Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 37(11): e23256, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37823685

RESUMO

The complement system is a complex, tightly regulated protein cascade involved in pathogen defense and the pathogenesis of several diseases. Thus, the development of complement modulators has risen as a potential treatment for complement-driven inflammatory pathologies. The enzymatically inactive MAP-2 has been reported to inhibit the lectin pathway by competing with its homologous serine protease MASP-2. The membrane-bound complement inhibitor CD55 acts on the C3/C5 convertase level. Here, we fused MAP-2 to the four N-terminal domains of CD55 generating a targeted chimeric inhibitor to modulate complement activation at two different levels of the complement cascade. Its biological properties were compared in vitro with the parent molecules. While MAP-2 and CD55 alone showed a minor inhibition of the three complement pathways when co-incubated with serum (IC50MAP-2+CD55 1-4 = 60.98, 36.10, and 97.01 nM on the classical, lectin, and alternative pathways, respectively), MAP-2:CD551-4 demonstrated a potent inhibitory activity (IC50MAP-2:CD55 1-4 = 2.94, 1.76, and 12.86 nM, respectively). This inhibitory activity was substantially enhanced when pre-complexes were formed with the lectin pathway recognition molecule mannose-binding lectin (IC50MAP-2:CD55 1-4 = 0.14 nM). MAP-2:CD551-4 was also effective at protecting sensitized sheep erythrocytes in a classical hemolytic assay (CH50 = 13.35 nM). Finally, the chimeric inhibitor reduced neutrophil activation in full blood after stimulation with Aspergillus fumigatus conidia, as well as phagocytosis of conidia by isolated activated neutrophils. Our results demonstrate that MAP-2:CD551-4 is a potent complement inhibitor reinforcing the idea that engineered fusion proteins are a promising design strategy for identifying and developing drug candidates to treat complement-mediated diseases.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento , Animais , Ovinos , Antígenos CD55/farmacologia , Lectinas/metabolismo , Fatores de Transcrição , Inativadores do Complemento , Serina Proteases Associadas a Proteína de Ligação a Manose/metabolismo
2.
Oncol Rep ; 42(6): 2686-2693, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31578581

RESUMO

In recent years, efforts to treat cancer by improving the immune function of patients have received a great deal of attention. As part of the immune system, complement is also under such evaluation. Among the many components of the complement system, complement decay accelerating factor (CD55 or DAF) is known to inhibit complement­mediated cell lysis. However, little is known about the role of CD55 in terms of cancer therapy. The present study aimed to demonstrate that increased levels of CD55 are strongly correlated with the progression of colorectal cancer. A novel CD55 chimeric monoclonal antibody was developed that may boost the immune response, thereby suppressing cancer. The CD55 antibody treatment activated complement and therefore suppressed the proliferation, invasion and migration of colorectal cancer cells. This tumoricidal activity is partly explained by the inflammatory response via the activation of proinflammatory cytokines. In addition, the CD55 antibody treatment synergistically enhanced the tumoricidal activity of 5­FU in colorectal cancer cells, suggesting that combined treatment may be a better strategy in colorectal cancer therapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos CD55/genética , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Antígenos CD55/imunologia , Antígenos CD55/farmacologia , Movimento Celular/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Proteínas do Sistema Complemento/genética , Citotoxicidade Imunológica/efeitos dos fármacos , Sinergismo Farmacológico , Fluoruracila/farmacologia , Células HT29 , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica
3.
FASEB J ; 33(11): 12723-12734, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31469600

RESUMO

Dysregulation of the complement system is involved in the pathogenesis of several diseases, and its inhibition has been shown to be a feasible therapeutic option. Therefore, there is an interest in the development of complement modulators to treat complement activation-related inflammatory pathologies. Mannose-binding lectin (MBL)/ficolin/collectin-associated protein-1 (MAP-1) is a regulatory molecule of the lectin pathway (LP), whereas complement receptor 1 (CD35) and decay-accelerating factor (CD55) are membrane-anchored regulators with effects on the central effector molecule C3. In this study, we developed 2 novel soluble chimeric inhibitors by fusing MAP-1 to the 3 first domains of CD35 (CD351-3) or the 4 domains of CD55 (CD551-4) to modulate the complement cascade at 2 different stages. The constructs showed biologic properties similar to those of the parent molecules. In functional complement activation assays, the constructs were very efficient in inhibiting LP activation at the level of C3 and in the formation of terminal complement complex. This activity was enhanced when coincubated with recombinant LP recognition molecules MBL and ficolin-3. Recombinant MAP-1 fusion proteins, combined with recombinant LP recognition molecules to target sites of inflammation, represent a novel and effective therapeutic approach involving the initiation and the central and terminal effector functions of the complement cascade.-Pérez-Alós, L., Bayarri-Olmos, R., Skjoedt, M.-O., Garred, P. Combining MAP-1:CD35 or MAP-1:CD55 fusion proteins with pattern-recognition molecules as novel targeted modulators of the complement cascade.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Reguladoras de Apoptose , Antígenos CD55 , Ativação do Complemento/efeitos dos fármacos , Complemento C3 , Receptores de Complemento 3b , Receptores de Reconhecimento de Padrão , Proteínas Recombinantes de Fusão , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/farmacologia , Animais , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/farmacologia , Antígenos CD55/química , Antígenos CD55/genética , Antígenos CD55/farmacologia , Células CHO , Complemento C3/química , Complemento C3/metabolismo , Cricetulus , Humanos , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Receptores de Reconhecimento de Padrão/química , Receptores de Reconhecimento de Padrão/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia
4.
J Surg Res ; 179(1): 153-67, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23122671

RESUMO

BACKGROUND: Complement is invariably activated during trauma and contributes to tissue injury. Recombinant human decay-accelerating factor (DAF), a complement regulatory protein that inhibits both classical and alternative pathways, improves survival and reduces tissue damage in animal models of tissue injury. The extent to which DAF may facilitate resuscitation in hemorrhaged large animals is not known. METHODS: Male Yorkshire swine assigned to one of six groups were subjected to controlled, isobaric hemorrhage over 15 min to a target mean arterial pressure (MAP) of 35 mm Hg. Hypotension was maintained for 20 min followed by a bolus intravenous injection of DAF or vehicle followed by Hextend resuscitation. Animals were observed for 3 h after hypotensive Hextend resuscitation. Survival, blood chemistry, and physiological parameters were recorded. Additionally, tissue from lung, small intestine, liver, and kidney were subjected to histopathologic evaluation and tissue deposition of complement proteins was determined by immunohistochemistry, dot-blot, and Western blot analyses. RESULTS: Administration of DAF (25 µg/kg) to animals subjected to hemorrhage prior to Hextend infusion significantly improved survival (73% versus 27%); protected gut, lung, liver, and kidney tissue from damage; and resulted in reduced resuscitation fluid requirements when compared with animals subjected to hemorrhage and resuscitation with Hextend alone. Animals treated with a higher dose of DAF (50 µg/kg) followed by Hextend fluid resuscitation did not experience the same benefit, suggesting a narrow therapeutic range for use of DAF as adjunct to Hextend fluid. CONCLUSION: DAF improved survival and reduced early Hextend fluid resuscitation requirements in swine subjected to hemorrhagic shock. These benefits are attributed to decreased complement deposition and limited organ damage.


Assuntos
Antígenos CD55/farmacologia , Intestino Delgado/efeitos dos fármacos , Fígado/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Ressuscitação , Choque Hemorrágico/complicações , Animais , Relação Dose-Resposta a Droga , Hidratação , Derivados de Hidroxietil Amido/uso terapêutico , Intestino Delgado/patologia , Rim/efeitos dos fármacos , Rim/patologia , Fígado/patologia , Pulmão/patologia , Masculino , Modelos Animais , Ressuscitação/métodos , Choque Hemorrágico/mortalidade , Taxa de Sobrevida , Suínos , Resultado do Tratamento
5.
Muscle Nerve ; 45(5): 668-675, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22499093

RESUMO

INTRODUCTION: Autoantibody-induced complement activation, which causes disruption of the postsynaptic membrane, is recognized as a key pathogenic factor in myasthenia gravis (MG). Therefore, specific targeting of complement inhibitors to the site of complement activation is a potential therapeutic strategy for treatment of MG. METHODS: We assessed expression of single-chain antibody fragment-decay accelerating factor (scFv-DAF), comprising a single-chain fragment scFv1956 based on the rat complement inhibitor DAF in prokaryotic systems, and studied its inhibitory effect on complement deposition in vitro. RESULTS: The recombinant conjugate scFv-DAF completely retained the wild-type binding activity of scFv1956 to AChR and inhibited complement activation of DAF in vitro. CONCLUSIONS: We found that scFv-DAF could bind specifically to TE671 cells, and it is significantly more potent at inhibiting complement deposition than the untargeted parent molecule DAF. scFv-DAF may be a candidate for in vivo protection of the AChR in MG.


Assuntos
Antígenos CD55/farmacologia , Proteínas do Sistema Complemento/farmacologia , Receptores Colinérgicos/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Animais , Linhagem Celular Transformada , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Citometria de Fluxo , Humanos , Ligação Proteica/efeitos dos fármacos , Ratos , Rabdomiossarcoma/patologia , Anticorpos de Cadeia Única/metabolismo
6.
Immunobiology ; 216(4): 528-34, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20851495

RESUMO

Despite multiple reports on autoantibody-initiated complement activation in autoimmune hepatitis (AIH), how does the humoral immunity contribute to the pathogenesis of AIH remained unclear. In this report, by adoptively transferring a polyclonal rabbit anti-OVA antibody into Hep-OVA Tg mice in which OVA is selectively expressed on the surface of hepatocytes, we found that excessive complement activation initiated by the autoantibody overwhelmed the protection of intrinsic cell surface complement regulators, and induced hepatocytes injury both in vitro and in vivo. The anti-OVA antibody induced hepatic injury in Hep-OVA Tg but not WT C57BL/6 mice as assessed by serum ALT levels and liver histopathology. Immunohistochemical analyses showed that after the antibody administration, there was massive complement activation on anti-OVA IgG coated hepatocytes in Hep-OVA Tg mice, but not in WT mice. Consistent with these results, depleting complement by cobra venom factor (CVF) prior to antibody injections protected Hep-OVA Tg mice from anti-OVA IgG induced hepatic injury. In addition, treating Hep-OVA Tg mice with recombinant mouse decay accelerating factor, a native complement inhibitor, protected them from autoantibody induced hepatitis. These results suggest that complement could play a pivotal role in liver specific autoantibody mediated hepatocyte injury in AIH, and that complement inhibitors could be, in principle, developed as novel therapeutics against AIH.


Assuntos
Autoanticorpos/imunologia , Proteínas do Sistema Complemento/metabolismo , Hepatite Autoimune/imunologia , Hepatite Autoimune/patologia , Hepatócitos , Adjuvantes Imunológicos/farmacologia , Animais , Antígenos CD55/metabolismo , Antígenos CD55/farmacologia , Antígenos CD59/metabolismo , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Modelos Animais de Doenças , Hepatócitos/imunologia , Hepatócitos/patologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Complemento/metabolismo , Receptores de Complemento 3b
7.
J Surg Res ; 167(2): e103-15, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20189588

RESUMO

BACKGROUND: C-reactive protein (CRP) is an acute pro-inflammatory mediator that has been demonstrated to enhance ischemia/reperfusion (IR) injury by virtue of activating the complement system. CRP is able to interact with complement proteins such as C1q, complement factor H, and C4b-binding protein. Since complement activation is central in the expression of tissue injury following IR, we have investigated the effects of human decay-accelerating factor (DAF), a complement inhibitor, on CRP-potentiated complement activation and tissue injury in mice subjected to mesenteric IR. MATERIALS AND METHODS: Male C57B1/6 mice were allocated into eight groups: (1) Sham-operated group without IR injury; (2) CRP+Sham group; (3) IR group; (4) CRP+IR group; (5) DAF group; (6) CRP+DAF group; (7) IR+DAF group, and (8) CRP+IR+DAF group. Intestinal and lung injury, neutrophil infiltration, myeloperoxidase (MPO) expression, complement component deposition, and interleukin-6 (IL-6) production were assessed for each treatment group of mice. RESULTS: We report that administration of DAF significantly attenuates the CRP-enhanced intestinal injury as well as remote lung damages following acute mesenteric IR in mice, while DAF inhibits complement activation, suppresses neutrophil infiltration, and reduces IL-6 production. CONCLUSIONS: Our study suggests that inhibition complement activation with DAF may prove useful for the treatment of post-ischemic inflammatory injuries associated with an increased production of CRP.


Assuntos
Proteína C-Reativa/metabolismo , Antígenos CD55/uso terapêutico , Mesentério/metabolismo , Mesentério/fisiopatologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Animais , Antígenos CD55/administração & dosagem , Antígenos CD55/farmacologia , Complemento C5a/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Injeções Intravenosas , Interleucina-6/metabolismo , Masculino , Mesentério/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Infiltração de Neutrófilos/efeitos dos fármacos , Peroxidase/metabolismo , Traumatismo por Reperfusão/fisiopatologia
8.
J Neuroinflammation ; 7: 24, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20380727

RESUMO

BACKGROUND: Activated complement system is known to mediate neuroinflammation and neurodegeneration following exposure to hypoxic-ischemic insults. Therefore, inhibition of the complement activation cascade may represent a potential therapeutic strategy for the management of ischemic brain injury. Decay-accelerating factor (DAF, also known as CD55) inhibits complement activation by suppressing the function of C3/C5 convertases, thereby limiting local generation or deposition of C3a/C5a and membrane attack complex (MAC or C5b-9) production. The present study investigates the ability of DAF to protect primary cultured neuronal cells subjected to sodium cyanide (NaCN)-induced hypoxia from degeneration and apoptosis. METHODS: Cultured primary cortical neurons from embryonic Sprague-Dawley rats were assigned one of four groups: control, DAF treatment alone, hypoxic, or hypoxic treated with DAF. Hypoxic cultures were exposed to NaCN for 1 hour, rinsed, followed by 24 hour exposure to 200 ng/ml of recombinant human DAF in normal medium. Human DAF was used in the present study and it has been shown to effectively regulate complement activation in rats. Neuronal cell function, morphology and viability were investigated by measuring plateau depolarization potential, counting the number dendritic spines, and observing TUNEL and MTT assays. Complement C3, C3a, C3a receptor (R) production, C3a-C3aR interaction and MAC formation were assessed along with the generation of activated caspase-9, activated caspase-3, and activated Src. RESULTS: When compared to controls, hypoxic cells had fewer dendritic spines, reduced plateau depolarization accompanied by increased apoptotic activity and accumulation of MAC, as well as up-regulation of C3, C3a and C3aR, enhancement of C3a-C3aR engagement, and elevated caspase and Src activity. Treatment of hypoxic cells with 200 ng/ml of recombinant human DAF resulted in attenuation of neuronal apoptosis and exerted significant protection against neuronal dendritic spine loss and plateau depolarization reduction. Furthermore, treatment with DAF resulted in decreased accumulation of C3a, MAC, C3a-C3aR interaction, caspase-9, activated caspase-3, and pTyr416-Src (activated Src) tyrosine kinase. CONCLUSION: DAF was found to reduce neuronal cell death and apoptosis in NaCN induced hypoxia. This effect is attributed to the ability of DAF to limit complement activation and inhibit the activity of Src and caspases 9 and 3. This study supports the inhibiting of complement as a neuroprotective strategy against CNS ischemia/reperfusion injury.


Assuntos
Antígenos CD55/farmacologia , Hipóxia Celular/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3/metabolismo , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Complemento C3a/metabolismo , Proteínas do Sistema Complemento/fisiologia , Dendritos/fisiologia , Imunofluorescência , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Receptores de Complemento/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
9.
Sheng Wu Gong Cheng Xue Bao ; 24(2): 220-5, 2008 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-18464604

RESUMO

Recombinant expression vector pcDNA3-DAFMCP-DP containing human membrane complement regulatory proteins (hCRPs) decay accelerating factor (DAF) and membrane cofactor protein (MCP) cDNA was constructed by using two independent promoters. After transfected into NIH3T3 cells by calcium phosphate-DNA precipitate method, NIH3T3 pcDNA3-DAFMCP-DP transfectants were obtained by G418 selection. Extraneous genes integration was identified by PCR. The co-expression of human DAF and MCP at both mRNA and protein levels was confirmed by using RT-PCR and Western blot analysis. Human DAF and MCP cDNA were integrated into NIH3T3 pcDNA3-DAFMCP-DP genomic DNA after continuous 30 times passages, indicating that NIH3T3 pcDNA3-DAFMCP-DP were stable cell lines. Human C-mediated cytolysis assays showed that NIH3T3 cells transfected stably with pcDNA3-DAF, pcDNA3-MCP, and pcDNA3-DAFMCP-DP were protected from C-mediated damage and co-expressed human DAF and MCP provided more excellent protection against C-mediated attack, which was compared with either DAF or MCP alone. These results suggest that the dicistronic vector could improve the efficiency of multi-gene delivery and benefit the synergic effect of human membrane complement regulatory proteins DAF and MCP.


Assuntos
Antígenos CD55/biossíntese , Rejeição de Enxerto/prevenção & controle , Proteína Cofatora de Membrana/biossíntese , Células 3T3 , Animais , Antígenos CD55/genética , Antígenos CD55/farmacologia , DNA Complementar/genética , Sinergismo Farmacológico , Humanos , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/farmacologia , Camundongos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Transfecção
10.
Transplant Proc ; 40(2): 551-3, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18374126

RESUMO

UNLABELLED: Swine tissue can express antigens similar to human A/B blood types. We evaluated whether the variation in human blood type influences the human xenoreactive antibody-mediated cytotoxicity and modifies the protective effect of human decay-accelerating factor (hDAF) exogene, a complement activation regulator, on swine endothelium. METHODS: Pig aortic endothelial cells were harvested form normal and hDAF transgenic pigs. Cellular viability was evaluated with an MTT assay. RESULTS: As compared with that of other human blood types, human serum from blood type O donors induced more prominent cytotoxicity on swine endothelial cells both from hDAF transgenic or normal pigs (P < .05). In addition, this difference of xenoreactive antibody-induced cytotoxicity between treatment with O and other human blood type sera was more evident in hDAF transgenic swine endothelial cells than those of normal pigs (P < .05). The hDAF exogene can significantly protect the endothelial cells from human xenoreactive antibody-mediated cytotoxicty when treated with human serum from AB blood type (P < .05). Our data demonstrated that human ABO blood type significantly affected human xenoreactive antibody-induced cytotoxicity, which may modulate the protective effect of hDAF exogene expression on swine endothelial cells.


Assuntos
Sistema ABO de Grupos Sanguíneos/imunologia , Anticorpos Heterófilos/imunologia , Antígenos CD55/genética , Antígenos CD55/farmacologia , Sobrevivência Celular/imunologia , Endotélio Vascular/imunologia , Animais , Endotélio Vascular/efeitos dos fármacos , Humanos , Modelos Biológicos , Suínos , Transplante Heterólogo
11.
Clin Immunol ; 124(3): 311-27, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17631050

RESUMO

Complement activation contributes to the expression of local and remote organ injury in animal models of ischemia-reperfusion (IR). We demonstrate here that a soluble form of decay-accelerating factor (DAF) protects normal C57Bl/6 and autoimmunity-prone B6.MRL/lpr mice subjected to hindlimb IR from remote intestinal and lung injury without affecting the degree of local skeletal muscle injury. In addition, DAF treatment attenuates remote organ injury in mice subjected to mesenteric IR. Soluble DAF allowed the deposition of complement 3 in local and remote injury sites while it limited the presence of terminal membrane attack complex and did not increase animal susceptibility to sepsis. These data provide evidence that soluble DAF might offer clinical benefit to patients suffering remote intestinal or lung damage in response to muscle or other organ injury.


Assuntos
Antígenos CD55/farmacologia , Intestinos/fisiopatologia , Pulmão/fisiopatologia , Músculo Esquelético/fisiopatologia , Traumatismo por Reperfusão/terapia , Animais , Modelos Animais de Doenças , Membro Posterior/irrigação sanguínea , Membro Posterior/fisiopatologia , Intestinos/irrigação sanguínea , Intestinos/patologia , Pulmão/patologia , Mesentério/irrigação sanguínea , Mesentério/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/patologia , Sepse/fisiopatologia , Solubilidade , Taxa de Sobrevida , Distribuição Tecidual
12.
Proc Natl Acad Sci U S A ; 104(1): 240-5, 2007 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17182750

RESUMO

Hemolytic uremic syndrome (HUS) is an important cause of acute renal failure in children. Mutations in one or more genes encoding complement-regulatory proteins have been reported in approximately one-third of nondiarrheal, atypical HUS (aHUS) patients, suggesting a defect in the protection of cell surfaces against complement activation in susceptible individuals. Here, we identified a subgroup of aHUS patients showing persistent activation of the complement alternative pathway and found within this subgroup two families with mutations in the gene encoding factor B (BF), a zymogen that carries the catalytic site of the complement alternative pathway convertase (C3bBb). Functional analyses demonstrated that F286L and K323E aHUS-associated BF mutations are gain-of-function mutations that result in enhanced formation of the C3bBb convertase or increased resistance to inactivation by complement regulators. These data expand our understanding of the genetic factors conferring predisposition to aHUS, demonstrate the critical role of the alternative complement pathway in the pathogenesis of aHUS, and provide support for the use of complement-inhibition therapies to prevent or reduce tissue damage caused by dysregulated complement activation.


Assuntos
Fator B do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Mutação , Antígenos CD55/farmacologia , Fator B do Complemento/química , Fator B do Complemento/fisiologia , Fator H do Complemento/farmacologia , Via Alternativa do Complemento , Síndrome Hemolítico-Urêmica/etiologia , Síndrome Hemolítico-Urêmica/imunologia , Humanos , Relação Estrutura-Atividade
13.
Xenotransplantation ; 13(5): 433-9, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16925667

RESUMO

BACKGROUND: It is difficult to produce a transgenic animal with high expression of decay-accelerating factor (CD55: DAF) or other molecules. The purpose of this study was to assess the effect of tandem forms of DAF on a xenogeneic cell membrane against human complement. METHODS: cDNAs of the delta-Short Consensus Repeat (SCR) 1-DAF, the double-DAF, the triple-DAF, and the tetra-DAF with a FLAG-tag were established. Chinese hamster ovary (CHO) cell lines and a pig endothelial cell (PEC) line expressing these molecules were established. The amelioration of complement-mediated lysis by the transfectant molecules on these cells was examined. The CHO cell transfectants were also incubated with normal human serum, and the amount of C3 deposited was determined by FACS analysis. RESULTS: Stable CHO cells and PEC transfectants, in which each molecule was clearly expressed, and Western blots showed that each band corresponded to the expected molecular weight. The extent of amelioration of complement-mediated lysis by these four molecules was then examined. A clear tendency was found, as follows: The higher the tandem number of DAF, the greater was the effect on cytotoxicity. Additional experiments focusing on triple-DAF and tetra-DAF did not indicate any significant difference in complement-mediated lysis. Consistent with the complement-regulatory ability, the inhibitory effect of the deposition of C3 fragments by these molecules was closely related to the degree of amelioration. CONCLUSION: These data indicate that tandem DAF, especially a triple-DAF, is a very effective form for protecting against complement activation.


Assuntos
Antígenos CD55/farmacologia , Ativação do Complemento/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , Transplante Heterólogo/imunologia , Animais , Antígenos CD55/genética , Células CHO , Sequência Consenso , Cricetinae , DNA Complementar , Citometria de Fluxo , Sequências Repetitivas de Ácido Nucleico , Suínos , Transfecção
14.
Immunohematology ; 21(2): 39-47, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15954803

RESUMO

The antigens of the Cromer blood group system are located on the protein decay-accelerating factor (DAF). This system consists of ten high-prevalence and three low-prevalence antigens; the molecular basis for all of these antigens is a single nucleotide polymorphism in the DAF gene. DAF is a 70,000-Da plasma membrane protein that is widely distributed on all blood cells and on endothelial and epithelial tissues. The physiological role of DAF is to inhibit the complement cascade at the level of the critical C3 convertase step. By this mechanism,DAF acts to protect autologous cells and tissues from complement-mediated damage and hence can play a role in preventing or modulating autoimmune disease and inflammation. The use of recombinant DAF as a therapeutic agent in autoimmunity and inflammation, and of DAF transgenic animals in xenotransplantation, is being actively investigated. Additionally, DAF serves as a receptor for certain strains of Escherichia coli and certain types of enteroviruses. The DAF protein that contains the Cromer antigens serves important roles in health and disease.


Assuntos
Antígenos de Grupos Sanguíneos/fisiologia , Antígenos CD55/fisiologia , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/farmacologia , Antígenos CD55/química , Antígenos CD55/farmacologia , Humanos , Modelos Biológicos , Conformação Proteica
15.
Protein Sci ; 13(9): 2406-15, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15322283

RESUMO

Decay-accelerating factor (DAF, CD55) is a glycophosphatidyl inositol-anchored glycoprotein that regulates the activity of C3 and C5 convertases. In addition to understanding the mechanism of complement inhibition by DAF through structural studies, there is also an interest in the possible therapeutic potential of the molecule. In this report we describe the cloning, expression in Escherichia coli, isolation and membrane-targeting modification of the four short consensus repeat domains of soluble human DAF with an additional C-terminal cysteine residue to permit site-specific modification. The purified refolded recombinant protein was active against both classical and alternative pathway assays of complement activation and had similar biological activity to soluble human DAF expressed in Pichia pastoris. Modification with a membrane-localizing peptide restored cell binding and gave a large increase in antihemolytic potency. These data suggested that the recombinant DAF was correctly folded and suitable for structural studies as well as being the basis for a DAF-derived therapeutic. Crystals of the E. coli-derived protein were obtained and diffracted to 2.2 A, thus permitting the first detailed X-ray crystallography studies on a functionally active human complement regulator protein with direct therapeutic potential.


Assuntos
Antígenos CD55/química , Antígenos CD55/metabolismo , Antígenos CD55/farmacologia , Membrana Celular/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD55/genética , Células Cultivadas , Ativação do Complemento/efeitos dos fármacos , Complemento C3a/antagonistas & inibidores , Cristalização , Escherichia coli/genética , Cobaias , Hemólise/efeitos dos fármacos , Humanos , Corpos de Inclusão/genética , Concentração Inibidora 50 , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Coelhos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Difração de Raios X
16.
Transplantation ; 75(5): 697-702, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12640312

RESUMO

BACKGROUND: Protection against complement activation may reduce acute islet damage in pig-to-human islet xenotransplantation. Expression of the human complement regulatory proteins decay-accelerating factor (DAF, CD55) or CD59 was induced on intact adult porcine islets (APIs) by adenoviral transduction. The functional capacity of the transgenes was examined in vitro after exposure to fresh human serum. METHODS: Intact APIs were transduced with adenoviral vectors Ad.hDAF or Ad.hCD59 or a control vector. After 3 days, the islets were trypsin dissociated to a single-cell suspension. A cytotoxicity assay was performed in which the islet cells were incubated with human complement active AB serum. Flow cytometry and immunohistochemistry were used to evaluate transgene expression. RESULTS: APIs could be transduced to express hDAF or hCD59. Flow cytometry analysis of islet single cells revealed that only a fraction of the cells expressed the transgene; immunohistochemical staining of transduced islets demonstrated that mainly cells located in the periphery of the islets were expressing the protein. Cells from nontransduced islets or islets expressing the control protein were sensitive to lysis in human sera (66+/-4.0% and 73+/-3.7% cytotoxicity, respectively). Single cells from islets transduced with hDAF and hCD59 were partially protected from lysis. Islet cells expressing hCD59 were slightly less sensitive to lysis (33+/-3.3%) than cells expressing hDAF (45+/-3.5%). CONCLUSIONS: These data show that intact pig islets can be transduced to express human regulators of complement activation on the surface and that pig islet cells expressing hDAF or hCD59 are less sensitive to complement-mediated lysis.


Assuntos
Fenômenos Fisiológicos Sanguíneos , Antígenos CD55/farmacologia , Antígenos CD59/farmacologia , Proteínas do Sistema Complemento/fisiologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Adenoviridae/genética , Animais , Antígenos CD55/genética , Antígenos CD55/metabolismo , Antígenos CD59/genética , Antígenos CD59/metabolismo , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Técnicas In Vitro , Suínos
17.
Histol Histopathol ; 17(4): 1085-94, 2002 10.
Artigo em Inglês | MEDLINE | ID: mdl-12371136

RESUMO

The limited and inadequate availability of organs from human donors has resulted in the utilisation of xenografts as an alternative tool. Nevertheless, hyperacute rejection (HAR) following xenograft determines the loss of the transplanted organ. The "primum movens" is the activation of the complement pathway mediated by the binding of natural xenogenic antibodies to the endothelium of the graft, followed by the lysis of the endothelial cells with subsequent oedema, thrombosis and necrosis of the transplanted organ. In this work we describe morphological and biomolecular observations of isolated human-decay accelerating factor (h-DAF, CD55) transgenic pig hearts, after perfusion for four hours with human blood. H-DAF is a membrane glycoprotein inhibiting the complement activation in humans. We describe considerably reduced damages in transgenic hearts, compared to controls. The cardiac function resulted preserved. Our data are in agreement with what was already shown by other groups using different experimental models. In conclusion, we encourage the use of new sources of transgenic animals, pointing out the importance of morphological analysis in evaluation of xenograft.


Assuntos
Antígenos CD55/farmacologia , Transplante de Coração/fisiologia , Transplante Heterólogo/fisiologia , Animais , Western Blotting , Circulação Coronária/fisiologia , Rejeição de Enxerto/fisiopatologia , Humanos , Imuno-Histoquímica , Microscopia Eletrônica , Tamanho do Órgão/fisiologia , Suínos
18.
Blood ; 98(9): 2771-7, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11675350

RESUMO

Mechanisms involving the in vitro effect of rituximab in cells from 55 patients with B-cell lymphoproliferative disorders were investigated. No cytotoxic effect was observed when cells were incubated with rituximab alone, but in the presence of human AB serum rituximab induced complement-dependent cell death (R-CDC). A cytotoxic effect was observed in cells from 9 of 33 patients with B-cell chronic lymphocytic leukemia, 16 of 16 patients with mantle-cell lymphoma, 4 of 4 patients with follicular lymphoma, and 2 of 2 patients with hairy-cell leukemia. R-CDC was observed in cells from patients expressing more than 50 x 10(3) CD20 molecules per cell, and directly correlated with the number of CD20 molecules per cell. Preincubation with anti-CD59 increased the cytotoxic effect of rituximab and sensitized cells from nonsensitive cases. Neither cleavage of poly-ADP ribose polymerase (PARP) nor activation of caspase-3 was observed in R-CDC. In addition, no cells with a hypodiploid DNA content were detected and R-CDC was not prevented by a broad-spectrum caspase inhibitor, suggesting a caspase-independent mechanism. Incubation with rituximab in the presence of AB serum induced a rapid and intense production of reactive oxygen species (ROS). R-CDC was blocked by the incubation of cells with N-acetyl-L-cysteine (NAC) or Tiron, 2 ROS scavengers, indicating that the cytotoxic effect was due to the generation of superoxide (O) radicals. In conclusion, the results of the present study suggest that CD20, CD59, and complement have a role in the in vitro cytotoxic effect of rituximab, which is mediated by a caspase-independent process that involves ROS generation.


Assuntos
Anticorpos Monoclonais/farmacologia , Linfócitos B/efeitos dos fármacos , Caspases/farmacologia , Morte Celular/efeitos dos fármacos , Proteínas do Sistema Complemento/farmacologia , Transtornos Linfoproliferativos/sangue , Espécies Reativas de Oxigênio/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais Murinos , Antígenos CD20/análise , Antineoplásicos/farmacologia , Linfócitos B/enzimologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Antígenos CD55/farmacologia , Antígenos CD59/farmacologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Transtornos Linfoproliferativos/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Rituximab
19.
Zhonghua Yi Xue Za Zhi ; 81(24): 1523-5, 2001 Dec 25.
Artigo em Chinês | MEDLINE | ID: mdl-16200781

RESUMO

OBJECTIVE: To observe the protective effect of human decay accelerating factor (DAF) expressed in mouse myeloma cells against the injury of human serum complement. METHODS: Mouse myeloma cells were transfected with human DAF cDNA and then put into normal human serum culture to observe if they were injured. RESULTS: The mouse cells containing human DAF cDNA and expressing human DAF survived for more than 48 hours and passed on from generation to generation in the culture medium containing human normal serum. However, the control mouse cells died in 1 - 4 hours. CONCLUSION: Human DAF protects mouse cells from injuring by human complement.


Assuntos
Antígenos CD55/farmacologia , Mieloma Múltiplo/metabolismo , Substâncias Protetoras/farmacologia , Transfecção , Animais , Antígenos CD55/genética , Proteínas do Sistema Complemento/imunologia , DNA Complementar/genética , Rejeição de Enxerto/imunologia , Humanos , Camundongos , Mieloma Múltiplo/patologia , Células Tumorais Cultivadas
20.
Transplantation ; 70(7): 989-98, 2000 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11045632

RESUMO

BACKGROUND: It is not known whether the pig liver is capable of functioning efficiently when transplanted into a primate, neither is there experience in transplanting a liver from a transgenic pigs expressing the human complement regulator human complement regulator decay accelerating factor (h-DAF) into a baboon. The objective of this study was to determine whether the porcine liver would support the metabolic functions of non-human primates and to establish the effect of hDAF expression in the prevention of hyperacute rejection of porcine livers transplanted into primates. METHODS: Five orthotopic liver xenotransplants from pig to baboon were carried out: three from unmodified pigs and two using livers from h-DAF transgenic pigs. FINDINGS: The three control animals transplanted with livers from unmodified pigs survived for less than 12 hr. Baboons transplanted with livers from h-DAF transgenic pigs survived for 4 and 8 days. Hyperacute rejection was not detected in the baboons transplanted with hDAF transgenic pig livers; however, it was demonstrated in the three transplants from unmodified pigs. Baboons transplanted with livers from h-DAF transgenic pigs were extubated at postoperative day 1 and were awake and able to eat and drink. In the recipients of hDAF transgenic pig livers the clotting parameters reached nearly normal levels at day 2 after transplantation and remained normal up to the end of the experiments. In these hDAF liver recipients, porcine fibrinogen was first detected in the baboon plasma 2 hr postreperfusion, and was present up to the end of the experiments. One animal was euthanized at day 8 after development of sepsis and coagulopathy, the other animal arrested at day 4, after an episode of vomiting and aspiration. The postmortem examination of the hDAF transgenic liver xenografts did not demonstrate rejection. INTERPRETATION: The livers from h-DAF transgenic pigs did not undergo hyperacute rejection after orthotopic xenotransplantation in baboons. When HAR is abrogated, the porcine liver maintains sufficient coagulation and protein levels in the baboon up to 8 days after OLT.


Assuntos
Antígenos CD55/farmacologia , Transplante de Fígado/imunologia , Transplante Heterólogo/imunologia , Transplante Heterólogo/fisiologia , Doença Aguda , Animais , Animais Geneticamente Modificados , Fatores de Coagulação Sanguínea/análise , Complemento C3/metabolismo , Complemento C4/metabolismo , Ensaio de Atividade Hemolítica de Complemento , Rejeição de Enxerto/prevenção & controle , Humanos , Fígado/patologia , Transplante de Fígado/mortalidade , Transplante de Fígado/patologia , Papio , Taxa de Sobrevida , Suínos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...