convertase. These results indicate that CR1 encompasses the functional capabilities of both proteins. They also raise a number of interesting genetic and structural questions in regard to these complement regulatory proteins, because C4bp is thought to have multiple C4b binding domains, whereas H is reported to bind one C3b. DAF was an approximately fourfold better inhibitor of the alternative pathway convertase than CR1 or H, but was a fourfold less efficient inhibitor of the classical pathway convertase than CR1 or C4bp. The effective inhibitory capacity of DAF in these fluid phase assay systems suggests that the DAF substrate specificity is for the convertases. Fluid phase CR1 was twofold less efficient than H in serving as a co-factor for the first cleavage of fluid phase C3b, and hardly mediated the second cleavage. These data are in contrast to the co-factor activity of CR1 on a cell membrane, and provide additional evidence for the local environment being a critical modulator of the function of proteins that regulate the activation of C3.


Assuntos
Enzimas Ativadoras do Complemento/fisiologia , Ativação do Complemento , Convertases de Complemento C3-C5/fisiologia , Proteínas Inativadoras do Complemento C3b/fisiologia , Polimorfismo Genético , Receptores de Complemento/isolamento & purificação , Convertases de Complemento C3-C5/antagonistas & inibidores , Fator H do Complemento , Eletroforese em Gel de Poliacrilamida , Variação Genética , Humanos , Focalização Isoelétrica , Fenótipo , Receptores de Complemento/genética , Receptores de Complemento/fisiologia , Receptores de Complemento 3b
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...