Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 210(10): 1543-1551, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36988282

RESUMO

Complement factor D (FD) is a rate-limiting enzyme of the alternative pathway (AP). Recent studies have suggested that it is synthesized as an inactive precursor and that its conversion to enzymatically active FD is catalyzed by mannan-binding lectin-associated serine protease 3 (MASP3). However, whether MASP3 is essential for AP complement activity remains uncertain. It has been shown that Masp1/3 gene knockout did not prevent AP complement overactivation in a factor H-knockout mouse, and a human patient lacking MASP3 still retained AP complement activity. In this study, we have assessed AP complement activity in a Masp3-knockout mouse generated by CRISPR/Cas9 editing of the Masp1/3 gene. We confirmed specific Masp3 gene inactivation by showing intact MASP1 protein expression and absence of mature FD in the mutant mice. Using several assays, including LPS- and zymosan-induced C3b deposition and rabbit RBC lysis tests, we detected plasma concentration-dependent AP complement activity in Masp3 gene-inactivated mice. Thus, although not measurable in 5% plasma, significant AP complement activity was detected in 20-50% plasma of Masp3 gene-inactivated mice. Furthermore, whereas FD gene deletion provided more than 90% protection of CD55/Crry-deficient RBCs from AP complement-mediated extravascular hemolysis, Masp3 gene deletion only provided 30% protection in the same study. We also found pro-FD to possess intrinsic catalytic activity, albeit at a much lower level than mature FD. Our data suggest that MASP3 deficiency reduces but does not abrogate AP complement activity and that this is explained by intrinsic pro-FD activity, which can be physiologically relevant in vivo.


Assuntos
Lectina de Ligação a Manose , Serina Proteases Associadas a Proteína de Ligação a Manose , Animais , Humanos , Camundongos , Coelhos , Fator D do Complemento/metabolismo , Via Alternativa do Complemento/fisiologia , Lectina de Ligação a Manose da Via do Complemento , Proteínas do Sistema Complemento , Camundongos Knockout , Serina Proteases Associadas a Proteína de Ligação a Manose/genética
2.
Sci Rep ; 12(1): 13658, 2022 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-35953544

RESUMO

A complement effect on homeostasis during infection is determined by both cytotoxic (activate complement component 5 (C5a) terminal cytotoxic complex (TCC)), and cytoprotective elements (complement factor H (FH), as well as apolipoprotein E (ApoE)). Here, we investigated the gap in knowledge in their blood milieu during SARS-CoV-2 infection with respect to the viral burden, level of tissue necrosis, and immunological response. 101 patients hospitalized with a PCR-confirmed diagnosis of COVID-19 had blood collected at H1 (48 h), H2 (3-4 Days), H3 (5-7 days), H4 (more than 7 days up to 93 days). Pre-existing conditions, treatment, the incidence of cerebrovascular events (CVA), a history of deep venous thrombosis (DVT) and pulmonary embolism (PE), and mortality was collected using electronic medical records. Plasma C5a, TCC, FH, and ApoE were considered as a complement milieu. Tissue necrosis (HMGB1, RAGE), non-specific inflammatory responses (IL-6, C-reactive protein), overall viral burden (SARS-CoV-2 spike protein), and specific immune responses (IgG, IgA, IgM directed αS- & N-proteins) were assessed simultaneously. C5a remained elevated across all time points, with the peak at 5-7 days. Studied elements of complement coalesced around three clusters: #0 (↑↑↑C5a, ↑↑TCC, ↓↓ApoE), #1 ↑C5a, ↑TCC, ↑↑↑FH); #2 (↑C5a, ↑TCC, ↑FH, ↑↑↑ApoE). The decline in FH and ApoE was a predictor of death, while TCC and C5a correlated with patient length of stay, APACHE, and CRP. Increased levels of C5a (Δ = 122.64; p = 0.0294; data not shown) and diminished levels of FH (Δ = 836,969; p = 0.0285; data not shown) co-existed with CVA incidence. C5a correlated storngly with blood RAGE and HMGB1, but not with viral load and immunological responsiveness. Remdesivir positively affected FH preservation, while convalescent plasma treatment elevated C5a levels. Three clusters of complement activation demonstrated a various milieu of ApoE & FH vs C5a & TCC in COVID-19 patients. Complement activation is linked to increased necrosis markers but not to viral burden or immune system response.


Assuntos
COVID-19 , Proteína HMGB1 , Apolipoproteínas E/genética , Proteína C-Reativa , COVID-19/terapia , Ativação do Complemento , Complemento C5a , Fator H do Complemento , Humanos , Imunização Passiva , Necrose , Fatores de Proteção , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Soroterapia para COVID-19
3.
Front Cardiovasc Med ; 9: 983617, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36606279

RESUMO

Background: Heart surgery results in complement activation with the potential for collateral end-organ damage, especially if the protective elements (complement factor H, Apolipoprotein J) are inadequate. Here, we have investigated if peri-operative stress results in an imbalance between complement activation and its protective mechanisms up to 3 months after heart surgery. Methods: 101 patients scheduled for non-emergent cardiac surgery donated blood before the procedure (tbaseline), and 24 h (t24h ), 7 days (t7d ) and 3 months (t3m ) after. Complement activation was measured as a serum level of soluble activated component 5 (sC5a) and soluble terminal complement complex (sTCC). Simultaneously, protective complement factor H (CfH), and apolipoprotein J (ApoJ) were measured. Inflammatory responses were quantified using C-reactive protein (CRP) and interleukin-6 (IL-6). Details regarding anesthesia, intensive care unit (ICU) stay, pre-existing conditions, the incidence of postoperative complications, and mortality were collected from medical records. Results: C5a declined at t24h to rebound at t7d and t3m . sTCC was significantly depressed at t24h and returned to baseline at later time points. In contrast, CfH and ApoJ were depressed at t3m . Milieu of complement factors aligned along two longitudinal patterns:cluster#1 (C5a/sTTC continuously increasing and CfH/ApoJ preserved at tbaseline) and cluster#2 (transient sC5a/sTTC increase and progressive decline of CfH). Most patients belonged to cluster #1 at t24h (68%), t7d (74%) and t3m (72%). sTCC correlated with APACHE1h (r 2 =-0.25; p < 0.031) and APACHE24h (r 2 = 0.27; p < 0.049). IL-6 correlated with C5a (r 2 =-0.28; p < 0.042) and sTTC (r 2 =-0.28; p < 0.015). Peri-operative administration of acetaminophen and aspirin altered the complement elements. Prolonged hospital stay correlated with elevated C5a [t (78) = 2.03; p = 0.048] and sTTC serum levels [U (73) = 2.07; p = 0.037]. Patients with stroke had a decreased serum level of C5a at t7d and t3m. Conclusion: There is a significant decrease in complement protective factors 3 months after cardiac surgery, while C5a seems to be slightly elevated, suggesting that cardiac surgery affects complement milieu long into recovery.

4.
Kidney Int ; 96(1): 67-79, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30910380

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a form of thrombotic microangiopathy (TMA) caused by dysregulated complement activation. Clinically, aHUS is effectively treated by an anti-C5 monoclonal antibody (mAb) but whether the disease is mediated by the C5a receptor (C5aR) or C5b-9 pathway, or both, is unknown. Here we address this in a factor H mutant mouse (FHR/R) which developed complement-mediated TMA as well as macrovascular thrombosis caused by an aHUS-related factor H point mutation (mouse W1206R, corresponding to human W1183R). C5 deficiency and anti-C5 mAb treatment blocked all disease manifestations in FHR/R mice. C5aR1 gene deficiency prevented macrovascular thrombosis in various organs but did not improve survival or reduce renal TMA. Conversely, C6 or C9 deficiency significantly improved survival and markedly diminished renal TMA but did not prevent macrovascular thrombosis. Interestingly, as they aged both FHR/R C6-/- and FHR/R C9-/- mice developed glomerular disease reminiscent of C3 glomerulonephritis. Thus, C5aR and C5b-9 pathways drove different aspects of disease in FHR/R mice with the C5aR pathway being responsible for macrovascular thrombosis and chronic inflammatory injury while the C5b-9 pathway caused renal TMA. Our data provide new understanding of the pathogenesis of complement-mediated TMA and macrovascular thrombosis in FHR/R mice and suggest that C5 blockade is more effective for the treatment of aHUS than selectively targeting the C5aR or C5b-9 pathway alone.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/imunologia , Fator H do Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Glomérulos Renais/patologia , Receptor da Anafilatoxina C5a/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/patologia , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/genética , Ativação do Complemento/imunologia , Complemento C6/genética , Complemento C6/imunologia , Complemento C6/metabolismo , Fator H do Complemento/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/ultraestrutura , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Mutação Puntual , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/metabolismo
5.
Invest Ophthalmol Vis Sci ; 59(8): 3405-3415, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-30025090

RESUMO

Purpose: Dense deposit disease (DDD) is caused by dysregulation of the alternative pathway of the complement cascade and characterized by electron-dense deposits in the kidney glomerular basement membrane (GBM) and drusen in Bruch's membrane (BrM). Complement factor H (fH) and factor properdin (fP) regulate complement activation; fH inhibits alternative pathway (AP) activation, whereas fP promotes it. We report pathologic changes in eyes of an fH and fP double-mutant mouse, which we previously showed have dense deposits in the GBM and early mortality from nephropathy. Methods: fHm/m, fP-/-, and fHm/m/fP-/- mice were generated on a C57BL/6-129J background. Fundus imaging at 8 weeks of age was followed by analysis via light and electron microscopy. Retinal function was assessed by electroretinography (ERG). Complement levels and localization were tested by immunohistochemistry and ELISA. Retinas of fHm/m/fP-/- mice treated with intraperitoneal injections of an anti-C5 antibody were compared to those of age- and genotype-matched mice injected with an isotype control antibody. Results: fHm/m/fP-/- mice suffered early-onset retinal hypopigmented spots detected using in vivo retinal photography, and histologic examination showed basal laminar deposits (BLamD), degeneration of the photoreceptors, and RPE vacuolization. ERG showed diminished retinal function. The anti-C5 antibody was retina-protective. Conclusions: This unique mouse represents a new model of complement-mediated rapid-onset DDD, and could be useful in exploring the pathologic changes associated with BLamD in age-related macular degeneration.


Assuntos
Lâmina Basilar da Corioide/patologia , Fator H do Complemento/metabolismo , Modelos Animais de Doenças , Glomerulonefrite Membranoproliferativa/patologia , Properdina/metabolismo , Doenças Retinianas/patologia , Epitélio Pigmentado da Retina/patologia , Animais , Eletrorretinografia , Ensaio de Imunoadsorção Enzimática , Glomerulonefrite Membranoproliferativa/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Doenças Retinianas/metabolismo
6.
J Am Soc Nephrol ; 29(8): 2053-2059, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29895552

RESUMO

Background C3 glomerulopathy (C3G) is a life-threatening kidney disease caused by dysregulation of the alternative pathway of complement (AP) activation. No approved specific therapy is available for C3G, although an anti-C5 mAb has been used off-label in some patients with C3G, with mixed results. Thus, there is an unmet medical need to develop other inhibitors of complement for C3G.Methods We used a murine model of lethal C3G to test the potential efficacy of an Fc fusion protein of complement receptor of the Ig superfamily (CRIg-Fc) in the treatment of C3G. CRIg-Fc binds C3b and inhibits C3 and C5 convertases of the AP. Mice with mutations in the factor H and properdin genes (FHm/mP-/-) develop early-onset C3G, with AP consumption, high proteinuria, and lethal crescentic GN.Results Treatment of FHm/mP-/- mice with CRIg-Fc, but not a control IgG, inhibited AP activation and diminished the consumption of plasma C3, factor B, and C5. CRIg-Fc-treated FHm/mP-/- mice also had significantly improved survival and reduced proteinuria, hematuria, BUN, glomerular C3 fragment, C9 and fibrin deposition, and GN pathology scores.Conclusions Therapeutics developed on the basis of the mechanism of action of soluble CRIg may be effective for the treatment of C3G and should be explored clinically.


Assuntos
Complemento C3/antagonistas & inibidores , Complemento C3/genética , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/prevenção & controle , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Receptores de Complemento/genética , Análise de Variância , Animais , Biópsia por Agulha , Western Blotting , Ativação do Complemento , Fator B do Complemento/imunologia , Fator B do Complemento/metabolismo , Modelos Animais de Doenças , Glomerulonefrite por IGA/patologia , Imuno-Histoquímica , Testes de Função Renal , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Receptores de Complemento/metabolismo , Taxa de Sobrevida
7.
J Immunol ; 201(3): 1021-1029, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29898960

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is a serious blood disorder characterized by dysregulated complement activation on blood cells. Eculizumab, the current standard therapy and a humanized anti-C5 mAb, relieves anemia and thrombosis symptoms of PNH patients by preventing complement-dependent intravascular hemolysis (IVH). However, up to 20% of PNH patients on long-term eculizumab treatment still suffer from significant anemia and are transfusion dependent because of extravascular hemolysis (EVH) of C3-opsonized PNH erythrocytes. In this study, we show that function-blocking anti-properdin (P) mAbs dose-dependently inhibited autologous, complement-mediated hemolysis induced by factor H dysfunction. Furthermore, anti-human P (hP) mAbs potently and dose-dependently inhibited acidified serum-induced hemolysis of PNH erythrocytes (Ham test). In contrast to erythrocytes rescued by anti-C5 mAb, nonlysed PNH erythrocytes rescued by anti-P mAb incurred no activated C3 fragment deposition on their surface. These results suggested that anti-P mAbs may prevent EVH as well as IVH of PNH erythrocytes. To test the in vivo efficacy of anti-hP mAbs in preventing EVH, we generated a P humanized mouse by transgenic expression of hP in P knockout mice (hP-Tg/P-/-). In a murine EVH model, complement-susceptible erythrocytes were completely eliminated within 3 d in control mAb-treated hP-Tg/P-/- mice, whereas such cells were protected and persisted in hP-Tg/P-/- mice treated with an anti-hP mAb. Collectively, these data suggest that anti-P mAbs can inhibit both IVH and EVH mediated by complement and may offer improved efficacy over eculizumab, the current standard therapy for PNH.


Assuntos
Anticorpos Monoclonais/imunologia , Ativação do Complemento/imunologia , Hemólise/imunologia , Properdina/imunologia , Animais , Anticorpos Bloqueadores/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Eritrócitos/imunologia , Feminino , Hemoglobinúria Paroxística/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
J Am Soc Nephrol ; 29(7): 1928-1937, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29858280

RESUMO

Background Properdin (P) is a positive regulator of the alternative pathway of complement activation. Although P inhibition is expected and has been shown to ameliorate the alternative pathway of complement-mediated tissue injury in several disease models, it unexpectedly exacerbated renal injury in a murine model of C3 glomerulopathy. The role of P in atypical hemolytic uremic syndrome (aHUS) is uncertain.Methods We blocked P function by genetic deletion or mAb-mediated inhibition in mice carrying a factor H (FH) point mutation, W1206R (FHR/R), that causes aHUS and systemic thrombophilia with high mortality.Results P deficiency completely rescued FHR/R mice from premature death and prevented thrombocytopenia, hemolytic anemia, and renal disease. It also eliminated macrovessel thrombi that were prevalent in FHR/R mice. All mice that received a function-blocking anti-P mAb for 8 weeks survived the experimental period and appeared grossly healthy. Platelet counts and hemoglobin levels were significantly improved in FHR/R mice after 4 weeks of anti-P mAb treatment. One half of the FHR/R mice treated with an isotype control mAb but none of the anti-P mAb-treated mice developed stroke-related neurologic disease. Anti-P mAb-treated FHR/R mice showed largely normal renal histology, and residual liver thrombi were detected in only three of 15 treated mice.Conclusions These results contrast with the detrimental effect of P inhibition observed in a murine model of C3 glomerulopathy and suggest that P contributes critically to aHUS pathogenesis. Inhibition of P in aHUS may be of therapeutic benefit.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Complemento C3/metabolismo , Complemento C9/metabolismo , Properdina/genética , Trombofilia/genética , Animais , Anticorpos Monoclonais/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Síndrome Hemolítico-Urêmica Atípica/prevenção & controle , Fator H do Complemento/genética , Via Alternativa do Complemento , Feminino , Fibrina/metabolismo , Hemoglobinas/metabolismo , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Knockout , Contagem de Plaquetas , Properdina/deficiência , Properdina/imunologia , Trombofilia/prevenção & controle , Trombose/prevenção & controle
9.
Kidney Int ; 91(6): 1386-1397, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28139294

RESUMO

C3 glomerulopathy is a potentially life-threatening disease of the kidney caused by dysregulated alternative pathway complement activation. The specific complement mediator(s) responsible for kidney injury in C3 glomerulopathy are yet to be defined and no specific therapy is currently available. We previously developed a mouse model of lethal C3 glomerulopathy with factor H and properdin gene double mutations. Therefore, we used this model to examine the role of C5 and C5a receptor (C5aR) in the pathogenesis of the disease. Disease severity in these factor H/properdin double-mutant mice was found to be correlated with plasma C5 levels, and prophylactic anti-C5 mAb therapy was effective in preventing lethal C3 glomerulopathy. When given to these double-mutant mice that had already developed active disease with severe proteinuria, anti-C5 mAb treatment also prevented death in half of the mice. Deficiency of C5aR significantly reduced disease severity, suggesting that C5aR-mediated inflammation contributed to C3 glomerulopathy. Thus, C5 and C5aR have a critical role in C3 glomerulopathy. Hence, early intervention targeting these pathways may be an effective therapeutic strategy for patients with C3 glomerulopathy.


Assuntos
Anticorpos Monoclonais/farmacologia , Complemento C3/metabolismo , Complemento C5/antagonistas & inibidores , Inativadores do Complemento/farmacologia , Via Alternativa do Complemento/efeitos dos fármacos , Glomerulonefrite/prevenção & controle , Rim/efeitos dos fármacos , Insuficiência Renal/prevenção & controle , Animais , Complemento C3/imunologia , Complemento C5/imunologia , Complemento C5/metabolismo , Fator H do Complemento/deficiência , Fator H do Complemento/genética , Modelos Animais de Doenças , Predisposição Genética para Doença , Glomerulonefrite/genética , Glomerulonefrite/imunologia , Glomerulonefrite/metabolismo , Rim/imunologia , Rim/metabolismo , Rim/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Properdina/deficiência , Properdina/genética , Proteinúria/imunologia , Proteinúria/metabolismo , Proteinúria/prevenção & controle , Receptor da Anafilatoxina C5a/deficiência , Receptor da Anafilatoxina C5a/genética , Insuficiência Renal/genética , Insuficiência Renal/imunologia , Insuficiência Renal/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
10.
Blood ; 129(9): 1184-1196, 2017 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-28057640

RESUMO

Complement plays a key role in host defense, but its dysregulation can cause autologous tissue injury. Complement activation is normally controlled by regulatory proteins, including factor H (FH) in plasma and membrane cofactor protein (MCP) on the cell surface. Mutations in FH and MCP are linked to atypical hemolytic uremic syndrome, a type of thrombotic microangiopathy (TMA) that causes renal failure. We describe here that disruption of FH function on the cell surface can also lead to disseminated complement-dependent macrovascular thrombosis. By gene targeting, we introduced a point mutation (W1206R) into murine FH that impaired its interaction with host cells but did not affect its plasma complement-regulating activity. Homozygous mutant mice carrying this mutation developed renal TMA as well as systemic thrombophilia involving large blood vessels in multiple organs, including liver, lung, spleen, and kidney. Approximately 30% of mutant mice displayed symptoms of stroke and ischemic retinopathy, and 48% died prematurely. Genetic deficiency of complement C3 and factor D prevented both the systemic thrombophilia and renal TMA phenotypes. These results demonstrate a causal relationship between complement dysregulation and systemic angiopathy and suggest that complement activation may contribute to various human thrombotic disorders involving both the micro- and macrovasculature.


Assuntos
Fator H do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Trombofilia/genética , Animais , Western Blotting , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação Puntual
11.
Immunobiology ; 221(10): 1080-90, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27371974

RESUMO

Remarkable advances have been made over past decades in understanding the pathogenesis of complement-mediated diseases. This has led to development of new therapies for, and in some cases re-classification of, complement-driven diseases. This success is due to not only insight from human patients but also studies using transgenic animal models. Animal models that mimic human diseases are useful tools to understand the mechanism of disease and develop new therapies but there are also limitations due to species differences in their complement systems. This review provides a summary of transgenic animal models for three human diseases that are at the forefront of anti-complement therapy, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy (C3G). They are discussed here as examples to highlight the values and limitations of animal modeling in complement-driven diseases.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Suscetibilidade a Doenças , Animais , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Síndrome Hemolítico-Urêmica Atípica/etiologia , Síndrome Hemolítico-Urêmica Atípica/metabolismo , Ativação do Complemento/imunologia , Inativadores do Complemento/farmacologia , Inativadores do Complemento/uso terapêutico , Modelos Animais de Doenças , Hemoglobinúria Paroxística/tratamento farmacológico , Hemoglobinúria Paroxística/etiologia , Hemoglobinúria Paroxística/metabolismo , Humanos , Camundongos Transgênicos , Nefrose/tratamento farmacológico , Nefrose/etiologia , Nefrose/metabolismo
12.
J Immunol ; 195(3): 1171-81, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26116506

RESUMO

Complement is implicated in asthma pathogenesis, but its mechanism of action in this disease remains incompletely understood. In this study, we investigated the role of properdin (P), a positive alternative pathway complement regulator, in allergen-induced airway inflammation. Allergen challenge stimulated P release into the airways of asthmatic patients, and P levels positively correlated with proinflammatory cytokines in human bronchoalveolar lavage (BAL). High levels of P were also detected in the BAL of OVA-sensitized and challenged but not naive mice. Compared with wild-type (WT) mice, P-deficient (P(-/-)) mice had markedly reduced total and eosinophil cell counts in BAL and significantly attenuated airway hyperresponsiveness to methacholine. Ab blocking of P at both sensitization and challenge phases or at challenge phase alone, but not at sensitization phase alone, reduced airway inflammation. Conversely, intranasal reconstitution of P to P(-/-) mice at the challenge phase restored airway inflammation to wild-type levels. Notably, C3a levels in the BAL of OVA-challenged P(-/-) mice were significantly lower than in wild-type mice, and intranasal coadministration of an anti-C3a mAb with P to P(-/-) mice prevented restoration of airway inflammation. These results show that P plays a key role in allergen-induced airway inflammation and represents a potential therapeutic target for human asthma.


Assuntos
Asma/imunologia , Complemento C3a/biossíntese , Properdina/imunologia , Células Th17/imunologia , Células Th2/imunologia , Animais , Asma/induzido quimicamente , Asma/patologia , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Complemento C3a/imunologia , Eosinófilos/imunologia , Humanos , Inflamação/imunologia , Contagem de Leucócitos , Pulmão/imunologia , Pulmão/patologia , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Properdina/genética
13.
Kidney Int ; 86(4): 726-37, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24850152

RESUMO

The murine cell surface protein Crry (complement receptor 1-related protein/gene y) is a key complement regulator with similar activities to human membrane cofactor protein (MCP) and decay-accelerating factor. MCP has a critical role in preventing complement-mediated tissue injury and its mutation has been implicated in several human kidney diseases. The study of Crry in mice has relevance to understanding MCP activity in human diseases; however, such efforts have been hampered by the embryonic lethality phenotype of Crry gene knockout. Here we used a conditional gene-targeting approach and deleted Crry from the mouse proximal tubular epithelial cells where Crry is prominently expressed. Absence of Crry from proximal tubular epithelial cells resulted in spontaneous C3 deposition on the basolateral surface but no apparent renal disease in unchallenged mice. However, mice deficient in Crry on proximal tubular epithelial cells developed exacerbated renal injury when subjected to renal ischemia-reperfusion, showing increased blood urea nitrogen levels, higher tubular injury scores, more tubular epithelial cell apoptosis, and inflammatory infiltrates. Renal ischemia-reperfusion injury in the Crry conditional knockout mice was prevented by blocking C3 and C5 activation using an anti-properdin or anti-C5 monoclonal antibody (mAb), respectively. Thus, Crry has a critical role in protecting proximal tubular epithelial cells during ischemia-reperfusion challenge. Our results highlight the latent risk for inflammatory kidney injury associated with defects in membrane complement regulators.


Assuntos
Predisposição Genética para Doença , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Animais , Anticorpos Monoclonais/farmacologia , Apoptose , Nitrogênio da Ureia Sanguínea , Ativação do Complemento/efeitos dos fármacos , Complemento C3/metabolismo , Complemento C5/imunologia , Complemento C5/metabolismo , Células Epiteliais , Técnicas de Inativação de Genes , Túbulos Renais Proximais , Camundongos , Camundongos Knockout , Fagócitos , Properdina/imunologia , Receptores de Complemento 3b
14.
J Immunol ; 190(7): 3552-9, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23427256

RESUMO

Complement is implicated in the pathogenesis of ischemia-reperfusion injury (IRI). The activation pathway(s) and effector(s) of complement in IRI may be organ specific and remain to be fully characterized. We previously developed a renal IRI model in decay-accelerating factor (DAF) and CD59 double-knockout (DAF(-/-)CD59(-/-)) mice. In this study, we used this model to dissect the pathway(s) by which complement is activated in renal IRI and to evaluate whether C3aR- or C5aR-mediated inflammation or the membrane attack complex was pathogenic. We crossed DAF(-/-)CD59(-/-) mice with mice deficient in various complement components or receptors including C3, C4, factor B (fB), factor properdin (fP), mannose-binding lectin, C3aR, C5aR, or Ig and assessed renal IRI in the resulting mutant strains. We found that deletion of C3, fB, fP, C3aR, or C5aR significantly ameliorated renal IRI in DAF(-/-)CD59(-/-) mice, whereas deficiency of C4, Ig, or mannose-binding lectin had no effect. Treatment of DAF(-/-)CD59(-/-) mice with an anti-C5 mAb reduced renal IRI to a greater degree than did C5aR deficiency. We also generated and tested a function-blocking anti-mouse fP mAb and showed it to ameliorate renal IRI when given to DAF(-/-)CD59(-/-) mice 24 h before, but not 4 or 8 h after, ischemia/reperfusion. These results suggest that complement is activated via the alternative pathway during the early phase of reperfusion, and both anaphylatoxin-mediated inflammation and the membrane attack complex contribute to tissue injury. Further, they demonstrate a critical role for properdin and support its therapeutic targeting in renal IRI.


Assuntos
Antígenos CD55/genética , Antígenos CD59/genética , Rim/irrigação sanguínea , Properdina/antagonistas & inibidores , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Complemento C3/deficiência , Complemento C3/imunologia , C3 Convertase da Via Alternativa do Complemento , Fator B do Complemento/genética , Fator B do Complemento/imunologia , Modelos Animais de Doenças , Rim/patologia , Masculino , Camundongos , Camundongos Knockout , Properdina/genética , Properdina/imunologia , Traumatismo por Reperfusão/patologia
15.
J Am Soc Nephrol ; 24(1): 53-65, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23204401

RESUMO

Factor H (fH) and properdin both modulate complement; however, fH inhibits activation, and properdin promotes activation of the alternative pathway of complement. Mutations in fH associate with several human kidney diseases, but whether inhibiting properdin would be beneficial in these diseases is unknown. Here, we found that either genetic or pharmacological blockade of properdin, which we expected to be therapeutic, converted the mild C3 GN of an fH-mutant mouse to a lethal C3 GN with features of human dense deposit disease. We attributed this phenotypic change to a differential effect of properdin on the dynamics of alternative pathway complement activation in the fluid phase and the cell surface in the fH-mutant mice. Thus, in fH mutation-related C3 glomerulopathy, additional factors that impact the activation of the alternative pathway of complement critically determine the nature and severity of kidney pathology. These results show that therapeutic manipulation of the complement system requires rigorous disease-specific target validation.


Assuntos
Glomerulonefrite Membranoproliferativa/genética , Nefropatias/genética , Properdina/deficiência , Animais , Complemento C3/metabolismo , Fator H do Complemento/deficiência , Fator H do Complemento/genética , Via Alternativa do Complemento , Modelos Animais de Doenças , Glomerulonefrite Membranoproliferativa/metabolismo , Glomerulonefrite Membranoproliferativa/patologia , Doenças da Deficiência Hereditária de Complemento , Humanos , Glomérulos Renais/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Mutação
16.
J Insect Physiol ; 56(9): 1224-31, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20361975

RESUMO

The insect development is intricately controlled by morphogenetic hormones, juvenile hormone (JH) and 20-hydroxyecdysone (20E) through the regulation of gene/protein expression. The role of hexamerins in the metamorphosis of insects and reproduction and their control by 20E at the gene level has been widely reported in insects. In the present study we for the first time report the role of ecdysteroids in the regulation of hexamerin synthesis in a lepidopteran insect Corcyra cephalonica. The hormonal studies were carried out using the normal and the thorax-ligated insects with both 20E and its non-steroidal agonist RH-5992. The in vitro as well as in vivo studies showed a stimulatory effect of 20E and its agonist on the hexamerin synthesis including arylphorin (Hex 2), whereas hormone blockade with azadirachtin caused a time dependent reduction in synthesis. The northern analysis using Hex 2b cDNA as probe too confirmed the above result. This was followed by the cloning of the Hex 2b gene. The full length of the genomic clone was found to be 3.5kb long and has four exons interspersed by three introns. The genome walking analysis revealed the presence of a steroid hormone binding sequence "Ecdysone response element" (EcRE) in the 5' untranscribed region (UTR) of the gene. The data presented in this paper clearly suggest that hexamerin synthesis in C. cephalonica is transcriptionally regulated by 20E.


Assuntos
Ecdisterona/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Insetos/biossíntese , Mariposas/metabolismo , Animais , Sequência de Bases , Northern Blotting , Clonagem Molecular , Primers do DNA/genética , Ecdisterona/agonistas , Eletroforese em Gel de Poliacrilamida , Componentes do Gene , Regulação da Expressão Gênica/efeitos dos fármacos , Hidrazinas/farmacologia , Imunoprecipitação , Proteínas de Insetos/genética , Inseticidas/farmacologia , Limoninas/farmacologia , Metamorfose Biológica/fisiologia , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Reprodução/fisiologia , Elementos de Resposta/genética , Análise de Sequência de DNA
17.
J Insect Physiol ; 56(9): 1071-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20230828

RESUMO

Selective receptor mediated uptake is a widely prevalent mechanism in insects by which important macromolecules are acquired. Among the various proteins sequestered by the insect fat body, the larval hexamerins form the major group. In the present work full length cDNA (2.6kb) of hexamerin receptor with an ORF of 2.4kb was cloned from the larval fat body of rice moth, Corcyra cephalonica. This was followed by the recombinant expression of truncated N-terminal sequence of putative hexamerin receptor and the confirmation of the expressed recombinant protein as the truncated hexamerin receptor by ligand blot analysis. Apart from this we also analyzed other hexamerin sequestering tissues like salivary gland, male accessory reproductive gland and ovary for the presence of hexamerin receptor. We found that the receptor in these tissues was similar in size and mode of activation to that of fat body hexamerin receptor, thus cementing the fact that identical hexamerin receptors are present in all the hexamerin sequestering tissues in the rice moth.


Assuntos
Proteínas de Transporte/genética , Corpo Adiposo/metabolismo , Proteínas de Insetos/genética , Mariposas/genética , Animais , Western Blotting , Proteínas de Transporte/metabolismo , Clonagem Molecular , Primers do DNA/genética , DNA Complementar/genética , Ecdisterona/metabolismo , Eletroforese em Gel de Poliacrilamida , Feminino , Perfilação da Expressão Gênica , Genitália/metabolismo , Proteínas de Insetos/metabolismo , Masculino , Reação em Cadeia da Polimerase , Proteínas Recombinantes/metabolismo , Glândulas Salivares/metabolismo
18.
Artigo em Inglês | MEDLINE | ID: mdl-20138238

RESUMO

Glutathione S-transferases (GSTs) are a large family of multifunctional enzymes, known for their role in cellular detoxification. Here we report a cytosolic GST with optimal activity at alkaline pH (8.3) from the visceral fat body of late-last instar (LLI) larvae of a lepidopteran insect rice moth Corcyra cephalonica. All previously known GSTs are active between pH 6.0 to 6.5. Purification and characterization revealed the Corcyra cephalonica GST (CcGST) as a 23-kDa protein. HPLC and 2D analysis showed a single isoform of the protein in the LLI visceral fat body. Degenerate primer based method identified a 701-nucleotide cDNA and the longest open reading frame contained 216 amino acids. Multiple sequence and structural alignment showed close similarity with delta-class GSTs. CcGST is present mainly in the fat body with highest activity at the late-last instar larval stage. Juvenile hormone (JH) negatively inhibits the CcGST activity both ex vivo and in vivo. We speculate that high expression and activity of CcGST in the fat body of the late-last instar larvae, when endogenous JH titer is low may have role in the insect post-embryonic development unrelated to their previously known function.


Assuntos
Glutationa Transferase/química , Proteínas de Insetos/química , Mariposas/enzimologia , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA Complementar/metabolismo , Glutationa Transferase/isolamento & purificação , Proteínas de Insetos/isolamento & purificação , Hormônios Juvenis/farmacologia , Modelos Moleculares , Dados de Sequência Molecular , Mariposas/crescimento & desenvolvimento , Conformação Proteica
19.
Protein Expr Purif ; 67(1): 53-60, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19383546

RESUMO

The apoptotic protease activating factor (Apaf-1) is a protein that binds to cytochrome c, and in the presence of dATP/ATP oligomerizes to assume the role of an adaptor platform for activating the caspase-9 zymogen. In order to study the biochemical and structural details of Apaf-1 function, we have generated an expression construct from pcDNA 3-Apaf-1XL for production of the WD40 domain ((WD40)Apaf-1) in Escherichia coli. The WD40 domain expressed contains 825 amino acids in addition to an N-terminal His(6) tag derived from the cloning vector. The expressed protein is invariably localized in the inclusion body fraction of E. coli. A simple protocol involving Sephadex G100 chromatography developed for purifying the protein starting from inclusion bodies has allowed protein recovery in highly pure form. Basic fluorescence and CD spectra indicate that the refolded protein has extensive secondary and tertiary structures. Immunoprecipitation studies have provided qualitative information about the binding interaction of (WD40)Apaf-1 and cytochrome c. The binding interaction has been quantified by spectrophotometric titration of cytochrome c with recombinant (WD40)Apaf-1. The results demonstrate a weak binding for cytochrome c and (WD40)Apaf-1 interaction, the binding affinity being 390 nM. The analysis indicates a 2:1 or possibly even 3:1 stoichiometry for cytochrome c and (WD40)Apaf-1 binding interaction.


Assuntos
Fator Apoptótico 1 Ativador de Proteases/metabolismo , Escherichia coli/genética , Estrutura Terciária de Proteína , Fator Apoptótico 1 Ativador de Proteases/química , Fator Apoptótico 1 Ativador de Proteases/genética , Cromatografia em Gel , Citocromos c/metabolismo , Dextranos , Ditiotreitol/química , Humanos , Concentração de Íons de Hidrogênio , Corpos de Inclusão/química , Espectrometria de Massas , Conformação Proteica , Dobramento de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Espectrofotometria
20.
Insect Biochem Mol Biol ; 38(3): 307-19, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18252245

RESUMO

Hexamerins are stage specifically sequestered during the non-feeding stages mainly by the fat body cells from hemolymph through ecdysteroid regulated receptor-mediated endocytosis. 20-Hydroxyecdysone (20E) stimulates the tyrosine kinase-mediated phosphorylation of the 120kDa hexamerin receptor in the rice moth, Corcyra cephalonica. Here, we demonstrate that phosphorylation of the hexamerin receptor by HP19-regulated-20E-dependent-tyrosine kinase is a critical regulator for its activation, and is required for hexamerin uptake. Hexamerin receptor is phosphorylated only in the hexamerin sequestering tissues. The receptor phosphorylation is a prerequisite for hexamerin uptake and both phosphorylation and concomitant uptake are developmentally regulated. In addition, endogenous fat body tyrosine kinase activity is also developmentally and hormonally regulated. 20E induces the tyrosine kinase activity both in vivo as well as ex vivo, and the receptor phosphorylation is likely an extra-cellular event. The hemolymph protein, HP19 inhibits the 20E-induced phosphorylation by inhibiting tyrosine kinase activity. These inhibitions are rapid in homogenate preparations and are unaffected by the inhibitors of transcription and translation. We propose that hexamerin sequestration is negatively regulated by active HP19 at the feeding larval stage, thus preventing the uptake. During the non-feeding pupal stage, high ecdysteroid titer and negligible HP19 activity, positively regulates the receptor phosphorylation resulting in hexamerin uptake. These studies are therefore the first evidence of hexamerin uptake regulated by the orchestration of 20E and HP19 at a nongenomic level.


Assuntos
Proteínas de Transporte/metabolismo , Ecdisterona/metabolismo , Hemolinfa/metabolismo , Proteínas de Insetos/metabolismo , Proteínas Quinases/metabolismo , Animais , Ecdisterona/farmacologia , Larva/metabolismo , Mariposas , Fosforilação/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...