Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Curr Opin Nephrol Hypertens ; 26(2): 123-128, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27977428

RESUMO

PURPOSE OF REVIEW: The complement system represents one of the more primitive forms of innate immunity. It has increasingly been found to contribute to pathologies in the native and transplanted kidney. We provide a concise review of the physiology of the complement cascade, and discuss current and upcoming complement-based therapies. RECENT FINDINGS: Current agents in clinical use either bind to complement components directly or prevent complement from binding to antibodies affixed to the endothelial surface. These include C1 esterase inhibitors, anti-C5 mAbs, anti-CD20 mAbs, and proteasome inhibitors. Treatment continues to show efficacy in the atypical hemolytic uremic syndrome and antibody-mediated rejection. Promising agents not currently available include CCX168, TP10, AMY-101, factor D inhibitors, coversin, and compstatin. Several new trials are targeting complement inhibition to treat antineutrophilic cystoplasmic antibody (ANCA)-associated vasculitis, C3 glomerulopathy, thrombotic microangiopathy, and IgA nephropathy. New agents for the treatment of the atypical hemolytic uremic syndrome are also in development. SUMMARY: Complement-based therapies are being considered for targeted therapy in the atypical hemolytic uremic syndrome and antibody-mediated rejection, C3 glomerulopathy, and ANCA-associated vasculitis. A few agents are currently in use as orphan drugs. A number of other drugs are in clinical trials and, overall, are showing promising preliminary results.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Proteínas do Sistema Complemento/metabolismo , Compostos de Anilina/uso terapêutico , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/tratamento farmacológico , Antígenos CD20/imunologia , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Ativação do Complemento/efeitos dos fármacos , Complemento C5/antagonistas & inibidores , Complemento C5/imunologia , Fator D do Complemento/antagonistas & inibidores , Glomerulonefrite por IGA/tratamento farmacológico , Humanos , Ácidos Nipecóticos/uso terapêutico , Peptídeos Cíclicos/uso terapêutico , Inibidores de Proteassoma/uso terapêutico , Receptores de Complemento/uso terapêutico , Microangiopatias Trombóticas/tratamento farmacológico
2.
Circulation ; 131(13): 1171-80, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25825397

RESUMO

BACKGROUND: Natural IgM antibodies represent a class of innate pattern recognition receptors that recognize danger-associated molecular patterns expressed on stressed or dying cells. They play important roles in tissue homeostasis by disposing of prenecrotic cells and suppressing inflammation. However, ischemic insult leads to a pathogenic level of IgM binding and complement activation, resulting in inflammation and injury. We investigate the role of self-reactive IgM in the unique setting of transplantation where the donor organ undergoes both cold and warm ischemia and global ischemic insult. METHODS AND RESULTS: By transplanting hearts from wild-type donor mice into antibody-deficient mice reconstituted with specific self-reactive IgM monoclonal antibodies, we identified neoepitopes expressed after transplantation and demonstrated a key role for IgM recognition of these epitopes in graft injury. With this information, we developed and characterized a therapeutic strategy that exploited the postischemia recognition system of natural antibodies. On the basis of neoepitope identification, we constructed an anti-annexin IV single-chain antibody (scFv) and an scFv linked to Crry, an inhibitor of C3 activation (scFv-Crry). In an allograft transplantation model in which recipients contain a full natural antibody repertoire, both constructs blocked graft IgM binding and complement activation and significantly reduced graft inflammation and injury. Furthermore, scFv-Crry specifically targeted to the transplanted heart and, unlike complement deficiency, did not affect immunity to infection, an important consideration for immunosuppressed transplant recipients. CONCLUSIONS: We identified pathophysiologically important epitopes expressed within the heart after transplantation and described a novel translatable strategy for targeted complement inhibition that has several advantages over currently available approaches.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Transplante de Coração/efeitos adversos , Imunoglobulina M/uso terapêutico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Reperfusão Miocárdica/efeitos adversos , Receptores de Complemento/uso terapêutico , Tolerância a Antígenos Próprios/imunologia , Anticorpos de Cadeia Única/uso terapêutico , Animais , Anexina A4/imunologia , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Ativação do Complemento , Epitopos/imunologia , Genes Sintéticos , Proteínas de Homeodomínio/genética , Imunoglobulina M/deficiência , Imunoglobulina M/genética , Imunoglobulina M/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/imunologia , Miocárdio/imunologia , Especificidade de Órgãos , Fosfolipídeos/imunologia , Receptores de Complemento/genética , Receptores de Complemento 3b , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Tolerância ao Transplante
3.
Int J Biol Macromol ; 260(Pt 2): 129357, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38216011

RESUMO

Osteoporosis is a prevalent systemic skeletal disorder, particularly affecting postmenopausal women, primarily due to excessive production and activation of osteoclasts. However, the current anti-osteoporotic drugs utilized in clinical practice may lead to certain side effects. Therefore, it is necessary to further unravel the potential mechanisms regulating the osteoclast differentiation and to identify novel targets for osteoporosis treatment. This study revealed the most significant decline in VSIG4 expression among the VSIG family members. VSIG4 overexpression significantly inhibited RANKL-induced osteoclastogenesis and bone resorption function. Mechanistically, both western blot and immunofluorescence assay results demonstrated that VSIG4 overexpression attenuated the expression of osteoclast marker genes and dampened the activation of MAPK and NF-κB signaling pathways. Furthermore, VSIG4 overexpression could inhibit the generation of reactive oxygen species (ROS) and stimulate the expression of Nrf2 along with its downstream antioxidant enzymes via interaction with Keap1. Notably, a potent Nrf2 inhibitor, ML385, could reverse the inhibitory effect of VSIG4 on osteoclast differentiation. In line with these findings, VSIG4 overexpression also mitigated bone loss induced by OVX and attenuated the activation of osteoclasts in vivo. In conclusion, our results suggest that VSIG4 holds promise as a novel target for addressing postmenopausal osteoporosis. This is achieved by suppressing osteoclast formation via enhancing Nrf2-dependent antioxidant response against reactive oxygen species production.


Assuntos
Osteogênese , Osteoporose , Feminino , Humanos , Espécies Reativas de Oxigênio/metabolismo , Antioxidantes/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Osteoclastos , NF-kappa B/metabolismo , Osteoporose/tratamento farmacológico , Osteoporose/metabolismo , Diferenciação Celular , Receptores de Complemento/metabolismo , Receptores de Complemento/uso terapêutico
5.
J Surg Res ; 167(2): e131-6, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19691988

RESUMO

Complement activation has been demonstrated to contribute significantly to the expression of IR-induced tissue damage. Each of the three complement pathways, classic, alternative, and lectin, has been implicated in the instigation of tissue pathology. In this study, we used a selective inhibitor of the alternative pathway, that is, a soluble form of complement receptor of the immunoglobulin superfamily (CRIg-Fc) to determine whether it can prevent IR tissue injury. We demonstrate that treatment of C57B1/6 mice prior to mesenteric IR prevents local (intestinal) and remote (lung) injury by limiting deposition of complement and entry of polymorphonuclear cells to the sites of injury. Our results show that CRIg-Fc represents a candidate to limit IR injury as it occurs in various clinical conditions.


Assuntos
Via Alternativa do Complemento/fisiologia , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Intestino Delgado/irrigação sanguínea , Intestino Delgado/fisiopatologia , Receptores de Complemento/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/fisiopatologia , Lesão Pulmonar Aguda/prevenção & controle , Animais , Linfócitos B/patologia , Movimento Celular/efeitos dos fármacos , Via Alternativa do Complemento/efeitos dos fármacos , Fragmentos Fc das Imunoglobulinas/farmacologia , Intestino Delgado/patologia , Mesentério/irrigação sanguínea , Mesentério/patologia , Mesentério/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Infiltração de Neutrófilos/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Linfócitos T/patologia
6.
Virol J ; 7: 30, 2010 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-20144216

RESUMO

BACKGROUND: Influenza is a respiratory disease that seriously threatens human health. In fact, influenza virus itself does not make critical contribution to mortality induced by influenza, but "cytokine storm" produced by the excessive immune response triggered by the virus can result in inflammatory reaction of lung tissues and fatal lung tissue injury, and thus increase influenza mortality. Therefore, besides antiviral drugs, immunosuppression drugs should also be included in infection treatment. PRESENTATION OF THE HYPOTHESIS: Complement is the center of inflammatory reaction. If complement system is over activated, the body will have strong inflammatory reaction or tissue injury, resulting in pathological process. Many studies have proved that, inflammatory injury of lung tissues caused by influenza virus is closely related to complement activation. Therefore, inhibiting complement activation can significantly reduce inflammatory injury in lung tissues. As complement is both a physiological defense and pathological damage medium, systematic inhibition may result in side effects including infection. Therefore, we design targeting complement inhibitors for complement activation sites, i.e. with CR2 as targeting vector, complement inhibitors like CD59 and Crry are targeted to inflammatory sites to specially inhibit the complement activation in local injury, thus local inflammatory reaction is inhibited. TESTING THE HYPOTHESIS: CR2-CD59 and CR2-Crry targeting complement inhibitors are fusion-expressed, and their biological activity is examined via in vivo and in vitro tests. CR2 targeting complement inhibitors are used to treat mouse influenza viral pneumonia model, with PBS treatment group as the control. The survival and lung tissue injury of the mice is observed and the effect of CR2 targeting complement inhibitors on pneumonia induced by influenza virus is evaluated. IMPLICATIONS OF THE HYPOTHESIS: CR2 targeting complement inhibitors are expected to be ideal drugs for viral pneumonia.


Assuntos
Imunossupressores/uso terapêutico , Influenza Humana/imunologia , Influenza Humana/patologia , Pulmão/patologia , Orthomyxoviridae/imunologia , Receptores de Complemento 3d/antagonistas & inibidores , Animais , Produtos Biológicos/genética , Produtos Biológicos/uso terapêutico , Antígenos CD59/genética , Antígenos CD59/uso terapêutico , Humanos , Influenza Humana/tratamento farmacológico , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/patologia , Receptores de Complemento/genética , Receptores de Complemento/uso terapêutico , Receptores de Complemento 3b , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Análise de Sobrevida
7.
Spinal Cord ; 48(2): 105-11, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19736560

RESUMO

STUDY DESIGN: Spinal cord injured rat model, treated with soluble complement receptor type 1 (sCR1). SETTING: Experimental Animal Department of China Medical University, Shenyang, China. OBJECTIVES: Soluble CR1 is a powerful inhibitor of complement activation. In this study, we investigate the effectiveness of sCR1 on spinal cord injury (SCI) in rats. METHODS: Spinal cord injury was induced in Sprague-Dawley rats. Three experimental groups were examined; the sCR1 group was administered sCR1 at 1 h after the SCI, whereas the control group was administered saline at 1 h after SCI and the sham group underwent a sham operation without SCI or administration. The expressions of C9 and CD59 in the injured spinal cords were evaluated by immunohistochemistry, and numbers of positive cells counted. Furthermore, myeloperoxidase (MPO) activity and motor function were evaluated in each group. RESULTS: At all postoperative time points, the numbers of C9- and CD59-positive cells in the sCR1 group were reduced compared with the control group and MPO activity was significantly decreased compared with both other groups. Moreover, the Basso, Beattie and Bresnahan score for the sCR1 group was significantly improved as compared with that of the control group after 7 days postoperatively. CONCLUSION: Soluble CR1 decreases inflammation reactions by inhibiting activation of the complement system and improves motor function after acute SCI.


Assuntos
Ativação do Complemento/fisiologia , Neurônios Motores/fisiologia , Receptores de Complemento/uso terapêutico , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/fisiopatologia , Animais , Antígenos CD59/fisiologia , Complemento C9/fisiologia , Modelos Animais , Neurônios Motores/patologia , Peroxidase/fisiologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/patologia , Resultado do Tratamento
8.
Neurobiol Dis ; 34(3): 450-6, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19281846

RESUMO

Decreased activity of cAMP responsive element-binding protein (CREB) is thought to contribute to the death of striatal medium spiny neurons in Huntington's disease (HD). Therefore, therapies that increase levels of activated CREB, may be effective in fighting neurodegeneration in HD. In this study, we sought to determine whether the phosphodiesterase type 10 (PDE10A) inhibitor TP10 exerts a neuroprotective effect in an excitotoxic model of HD. Rats were surgically administered with quinolinic acid into striatum and subsequently treated with TP10 daily for two or eight weeks. After 2 weeks of TP10 treatment, striatal lesion size was 52% smaller and the surviving cell number was several times higher than in the vehicle-treated group. These beneficial effects of TP10 were maintained through 8 weeks. TP10 treatment also increased significantly the levels of activated CREB in the striatal spiny neurons, which is hypothesized to be a contributing mechanism for the neuroprotective effect. Our findings suggest PDE10A inhibition as a novel neuroprotective approach to the treatment of HD and confirm the importance of phosphodiesterase inhibition in fighting the disease.


Assuntos
Doença de Huntington/induzido quimicamente , Doença de Huntington/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Diester Fosfórico Hidrolases/metabolismo , Receptores de Complemento/uso terapêutico , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Doença de Huntington/patologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fosforilação , Ácido Quinolínico , Ratos , Ratos Wistar
9.
Science ; 285(5427): 595-9, 1999 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-10417391

RESUMO

Glycoprotein adhesion receptors such as selectins contribute to tissue injury in stroke. Ischemic neurons strongly expressed C1q, which may target them for complement-mediated attack or C1qRp-mediated clearance. A hybrid molecule was used to simultaneously inhibit both complement activation and selectin-mediated adhesion. The extracellular domain of soluble complement receptor-1 (sCR1) was sialyl Lewis x glycosylated (sCR1sLex) to inhibit complement activation and endothelial-platelet-leukocyte interactions. sCR1 and sCR1sLex colocalized to ischemic cerebral microvessels and C1q-expressing neurons, inhibited neutrophil and platelet accumulation, and reduced cerebral infarct volumes. Additional benefit was conferred by sialyl Lewis x glycosylation of the unmodified parent sCR1 molecule.


Assuntos
Transtornos Cerebrovasculares/tratamento farmacológico , Ataque Isquêmico Transitório/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Oligossacarídeos/uso terapêutico , Receptores de Complemento/uso terapêutico , Animais , Plaquetas/fisiologia , Adesão Celular , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/imunologia , Córtex Cerebral/metabolismo , Infarto Cerebral/tratamento farmacológico , Circulação Cerebrovascular , Transtornos Cerebrovasculares/imunologia , Transtornos Cerebrovasculares/fisiopatologia , Ativação do Complemento , Complemento C1q/metabolismo , Glicosilação , Humanos , Ataque Isquêmico Transitório/imunologia , Ataque Isquêmico Transitório/fisiopatologia , Leucócitos/fisiologia , Camundongos , Neurônios/imunologia , Neurônios/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/efeitos adversos , Fármacos Neuroprotetores/metabolismo , Neutrófilos/fisiologia , Oligossacarídeos/administração & dosagem , Oligossacarídeos/efeitos adversos , Oligossacarídeos/metabolismo , Adesividade Plaquetária , Receptores de Complemento/administração & dosagem , Receptores de Complemento/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/metabolismo , Selectinas/metabolismo , Antígeno Sialil Lewis X , Fatores de Tempo
10.
Circulation ; 116(11 Suppl): I83-8, 2007 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-17846331

RESUMO

BACKGROUND: TP10, a potent inhibitor of complement activation during cardiopulmonary bypass (CPB) has been shown to significantly reduce the incidence of death and myocardial infarction (MI) in high-risk male patients undergoing cardiac surgery. However, the effect of TP10 in females was undefined because of the limited number of females studied. To examine the possibility of a gender effect, this phase 2 multi-center trial was undertaken to determine whether TP10 would also limit ischemic damage in a larger sample size of high-risk females undergoing cardiac surgery on cardiopulmonary bypass (CPB). METHODS AND RESULTS: This prospective, double-blind, placebo-controlled, multi-center trial involved 297 high-risk (urgent surgery, CABG + Valve, reoperations, ejection fraction <30%) female patients randomized to receive a 5 mg/kg dose of TP10 (n=150) or placebo (n=147) as a 30-minute intravenous infusion before surgery. The primary end point was the incidence of death or MI at 28 days after surgery. Complement activation was assessed by levels of CH50 and SC5b-9 during and after CPB. TP10 was well tolerated and there were no differences in the safety profiles of the 2 groups. Although TP10 effectively suppressed complement activation (at 2 hours after CPB CH50 (mean+SD % change from baseline) 50+/-17% placebo versus 4+/-14% TP10; P=0.0001; SC5b-9 (ng/mL) 917+/-1067 placebo versus 204+/-79 TP10; P=0.0001), there was no difference in the primary end point between the groups (17% placebo versus 21% TP10; P=0.2550). CONCLUSIONS: The benefits of TP10 appear to be gender-related. and mechanisms other than complement activation may be responsible for myocardial injury in high-risk female patients during cardiac surgery on CPB.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Proteínas do Sistema Complemento/metabolismo , Receptores de Complemento/uso terapêutico , Caracteres Sexuais , Idoso , Procedimentos Cirúrgicos Cardíacos/métodos , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
11.
Nat Clin Pract Nephrol ; 3(5): 277-86, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17457361

RESUMO

The fact that the complement system is activated during immune-complex glomerular disease has been known for nearly 50 years. Detection of complement deposition in the glomerulus using immunochemistry has become an important element of the histological analysis of renal biopsies, and is key to the diagnosis of many types of glomerulonephritis. In recent years it has become evident that complement activation is involved in the pathogenesis of other types of renal disease; complement activation is implicated in transplant injury, atypical hemolytic uremic syndrome and progressive tubulointerstitial fibrosis. Emergence of this evidence has provided insight into how these diseases develop, and has yielded useful diagnostic tools and potential targets for therapeutic intervention. Clinicians have, by using plasma-based therapies, unknowingly treated abnormalities of the complement system in renal patients for many years. Advances in antibody and protein technologies have led to the development of complement inhibitors that have been used in phase III clinical studies. More-specific agents and applications are likely to be developed over the coming years and are discussed in this Review.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Nefropatias/imunologia , Animais , Proteínas do Sistema Complemento/genética , Humanos , Nefropatias/genética , Transplante de Rim , Receptores de Complemento/uso terapêutico
12.
Zhonghua Xin Xue Guan Bing Za Zhi ; 35(11): 1037-40, 2007 Nov.
Artigo em Zh | MEDLINE | ID: mdl-18269827

RESUMO

OBJECTIVE: To investigate the protective effects of recombinant SCR15-18 domain of human soluble complement receptor type 1 (sCR1-SCR-15-18) in rats underwent myocardial ischemia and reperfusion (I/R). METHODS: Sprague-Dawley rats were randomly divided into three groups (n = 12 each group): sham (SO); 30 min ischemia/3h reperfusion (I/R) and I/R plus sCR1-SCR15-18 (15 mg/kg before I/R, sCR1). Serum LDH, CK and cardiac myeloperoxidase (MPO) activity were measured. Infarct size, myocardial histopathological changes and myocardial C3c were also compared among groups. RESULTS: Infarct size [(16.1 +/- 3.3)% vs. (22.9 +/- 3.0)%, infarct zone/left ventricular mass, P < 0. 05] and CK [(2532.5 +/- 597.1) U/L vs. (3400.9 +/- 534.9) U/L, P < 0. 05] and LDH [(5436.2 +/- 611.3) U/L vs. (6572.0 +/- 476.3) U/L, P < 0. 05] as well as MPO activity in infarct zone [(0.81 +/- 0.14) U/g vs. (1.12 +/- 0.13) U/g, P < 0.05] were significantly decreased post sCR1 compared to I/R group. sCR1 also significantly attenuated histological myocardial injury and reduced the deposition of C3c in infarct zone. CONCLUSION: sCR1-SCR15-18 protein exerts cardioprotective effects in this rat I/R model.


Assuntos
Traumatismo por Reperfusão Miocárdica/prevenção & controle , Receptores de Complemento/uso terapêutico , Animais , Creatina Quinase/metabolismo , Modelos Animais de Doenças , Humanos , L-Lactato Desidrogenase/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Peroxidase/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Complemento 3b/metabolismo , Proteínas Recombinantes/uso terapêutico
13.
Biochim Biophys Acta ; 1639(3): 169-76, 2003 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-14636948

RESUMO

Changes in brain water and cerebral volume can lead to brain edema that may be one of the underlying causes of death in many neurological diseases. Cerebral water content is regulated by aquaporin 4 (AQ4) present in astrocytic end feet and around blood vessels. In systemic lupus erythematosus (SLE), magnetic resonance imaging (MRI) studies of the brain have demonstrated lesions with the prominent appearance of edema. Activation of complement may play a significant role in the pathogenesis of lupus cerebritis by causing inflammation that can lead to edema. In this study, the well-established MRL/lpr lupus mouse model was used to evaluate the role of complement in lupus cerebritis. IgG and C1q colocalized in perivascular deposits indicating that the blood-brain barrier was compromised. Both RNA and protein expressions of AQ4 were significantly increased in brains of MRL/lpr mice. Chronic administration of the soluble complement inhibitor, Crry-Ig, reduced inflammation as measured by decreased accumulation of IgG. In contrast to control MRL/lpr mice, AQ4 expression in complement inhibited MRL/lpr mice was not changed relative to untreated congenic controls. These results illustrate that complement activation in brains of lupus mice leads to enhanced AQ4 expression and inflammation. It is conceivable that increased AQ4 expression results in cerebral edema and hence complement inhibition may provide a new therapeutic option in inflammatory cerebral disorders such as lupus cerebritis.


Assuntos
Aquaporinas/genética , Proteínas Inativadoras do Complemento/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Imunoglobulina G , Inflamação/prevenção & controle , Vasculite Associada ao Lúpus do Sistema Nervoso Central/tratamento farmacológico , Receptores de Complemento/uso terapêutico , Animais , Aquaporina 4 , Aquaporinas/antagonistas & inibidores , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Sequência de Bases , Barreira Hematoencefálica , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Primers do DNA , Modelos Animais de Doenças , Vasculite Associada ao Lúpus do Sistema Nervoso Central/imunologia , Vasculite Associada ao Lúpus do Sistema Nervoso Central/patologia , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos MRL lpr , Análise de Sequência com Séries de Oligonucleotídeos , Sondas de Oligonucleotídeos , Receptores de Complemento 3b , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Circulation ; 110(11 Suppl 1): II274-9, 2004 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-15364875

RESUMO

BACKGROUND: This study was undertaken to determine whether soluble human complement receptor type 1 (TP10), a potent inhibitor of complement activation, would reduce morbidity and mortality in high-risk patients undergoing cardiac surgery on cardiopulmonary bypass (CPB). METHODS: This was a randomized multicenter, prospective, placebo-controlled, double-blind study in which 564 high-risk patients undergoing cardiac surgery on CPB received an intravenous bolus of TP10 (1, 3, 5, 10 mg/kg) or placebo immediately before CPB. The primary endpoint was the composite events of death, myocardial infarction (MI), prolonged (> or =24 hours) intra-aortic balloon pump support (IABP), and prolonged intubation. RESULTS: TP10 significantly inhibited complement activity after 10 to 15 minutes of CPB and this inhibition persisted for 3 days postoperatively. However, there was no difference in the primary endpoint between the 2 groups (33.7% placebo versus 31.4% TP10; P=0.31). The primary composite endpoint was, however, reduced in all male TP10 patients by 30% (P=0.025). TP10 reduced the incidence of death or MI in males by 36% (P=0.026), the incidence of death or MI in CABG males by 43% (P=0.043) and the need for prolonged IABP support in male CABG and valve patients by 100% (P=0.019). There was, however, no improvement seen in female TP10 patients. There were no significant differences in adverse events between the groups. CONCLUSIONS: TP10 effectively inhibits complement activation during CPB; however, this was not associated with an improvement in the primary endpoint of the study. Nevertheless, TP10 did significantly decrease the incidence of mortality and MI in male patients.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Ponte Cardiopulmonar/efeitos adversos , Ativação do Complemento/efeitos dos fármacos , Isquemia Miocárdica/prevenção & controle , Receptores de Complemento/uso terapêutico , Idoso , Procedimentos Cirúrgicos Cardíacos/mortalidade , Complemento C3a/biossíntese , Complexo de Ataque à Membrana do Sistema Complemento/biossíntese , Método Duplo-Cego , Feminino , Humanos , Infecções/epidemiologia , Injeções Intravenosas , Balão Intra-Aórtico/estatística & dados numéricos , Tempo de Internação/estatística & dados numéricos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/epidemiologia , Infarto do Miocárdio/etiologia , Isquemia Miocárdica/etiologia , Isquemia Miocárdica/imunologia , Estudos Prospectivos , Receptores de Complemento/administração & dosagem , Fatores Sexuais , Solubilidade , Resultado do Tratamento
15.
Circulation ; 101(5): 541-6, 2000 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-10662752

RESUMO

BACKGROUND: Host defense system activation occurs with cardiopulmonary bypass (CPB) and is thought to contribute to the pathophysiological consequences of CPB. Complement inhibition effects on the post-CPB syndrome were tested with soluble complement receptor-1 (sCR1). METHODS AND RESULTS: Twenty neonatal pigs (weight 1.8 to 2.8 kg) were randomized to control and sCR1-treated groups. LV pressure and volume, left atrial pressure, pulmonary artery pressure and flow, and respiratory system compliance and resistance were measured. Preload recruitable stroke work, isovolumic diastolic relaxation time constant (tau), and pulmonary vascular resistance were determined. Pre-CPB measures were not statistically significantly different between the 2 groups. After CPB, preload recruitable stroke work was significantly higher in the sCR1 group (n=5, 46.8+/-3.2x10(3) vs n=6, 34.3+/-3.7x10(3) erg/cm(3), P=0.042); tau was significantly lower in the sCR1 group (26.4+/-1.5, 42.4+/-6. 6 ms, P=0.003); pulmonary vascular resistance was significantly lower in the sCR1 group (5860+/-1360 vs 12 170+/-1200 dyn. s/cm(5), P=0.009); arterial PO(2) in 100% FIO(2) was significantly higher in the sCR1 group (406+/-63 vs 148+/-33 mm Hg, P=0.01); lung compliance and airway resistance did not differ significantly. The post-CPB Hill coefficient of atrial myocardium was higher in the sCR1 group (2.88+/-0.29 vs 1.88+/-0.16, P=0.023). CONCLUSIONS: sCR1 meaningfully moderates the post-CPB syndrome, supporting the hypothesis that complement activation contributes to this syndrome.


Assuntos
Ponte Cardiopulmonar/efeitos adversos , Cardiopatias/prevenção & controle , Pneumopatias/prevenção & controle , Complicações Pós-Operatórias/prevenção & controle , Substâncias Protetoras/uso terapêutico , Receptores de Complemento/uso terapêutico , Citoesqueleto de Actina/química , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Cálcio/metabolismo , Cardiopatias/etiologia , Cardiopatias/fisiopatologia , Testes de Função Cardíaca , Pneumopatias/etiologia , Pneumopatias/fisiopatologia , Oxigênio/metabolismo , Conformação Proteica , Testes de Função Respiratória , Suínos , Fatores de Tempo
16.
Trends Mol Med ; 8(9): 430-6, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12223314

RESUMO

A great deal of information has accumulated implicating the complement system in several human disease processes. Although some of this information is circumstantial, protein inhibitors of the complement system have been developed and applied successfully to experimental disease models in animals. Two inhibitors, soluble complement receptor 1 (sCR1) and anti-C5 monoclonal antibody, are now being investigated in a variety of clinical conditions such as systemic lupus erythematosus and rheumatoid arthritis (RA), diseases for which current therapy has changed little and remains unsatisfactory. Preliminary successes in Phase II clinical trials of RA have provided optimism that complement inhibition might prove useful in these diseases and become part of standard medical therapy.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/patologia , Proteínas Inativadoras do Complemento/uso terapêutico , Proteínas do Sistema Complemento/metabolismo , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Animais , Ensaios Clínicos como Assunto , Humanos , Lúpus Eritematoso Sistêmico/patologia , Receptores de Complemento/uso terapêutico
17.
J Thorac Cardiovasc Surg ; 129(2): 423-8, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15678055

RESUMO

OBJECTIVE: Complement activation has been shown to play a significant role in ischemia-reperfusion injury after lung transplantation. TP-10 (soluble complement receptor 1 inhibitor) inhibits the activation of complement by inactivating C3a and C5a convertases. This was a clinical trial of TP-10 to reduce ischemia-reperfusion injury in lung transplantation. METHODS: In a randomized, double-blinded, multicenter, placebo-controlled trial, 59 patients from four lung transplant programs received TP-10 (10 mg/kg, n = 28) or placebo (n = 31) before reperfusion. This dose achieved 90% complement inhibition for 24 hours, and activity had returned toward normal by 72 hours. RESULTS: At 24 hours, 14 of 28 patients in the TP-10 group (50%) were extubated, whereas only 6 of 31 patients in the placebo group (19%) were (P = .01). The total times on the ventilator and in the intensive care unit both tended to be shorter in the TP-10 group, but these differences did not achieve statistical significance. Among patients requiring cardiopulmonary bypass (n = 5 in placebo group and n = 7 in TP-10 group), the mean duration of mechanical ventilation was reduced by 11 days in the TP-10 group (10.6 +/- 5.0 days vs 21.5 +/- 5.9 days in placebo group, P = .2). Operative deaths, incidences of infection and rejection, and length of hospital stay were not significantly different between the two groups. CONCLUSIONS: Short-term complement inhibition with TP-10 led to early extubation in a significantly higher proportion of lung transplant recipients. The effect of TP-10 was greater among patients undergoing cardiopulmonary bypass, with a large reduction in ventilator days. Complement inhibition thus significantly decreases the duration of mechanical ventilation and could be useful in improving the outcome of lung transplant recipients.


Assuntos
Transplante de Pulmão , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/prevenção & controle , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/prevenção & controle , Adolescente , Adulto , Idoso , Biomarcadores/sangue , Ponte Cardiopulmonar , Proteínas Inativadoras do Complemento/antagonistas & inibidores , Proteínas Inativadoras do Complemento/uso terapêutico , Proteínas do Sistema Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/metabolismo , Método Duplo-Cego , Feminino , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/mortalidade , Rejeição de Enxerto/prevenção & controle , Humanos , Tempo de Internação , Pneumopatias/mortalidade , Pneumopatias/cirurgia , Masculino , Pessoa de Meia-Idade , América do Norte , Consumo de Oxigênio/efeitos dos fármacos , Complicações Pós-Operatórias/mortalidade , Receptores de Complemento/antagonistas & inibidores , Receptores de Complemento/uso terapêutico , Traumatismo por Reperfusão/mortalidade , Respiração Artificial , Infecção da Ferida Cirúrgica/etiologia , Infecção da Ferida Cirúrgica/mortalidade , Infecção da Ferida Cirúrgica/prevenção & controle , Análise de Sobrevida , Resultado do Tratamento
18.
Chin J Traumatol ; 8(1): 49-53, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15676091

RESUMO

OBJECTIVE: To determine the effects of recombinant soluble complement receptor type I (sCR1) on the immune inflammatory reaction in acute spinal cord injury tissue of rats and its protective effects. METHODS: SD rat models of acute spinal cord injury were prepared by modified Allen's method. The motor function of the rat lower extremities in sCR1 group and normal saline (NS) group was evaluated by the tiltboard experiment at 12 h, 1 d, 3 d, 7 d, and 14 d. The neutrophil infiltration and C3c positive expression were observed. The myeloperoxidase activity was assessed in the injury tissue at 12 h, 1 d, 3 d, 7 d, and 14 d after injury in the two groups. RESULTS: The motor function of rat in sCR1 group at 3 d, 7 d, and 14 d was obviously better than that in NS group (P<0.01, P<0.01, P<0.01). C3c positive expression in sCR1 group at each time point after injury was obviously less than that in NS group (P<0.01). The myeloperoxidase activity in sCR1 group at each time point after injury was obviously less than that in NS group (P<0.01). CONCLUSIONS: Recombinant soluble complement receptor type I (sCR1) can lessen the immune inflammatory reaction in acute spinal cord injury tissue and relieve secondary spinal cord injury by inhibiting the activation of the complement system.


Assuntos
Receptores de Complemento/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Modelos Animais de Doenças , Imuno-Histoquímica , Inflamação , Peroxidase/biossíntese , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/enzimologia , Traumatismos da Medula Espinal/patologia
19.
Mol Immunol ; 38(2-3): 249-55, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11532286

RESUMO

Undesirable complement activation contributes to the pathology of many human diseases by damaging tissue and promoting inflammation. Because complement-mediated damage is caused by the deposition of complement components on the cell surface, several strategies have been devised to target complement regulator proteins to cell membranes. These strategies have resulted in engineered proteins that have improved potency in vitro and enhanced therapeutic benefit in animal models of disease. One membrane-targeted complement inhibitor has now entered clinical development and this class of second-generation agents may provide effective therapies for the treatment of a variety of disease states.


Assuntos
Membrana Celular/imunologia , Proteínas Inativadoras do Complemento/uso terapêutico , Antígenos CD59 , Sistemas de Liberação de Medicamentos , Receptores de Complemento/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA