Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Cardiovasc Disord ; 24(1): 417, 2024 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-39127656

RESUMO

Mutations in fibrillin 1 (FBN1) is the main cause of Marfan syndrome (MFS) with thoracic aortic aneurysm (TAA) as the main complication. Activation of the complement system plays a key role in the formation of thoracic and abdominal aortic aneurysms. However, the role of the complement system in MFS-associated aortic aneurysms remains unclear. In this study, we observed increased levels of complement C3a and C5a in the plasma of MFS patients and mouse, and the increased deposition of the activated complement system product C3b/iC3b was also observed in the elastic fiber rupture zone of 3-month-old MFS mice. The expression of C3a receptor (C3aR) was increased in MFS aortas, and recombinant C3a promoted the expression of cytokines in macrophages. The administration of a C3aR antagonist (C3aRA) attenuated the development of thoracic aortic aneurysms in MFS mice. The increased inflammation response and matrix metalloproteinases activities were also attenuated by C3aRA treatment in MFS mice. Therefore, these findings indicate that the complement C3a/C3aR inhibition alleviates the formation of aortic aneurysm in Marfan syndrome mice.


Assuntos
Adipocinas , Aneurisma da Aorta Torácica , Complemento C3a , Fibrilina-1 , Síndrome de Marfan , Receptores de Complemento , Animais , Feminino , Humanos , Masculino , Camundongos , Adipocinas/genética , Aorta Torácica/patologia , Aneurisma da Aorta Torácica/prevenção & controle , Aneurisma da Aorta Torácica/etiologia , Aneurisma da Aorta Torácica/metabolismo , Aneurisma da Aorta Torácica/patologia , Complemento C3a/antagonistas & inibidores , Complemento C3a/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Fibrilina-1/genética , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Síndrome de Marfan/complicações , Síndrome de Marfan/genética , Síndrome de Marfan/tratamento farmacológico , Camundongos Endogâmicos C57BL , Receptores de Complemento/antagonistas & inibidores , Receptores Acoplados a Proteínas G , Transdução de Sinais
2.
Hum Immunol ; 82(4): 264-269, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33632561

RESUMO

The novel Coronavirus SARS-CoV-2 is the viral pathogen responsible for the ongoing global pandemic, COVID-19 (Coronavirus disease 2019). To date, the data recorded indicate 1.62 Mln deaths and 72.8 Mln people infected (WHO situation report Dec 2020). On December 27, the first anti-COVID-19 vaccinations started in Europe. There are no direct antivirals against SARS-CoV-2. Understanding the pathophysiological and inflammatory/immunological processes of SARS-CoV-2 infection is essential to identify new drug therapies. In the most severe COVID-19 cases, an unregulated immunological/inflammatory system results in organ injury that can be fatal to the host in some cases. Pharmacologic approaches to normalize the unregulated inflammatory/immunologic response is an important therapeutic solution. Evidence associates a non-regulation of the "complement system" as one of the causes of generalized inflammation causing multi-organ dysfunction. Serum levels of a complement cascade mediator, factor "C5a", have been found in high concentrations in the blood of COVID-19 patients with severe disease. In this article we discuss the correlation between complement system and COVID-19 infection and pharmacological solutions directed to regulate.


Assuntos
Tratamento Farmacológico da COVID-19 , Ativação do Complemento/efeitos dos fármacos , Complemento C3a/antagonistas & inibidores , Complemento C5a/antagonistas & inibidores , Inativadores do Complemento/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , COVID-19/patologia , COVID-19/fisiopatologia , Ativação do Complemento/imunologia , Complemento C3a/imunologia , Complemento C5a/imunologia , Humanos , SARS-CoV-2/imunologia
3.
Inflammation ; 43(6): 2128-2136, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32617860

RESUMO

Ulcerative colitis (UC) is a serious digestive system disease. Furthermore, the activation of C3a/C3aR axis promoted the expression of caspase-11. And higher levels of caspase-11 could induce the pyroptosis and inflammation of cells. However, some studies suggested that caspase-11 could promote and suppress the inflammation during the development of UC. In addition, whether C3a/C3aR axis could affect the development of UC by modulating the expression of caspase-11 is unclear. We established the UC rat model in this study. Next, the C3aR inhibitor was used to treat these rats at diverse stages of UC. Next, the HE staining was performed to detect the intestinal damage. ELISA was performed to reveal the expression of IL-6 and TNF-α in different stages of UC. Western blotting was used to detect the expression of caspase-11 and C3aR in different stages of UC. Stimulation of C3aR inhibitor in early stage of UC promoted the expression of C3aR and caspase-11 in later stage of UC. Treatment of C3aR inhibitor in later stage of UC inhibited the expression of C3aR and caspase-11 in later stage of UC. Furthermore, application of C3aR inhibitor in early stage of UC aggravates the damage of colon tissue and enhanced the secretion of TNF-α and IL-6 in the later stage of UC. Treatment of C3aR inhibitor in later stage of UC relieved the symptoms of UC and suppressed the production of TNF-α and IL-6 in the later stage of UC. Application of C3aR inhibitor in early stage of UC induced the poor prognosis of UC by upregulating the expression of caspase-11. Treatment of C3aR inhibitor in later stage of UC relieved the symptoms of UC and lead to the favorable prognosis of UC by inhibiting the expression of caspase-11.


Assuntos
Caspases/metabolismo , Colite Ulcerativa/diagnóstico , Complemento C3a/antagonistas & inibidores , Regulação da Expressão Gênica , Piroptose , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Animais , Colite Ulcerativa/metabolismo , Inflamação , Interleucina-6/metabolismo , Intestinos/patologia , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Proteínas de Ligação a Fosfato/biossíntese , Prognóstico , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio , Fator de Necrose Tumoral alfa/biossíntese , Regulação para Cima
4.
Biotechnol Bioeng ; 117(6): 1904-1908, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32068245

RESUMO

Complement component 3a (C3a) plays a crucial role in the immune response and host defense, but it is also involved in pro-inflammatory responses, causing many inflammatory disorders. Blockade of C3a has been regarded as a potent therapeutic strategy for inflammatory diseases. Here, we present the development of a human C3a (hC3a)-specific protein binder, which effectively inhibits pro-inflammatory responses. The protein binder, which is composed of leucine-rich repeat modules, was selected against hC3a through phage display, and its binding affinity was matured up to 600 pM by further expanding the binding interface in a module-by-module manner. The developed protein binder was shown to have more than 10-fold higher specificity to hC3a compared with human C5a, exhibiting a remarkable suppression effect on pro-inflammatory response in monocyte, by blocking the interaction between hC3a and its receptor. The hC3a-specific protein binder is likely to have a therapeutic potential for C3a-mediated inflammatory diseases.


Assuntos
Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Complemento C3a/antagonistas & inibidores , Inflamação/tratamento farmacológico , Leucina/análogos & derivados , Leucina/farmacologia , Células Cultivadas , Ativação do Complemento/efeitos dos fármacos , Complemento C3a/imunologia , Humanos , Inflamação/imunologia , Modelos Moleculares
5.
PLoS One ; 13(8): e0202594, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30133517

RESUMO

BACKGROUND AND OBJECTIVE: Complement activation as an early and important inflammatory process contributes to multiple organ dysfunction after trauma. We have recently shown that complement inhibition by decay-accelerating factor (DAF) protects brain from blast-overpressure (BOP)-induced damage. This study was conducted to determine the effect of DAF on acute lung injury induced by BOP exposure and to elucidate its possible mechanisms of action. METHODS: Anesthetized adult male Sprague-Daley rats were exposed to BOP (120 kPa) from a compressed air-driven shock tube. Rats were randomly assigned to three experimental groups: 1) Control (no BOP and no DAF treatment), 2) BOP (120 kPa BOP exposure), and 3) BOP followed by treatment with rhDAF (500µg/kg, i.v) at 30 minutes after blast. After a recovery period of 3, 24, or 48 hours, animals were euthanized followed by the collection of blood and tissues at each time point. Samples were subjected to the assessment of cytokines and histopathology as well as for the interaction of high-mobility-group box 1 (HMGB1) protein, NF-κB, receptor for advanced glycation end products (RAGE), C3a, and C3aR. RESULTS: BOP exposure significantly increased in the production of systemic pro- and anti-inflammatory cytokines, and obvious pathological changes as characterized by pulmonary edema, inflammation, endothelial damage and hemorrhage in the lungs. These alterations were ameliorated by early administration of rhDAF. The rhDAF treatment not only significantly reduced the expression levels of HMGB1, RAGE, NF-κB, C3a, and C3aR, but also reversed the interaction of C3a-C3aR and nuclear translocation of HMGB1 in the lungs. CONCLUSIONS: Our findings indicate that early administration of DAF efficiently inhibits systemic and local inflammation, and mitigates blast-induced lung injury. The underlying mechanism might be attributed to its potential modulation of C3a-C3aR-HMGB1-transcriptional factor axis. Therefore, complement and/or HMGB1 may be potential therapeutic targets in amelioration of acute lung injury after blast injury.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Traumatismos por Explosões/tratamento farmacológico , Antígenos CD55/administração & dosagem , Proteína HMGB1/genética , Inflamação/tratamento farmacológico , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/fisiopatologia , Animais , Traumatismos por Explosões/genética , Traumatismos por Explosões/patologia , Ativação do Complemento/efeitos dos fármacos , Complemento C3a/antagonistas & inibidores , Modelos Animais de Doenças , Humanos , Inflamação/genética , Inflamação/fisiopatologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/fisiopatologia , NF-kappa B/genética , Pressão/efeitos adversos , Ratos , Ratos Sprague-Dawley
6.
Sci Rep ; 8(1): 9679, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29946065

RESUMO

The mechanisms that connect complement system activation and basal deposit formation in early stages of age-related macular degeneration (AMD) are insufficiently understood, which complicates the design of efficient therapies to prevent disease progression. Using human fetal (hf) retinal pigment epithelial (RPE) cells, we have established an in vitro model to investigate the effect of complement C3a on RPE cells and its role in the formation of sub-RPE deposits. The results of these studies revealed that C3a produced after C3 activation is sufficient to induce the formation of sub-RPE deposits via complement-driven proteasome inhibition. C3a binds the C3a receptor (C3aR), stimulates deposition of collagens IV and VI underneath the RPE, and impairs the extracellular matrix (ECM) turnover by increased MMP-2 activity, all mediated by downregulation of the ubiquitin proteasome pathway (UPP). The formation of basal deposits can be prevented by the addition of a C3aR antagonist, which restores the UPP activity and ECM turnover. These findings indicate that the cell-based model can be used to test potential therapeutic agents in vitro. The data suggest that modulation of C3aR-mediated events could be a therapeutic approach for treatment of early AMD.


Assuntos
Complemento C3a/metabolismo , Degeneração Macular/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Anafilatoxinas/antagonistas & inibidores , Anafilatoxinas/metabolismo , Arginina/análogos & derivados , Arginina/farmacologia , Compostos Benzidrílicos/farmacologia , Células Cultivadas , Ativação do Complemento/efeitos dos fármacos , Complemento C3a/antagonistas & inibidores , Inibidores de Cisteína Proteinase/farmacologia , Ensaio de Imunoadsorção Enzimática , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Leupeptinas/farmacologia , Metaloproteinase 2 da Matriz/metabolismo , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Epitélio Pigmentado da Retina/efeitos dos fármacos
7.
Sci Rep ; 8(1): 5165, 2018 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-29581525

RESUMO

Deficiency of ribosomal proteins (RPs) leads to Diamond Blackfan Anemia (DBA) associated with anemia, congenital defects, and cancer. While p53 activation is responsible for many features of DBA, the role of immune system is less defined. The Innate immune system can be activated by endogenous nucleic acids from non-processed pre-rRNAs, DNA damage, and apoptosis that occurs in DBA. Recognition by toll like receptors (TLRs) and Mda5-like sensors induces interferons (IFNs) and inflammation. Dying cells can also activate complement system. Therefore we analyzed the status of these pathways in RP-deficient zebrafish and found upregulation of interferon, inflammatory cytokines and mediators, and complement. We also found upregulation of receptors signaling to IFNs including Mda5, Tlr3, and Tlr9. TGFb family member activin was also upregulated in RP-deficient zebrafish and in RPS19-deficient human cells, which include a lymphoid cell line from a DBA patient, and fetal liver cells and K562 cells transduced with RPS19 shRNA. Treatment of RP-deficient zebrafish with a TLR3 inhibitor decreased IFNs activation, acute phase response, and apoptosis and improved their hematopoiesis and morphology. Inhibitors of complement and activin also had beneficial effects. Our studies suggest that innate immune system contributes to the phenotype of RPS19-deficient zebrafish and human cells.


Assuntos
Anemia de Diamond-Blackfan/imunologia , Anemia de Diamond-Blackfan/metabolismo , Imunidade Inata/fisiologia , Peixe-Zebra/imunologia , Peixe-Zebra/metabolismo , Receptores de Ativinas/antagonistas & inibidores , Ativinas/metabolismo , Animais , Arginina/análogos & derivados , Arginina/farmacologia , Benzamidas/farmacologia , Compostos Benzidrílicos/farmacologia , Complemento C3a/antagonistas & inibidores , Complemento C3a/metabolismo , Dioxóis/farmacologia , Modelos Animais de Doenças , Humanos , Interferons/metabolismo , Células K562 , RNA Interferente Pequeno/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/antagonistas & inibidores , Proteínas Ribossômicas/metabolismo , Receptor 3 Toll-Like/antagonistas & inibidores , Receptor 3 Toll-Like/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/metabolismo
8.
Mol Immunol ; 93: 266-277, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28860090

RESUMO

Candida albicans the most frequently isolated clinical fungal pathogen can cause local as well as systemic and life-threatening infections particularly in immune-compromised individuals. A better and more detailed understanding how C. albicans evades human immune attack is therefore needed for identifying fungal immune-evasive proteins and develop new therapies. Here, we identified Pra1, the pH-regulated C. albicans antigen as a hierarchical complement inhibitor that targets C3, the central human complement component. Pra1 cleaved C3 at a unique site and further inhibited effector function of the activation fragments. The newly formed C3a-like peptide lacked the C-terminal arginine residue needed for C3a-receptor binding and activation. Moreover, Pra1 also blocked C3a-like antifungal activity as shown in survival assays, and the C3b-like molecule formed by Pra1 was degraded by the host protease Factor I. Pra1 also bound to C3a and C3b generated by human convertases and blocked their effector functions, like C3a antifungal activity shown by fungal survival, blocked C3a binding to human C3a receptor-expressing HEK cells, activation of Fura2-AM loaded cells, intracellular Ca2+ signaling, IL-8 release, C3b deposition, as well as opsonophagocytosis and killing by human neutrophils. Thus, upon infection C. albicans uses Pra1 to destroy C3 and to disrupt host complement attack. In conclusion, candida Pra1 represents the first fungal C3-cleaving protease identified and functions as a fungal master regulator of innate immunity and as a central fungal immune-escape protein.


Assuntos
Candida albicans/enzimologia , Complemento C3/antagonistas & inibidores , Proteínas Fúngicas/fisiologia , Sequência de Aminoácidos , Ligação Competitiva , Sinalização do Cálcio/efeitos dos fármacos , Candida albicans/efeitos dos fármacos , Candida albicans/imunologia , Linhagem Celular , Complemento C3/imunologia , Complemento C3/metabolismo , Complemento C3/farmacologia , Complemento C3a/antagonistas & inibidores , Complemento C3a/farmacologia , Complemento C3b/antagonistas & inibidores , Complemento C3b/farmacologia , Proteínas Fúngicas/antagonistas & inibidores , Proteínas Fúngicas/farmacologia , Células HEK293 , Humanos , Interleucina-8/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/fisiologia , Proteínas Opsonizantes/imunologia , Fragmentos de Peptídeos/metabolismo , Fagocitose/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Proteólise , Receptores de Complemento/antagonistas & inibidores , Receptores de Complemento/metabolismo , Virulência/imunologia
9.
Nat Commun ; 8(1): 2078, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29233958

RESUMO

Regeneration of skeletal muscle following injury is accompanied by transient inflammation. Here we show that complement is activated in skeletal muscle injury and plays a key role during regeneration. Genetic ablation of complement C3 or its inactivation with Cobra Venom Factor (CVF) result in impaired muscle regeneration following cardiotoxin-induced injury in mice. The effect of complement in muscle regeneration is mediated by the alternative pathway and C3a receptor (C3aR) signaling, as deletion of Cfb, a key alternative pathway component, or C3aR leads to impaired regeneration and reduced monocyte/macrophage infiltration. Monocytes from C3aR-deficient mice express a reduced level of adhesion molecules, cytokines and genes associated with antigen processing and presentation. Exogenous administration of recombinant CCL5 to C3aR-deficient mice rescues the defects in inflammatory cell recruitment and regeneration. These findings reveal an important role of complement C3a in skeletal muscle regeneration, and suggest that manipulating complement system may produce therapeutic benefit in muscle injury and regeneration.


Assuntos
Complemento C3a/fisiologia , Inflamação/imunologia , Monócitos/fisiologia , Músculo Esquelético/fisiologia , Receptores de Complemento/fisiologia , Regeneração/imunologia , Animais , Transplante de Medula Óssea , Cardiotoxinas/toxicidade , Movimento Celular/fisiologia , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimera/fisiologia , Complemento C3a/antagonistas & inibidores , Via Alternativa do Complemento/fisiologia , Modelos Animais de Doenças , Venenos Elapídicos/farmacologia , Humanos , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Receptores de Complemento/deficiência , Regeneração/efeitos dos fármacos , Transdução de Sinais/fisiologia , Cicatrização/fisiologia
10.
J Immunol ; 199(3): 1069-1085, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28687659

RESUMO

Inflammatory processes play a key role in pathophysiology of many neurologic diseases/trauma, but the effect of immune cells and factors on neurotransplantation strategies remains unclear. We hypothesized that cellular and humoral components of innate immunity alter fate and migration of human neural stem cells (hNSC). In these experiments, conditioned media collected from polymorphonuclear leukocytes (PMN) selectively increased hNSC astrogliogenesis and promoted cell migration in vitro. PMN were shown to generate C1q and C3a; exposure of hNSC to PMN-synthesized concentrations of these complement proteins promoted astrogliogenesis and cell migration. Furthermore, in vitro, Abs directed against C1q and C3a reversed the fate and migration effects observed. In a proof-of-concept in vivo experiment, blockade of C1q and C3a transiently altered hNSC migration and reversed astroglial fate after spinal cord injury. Collectively, these data suggest that modulation of the innate/humoral inflammatory microenvironment may impact the potential of cell-based therapies for recovery and repair following CNS pathology.


Assuntos
Astrócitos/fisiologia , Diferenciação Celular/fisiologia , Complemento C1q/biossíntese , Complemento C3a/biossíntese , Células-Tronco Neurais/fisiologia , Neutrófilos/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Movimento Celular , Células Cultivadas , Complemento C1q/antagonistas & inibidores , Complemento C1q/genética , Complemento C1q/imunologia , Complemento C3a/antagonistas & inibidores , Complemento C3a/genética , Complemento C3a/imunologia , Meios de Cultivo Condicionados , Humanos , Imunidade Inata , Camundongos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/imunologia , Neutrófilos/imunologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia
11.
Wound Repair Regen ; 25(1): 41-50, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28019718

RESUMO

Larvae of the blowfly Lucilia sericata facilitate wound healing by removing dead tissue and biofilms from non-healing and necrotic wounds. Another beneficial action of larvae and their excretions/secretions (ES) is down-regulation of excessive inflammation. As prolonged complement activation is key to excessive inflammation, the aim of this study was to elucidate the mechanisms underlying the anti-complement activities of ES. Results revealed that heat sensitive serine proteases in ES degrade multiple complement proteins in all steps of the three complement activation pathways. Importantly, C3a and C5a-major activators of inflammation-were also degraded by ES and pretreatment of these factors with ES completely blocked their ability to induce activation of human neutrophils. Pre-exposure of the neutrophils to ES did not affect their responsiveness to C3a/C5a and fMLP, indicating that the receptors for these activators on neutrophils were not affected by ES. Surprisingly, heat and serine protease inhibitor pretreatment did not affect the ability of ES to inhibit C5b-9 complex formation despite degrading complement proteins, indicating a second complement-inhibiting molecule in ES. Heated ES was as effective as intact ES in inhibiting C3 deposition upon activation of the alternative pathway, but was significantly less effective in wells with a classical or lectin pathway-specific coating. Unfortunately, the molecules affecting the complement system could not be identified due to an insufficient database for L. sericata. Together, larval ES inhibit complement activation by two different mechanisms and down-regulate the C3a/C5a-mediated neutrophil activation. This attenuates the inflammatory process, which may facilitate wound healing.


Assuntos
Complemento C3a/antagonistas & inibidores , Complemento C5a/antagonistas & inibidores , Dípteros/metabolismo , Inflamação/fisiopatologia , Larva/fisiologia , Cicatrização/fisiologia , Animais , Secreções Corporais/metabolismo , Ativação do Complemento/fisiologia , Complemento C3a/metabolismo , Complemento C5a/metabolismo , Desbridamento/métodos , Regulação para Baixo , Inflamação/metabolismo , Ativação de Neutrófilo/fisiologia
12.
J Thromb Haemost ; 13(6): 1090-102, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25851247

RESUMO

BACKGROUND AND OBJECTIVES: Carboxypeptidase B2 (CPB2) is a basic carboxypeptidase with fibrin and complement C3a and C5a as physiological substrates. We hypothesized that in polymicrobial sepsis, CPB2-deficient mice would have sustained C5a activity, leading to disease exacerbation. METHODS: Polymicrobial sepsis was induced by cecal ligation and puncture (CLP). RESULTS: Contrary to our hypothesis, Cpb2(-/-) mice had significantly improved survival, with reduced lung edema, less liver and kidney damage, and less disseminated intravascular coagulation. Hepatic pro-CPB2 was induced by CLP, leading to increased pro-CPB2 levels. Thrombomodulin present on mesothelium supported thrombin activation of pro-CPB2. Both wild-type and Cpb2(-/-) animals treated with a C5a receptor antagonist had improved survival, demonstrating that C5a was detrimental in this model. Treatment with a fibrinolysis inhibitor, tranexamic acid, caused a decrease in survival in both genotypes; however, the Cpb2(-/-) animals retained their survival advantage. Administration of a C3a receptor antagonist exacerbated the disease in both wild-type and Cpb2(-/-) mice and eliminated the survival advantage of Cpb2(-/-) mice. C5a receptor is expressed in both peritoneal macrophages and neutrophils; in contrast, C3a receptor expression is restricted to peritoneal macrophages, and C3a induced signaling in macrophages but not neutrophils. CONCLUSIONS: While C5a exacerbates the peritonitis, resulting in a deleterious generalized inflammatory state, C3a activation of peritoneal macrophages may limit the initial infection following CLP, thereby playing a diametrically opposing protective role in this polymicrobial sepsis model.


Assuntos
Carboxipeptidase B2/deficiência , Complemento C3a/metabolismo , Complemento C5a/metabolismo , Peritonite/enzimologia , Sepse/enzimologia , Animais , Antifibrinolíticos/farmacologia , Transtornos da Coagulação Sanguínea/enzimologia , Transtornos da Coagulação Sanguínea/genética , Transtornos da Coagulação Sanguínea/imunologia , Transtornos da Coagulação Sanguínea/microbiologia , Carboxipeptidase B2/genética , Ceco/microbiologia , Ceco/cirurgia , Células Cultivadas , Complemento C3a/antagonistas & inibidores , Complemento C3a/imunologia , Complemento C5a/antagonistas & inibidores , Complemento C5a/imunologia , Modelos Animais de Doenças , Ativação Enzimática , Fibrina/metabolismo , Mediadores da Inflamação/sangue , Leucopenia/enzimologia , Leucopenia/genética , Leucopenia/imunologia , Leucopenia/microbiologia , Ligadura , Fígado/enzimologia , Fígado/imunologia , Fígado/microbiologia , Ativação de Macrófagos , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritonite/genética , Peritonite/imunologia , Peritonite/microbiologia , Fatores de Proteção , Punções , Fatores de Risco , Sepse/genética , Sepse/imunologia , Sepse/microbiologia , Trombina/metabolismo , Trombomodulina/metabolismo , Fatores de Tempo
13.
J Pharmacol Exp Ther ; 351(3): 527-37, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25232192

RESUMO

Anti-factor D (AFD; FCFD4514S, lampalizumab) is a humanized IgG Fab fragment directed against factor D (fD), a rate-limiting serine protease in the alternative complement pathway (AP). Evaluation of AFD as a potential intravitreal (IVT) therapeutic for dry age-related macular degeneration patients with geographic atrophy (GA) is ongoing. However, it is unclear whether IVT administration of AFD can affect systemic AP activation and potentially compromise host-immune responses. We characterized the pharmacologic properties of AFD and assessed the effects of AFD administered IVT (2 or 20 mg) or intravenous (0.2, 2, or 20 mg) on systemic complement activity in cynomolgus monkeys. For the IVT groups, serum AP activity was reduced for the 20 mg dose group between 2 and 6 hours postinjection. For the intravenous groups, AFD inhibited systemic AP activity for periods of time ranging from 5 minutes (0.2 mg group) to 3 hours (20 mg group). Interestingly, the concentrations of total serum fD increased up to 10-fold relative to predose levels following administration of AFD. Furthermore, AFD was found to inhibit systemic AP activity only when the molar concentration of AFD exceeded that of fD. This occurred in cynomolgus monkeys at serum AFD levels ≥2 µg/ml, a concentration 8-fold greater than the maximum serum concentration observed following a single 10 mg IVT dose in a clinical investigation in patients with GA. Based on these findings, the low levels of serum AFD resulting from IVT administration of a clinically relevant dose are not expected to appreciably affect systemic AP activity.


Assuntos
Complemento C3a/antagonistas & inibidores , Fator D do Complemento/antagonistas & inibidores , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Degeneração Macular/tratamento farmacológico , Animais , Bovinos , Complemento C3a/imunologia , Fator D do Complemento/imunologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Injeções Intravítreas , Macaca fascicularis , Degeneração Macular/sangue , Degeneração Macular/imunologia , Masculino , Camundongos , Resultado do Tratamento
14.
J Pharmacol Exp Ther ; 351(2): 344-51, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25150279

RESUMO

Early-onset pre-eclampsia is characterized by decreased placental perfusion, new-onset hypertension, angiogenic imbalance, and endothelial dysfunction associated with excessive activation of the innate immune complement system. Although our previous studies demonstrated that inhibition of complement activation attenuates placental ischemia-induced hypertension using the rat reduced uterine perfusion pressure (RUPP) model, the important product(s) of complement activation has yet to be identified. We hypothesized that antagonism of receptors for complement activation products C3a and C5a would improve vascular function and attenuate RUPP hypertension. On gestational day (GD) 14, rats underwent sham surgery or vascular clip placement on ovarian arteries and abdominal aorta (RUPP). Rats were treated once daily with the C5a receptor antagonist (C5aRA), PMX51 (acetyl-F-[Orn-P-(D-Cha)-WR]), the C3a receptor antagonist (C3aRA), SB290157 (N(2)-[(2,2-diphenylethoxy)acetyl]-l-arginine), or vehicle from GD 14-18. Both the C3aRA and C5aRA attenuated placental ischemia-induced hypertension without affecting the decreased fetal weight or decreased concentration of free circulating vascular endothelial growth factor (VEGF) also present in this model. The C5aRA, but not the C3aRA, attenuated placental ischemia-induced increase in heart rate and impaired endothelial-dependent relaxation. The C3aRA abrogated the acute pressor response to C3a peptide injection, but it also unexpectedly attenuated the placental ischemia-induced increase in C3a, suggesting nonreceptor-mediated effects. Overall, these results indicate that both C3a and C5a are important products of complement activation that mediate the hypertension regardless of the reduction in free plasma VEGF. The mechanism by which C3a contributes to placental ischemia-induced hypertension appears to be distinct from that of C5a, and management of pregnancy-induced hypertension is likely to require a broad anti-inflammatory approach.


Assuntos
Sistema Cardiovascular/imunologia , Sistema Cardiovascular/fisiopatologia , Ativação do Complemento/imunologia , Complemento C3a/imunologia , Complemento C5a/imunologia , Hipertensão/imunologia , Hipertensão/fisiopatologia , Animais , Complemento C3a/antagonistas & inibidores , Complemento C5a/antagonistas & inibidores , Modelos Animais de Doenças , Endotélio Vascular/imunologia , Endotélio Vascular/fisiopatologia , Feminino , Frequência Cardíaca/imunologia , Isquemia/imunologia , Isquemia/fisiopatologia , Placenta/imunologia , Gravidez , Ratos , Receptores de Complemento/imunologia , Fator A de Crescimento do Endotélio Vascular/imunologia
15.
Med Hypotheses ; 80(1): 65-6, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23151526

RESUMO

Antiphospholipid syndrome is a pregnancy related, systemic autoimmune disorder in which antibodies directed against cell membrane phospholipids with multiple venous/arterial thromboses. Though the etiology of antiphospholipid syndrome has not been identified exactly, experimental evidences suggest the possible mechanisms involved in the pathogenicity of this syndrome. Antiphospholipid antibodies mediate deposition of complement proteins in placenta and over expression of tissue factor on the surface of neutrophils which are reported to be the prominent cause of prothrombotic phenotype. The activation complement components C3 and C5 by antiphospholipid antibodies would eventually activates blood coagulation pathway that leads to thrombosis. Inhibition of activated complement components C3a and C5a by anticomplement agents protects from antiphospholipid antibody mediated fetal loss. Since the interference of complement pathway may lead to deleterious effects, we hypothesis that local inhibition of complement proteins C3a and C5a at placenta would reduce the risk of antiphospholipid antibody mediated placental thrombosis and pregnancy complications.


Assuntos
Anticorpos Antifosfolipídeos/metabolismo , Síndrome Antifosfolipídica/tratamento farmacológico , Proteínas Inativadoras do Complemento/farmacologia , Morte Fetal/prevenção & controle , Regulação da Expressão Gênica/imunologia , Síndrome Antifosfolipídica/complicações , Complemento C3a/antagonistas & inibidores , Complemento C5a/antagonistas & inibidores , Feminino , Morte Fetal/etiologia , Humanos , Modelos Biológicos , Placenta/metabolismo , Gravidez
16.
PLoS One ; 7(6): e38664, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761695

RESUMO

BACKGROUND AND PURPOSE: The restoration of blood-flow following cerebral ischemia incites a series of deleterious cascades that exacerbate neuronal injury. Pharmacologic inhibition of the C3a-receptor ameliorates cerebral injury by attenuating post-ischemic inflammation. Recent reports also implicate C3a in the modulation of tissue repair, suggesting that complement may influence both injury and recovery at later post-ischemic time-points. METHODS: To evaluate the effect of C3a-receptor antagonism on post-ischemic neurogenesis and neurological outcome in the subacute period of stroke, transient focal cerebral ischemia was induced in adult male C57BL/6 mice treated with multiple regimens of a C3a receptor antagonist (C3aRA). RESULTS: Low-dose C3aRA administration during the acute phase of stroke promotes neuroblast proliferation in the subventricular zone at 7 days. Additionally, the C3a receptor is expressed on T-lymphocytes within the ischemic territory at 7 days, and this cellular infiltrate is abrogated by C3aRA administration. Finally, C3aRA treatment confers robust histologic and functional neuroprotection at this delayed time-point. CONCLUSIONS: Targeted complement inhibition through low-dose antagonism of the C3a receptor promotes post-ischemic neuroblast proliferation in the SVZ. Furthermore, C3aRA administration suppresses T-lymphocyte infiltration and improves delayed functional and histologic outcome following reperfused stroke. Post-ischemic complement activation may be pharmacologically manipulated to yield an effective therapy for stroke.


Assuntos
Anti-Inflamatórios/farmacologia , Complemento C3a/antagonistas & inibidores , Inflamação/prevenção & controle , Neurogênese/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Receptores de Complemento/antagonistas & inibidores , Acidente Vascular Cerebral/prevenção & controle , Animais , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Complemento C3a/metabolismo , Modelos Animais de Doenças , Inflamação/etiologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia , Receptores de Complemento/metabolismo , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/mortalidade , Traumatismo por Reperfusão/prevenção & controle , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/mortalidade , Taxa de Sobrevida , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia
17.
J Immunol ; 188(11): 5694-705, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22539791

RESUMO

Allergen-specific IgE plays an essential role in the pathogenesis of allergic asthma. Although there has been increasing evidence suggesting the involvement of IL-17 in the disease, the relationship between IL-17 and IgE-mediated asthmatic responses has not yet been defined. In this study, we attempted to elucidate the contribution of IL-17 to an IgE-mediated late-phase asthmatic response and airway hyperresponsiveness (AHR). BALB/c mice passively sensitized with an OVA-specific IgE mAb were challenged with OVA intratracheally four times. The fourth challenge caused a late-phase increase in airway resistance associated with elevated levels of IL-17(+)CD4(+) cells in the lungs. Multiple treatments with a C3a receptor antagonist or anti-C3a mAb during the challenges inhibited the increase in IL-17(+)CD4(+) cells. Meanwhile, a single treatment with the antagonist or the mAb at the fourth challenge suppressed the late-phase increase in airway resistance, AHR, and infiltration by neutrophils in bronchoalveolar lavage fluid. Because IL-17 production in the lungs was significantly repressed by both treatments, the effect of an anti-IL-17 mAb was examined. The late-phase increase in airway resistance, AHR, and infiltration by neutrophils in bronchoalveolar lavage fluid was inhibited. Furthermore, an anti-Gr-1 mAb had a similar effect. Collectively, we found that IgE mediated the increase of IL-17(+)CD4(+) cells in the lungs caused by repeated Ag challenges via C3a. The mechanisms leading to the IgE-mediated late-phase asthmatic response and AHR are closely associated with neutrophilic inflammation through the production of IL-17 induced by C3a.


Assuntos
Asma/imunologia , Asma/patologia , Hiper-Reatividade Brônquica/imunologia , Complemento C3a/fisiologia , Imunoglobulina E/fisiologia , Interleucina-17/fisiologia , Neutrófilos/imunologia , Neutrófilos/patologia , Animais , Anticorpos Monoclonais/fisiologia , Asma/metabolismo , Hiper-Reatividade Brônquica/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Complemento C3a/antagonistas & inibidores , Complemento C3a/imunologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/fisiologia , Interleucina-17/biossíntese , Interleucina-17/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Complemento/antagonistas & inibidores , Fatores de Tempo
18.
J Immunol ; 186(5): 3120-9, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21270401

RESUMO

The complement system plays an important role in eliminating invading pathogens. Activation of complement results in C3b deposition (opsonization), phagocytosis, anaphylatoxin (C3a, C5a) release, and consequently cell lysis. Moraxella catarrhalis is a human respiratory pathogen commonly found in children with otitis media and in adults with chronic obstructive pulmonary disease. The species has evolved multiple complement evasion strategies, which among others involves the ubiquitous surface protein (Usp) family consisting of UspA1, A2, and A2 hybrid. In the present study, we found that the ability of M. catarrhalis to bind C3 correlated with UspA expression and that C3 binding contributed to serum resistance in a large number of clinical isolates. Recombinantly expressed UspA1 and A2 inhibit both the alternative and classical pathways, C3b deposition, and C3a generation when bound to the C3 molecule. We also revealed that the M. catarrhalis UspA-binding domain on C3b was located to C3d and that the major bacterial C3d-binding domains were within UspA1(299-452) and UspA2(165-318). The interaction with C3 was not species specific since UspA-expressing M. catarrhalis also bound mouse C3 that resulted in inhibition of the alternative pathway of mouse complement. Taken together, the binding of C3 to UspAs is an efficient strategy of Moraxella to block the activation of complement and to inhibit C3a-mediated inflammation.


Assuntos
Antígenos de Bactérias/fisiologia , Antígenos de Superfície/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Complemento C3d/metabolismo , Evasão da Resposta Imune/imunologia , Moraxella catarrhalis/imunologia , Adulto , Animais , Antígenos de Bactérias/metabolismo , Antígenos de Superfície/metabolismo , Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas da Membrana Bacteriana Externa/metabolismo , Criança , Ativação do Complemento/imunologia , Complemento C3a/antagonistas & inibidores , Complemento C3a/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Via Alternativa do Complemento/imunologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Camundongos , Moraxella catarrhalis/isolamento & purificação , Moraxella catarrhalis/patogenicidade , Infecções por Moraxellaceae/imunologia , Infecções por Moraxellaceae/microbiologia , Infecções por Moraxellaceae/patologia , Ligação Proteica/imunologia , Coelhos , Ovinos
19.
J Pharmacol Sci ; 112(1): 56-63, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20051658

RESUMO

It was investigated whether the C3a-receptor antagonist (C3aRA) SB 290157 was involved in the suppression of anti-OVA pAb-induced arthritis because it is well known that anaphylatoxin C3a plays a crucial role in the development of an effective inflammatory response during complement activation. Anti-OVA pAb-induced arthritis was induced in DBA/1J mice by administration of anti-OVA pAb 0.5 h prior to intra-articular (i.a.) injection of OVA (0 h). Two peaks of joint swelling were observed at 0.5 and 3 h. The role of C3aRA in arthritis was investigated by injection of SB 290157 at concentrations of 10 and 30 mg/kg at 0 and 2 h. The antagonist was able to reduce joint swelling only at 3 h, and about 50% inhibition of joint swelling was observed with the concentration of 30 mg/kg. The C3 level was significantly decreased at 3 h compared with naïve mice showing complement consumption. Furthermore, the C3 activation was observed and increased corresponding to the graded concentration of anti-OVA pAb. The results also revealed that the C3aRA was able to reduce the expression of IL-1beta in synovial tissue. Taken together, the results suggested that C3aRA may be effective in the inhibition of arthritis.


Assuntos
Anticorpos/toxicidade , Arginina/análogos & derivados , Artrite Experimental/prevenção & controle , Compostos Benzidrílicos/farmacologia , Compostos Benzidrílicos/uso terapêutico , Complemento C3a/antagonistas & inibidores , Óvulo/imunologia , Animais , Arginina/farmacologia , Arginina/uso terapêutico , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Complemento C3a/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos DBA , Ratos , Ratos Endogâmicos Lew , Receptores de Complemento/antagonistas & inibidores , Receptores de Complemento/imunologia , Receptores de Complemento/metabolismo
20.
Circ Res ; 105(11): 1094-101, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19815824

RESUMO

RATIONALE: Donor brain death (BD) is an unavoidable occurrence in heart transplantation and results in profound physiological derangements that render the heart more susceptible to ischemia/reperfusion injury in the recipient and likely has negative long-term consequences to allograft survival. OBJECTIVE: We developed a novel mouse model of BD and investigated the role of complement in BD-induced myocardial inflammation and injury. METHODS AND RESULTS: BD was induced by inflation of a balloon catheter in the cranial cavity. BD in wild-type mice resulted in a significant increase in serum concentrations of the complement activation product complement component (C)3a, and immunohistochemical analysis of heart sections demonstrated C3 deposition on the vascular endothelium and surrounding myocytes. Following induction of BD in complement (C3)-deficient mice, cardiac troponin levels, and histological evidence of injury were significantly reduced compared to wild-type mice. C3 deficiency was also associated with reduced myocardial leukocyte infiltration and reduced or absent expression of P-selectin, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, tumor necrosis factor-alpha, and interleukin-1beta. CONCLUSIONS: These data indicate an important role for complement in BD-induced inflammation and injury and suggest that a complement inhibitory strategy applied to the donor (in addition to the recipient) may provide graft protection.


Assuntos
Morte Encefálica/imunologia , Complemento C3a/imunologia , Transplante de Coração/imunologia , Traumatismo por Reperfusão Miocárdica/imunologia , Doadores de Tecidos , Animais , Morte Encefálica/patologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Ativação do Complemento/imunologia , Complemento C3a/antagonistas & inibidores , Modelos Animais de Doenças , Expressão Gênica/imunologia , Precondicionamento Isquêmico/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocardite/imunologia , Miocardite/patologia , Miocardite/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA