Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Vis ; 24: 518-535, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30090015

RESUMO

Purpose: Variants of complement factor genes, hypoxia and oxidative stress of the outer retina, and systemic hypertension affect the risk of age-related macular degeneration. Hypertension often results from the high intake of dietary salt that increases extracellular osmolarity. We determined the effects of extracellular hyperosmolarity, hypoxia, and oxidative stress on the expression of complement genes in cultured (dedifferentiated) human RPE cells and investigated the effects of C9 siRNA and C9 protein on RPE cells. Methods: Hyperosmolarity was induced by adding 100 mM NaCl or sucrose to the culture medium. Hypoxia was induced by culturing cells in 1% O2 or by adding the hypoxia mimetic CoCl2. Oxidative stress was induced by adding H2O2. Gene and protein expression levels were determined with real-time RT-PCR, western blot, and ELISA analyses. The expression of the nuclear factor of activated T cell 5 (NFAT5) and complement factor (C9) was knocked down with siRNA. Results: Extracellular hyperosmolarity, hypoxia, and oxidative stress strongly increased the transcription of the C9 gene, while the expression of the C3, C5, CFH, and CFB genes was moderately altered or not altered at all. Hyperosmolarity also induced a moderate increase in the cytosolic C9 protein level. The hyperosmotic C9 gene expression was reduced by inhibitors of the p38 MAPK, ERK1/2, JNK, and PI3K signal transduction pathways and of the transcription factors STAT3 and NFAT5. The hypoxic C9 gene expression was reduced by a STAT3 inhibitor. The knockdown of C9 with siRNA decreased the hypoxic vascular endothelial growth factor (VEGF) and NLRP3 gene expression, the hypoxic secretion of VEGF, and the hyperosmotic expression of the NLRP3 gene. Exogenous C9 protein inhibited the hyperosmotic expression of the C9 gene, the hypoxic and hyperosmotic VEGF gene expression, and the hyperosmotic expression of the NLRP3 gene. Both C9 siRNA and C9 protein inhibited inflammasome activation under hyperosmotic conditions, as indicated by the decrease in the cytosolic level of mature IL-1ß. Conclusions: The expression of the C9 gene in cultured RPE cells is highly induced by extracellular hyperosmolarity, hypoxia, and oxidative stress. The data may support the assumption that C9 gene expression may stimulate the expression of inflammatory (NLRP3) and angiogenic growth factors (VEGF) in RPE cells. Extracellular C9 protein may attenuate this effect, in part via negative regulation of the C9 mRNA level.


Assuntos
Cobalto/farmacologia , Complemento C9/genética , Células Epiteliais/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Cloreto de Sódio/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Complemento C3/genética , Complemento C3/imunologia , Complemento C5/genética , Complemento C5/imunologia , Complemento C9/antagonistas & inibidores , Complemento C9/imunologia , Fator B do Complemento/genética , Fator B do Complemento/imunologia , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Concentração Osmolar , Pressão Osmótica/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/imunologia , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Fator A de Crescimento do Endotélio Vascular/imunologia
2.
PLoS Negl Trop Dis ; 9(12): e0004310, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26720603

RESUMO

BACKGROUND: Trichinella spiralis expresses paramyosin (Ts-Pmy) as a defense mechanism. Ts-Pmy is a functional protein with binding activity to human complement C8 and C9 and thus plays a role in evading the attack of the host's immune system. In the present study, the binding activity of Ts-Pmy to human complement C1q and its ability to inhibit classical complement activation were investigated. METHODS AND FINDINGS: The binding of recombinant and natural Ts-Pmy to human C1q were determined by ELISA, Far Western blotting and immunoprecipitation, respectively. Binding of recombinant Ts-Pmy (rTs-Pmy) to C1q inhibited C1q binding to IgM and consequently inhibited C3 deposition. The lysis of antibody-sensitized erythrocytes (EAs) elicited by the classical complement pathway was also inhibited in the presence of rTs-Pmy. In addition to inhibiting classical complement activation, rTs-Pmy also suppressed C1q binding to THP-1-derived macrophages, thereby reducing C1q-induced macrophages migration. CONCLUSION: Our results suggest that T. spiralis paramyosin plays an important role in immune evasion by interfering with complement activation through binding to C1q in addition to C8 and C9.


Assuntos
Complemento C1q/imunologia , Via Clássica do Complemento/efeitos dos fármacos , Evasão da Resposta Imune/efeitos dos fármacos , Trichinella spiralis/imunologia , Triquinelose/imunologia , Tropomiosina/farmacologia , Animais , Complemento C1q/metabolismo , Complemento C8/antagonistas & inibidores , Complemento C8/imunologia , Complemento C9/antagonistas & inibidores , Complemento C9/imunologia , Eritrócitos/efeitos dos fármacos , Feminino , Hemólise , Humanos , Fatores Imunológicos/imunologia , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Proteínas Recombinantes , Tropomiosina/imunologia , Tropomiosina/metabolismo
3.
J Virol ; 87(10): 5858-67, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23487461

RESUMO

Hepatitis C virus (HCV) proteins inhibit complement component expression, which may attenuate immunity against infection. In this study, we examined whether HCV regulates the membrane attack complex (MAC) via complement component C9. MAC is composed of C5b to C9 (C5b-9) and mediates cell lysis of invaded pathogens. Liver biopsy specimens from chronically HCV-infected patients exhibited a lower level of C9 mRNA expression than liver biopsy specimens from unrelated disease or healthy control human liver RNA. Hepatocytes infected with cell culture-grown HCV or expressing HCV core protein also displayed significant repression of C9 mRNA and protein levels. Promoter analysis suggested that the T cell factor-4 (TCF-4E) transcription factor is responsible for HCV core-mediated C9 promoter regulation. Sera from chronically HCV-infected patients displayed a lower level of C5b-9 and a reduced antimicrobial effect on model organisms compared to unrelated patient sera or sera from healthy volunteers. Together, these results for C9 regulation by HCV core protein coupled with functional impairment of the membrane attack complex underscore HCV-mediated attenuation of immune mechanisms.


Assuntos
Complemento C9/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Hepacivirus/imunologia , Hepacivirus/patogenicidade , Biópsia , Perfilação da Expressão Gênica , Hepatite C Crônica/patologia , Humanos , Evasão da Resposta Imune , Fígado/patologia
4.
J Fish Biol ; 79(7): 2083-6, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22141908

RESUMO

Neutralizing complement C9 in grass carp Ctenopharyngodon idella sera with rabbit anti-C9 sera against fish complement C9, demonstrated that bactericidal activity against Aeromonas hydrophila of the C9-deficient fish sera was greatly impaired. These results indicated that the fish complement C9 plays a key role in pathogen killing through the lytic pathway.


Assuntos
Aeromonas hydrophila/fisiologia , Atividade Bactericida do Sangue , Carpas/imunologia , Complemento C9/imunologia , Animais , Carpas/microbiologia , Complemento C9/antagonistas & inibidores , Soros Imunes/metabolismo
5.
PLoS Negl Trop Dis ; 5(7): e1225, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21750743

RESUMO

BACKGROUND: Paramyosin is a thick myofibrillar protein found exclusively in invertebrates. Evidence suggested that paramyosin from helminths serves not only as a structural protein but also as an immunomodulatory agent. We previously reported that recombinant Trichinella spiralis paramyosin (Ts-Pmy) elicited a partial protective immunity in mice. In this study, the ability of Ts-Pmy to bind host complement components and protect against host complement attack was investigated. METHODS AND FINDINGS: In this study, the transcriptional and protein expression levels of Ts-Pmy were determined in T. spiralis newborn larva (NBL), muscle larva (ML) and adult worm developmental stages by RT-PCR and western blot analysis. Expression of Ts-Pmy at the outer membrane was observed in NBL and adult worms using immunogold electron microscopy and immunofluorescence staining. Functional analysis revealed that recombinant Ts-Pmy(rTs-Pmy) strongly bound to complement components C8 and C9 and inhibited the polymerization of C9 during the formation of the membrane attack complex (MAC). rTs-Pmy also inhibited the lysis of rabbit erythrocytes (E(R)) elicited by an alternative pathway-activated complement from guinea pig serum. Inhibition of native Ts-Pmy on the surface of NBL with a specific antiserum reduced larvae viability when under the attack of complement in vitro. In vivo passive transfer of anti-Ts-Pmy antiserum and complement-treated larvae into mice also significantly reduced the number of larvae that developed to ML. CONCLUSION: These studies suggest that the outer membrane form of T. spiralis paramyosin plays an important role in the evasion of the host complement attack.


Assuntos
Complemento C8/metabolismo , Complemento C9/metabolismo , Evasão da Resposta Imune , Trichinella spiralis/imunologia , Tropomiosina/metabolismo , Animais , Western Blotting , Complemento C8/antagonistas & inibidores , Complemento C8/imunologia , Complemento C9/antagonistas & inibidores , Complemento C9/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Eritrócitos , Feminino , Imunofluorescência , Perfilação da Expressão Gênica , Hemólise , Camundongos , Camundongos Endogâmicos ICR , Microscopia Imunoeletrônica , Dados de Sequência Molecular , Ligação Proteica , RNA de Helmintos/genética , RNA Mensageiro/genética , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Tropomiosina/imunologia
6.
J Immunol ; 186(12): 6657-60, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21572031

RESUMO

Cerebral malaria is the most severe complication of Plasmodium falciparum infection and accounts for a large number of malaria fatalities worldwide. Recent studies demonstrated that C5(-/-) mice are resistant to experimental cerebral malaria (ECM) and suggested that protection was due to loss of C5a-induced inflammation. Surprisingly, we observed that C5aR(-/-) mice were fully susceptible to disease, indicating that C5a is not required for ECM. C3aR(-/-) and C3aR(-/-) × C5aR(-/-) mice were equally susceptible to ECM as were wild-type mice, indicating that neither complement anaphylatoxin receptor is critical for ECM development. In contrast, C9 deposition in the brains of mice with ECM suggested an important role for the terminal complement pathway. Treatment with anti-C9 Ab significantly increased survival time and reduced mortality in ECM. Our data indicate that protection from ECM in C5(-/-) mice is mediated through inhibition of membrane attack complex formation and not through C5a-induced inflammation.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Malária Cerebral/etiologia , Animais , Encéfalo/imunologia , Complemento C5a/fisiologia , Complemento C9/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Suscetibilidade a Doenças , Inflamação , Malária Cerebral/imunologia , Camundongos , Camundongos Knockout , Receptor da Anafilatoxina C5a , Receptores de Complemento , Taxa de Sobrevida
7.
Infect Immun ; 71(11): 6402-10, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14573661

RESUMO

Larvae and adults of the parasitic blood fluke Schistosoma mansoni are resistant to killing by human complement. An earlier search by Parizade et al. for a schistosome complement inhibitor identified a 94-kDa surface protein which was named SCIP-1 (M. Parizade, R. Arnon, P. J. Lachmann, and Z. Fishelson, J. Exp. Med. 179:1625-1636, 1994). Following partial purification and analysis by mass spectrometry, we have determined SCIP-1 to be a surface-exposed form of the muscle protein paramyosin. As shown by immunofluorescence, anti-paramyosin antibodies label the surface of live schistosomula and adult worms. Like SCIP-1, purified native paramyosin reacts with a polyclonal rabbit anti-human CD59 antiserum, as shown by Western blot analysis. Also, the human complement components C8 and C9 bind to recombinant and native paramyosin. Analysis of paramyosin binding to fragments of C9 generated by thrombin or trypsin has demonstrated that paramyosin binds to C9 at a position located between Gly245 and Arg391. Paramyosin inhibited Zn(2+)-induced C9 polymerization and poly-C9 deposition onto rabbit erythrocytes (E(R)). In addition, paramyosin inhibited lysis of E(R) and of sensitized sheep erythrocytes by human complement. Finally, anti-paramyosin antibodies enhanced in vitro killing of schistosomula by normal and C4-depleted human complement. Taken together, these findings suggest that an exogenous form of S. mansoni paramyosin inhibits activation of the terminal pathway of complement and thus has an important immunomodulatory role in schistosomiasis.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Proteínas de Helminto/fisiologia , Schistosoma mansoni/imunologia , Tropomiosina/fisiologia , Animais , Antígenos CD59/fisiologia , Complemento C8/metabolismo , Complemento C9/antagonistas & inibidores , Complemento C9/química , Complemento C9/metabolismo , Imunofluorescência , Hemólise/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos ICR , Zinco/farmacologia
8.
Neurosci Lett ; 325(3): 175-8, 2002 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-12044649

RESUMO

To determine whether ischemic cerebral infarction is mediated in part by complement component C9, C9-deficient neonatal rats were subjected to unilateral cerebral ischemia. Brains were harvested 24 h later, stained with 2,3,5-triphenyl tetrazolium chloride, and cerebral infarct volumes were quantified by computer-based planimetry. Compared with buffer, prophylactic intraperitoneal (i.p.) administration of the complement inhibitors soluble complement receptor type 1 (sCR1), a molecular hybrid of sCR1 and the selectin inhibitor sialyl Lewis x (sCR1-sLex), or cobra venom factor did not affect the cerebral infarct volume. In contrast, i.p. human C9 (75 microg/g body weight) significantly increased the volume of infarct located 6 through 10 mm posterior to the frontal pole. Therefore, in the post-ischemic brain, C9 was neurotoxic and augmented the focal cerebral infarct volume.


Assuntos
Córtex Cerebral/imunologia , Infarto Cerebral/imunologia , Complemento C9/efeitos adversos , Traumatismo por Reperfusão/imunologia , Animais , Animais Recém-Nascidos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Infarto Cerebral/patologia , Complemento C9/administração & dosagem , Complemento C9/antagonistas & inibidores , Complemento C9/deficiência , Modelos Animais de Doenças , Ratos , Ratos Sprague-Dawley , Receptores de Complemento/metabolismo , Traumatismo por Reperfusão/patologia
9.
J Physiol ; 539(Pt 2): 537-45, 2002 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11882685

RESUMO

Activation of the complement system on the cell surface results in the insertion of pore forming membrane attack complexes (MAC, C5b-9). In order to protect themselves from the complement attack, the cells express several regulatory molecules, including the terminal complex regulator CD59 that inhibits assembly of the large MACs by inhibiting the insertion of additional C9 molecules into the C5b-9 complex. Using the whole cell patch clamp method, we were able to measure accumulation of homologous MACs in the membrane of CD59(-) human B-cells, which formed non-selective ion channels with a total conductance of 360 +/- 24 pS as measured at the beginning of the steady-state phase of the inward currents. C5b-8 and small-size MAC (MAC containing only a single C9) can also form ion channels. Nevertheless, in CD59(+) human B-cells in spite of small-size MAC formation, an ion current could not be detected. In addition, restoring CD59 to the membrane of the CD59(-) cells inhibited the serum-evoked inward current. The ion channels formed by the small-size MAC were therefore sealed, indicating that CD59 directly interfered with the pore formation of C5b-8 as well as that of small-size C5b-9. These results offer an explanation as to why CD59-expressing cells are not leaky in spite of a buildup of homologous C5b-8 and small-size MAC. Our experiments also confirmed that ion channel inhibition by CD59 is subject to homologous restriction and that CD59 cannot block the conductivity of MAC when generated by xenogenic (rabbit) serum.


Assuntos
Antígenos CD59/farmacologia , Complemento C9/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Proteínas do Sistema Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Complemento C9/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Citometria de Fluxo , Humanos , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/fisiologia , Técnicas de Patch-Clamp , Coelhos
10.
Biochemistry ; 38(21): 6807-16, 1999 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-10346902

RESUMO

The association of terminal complement proteins was investigated by analytical ultracentrifugation and multi-angle laser light scattering. Native C8 and C9 formed a heterodimer in solution of physiological ionic strength with a free-energy change DeltaG degrees of -8.3 kcal/mol and a dissociation constant Kd of 0.6 microM (at 20 degrees C) that was ionic strength- and temperature-dependent. A van't Hoff plot of the change in Kd was linear between 10 and 37 degrees C and yielded values of DeltaH degrees = -12.9 kcal/mol and DeltaS degrees = -15.9 cal mol-1 deg-1, suggesting that electrostatic forces play a prominent role in the interaction of C8 with C9. Native C8 also formed a heterodimer with C5, and low concentrations of polyionic ligands such as protamine and suramin inhibited the interaction. Suramin induced high-affinity trimerization of C8 (Kd = 0.10 microM at 20 degrees C) and dimerization of C9 (Kd = 0.86 microM at 20 degrees C). Suramin-induced C8 oligomerization may be the primary reason for the drug's ability to prevent complement-mediated hemolysis. Analysis of sedimentation equilibria and also of the fluorescence enhancement of suramin when bound to protein provided evidence for two suramin-binding sites on each C9 and three on each C8 in the oligomers. Oligomerization could be reversed by high suramin concentrations, but 8-aminonaphthalene-1,3,6- trisulfonate (ANTS2- ), which mimics half a suramin molecule, could not compete with suramin binding and oligomerization suggesting that the drug also binds nonionically to the proteins.


Assuntos
Complemento C8/química , Complemento C8/metabolismo , Complemento C9/química , Complemento C9/metabolismo , Suramina/química , Varredura Diferencial de Calorimetria , Centrifugação com Gradiente de Concentração , Complemento C8/antagonistas & inibidores , Complemento C9/antagonistas & inibidores , Proteínas Inativadoras do Complemento/química , Proteínas Inativadoras do Complemento/farmacologia , Humanos , Lasers , Luz , Substâncias Macromoleculares , Concentração Osmolar , Espalhamento de Radiação , Soluções , Espectrometria de Fluorescência , Suramina/farmacologia , Termodinâmica
11.
Biochemistry ; 35(10): 3263-9, 1996 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-8605162

RESUMO

CD59 antigen is a membrane glycoprotein that inhibits the activity of the C9 component of the C5b-9 membrane attack complex (MAC), thereby protecting human cells from lysis by human complement. The complement-inhibitory activity of CD59 is species-selective, and is most effective toward C9 derived from human or other primate plasma. The species-selective activity of CD59 was recently used to map the segment of human C9 that is recognized by this MAC inhibitor, using recombinant rabbit/human C9 chimeras that retain lytic function within the MAC [Husler, T., Lockert, D. H., Kaufman, K. M., Sodetz, J. M., & Sims, P. J. (1995) J. Biol. Chem. 270,3483-3486]. These experiments suggested that the CD59 recognition domain was contained between residues 334 and 415 in human C9. By analyzing the species-selective lytic activity of recombinant C9 with chimeric substitutions internal to this segment, we now demonstrate that the site in human C9 uniquely recognized by CD59 is centered on those residues contained between C9 Cys359/Cys384, with an additional contribution by residues C-terminal to this segment. Consistent with its role as a CD59 recognition domain, CD59 specifically bound a human C9-derived peptide corresponding to residues 359-384, and antibody (Fab) raised against this C9-derived peptide inhibited the lytic activity of human MAC. Mutant human C9 in which Ala was substituted for Cys359/384 was found to express normal lytic activity and to be fully inhibited by CD59. This suggests that the intrachain Cys359/Cys384 disulfide bond within C9 is not required to maintain the conformation of this segment of C9 for interaction with CD59.


Assuntos
Antígenos CD59/metabolismo , Complemento C9/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD59/farmacologia , Complemento C9/antagonistas & inibidores , Complemento C9/genética , Proteínas Inativadoras do Complemento/farmacologia , Complexo de Ataque à Membrana do Sistema Complemento , Dissulfetos , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Especificidade da Espécie , Relação Estrutura-Atividade , Transfecção
12.
Clin Exp Immunol ; 101(1): 136-41, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7542572

RESUMO

Vitronectin (complement S-protein), a multifunctional glycoprotein, inhibits complement-mediated cytolysis at two identified stages of terminal complement complex (TCC) formation: blocking of C5b-7 membrane binding, and prevention of C9 polymerization. However, the functional domain(s) of vitronectin involved in these reactions remains incompletely defined. In order to identify the complement inhibition site, a 12-kD heparin binding fragment and two other internal fragments (53 kD and 43 kD) of vitronectin were isolated after cyanogen bromide (CNBr) treatment of the native molecule. Potent inhibition of guinea pig erythrocyte (GPE) reactive lysis was demonstrated with native vitronectin, total CNBr digest and the 53-kD and 43-kD fragments, but only very poorly by the heparin binding 12-kD peptide. Similarly, the 43-kD fragment blocked the binding of C5b-7 to immobilized vitronectin, whereas the 12-kD fragment had no effect. These data localize the C5b-7 binding site to a 43-kD internal region. Further characterization of the fragments was carried out in an assay which detected C9 polymerization in the presence of C5b-8. Polymerized material was separated by PAGE, detected by autoradiography and quantified after excision from the gels. Results showed that polymerization did not occur in the presence of the 53-kD and 43-kD fragments. However, the 12-kD heparin binding fragment had no effect. It is proposed that prevention of C5b-8-induced C9 polymerization resides at a site in an internal region of the vitronectin molecule.


Assuntos
Proteínas Inativadoras do Complemento/metabolismo , Proteínas Inativadoras do Complemento/fisiologia , Glicoproteínas/metabolismo , Glicoproteínas/fisiologia , Heparina/metabolismo , Sequência de Aminoácidos , Complemento C5/antagonistas & inibidores , Complemento C5/metabolismo , Complemento C5b , Complemento C7/antagonistas & inibidores , Complemento C7/metabolismo , Complemento C9/antagonistas & inibidores , Complemento C9/metabolismo , Proteínas Inativadoras do Complemento/química , Brometo de Cianogênio/farmacologia , Glicoproteínas/química , Humanos , Dados de Sequência Molecular , Polímeros/metabolismo , Vitronectina
13.
Immunol Invest ; 22(2): 127-49, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8505069

RESUMO

The exact mechanism by which deposited C5b-9 complexes kill Gram-negative bacteria is unclear. It has been proposed that during complement activation the membrane attack complex triggers an energy dependent process in Gram-negative bacteria that mediates destruction of the inner membrane. This observation in part resulted from the survival of Gram-negative bacteria that were incubated with an uncoupler (DNP) or an inhibitor (KCN) of oxidative phosphorylation during complement activation. In a reexamination of this issue we employed potassium cyanide (KCN) to block energy dependent pathways and observed a dose dependent inhibition of C9 uptake on E. coli J5 during serum incubation, suggesting that cyanide was interfering with complement activation. To verify the effect on complement activation we chose specifically to study the effects of KCN on the C3 convertase of the classical pathway. Sensitized sheep erythrocytes were employed as our model system. This system allowed us to construct a series of stable intermediates that were used to test the effect of cyanide on the formation and activity of precursors of the classical pathway C3 convertase. The data illustrate that the concentrations of potassium cyanide that inhibit complement killing of J5 also inhibit C3 convertase activity on sensitized sheep erythrocytes. The results of this study refute the principal observation made by other investigators, that potassium cyanide protects bacteria from complement killing by inhibiting bacterial energy dependent pathways that spark inner membrane destruction. A better scenario is that the organisms survive because cyanide inhibits complement activation.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Convertases de Complemento C3-C5/efeitos dos fármacos , Complemento C9/antagonistas & inibidores , Escherichia coli/efeitos dos fármacos , Cianeto de Potássio/farmacologia , Animais , Convertases de Complemento C3-C5/antagonistas & inibidores , Citotoxicidade Imunológica , Relação Dose-Resposta a Droga , Escherichia coli/imunologia , Humanos
14.
Cell Immunol ; 126(1): 176-84, 1990 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1689219

RESUMO

Here we compare the properties of leukocyte antigens H19 and CD59 with those of the PI-linked 18,000-20,000 Mr molecules which inhibit lysis of human cells by the autologous terminal complement components C5b-9. H19, a 19,000 Mr protein found on human erythrocytes, monocytes, neutrophils, T-lymphocytes and other cells, is one of the ligands involved in the spontaneous rosette formation between human T-lymphocytes and erythrocytes. Recent evidence indicates that H19 also participates in T-cell activation. CD59 is a widely distributed 18,000-25,000 Mr protein anchored to the cell membrane by phosphatidylinositol (PI). The function of CD59 is unknown. Affinity-purified H19 incorporates into cell membranes and inhibits channel formation by human C5b-9 on guinea pig erythrocytes. Significant inhibition is achieved with picogram quantities of H19, corresponding to approximately 600 molecules per erythrocyte. H19 is most effective when C9 is limiting but quite active when C5b-7 or C8 are limiting, indicating that it may interact with several of the structurally related terminal complement components. The inhibitory activity is blocked by mAbs to either CD59 or to H19. H19 is PI-anchored: it is released from the cell membrane by treatment with PI-specific phospholipase C, and it is absent from cells from a patient with paroxysmal nocturnal hemoglobinuria (PNH). Analysis of PNH erythrocytes after treatment with terminal complement proteins shows that the H19-negative erythrocytes are more susceptible to C5b-9-mediated lysis. Treatment of normal human erythrocytes with either anti-H19 or anti-CD59 renders them more susceptible to lysis by human C5b-9. We conclude that H19 and CD59 are probably the same molecule and are identical or closely related to the recently described inhibitors of C5b-9 channel formation.


Assuntos
Antígenos de Diferenciação/fisiologia , Antígenos de Superfície/fisiologia , Complemento C5/antagonistas & inibidores , Complemento C9/antagonistas & inibidores , Proteínas Inativadoras do Complemento/fisiologia , Ativação Linfocitária , Linfócitos T/imunologia , Anticorpos Monoclonais/imunologia , Antígenos de Diferenciação/imunologia , Antígenos de Superfície/imunologia , Antígenos CD59 , Complemento C5b , Hemoglobinúria Paroxística/sangue , Hemólise
15.
Nature ; 322(6082): 831-4, 1986.
Artigo em Inglês | MEDLINE | ID: mdl-2427956

RESUMO

Cytolysis mediated by complement or cytolytic lymphocytes results in the formation of morphology similar lesions in the target membrane. These lesions, formed by the polymerization of C9 or perforin respectively, contribute the major killing action by causing osmotic lysis of the target cell. Following the suggestion of Mayer that the mechanisms of humoral and cell-mediated cytotoxicity might be related, studies into the morphology of the membrane lesions formed, and the proteins responsible for causing the lesions, have shown several similarities. While the lesion caused by natural and T-killer cells is a little larger than that caused by complement, its overall shape is similar and in both cases the cylindrical pore is formed by polymerization of a monomeric subunit, C9 (relative molecular mass, Mr = 71,000) for complement, and perforin (Mr = 66,000) for cell-mediated cytotoxicity. C9 has an absolute requirement for a receptor in the target membrane formed by the earlier membrane attack complex components, C5b, C6, C7 and C8 (ref. 8). For perforin, polymerization in a target membrane requires no receptor, specificity being derived from the specific recognition between killer and target cell. Both proteins can be made to polymerize in vitro by the addition of divalent cations (Zn2+ for C9 (ref. 16) and Ca2+ for perforin) and the resultant complexes closely resemble their physiological counterparts. Antibodies raised against lymphocyte-killed targets have also been shown to cross-react with complement proteins, but the antigenically related proteins were not determined in these studies. We show here using purified proteins that perforin, C9 and complexes involving C7 and C8 share a common antigenic determinant which is probably involved in polymerization.


Assuntos
Complemento C9/fisiologia , Proteínas do Sistema Complemento/fisiologia , Citotoxicidade Imunológica , Glicoproteínas de Membrana , Proteínas de Membrana/fisiologia , Linfócitos T Citotóxicos/imunologia , Sequência de Aminoácidos , Biopolímeros , Membrana Celular/ultraestrutura , Complemento C9/antagonistas & inibidores , Complemento C9/genética , Complexo de Ataque à Membrana do Sistema Complemento , Proteínas do Sistema Complemento/genética , Epitopos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Fragmentos de Peptídeos/farmacologia , Perforina , Proteínas Citotóxicas Formadoras de Poros , Receptores de LDL/genética , Homologia de Sequência do Ácido Nucleico , Suramina/farmacologia
16.
J Clin Lab Immunol ; 6(1): 7-11, 1981 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7265179

RESUMO

Further studies were carried out on the C9 deficiency (C9D). Her serum complement activity (CH50) was 15.7 units when assayed in high ionic strength buffer and 8.8 or less than 5.0 units when assayed in low ionic strength buffer containing glucose or sucrose, respectively. It was revealed that this buffer-dependent CH50 variation of C9D serum was due to the effect of the buffer on the spontaneous lysis of EAC1-8. The serum bactericidal activity of C9D was low, but the addition of specific antibody against bacteria increased the activity indicating an important role of antibody in the serum bactericidal activity. Neither C9 inactivator(s) nor antibody against C9 was detected in the serum, indicating that the case had a defect of C9 synthesis. However, the estimation of C9 levels in the sera of her family could not reveal the mode of inheritance of C9D.


Assuntos
Complemento C9/deficiência , Adulto , Atividade Bactericida do Sangue , Proteínas Sanguíneas/análise , Soluções Tampão/farmacologia , Complemento C9/antagonistas & inibidores , Complemento C9/genética , Via Clássica do Complemento , Feminino , Humanos
19.
J Immunol ; 115(6): 1558-65, 1975 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1184967

RESUMO

Sodium cyanate added to normal human serum or serum from patients with sickle-cell disease resulted in the functional inactivation of C3, C5, C6, C7, and the C3b inactivator, but not C8 and C9. Final concentrations as low as 0.5 mM in serum caused inactivation of 12 to 64% of the C3 after 8 hr at 37 degrees C. The activity of the inactivated C3, C5, and C3b inactivator was not restored by dialysis. Most of the functional activity of C3 in cyanate-treated sera was destroyed by very small quantities of 14C-labeled cyanate that was bound to the protein. C3 inactivation by cyanate occurred in heated sera (50 degrees C, 30 min) and sera treated with EDTA, probably indicating that one mechanism for inactivation was by a direct carbamylation reaction. Both C3 and C5 showed two anodal-migrating forms in two dimensional antigen-antibody crossed electrophoresis in some sera treated with low concentrations of cyanate. Measurements of circular dichroism of highly purified carbamylated C3 showed no detectable changes in structure even though most of the functional activity was destroyed. Purified, inactive C3 that was carbamylated with 14C-labeled cyanate was capable of binding to EAC142, but the resulting EAC1423 was weakly positive for immune adherence and negative for agglutination with anti-C3 antiserum. Unlabeled, cell-bound C3b on EAC142 was not susceptible to cyanate action as shown by no loss in immune adherence and positive agglutination with anti-C3 antiserum. The C3b inactivator was more susceptible to cyanate than C3 in a short time period, whereas both were inactivated after 8 hr. Since cyanate is currently being evaluated as a treatment for sickle-cell disease, the inactivation of C3 by the drug is an important consideration for such patients who are already deficient in C3 dependent heat-labile opsonins that aid in host defense.


Assuntos
Proteínas Inativadoras do Complemento , Cianatos/farmacologia , Anemia Falciforme/imunologia , Complemento C3/antagonistas & inibidores , Complemento C5/antagonistas & inibidores , Complemento C6/antagonistas & inibidores , Complemento C7/antagonistas & inibidores , Complemento C8/antagonistas & inibidores , Complemento C9/antagonistas & inibidores , Proteínas do Sistema Complemento/metabolismo , Hemólise , Humanos , Reação de Imunoaderência , Imunoeletroforese Bidimensional , Conformação Proteica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...