Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Transplant ; 8(6): 1129-42, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18444931

RESUMO

We explored whether a functionally blocking anti-C5 monoclonal antibody (mAb) combined with T- and B-cell immunosuppression can successfully prevent antibody-mediated (AMR) and cell-mediated rejection (CMR) in presensitized murine recipients of life-supporting kidney allografts. To mimic the urgent clinical features of AMR experienced by presensitized patients, we designed a murine model in which BALB/c recipients were presensitized with fully MHC-mismatched C3H donor skin grafts one week prior to C3H kidney transplantation. Presensitized recipients demonstrated high levels of circulating and intragraft antidonor antibodies and terminal complement activity, rejecting grafts within 8.5 +/- 1.3 days. Graft rejection was predominantly by AMR, characterized by interstitial hemorrhage, edema and glomerular/tubular necrosis, but also demonstrated moderate cellular infiltration, suggesting CMR involvement. Subtherapeutic treatment with cyclosporine (CsA) and LF15-0195 (LF) did not significantly delay rejection. Significantly, however, the addition of anti-C5 mAb to this CsA/LF regimen prevented terminal complement activity and inhibited both AMR and CMR, enabling indefinite (>100 days) kidney graft survival despite the persistence of antidonor antibodies. Long-term surviving kidney grafts expressed the protective proteins Bcl-x(S/L) and A-20 and demonstrated normal histology, suggestive of graft accommodation or tolerance. Thus, C5 blockade combined with routine immunosuppression offers a promising approach to prevent graft loss in presensitized patients.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complemento C5/antagonistas & inibidores , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Imunossupressores/uso terapêutico , Transplante de Rim/imunologia , Animais , Modelos Animais de Doenças , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/efeitos dos fármacos , Imunização , Transplante de Rim/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos BALB C
2.
Haematologica ; 92(3): e31-3, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17405753

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by intravascular hemolysis leading to anemia and other clinical manifestations. Transfusions are often required to support hemoglobin at tolerable levels. A PNH patient with aplastic anemia was treated with the complement inhibitor eculizumab, followed by concurrent treatment with recombinant human erythropoietin (rHuEpo). Eculizumab alone reduced hemolysis, increased PNH red blood cell (RBC) mass, and decreased transfusions. Addition of rHuEpo during eculizumab therapy, enhanced erythropoiesis, further increased PNH RBC mass and hemoglobin levels, and rendered the patient transfusion independent for more than two years. These data show that driving erythropoiesis during eculizumab treatment provided further benefit to a patient with PNH and underlying bone marrow failure.


Assuntos
Anemia Aplástica/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Complemento C5/antagonistas & inibidores , Eritropoese/efeitos dos fármacos , Eritropoetina/farmacologia , Hemoglobinúria Paroxística/tratamento farmacológico , Hemólise/efeitos dos fármacos , Anemia Aplástica/sangue , Anemia Aplástica/etiologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Transfusão de Sangue , Complemento C5/imunologia , Sinergismo Farmacológico , Índices de Eritrócitos/efeitos dos fármacos , Eritropoetina/uso terapêutico , Hemoglobinas/análise , Hemoglobinúria Paroxística/complicações , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes , Resultado do Tratamento
3.
Br J Surg ; 92(10): 1227-34, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16078298

RESUMO

BACKGROUND: Ruptured abdominal aortic aneurysm (RAAA) is associated with a systemic inflammatory response syndrome and multiple organ dysfunction. The potential role of a novel C5 complement inhibitor in attenuation of pathological complement activation and tissue injury was explored in a model of RAAA. METHODS: Anaesthetized rats were randomized to sham (control) or shock and clamp (SC) groups. Animals in the SC group underwent 1 h of haemorrhagic shock (mean arterial pressure 50 mmHg or less), 45 min of supramesenteric aortic clamping and 2 h of reperfusion. They were randomized to receive an intravenous bolus of a functionally blocking anti-C5 monoclonal antibody (C5 inhibitor), at a dose of 20 mg/kg, or saline. Lung injury was assessed by permeability to 125I-labelled albumin, tissue myeloperoxidase (MPO) activity, and semiquantitative reverse transcriptase-polymerase chain reaction (RT-PCR) for mRNAs encoding tumour necrosis factor (TNF) alpha and interleukin (IL) 6. RESULTS: The lung permeability index was significantly increased in the SC compared with the sham group (P = 0.032); this was prevented by the C5 inhibitor (P = 0.015). Lung MPO activity was significantly increased in the SC compared with the sham group (P < 0.001), and this increase was attenuated by treatment with the C5 inhibitor (P < 0.001). Semiquantitative RT-PCR in SC group demonstrated downregulation of TNF-alpha mRNA (P = 0.050) and upregulation of IL-6 mRNA (P < 0.001), which were both prevented by the C5 inhibitor (P = 0.014 and P < 0.001 respectively). CONCLUSION: These results indicated that C5 complement inhibition can reduce shock and acute lung injury in an experimental model of RAAA.


Assuntos
Aneurisma da Aorta Abdominal/imunologia , Ruptura Aórtica/imunologia , Complemento C5/antagonistas & inibidores , Síndrome do Desconforto Respiratório/prevenção & controle , Choque Hemorrágico/prevenção & controle , Animais , Aneurisma da Aorta Abdominal/enzimologia , Ruptura Aórtica/enzimologia , Pressão Sanguínea , Ativação do Complemento/imunologia , Interleucina-6/metabolismo , Masculino , Permeabilidade , Peroxidase/metabolismo , RNA/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Síndrome do Desconforto Respiratório/enzimologia , Síndrome do Desconforto Respiratório/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Choque Hemorrágico/imunologia , Fator de Necrose Tumoral alfa/metabolismo
4.
Lupus ; 13(5): 328-34, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15230287

RESUMO

The importance of the complement system in the pathophysiology of systemic lupus erythematosus (SLE) is clear although individual complement components play very different roles in the disease process. Early complement proteins are critical in the clearance of immune complexes and apoptotic bodies, and their absence predisposes individuals to SLE. Conversely, activation of terminal complement is associated with exacerbations of disease and damage to tissues and organs, particularly in lupus nephritis. Monoclonal antibodies that specifically inhibit terminal complement activation while preserving the critical functions of the early complement cascade have now been developed. These antibodies target the C5 complement protein, blocking its cleavage and the subsequent generation of potent proinflammatory molecules. Anti-C5 therapeutics have recently been investigated in an animal model of SLE and in a Phase I single dose study in humans. The results of these studies and the multiple roles of complement in SLE are discussed.


Assuntos
Proteínas Inativadoras do Complemento/uso terapêutico , Proteínas do Sistema Complemento/fisiologia , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Humanos
5.
J Immunol ; 167(4): 1996-2003, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11489981

RESUMO

A novel costimulatory molecule expressed on activated T cells, inducible costimulator (ICOS), and its ligand, B7-related protein-1 (B7RP-1), were recently identified. ICOS costimulation leads to the induction of Th2 cytokines without augmentation of IL-2 production, suggesting a role for ICOS in Th2 cell differentiation and expansion. In the present study, a soluble form of murine ICOS, ICOS-Ig, was used to block ICOS/B7RP-1 interactions in a Th2 model of allergic airway disease. In this model, mice are sensitized with inactivated Schistosoma mansoni eggs and are subsequently challenged with soluble S. mansoni egg Ag directly in the airways. Treatment of C57BL/6 mice with ICOS-Ig during sensitization and challenge attenuated airway inflammation, as demonstrated by a decrease in cellular infiltration into the lung tissue and airways, as well as by a decrease in local IL-5 production. These inhibitory effects were not due to a lack of T cell priming nor to a defect in Th2 differentiation. In addition, blockade of ICOS/B7RP-1 interactions during ex vivo restimulation of lung Th2 effector cells prevented cytokine production. Thus, blockade of ICOS signaling can significantly reduce airway inflammation without affecting Th2 differentiation in this model of allergic airway disease.


Assuntos
Antígenos de Diferenciação de Linfócitos T/fisiologia , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/patologia , Células Th2/citologia , Células Th2/imunologia , Animais , Antígenos de Diferenciação de Linfócitos T/administração & dosagem , Antígenos de Diferenciação de Linfócitos T/genética , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígenos de Helmintos/administração & dosagem , Antígeno B7-1/metabolismo , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/biossíntese , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Imunoglobulina E/sangue , Imunossupressores/administração & dosagem , Ligante Coestimulador de Linfócitos T Induzíveis , Proteína Coestimuladora de Linfócitos T Induzíveis , Inflamação/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Schistosoma mansoni/imunologia , Células Th2/metabolismo
6.
J Immunol ; 166(6): 4148-53, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11238665

RESUMO

Complement plays a significant role in mediating endothelial injury following oxidative stress. We have previously demonstrated that the lectin complement pathway (LCP), which is initiated by deposition of the mannose-binding lectin (MBL), is largely responsible for activating complement on endothelial cells following periods of oxidative stress. Identifying functional inhibitors that block MBL binding will be useful in characterizing the role of the LCP in disease models. The human cytokeratin peptide SFGSGFGGGY has been identified as a molecular mimic of N-acetyl-D-glucosamine (GlcNAc), a known ligand of MBL. Thus, we hypothesized that this peptide would specifically bind to MBL and functionally inhibit the LCP on endothelial cells following oxidative stress. Using a BIAcore 3000 optical biosensor, competition experiments were performed to demonstrate that the peptide SFGSGFGGGY inhibits binding of purified recombinant human MBL to GlcNAc in a concentration-dependent manner. Solution affinity data generated by BIAcore indicate this peptide binds to MBL with an affinity (K(D)) of 5 x 10(-5) mol/L. Pretreatment of human serum (30%) with the GlcNAc-mimicking peptide (10-50 microg/ml) significantly attenuated MBL and C3 deposition on human endothelial cells subjected to oxidative stress in a dose-dependent manner, as demonstrated by cell surface ELISA and confocal microscopy. Additionally, this decapeptide sequence attenuated complement-dependent VCAM-1 expression following oxidative stress. These data indicate that a short peptide sequence that mimics GlcNAc can specifically bind to MBL and functionally inhibit the proinflammatory action of the LCP on oxidatively stressed endothelial cells.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Queratinas/imunologia , Queratinas/metabolismo , Lectinas/metabolismo , Mananas/metabolismo , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Acetilglucosamina/metabolismo , Ligação Competitiva/imunologia , Linhagem Celular , Células Cultivadas , Colectinas , Ativação do Complemento/imunologia , Complemento C3/antagonistas & inibidores , Complemento C3/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Humanos , Imunossupressores/metabolismo , Mimetismo Molecular , Estresse Oxidativo/imunologia , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Ressonância de Plasmônio de Superfície , Transfecção
7.
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
8.
J Immunol ; 164(12): 6340-8, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10843688

RESUMO

Recent studies have suggested that soluble forms of B7-1 and B7-2 may exist, but transcripts that code for these molecules have not been previously described. In this study, we report the cloning and characterization of an alternatively spliced soluble form of porcine B7-1 (sB7-1) that lacks exons coding for both the transmembrane and cytoplasmic domains. Northern blot analysis of RNA from alveolar macrophages revealed an approximate 3:1 ratio of the transmembrane form of B7-1 mRNA relative to sB7-1 mRNA. Porcine B7-1 was present on the surface of both B and T cells following stimulation with PMA/ionomycin. A histidine-tagged form of porcine sB7-1 (sB7-1-His) interacted with both CD28 and CTLA-4, and effectively blocked IL-2 production from human responder cells stimulated with PHA and either porcine or human stimulator cells. In addition, sB7-1-His inhibited human T cell proliferation in response to porcine or human peripheral blood leukocytes. This study is the first report of an alternatively spliced form of B7 that codes for a soluble protein. Furthermore, these data demonstrate that porcine B7-1 interacts with the human receptors CD28 and CTLA-4, suggesting a potential role for this molecule in pig to human xenotransplantation. Possible physiological functions for the soluble form of B7-1 are discussed.


Assuntos
Processamento Alternativo/imunologia , Antígeno B7-1/química , Antígeno B7-1/fisiologia , Imunoconjugados , Abatacepte , Adulto , Sequência de Aminoácidos , Animais , Antígenos CD , Antígenos de Diferenciação/metabolismo , Antígenos Heterófilos/imunologia , Antígeno B7-1/biossíntese , Antígeno B7-1/genética , Sequência de Bases , Northern Blotting , Antígenos CD28/metabolismo , Antígeno CTLA-4 , Células Cultivadas , Clonagem Molecular , Dimerização , Humanos , Imunossupressores/farmacologia , Interleucina-2/antagonistas & inibidores , Interleucina-2/biossíntese , Isoantígenos/imunologia , Células Jurkat , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/química , Dados de Sequência Molecular , Ligação Proteica , Solubilidade , Suínos , Linfócitos T/imunologia , Linfócitos T/metabolismo
9.
Transplantation ; 69(8): 1659-67, 2000 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-10836378

RESUMO

BACKGROUND: Rapid leukocyte adherence to donor organ vasculature is a hallmark of hyperacute xenograft rejection. However, the molecular interactions required for leukocyte binding to vascular endothelium have not been characterized. METHODS AND RESULTS: Binding assays performed between human neutrophils and porcine aortic endothelial cells (PAEC) after exposure to human complement demonstrated that adhesion was mediated by both surface-bound C3b and C5b-9 activity. C5b-9-dependent adhesion was blocked by neuraminidase treatment of the neutrophils, suggesting that this binding was mediated by porcine P-selectin. Porcine P-selectin was isolated from a PAEC cDNA library. The porcine P-selectin primary sequence contained an open reading frame encoding 646 amino acids with 82% identity to human P-selectin. Recombinant soluble porcine P-selectin specifically bound to human neutrophils and HL-60 cells. Transfection of COS cells with the full-length porcine P-selectin cDNA resulted in surface expression of the protein and markedly increased the binding of human neutrophils to these cells. The binding of both soluble and COS-expressed porcine P-selectin to human neutrophils was blocked by pretreatment of the neutrophils with neuraminidase or the addition of EDTA. Finally, treatment of PAEC with human thrombin or normal human serum but not purified human C5a- or C8-deficient human serum resulted in the rapid expression of porcine P-selectin on the cell surface. CONCLUSIONS: This report establishes that porcine P-selectin supports the binding of human neutrophils to PAEC in vitro. Further, these data suggest that sublytic deposition of C5b-9 during hyperacute rejection results in the expression of porcine P-selectin, which may contribute to the rapid adhesion of neutrophils to porcine xenografts.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Endotélio Vascular/fisiologia , Leucócitos/fisiologia , Selectina-P/fisiologia , Suínos/imunologia , Sequência de Aminoácidos/genética , Animais , Aorta/citologia , Aorta/fisiologia , Células COS , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Células Cultivadas , Clonagem Molecular , Complemento C3b/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Endotélio Vascular/citologia , Humanos , Dados de Sequência Molecular , Neutrófilos/efeitos dos fármacos , Neutrófilos/fisiologia , Selectina-P/genética , Proteínas Recombinantes/farmacologia , Transfecção
11.
J Virol ; 72(6): 4650-6, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9573228

RESUMO

Human sera contain high levels of natural antibody (Ab) to Galalpha1-3Gal, a terminal glycosidic structure expressed on the surface of cells of mammals other than Old World primates. Incorporation of this determinant onto retroviral membranes by passage of viruses in cells encoding alpha-1-3-galactosyltransferase (GT) renders retroviruses sensitive to lysis by natural Ab and complement in normal human serum (NHS). Plasma membrane-budding viruses representing four additional virus groups were examined for their sensitivities to serum inactivation after passage through human cell lines that lack a functional GT or human cells expressing recombinant porcine GT. The inactivation of lymphocytic choriomeningitis virus (LCMV) by NHS directly correlated with host modification of the virus via expression of Galalpha1-3Gal and was blocked by incorporation of soluble Galalpha1-3Gal disaccharide into the inactivation assay. GT-deficient mice immunized to make high levels of Ab to Galalpha1-3Gal (anti-Gal Ab) were tested for resistance to LCMV passaged in GT-expressing cells. Resistance was not observed, but in vitro analyses of the mouse immune sera revealed that the antiviral activity of the sera was insufficient to eliminate LCMV infectivity on its natural targets of infection, macrophages, which express receptors for Ab and complement. Newcastle disease virus and vesicular stomatitis virus (VSV) were inactivated by NHS regardless of cell passage history, whereas Sindbis virus (SV) passaged in human cells resisted inactivation. Both VSV and SV passaged in Galalpha1-3Gal-expressing human cells incorporated this sugar moiety onto their major envelope glycoproteins. SV passaged in mouse cells expressing Galalpha1-3Gal was moderately sensitive to inactivation by NHS. These results indicate that enveloped viruses expressing Galalpha1-3Gal differ in their sensitivities to NHS and that a potent complement source, such as that in NHS, is required for efficient inactivation of sensitive viruses in vitro and in vivo.


Assuntos
Anticorpos Antivirais/imunologia , Formação de Anticorpos , Antígenos Virais/imunologia , Glicosídeos/imunologia , Herpesvirus Humano 1/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , Especificidade de Anticorpos , Linhagem Celular , Epitopos/imunologia , Humanos , Camundongos , Proteínas do Envelope Viral/imunologia
12.
J Immunol ; 159(9): 4356-61, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9379032

RESUMO

Retroviruses from various mammalian species, excluding humans, are effectively inactivated in normal human serum (NHS). Recent studies have shown that NHS inactivation of retroviruses occurs through natural Ab recognition of a terminal glycosidic moiety on the viral envelope that is acquired during replication in the host cell. This carbohydrate structure (the alpha-galactosyl epitope) is expressed on the cells of most mammals, with the exception of humans and other Old World primates. In this study, NHS sensitivity of HIV was assessed following viral propagation in human cells that were manipulated to express the alpha-galactosyl epitope. HUT-78 cells were transduced with an exogenous alpha1-3-galactosyl transferase gene, which codes for the terminal glycosyl transferase responsible for generation of the alpha-galactosyl epitope. The transduced HUT-78 cells expressed high levels of the alpha-galactosyl epitope on their membrane surface, rendering them sensitive to killing in NHS. Similarly, HIV passaged through these cells acquired the alpha-galactosyl epitope in association with the envelope glycoprotein gp120 and was also effectively inactivated in NHS. Viral inactivation was abolished by the addition of a synthetic disaccharide that contains the alpha-galactosyl epitope, indicating that virolysis is mediated by anti-alpha-galactosyl natural Ab. These results demonstrate that, like other retroviruses bearing the alpha-galactosyl epitope, HIV modified to express this epitope is inactivated in NHS. Furthermore, these data suggest that expression of the alpha-galactosyl epitope on the surface of viruses may have implications in the interspecies transmission of such viruses to humans.


Assuntos
Anticorpos Anti-HIV/imunologia , Antígenos HIV/imunologia , Soropositividade para HIV , HIV-1/imunologia , Epitopos Imunodominantes/imunologia , Animais , Linhagem Celular , Glicosilação , Anticorpos Anti-HIV/sangue , Humanos , Imunidade Celular , Epitopos Imunodominantes/genética , Suínos , Transfecção
13.
Mol Immunol ; 33(17-18): 1389-401, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9171898

RESUMO

Activation of the complement system contributes significantly to the pathogenesis of numerous acute and chronic diseases. Recently, a monoclonal antibody (5G1.1) that recognizes the human complement protein C5, has been shown to effectively block C5 cleavage, thereby preventing the generation of the pro-inflammatory complement components C5a and C5b-9. Humanized 5G1.1 antibody, Fab and scFv molecules have been produced by grafting the complementarity determining regions of 5G1.1 on to human framework regions. Competitive ELISA analysis indicated that no framework changes were required in the humanized variable regions for retention of high affinity binding to C5, even at framework positions predicted by computer modeling to influence CDR canonical structure. The humanized Fab and scFv molecules blocked complement-mediated lysis of chicken erythrocytes and porcine aortic endothelial cells in a dose-dependent fashion, with complete complement inhibition occurring at a three-fold molar excess, relative to the human C5 concentration. In contrast to a previously characterized anti-C5 scFv molecule, the humanized h5G1.1 scFv also effectively blocked C5a generation. Finally, an intact humanized h5G1.1 antibody blocked human complement lytic activity at concentrations identical to the original murine monoclonal antibody. These results demonstrate that humanized h5G1.1 and its recombinant derivatives retain both the affinity and blocking functions of the murine 5G1.1 antibody, and suggest that these molecules may serve as potent inhibitors of complement-mediated pathology in human inflammatory diseases.


Assuntos
Complemento C5/imunologia , Proteínas Inativadoras do Complemento/farmacologia , Fragmentos de Imunoglobulinas/farmacologia , Região Variável de Imunoglobulina/farmacologia , Proteínas Recombinantes de Fusão/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Bloqueadores/genética , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Aorta , Sequência de Bases , Ligação Competitiva/imunologia , Galinhas , Complemento C5a/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Ensaio de Imunoadsorção Enzimática , Eritrócitos/imunologia , Humanos , Região Variável de Imunoglobulina/genética , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/farmacologia , Suínos
14.
J Biol Chem ; 271(39): 23842-5, 1996 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-8798614

RESUMO

CD59 is a glycosylphosphatidylinositol-anchored membrane glycoprotein that serves as the principle cellular inhibitor of the C5b-9 membrane attack complex (MAC) of human complement. Approximately 50% of the total apparent mass of CD59 is attributable to glycosylation of a single Asn (Asn18). The deduced amino acid sequences of CD59 homologues identified in Old and New World primates as well as in rat reveal that the motif for N-linked glycosylation at the residue corresponding to Asn18 of human CD59 is invariably conserved, despite considerable sequence divergence elsewhere in the protein. Such conservation suggests that the post-translational modification at Asn18 has importance for either expression or normal function of CD59 at the cell surface. In this study, we specifically examined how deletion or transposition of the site of N-linked glycosylation in the CD59 polypeptide affects its MAC inhibitory function. Our data demonstrate that the inhibitory potency of CD59 is unaffected when glycosylation is transposed from Asn18 to another site in the polypeptide. Furthermore, we show that CD59 retains normal MAC regulatory function when mutated to eliminate all potential sites for N-linked glycosylation. These data suggest that the MAC inhibitory function of CD59 is entirely provided by residues exposed at the surface of the core polypeptide and that this core structure is not influenced by glycosylation at Asn18.


Assuntos
Antígenos CD59/química , Sequência de Aminoácidos , Asparagina/química , Antígenos CD59/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Análise Mutacional de DNA , Glicoproteínas/fisiologia , Glicosilação , Humanos , Dados de Sequência Molecular , Proteínas Recombinantes , Alinhamento de Sequência , Relação Estrutura-Atividade
16.
Hum Gene Ther ; 7(5): 619-26, 1996 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-8845387

RESUMO

The introduction of retroviral vector producer cells (VPC) into tumors as a means of increasing transduction efficiency has recently been employed in human gene therapy trials. However, the fate of these xenogeneic cells in humans is not well understood. In the present study, we used an in vitro model to examine the survival of commonly used VPC lines in serum from humans and various other species. VPC derived from the murine NIH-3T3 cell line, including PA317, Psi CRIP, and GP + E-86, were effectively killed in sera from Old World primates, including human and baboon. Conversely, the same murine cell lines survived exposure to sera from dog, rabbit, rat, and mouse. This pattern of serum killing parallels the occurrence of the anti-alpha-galactosyl natural antibody (Ab) found exclusively in Old World primates. The anti-alpha-galactosyl Ab targets the terminal glycosidic structure Gal alpha 1-3Gal beta 1-4GlcNAc-R (alpha-galactosyl epitope) found on the surface of mammalian cells, excluding Old World primates. All murine-derived VPC tested expressed high levels of the alpha-galactosyl epitope as determined by FACS analysis. VPC killing was complement-mediated, because preincubation of human serum with a functionally blocking anti-C5 mAb completely abolished cell lysis. Furthermore, addition of soluble galactose(alpha 1-3)galactose (Gal alpha 1-3Gal) to human serum or down-regulation of the alpha-galactosyl epitope on the surface of VPC effectively reduced VPC killing, indicating that complement activation by these cells is primarily initiated by natural antibody recognition of the alpha-galactosyl epitope. Finally, VPC incubated with human serum for 8 hr in the presence of complement inhibition continued to produce viable retroviral particles, thus demonstrating a correlation between VPC and particle survival. Taken together, these data suggest that elimination of the alpha-galactosyl epitope or complement blockade may provide a strategy to prolong the survival of VPC and the particles that they produce in vivo.


Assuntos
Anticorpos/imunologia , Sobrevivência Celular/genética , Proteínas do Sistema Complemento/imunologia , Galactose/imunologia , Vetores Genéticos , Retroviridae/genética , Animais , Sequência de Carboidratos , Células Cultivadas , Cães , Epitopos/imunologia , Citometria de Fluxo , Humanos , Camundongos , Dados de Sequência Molecular , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Papio/metabolismo , Coelhos , Ratos , Retroviridae/metabolismo
17.
Transplantation ; 60(11): 1299-306, 1995 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8525525

RESUMO

Vascular cell adhesion molecule (VCAM) is expressed on activated endothelial cells and binds to the alpha 4 beta 1 integrin receptor, very late antigen-4 (VLA-4), expressed on human lymphoid cells. Anti-VCAM mAbs have been shown to prolong allograft survival. To explore the role of porcine VCAM (pVCAM) in xenotransplantation, a recombinant secreted form of pVCAM (spVCAM) was expressed in 293-EBNA cells and purified by metal affinity chromatography. A human lymphoid cell line bound to spVCAM in a VLA-4-dependent manner. Using spVCAM as an immunogen, we developed three anti-pVCAM mAbs that reacted with cell surface pVCAM on porcine aortic endothelial cells (PAEC) but not to human VCAM on human umbilical vein endothelial cells. Pairwise interaction analysis indicated that these mAbs recognized distinct epitopes on pVCAM. Two anti-pVCAM mAbs, 2A2 and 3F4, inhibited the binding of Ramos cells to spVCAM, while the third, 5D11, did not. Similarly, mAbs 2A2 and 3F4 inhibited binding of Ramos cells or human peripheral blood T cells to activated PAEC. The extent of inhibition with mAbs 2A2 and 3F4 was comparable to the inhibition obtained with a blocking mAb to human VLA-4. These anti-pVCAM mAbs will provide a means to specifically block pVCAM in a xenograft setting and allow the determination of the role of pVCAM in a primary xenogeneic immune response.


Assuntos
Integrinas/metabolismo , Receptores de Retorno de Linfócitos/metabolismo , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Bloqueadores , Anticorpos Monoclonais/imunologia , Sequência de Bases , Adesão Celular , Chlorocebus aethiops , Primers do DNA/química , Humanos , Integrina alfa4beta1 , Dados de Sequência Molecular , Proteínas Recombinantes/imunologia , Especificidade da Espécie , Suínos , Linfócitos T/citologia , Molécula 1 de Adesão de Célula Vascular/imunologia
18.
J Exp Med ; 182(5): 1345-55, 1995 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-7595205

RESUMO

Type C retroviruses endogenous to various nonprimate species can infect human cells in vitro, yet the transmission of these viruses to humans is restricted. This has been attributed to direct binding of the complement component C1q to the viral envelope protein p15E, which leads to classical pathway-mediated virolysis in human serum. Here we report a novel mechanism of complement-mediated type C retrovirus inactivation that is initiated by the binding of "natural antibody" [Ab] (anti-alpha-galactosyl Ab) to the carbohydrate epitope Gal alpha 1-3Gal beta 1-4GlcNAc-R expressed on the retroviral envelope. Complement-mediated inactivation of amphotropic retroviral particles was found to be restricted to human and other Old World primate sera, which parallels the presence of anti-alpha-galactosyl natural Ab. Blockade or depletion of anti-alpha-galactosyl Ab in human serum prevented inactivation of both amphotropic and ecotropic murine retroviruses. Similarly, retrovirus was not killed by New World primate serum except in the presence of exogenous anti-alpha-galactosyl Ab. Enzyme-linked immunosorbent assays revealed that the alpha-galactosyl epitope was expressed on the surface of amphotropic and ecotropic retroviruses, and Western blot analysis further localized this epitope to the retroviral envelope glycoprotein gp70. Finally, down-regulation of this epitope on the surface of murine retroviral particle producer cells rendered them, as well as the particles liberated from these cells, resistant to inactivation by human serum complement. Our data suggest that anti-alpha-galactosyl Ab may provide a barrier for the horizontal transmission of retrovirus from species that express the alpha-galactosyl epitope to humans and to other Old World primates. Further, these data provide a mechanism for the generation of complement-resistant retroviral vectors for in vivo gene therapy applications where exposure to human complement is unavoidable.


Assuntos
Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Sangue/virologia , Cebidae/imunologia , Cercopithecidae/imunologia , Epitopos/imunologia , Galactose/imunologia , Vírus da Leucemia Murina/fisiologia , Proteínas Oncogênicas de Retroviridae/imunologia , Proteínas do Envelope Viral/imunologia , Células 3T3 , Animais , Anticorpos Antivirais/imunologia , Antígenos Virais/biossíntese , Sangue/imunologia , Sequência de Carboidratos , Cebidae/sangue , Cercopithecidae/sangue , Proteínas do Sistema Complemento/imunologia , Humanos , Imunidade Inata , Mamíferos/sangue , Mamíferos/imunologia , Camundongos , Dados de Sequência Molecular , Vírus da Leucemia Murina de Moloney/imunologia , Proteínas Oncogênicas de Retroviridae/biossíntese , Especificidade da Espécie , Proteínas do Envelope Viral/biossíntese
19.
Mol Immunol ; 32(16): 1183-95, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8559143

RESUMO

Complement activation has been implicated in the pathogenesis of several human diseases. Recently, a monoclonal antibody, (N19-8) that recognizes the human complement protein C5 has been shown to effectively block the cleavage of C5 into C5a and C5b, thereby blocking terminal complement activation. In this study, a recombinant N19-8 scFv antibody fragment was constructed from the N19-8 variable regions, and produced in both mammalian and bacterial cells. The N19-8 scFv bound human C5 and was as potent as the N19-8 monoclonal antibody at inhibiting human C5b-9-mediated hemolysis of chicken erythrocytes. In contrast, the N19-8 scFv only partially retained the ability of the N19-8 monoclonal antibody to inhibit C5a generation. To investigate the ability of the N19-8 scFv to inhibit complement-mediated tissue damage, complement-dependent myocardial injury was induced in isolated mouse hearts by perfusion with Krebs-Henseleit buffer containing 6% human plasma. The perfused hearts sustained extensive deposition of human C3 and C5b-9, resulting in increased coronary artery perfusion pressure, end-diastolic pressure, and a decrease in heart rate until the hearts ceased beating approximately 10 min after addition of plasma. Hearts treated with human plasma supplemented with either the N19-8 monoclonal antibody or the N19-8 scFv did not show any detectable changes in cardiac performance for at least 1 hr following the addition of plasma. Hearts treated with human plasma alone showed extensive deposition of C3 and C5b-9, while hearts treated with human plasma containing N19-8 scFv showed extensive deposition of C3, but no detectable deposition of C5b-9. Administration of a 100 mg bolus dose of N19-8 scFv to rhesus monkeys inhibited the serum hemolytic activity by at least 50% for up to 2 hr. Pharmacokinetic analysis of N19-8 scFv serum levels suggested a two-compartment model with a T1/2 alpha of 27 min. Together, these data suggest the recombinant N19-8 scFv is a potent inhibitor of the terminal complement cascade and may have potential in vivo applications where short duration inhibition of terminal complement activity is desirable.


Assuntos
Anticorpos Monoclonais/isolamento & purificação , Complemento C5/imunologia , Região Variável de Imunoglobulina/imunologia , Miocárdio/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Sequência de Bases , Pressão Sanguínea/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Complemento C5/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Humanos , Região Variável de Imunoglobulina/química , Macaca mulatta , Camundongos , Dados de Sequência Molecular , Miocárdio/patologia , Perfusão , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia
20.
J Virol ; 69(6): 3889-92, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7745740

RESUMO

The herpesvirus saimiri genome encodes a complement control protein homolog (CCPH). Stable mammalian cell transfectants expressing a recombinant transmembrane form of CCPH (mCCPH) or a 5'FLAG epitope-tagged mCCPH (5'FLAGmCCPH) conferred resistance to complement-mediated cell damage by inhibiting the lytic activity of human serum complement. The function of CCPH was further defined by showing that the mCCPH and the 5'FLAGmCCPH transfectants inhibited C3 convertase activity and effectively reduced cell surface deposition of the activated complement component, C3d.


Assuntos
Convertases de Complemento C3-C5/antagonistas & inibidores , Proteínas do Sistema Complemento/metabolismo , Herpesvirus Saimiriíneo 2/metabolismo , Glicoproteínas de Membrana/fisiologia , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Primers do DNA , Humanos , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...