Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.941
Filtrar
1.
Front Immunol ; 13: 746068, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154091

RESUMO

Antibody-induced complement activation may cause injury of the neuromuscular junction (NMJ) and is thus considered as a primary pathogenic factor in human myasthenia gravis (MG) and animal models of experimental autoimmune myasthenia gravis (EAMG). In this study, we tested whether CRIg/FH, a targeted complement inhibitor, could attenuate NMJ injury in rat MG models. We first demonstrated that CRIg/FH could inhibit complement-dependent cytotoxicity on human rhabdomyosarcoma TE671 cells induced by MG patient-derived IgG in vitro. Furthermore, we investigated the therapeutic effect of CRIg/FH in a passive and an active EAMG rodent model. In both models, administration of CRIg/FH could significantly reduce the complement-mediated end-plate damage and suppress the development of EAMG. In the active EAMG model, we also found that CRIg/FH treatment remarkably reduced the serum concentration of autoantibodies and of the cytokines including IFN-γ, IL-2, IL-6, and IL-17, and upregulated the percentage of Treg cells in the spleen, which was further verified in vitro. Therefore, our findings indicate that CRIg/FH may hold the potential for the treatment of MG via immune modulation.


Assuntos
Inativadores do Complemento/farmacologia , Imunomodulação/efeitos dos fármacos , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/prevenção & controle , Proteínas Recombinantes de Fusão/farmacologia , Animais , Autoanticorpos/imunologia , Autoimunidade , Diferenciação Celular , Linhagem Celular , Ativação do Complemento/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Humanos , Imunoglobulina G/imunologia , Mediadores da Inflamação/metabolismo , Ativação Linfocitária , Miastenia Gravis Autoimune Experimental/diagnóstico , Ratos , Índice de Gravidade de Doença , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
2.
PLoS One ; 17(2): e0264628, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35213675

RESUMO

BACKGROUND: The complement system plays an important role in pathophysiology of cardiovascular disease (CVD), and might be involved in accelerated atherogenesis in rheumatoid arthritis (RA). The role of complement activation in response to treatment, and in development of premature CVD in RA, is limited. Therefore, we examined the effects of methotrexate (MTX) and tumor necrosis factor inhibitors (TNFi) on complement activation using soluble terminal complement complex (TCC) levels in RA; and assessed associations between TCC and inflammatory and cardiovascular biomarkers. METHODS: We assessed 64 RA patients starting with MTX monotherapy (n = 34) or TNFi with or without MTX co-medication (TNFi±MTX, n = 30). ELISA was used to measure TCC in EDTA plasma. The patients were examined at baseline, after 6 weeks and 6 months of treatment. RESULTS: Median TCC was 1.10 CAU/mL, and 57 (89%) patients had TCC above the estimated upper reference limit (<0.70). Compared to baseline, TCC levels were significantly lower at 6-week visit (0.85 CAU/mL, p<0.0001), without significant differences between the two treatment regimens. Notably, sustained reduction in TCC was only achieved after 6 months on TNFi±MTX (0.80 CAU/mL, p = 0.006). Reductions in TCC after treatment were related to decreased C-reactive protein (CRP), erythrocyte sedimentation rate (ESR) and interleukin 6, and increased levels of total, high and low-density lipoprotein cholesterol. Similarly, baseline TCC was significantly related to baseline CRP, ESR and interleukin 6. Patients with endothelial dysfunction had higher baseline TCC than those without (median 1.4 versus 1.0 CAU/mL, p = 0.023). CONCLUSIONS: Patients with active RA had elevated TCC, indicating increased complement activation. TCC decreased with antirheumatic treatment already after 6 weeks. However, only treatment with TNFi±MTX led to sustained reduction in TCC during the 6-month follow-up period. RA patients with endothelial dysfunction had higher baseline TCC compared to those without, possibly reflecting involvement of complement in the atherosclerotic process in RA.


Assuntos
Antirreumáticos/farmacologia , Artrite Reumatoide/tratamento farmacológico , Ativação do Complemento/efeitos dos fármacos , Antirreumáticos/uso terapêutico , Sedimentação Sanguínea , Proteína C-Reativa/análise , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Complexo de Ataque à Membrana do Sistema Complemento/análise , Esquema de Medicação , Quimioterapia Combinada , Feminino , Humanos , Interleucina-6/sangue , Masculino , Metotrexato/farmacologia , Metotrexato/uso terapêutico , Pessoa de Meia-Idade , Resultado do Tratamento , Inibidores do Fator de Necrose Tumoral/farmacologia , Inibidores do Fator de Necrose Tumoral/uso terapêutico
3.
J Ethnopharmacol ; 288: 115004, 2022 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-35051603

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Esculetin is a bioactive compound of medicinal herb Hydrangea paniculata, and has showed anti-oxidation and anti-inflammation bioactivities. Renal local oxidative stress and inflammation are import contributors for progression of lupus nephritis (LN). AIM OF THE STUDY: In the present study, the renal protective effect of esculetin against LN was evaluated using MRL/lpr mice. MATERIALS AND METHODS: MRL/lpr mice were orally administrated with esculetin (20 mg/kg and 40 mg/kg) from 10 to 20 weeks and then renal function and kidney pathology were analyzed. RESULTS: Esculetin significantly attenuated renal impairment in MRL/lpr mice by reducing blood urea nitrogen (BUN), serum creatinine (Scr) and albuminuria, and ameliorated the glomerular hypertrophy, tubular interstitial fibrosis and mononuclear cell infiltration into interstitium. mRNA microarray suggested that esculetin could significantly down-regulate complement cascade, inflammation and fibrosis pathway, and up-regulate Nrf2-related anti-oxidation genes. Most surprising finding in the current study was that esculetin could inhibit the complement activation both in classical and alternative pathway using in vitro hemolysis assay, further enzyme assay suggested that esculetin blocked the C3 convertase (C4b2a) to exert this inhibitory capability. Molecular docking predicted that esculetin had four conventional hydrogen bonds interacting with C4b2a, and CDOCKER energy is relatively lower. Luciferase reporter gene demonstrated that esculetin could activate Nrf2 signaling pathway, and further flow cytometry confirmed that anti-oxidation bioactivity of esculetin was dependent on Nrf2 activation. On the other hand, esculetin could inhibit NFκB nuclear translocation and TGFß-smad3 profibrosis pathway. CONCLUSION: Esculetin shows beneficial effect on LN progression, and it may be a good natural leading compound for design of chemical compounds to treat LN.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Inflamação/tratamento farmacológico , Nefrite Lúpica/tratamento farmacológico , Umbeliferonas/farmacologia , Animais , Nitrogênio da Ureia Sanguínea , Creatinina/sangue , Progressão da Doença , Relação Dose-Resposta a Droga , Feminino , Hydrangea/química , Inflamação/patologia , Camundongos , Camundongos Endogâmicos MRL lpr , Simulação de Acoplamento Molecular , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Umbeliferonas/administração & dosagem , Umbeliferonas/isolamento & purificação
4.
Cells ; 10(12)2021 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-34944083

RESUMO

Considering the fact that many retinal diseases are yet to be cured, the pathomechanisms of these multifactorial diseases need to be investigated in more detail. Among others, oxidative stress and hypoxia are pathomechanisms that take place in retinal diseases, such as glaucoma, age-related macular degeneration, or diabetic retinopathy. In consideration of these diseases, it is also evidenced that the immune system, including the complement system and its activation, plays an important role. Suitable models to investigate neuroretinal diseases are organ cultures of porcine retina. Based on an established model, the role of the complement system was studied after the induction of oxidative stress or hypoxia. Both stressors led to a loss of retinal ganglion cells (RGCs) accompanied by apoptosis. Hypoxia activated the complement system as noted by higher C3+ and MAC+ cell numbers. In this model, activation of the complement cascade occurred via the classical pathway and the number of C1q+ microglia was increased. In oxidative stressed retinas, the complement system had no consideration, but strong inflammation took place, with elevated TNF, IL6, and IL8 mRNA expression levels. Together, this study shows that hypoxia and oxidative stress induce different mechanisms in the porcine retina inducing either the immune response or an inflammation. Our findings support the thesis that the immune system is involved in the development of retinal diseases. Furthermore, this study is evidence that both approaches seem suitable models to investigate undergoing pathomechanisms of several neuroretinal diseases.


Assuntos
Ativação do Complemento/imunologia , Via Clássica do Complemento/imunologia , Hipóxia/imunologia , Retina/imunologia , Retina/patologia , Células Ganglionares da Retina/patologia , Animais , Apoptose/efeitos dos fármacos , Cobalto/toxicidade , Ativação do Complemento/efeitos dos fármacos , Via Alternativa do Complemento/efeitos dos fármacos , Via Alternativa do Complemento/imunologia , Via Clássica do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/metabolismo , Peróxido de Hidrogênio/toxicidade , Lectinas/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Estresse Oxidativo/efeitos dos fármacos , Células Ganglionares da Retina/efeitos dos fármacos , Neurônios Retinianos/efeitos dos fármacos , Neurônios Retinianos/patologia , Estresse Fisiológico/efeitos dos fármacos , Suínos
5.
Front Immunol ; 12: 777932, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899737

RESUMO

Complement activation is key to anti-microbial defenses by directly acting on microbes and indirectly by triggering cellular immune responses. Complement activation may also contribute to the pathogenesis of numerous inflammatory and immunological diseases. Consequently, intense research focuses on developing therapeutics that block pathology-causing complement activation while preserving anti-microbial complement activities. However, the pace of research is slowed down significantly by the limitations of current tools for evaluating complement-targeting therapeutics. Moreover, the effects of potential therapeutic agents on innate immune cells, like neutrophils, are not fully understood. Here, we employ microfluidic assays and measure chemotaxis, phagocytosis, and swarming changes in human neutrophils ex vivo in response to various complement-targeting agents. We show that whereas complement factor 5 (C5) cleavage inhibitor eculizumab blocks all neutrophil anti-microbial functions, newer compounds like the C5 cleavage inhibitor RA101295 and C5a receptor antagonist avacopan inhibit chemotaxis and swarming while preserving neutrophil phagocytosis. These results highlight the utility of microfluidic neutrophil assays in evaluating potential complement-targeting therapeutics.


Assuntos
Compostos de Anilina/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/farmacologia , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas/instrumentação , Neutrófilos/efeitos dos fármacos , Ácidos Nipecóticos/farmacologia , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Complemento C3/farmacologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Complemento C5a/farmacologia , Humanos , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Fagocitose/efeitos dos fármacos , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Receptor da Anafilatoxina C5a/metabolismo
6.
Front Immunol ; 12: 785229, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899752

RESUMO

The complement system has long been recognized as a potential druggable target for a variety of inflammatory conditions. Very few complement inhibitors have been approved for clinical use, but a great number are in clinical development, nearly all of which systemically inhibit complement. There are benefits of targeting complement inhibition to sites of activation/disease in terms of efficacy and safety, and here we describe P-selectin targeted complement inhibitors, with and without a dual function of directly blocking P-selectin-mediated cell-adhesion. The constructs are characterized in vitro and in murine models of hindlimb ischemia/reperfusion injury and hindlimb transplantation. Both constructs specifically targeted to reperfused hindlimb and provided protection in the hindlimb ischemia/reperfusion injury model. The P-selectin blocking construct was the more efficacious, which correlated with less myeloid cell infiltration, but with similarly reduced levels of complement deposition. The blocking construct also improved tissue perfusion and, unlike the nonblocking construct, inhibited coagulation, raising the possibility of differential application of each construct, such as in thrombotic vs. hemorrhagic conditions. Similar outcomes were obtained with the blocking construct following vascularized composite graft transplantation, and treatment also significantly increased graft survival. This is outcome may be particularly pertinent in the context of vascularized composite allograft transplantation, since reduced ischemia reperfusion injury is linked to a less rigorous alloimmune response that may translate to the requirement of a less aggressive immunosuppressive regime for this normally nonlife-threatening procedure. In summary, we describe a new generation of targeted complement inhibitor with multi-functionality that includes targeting to vascular injury, P-selectin blockade, complement inhibition and anti-thrombotic activity. The constructs described also bound to both mouse and human P-selectin which may facilitate potential translation.


Assuntos
Adesão Celular/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/administração & dosagem , Membro Posterior/irrigação sanguínea , Membro Posterior/transplante , Selectina-P/antagonistas & inibidores , Receptores de Complemento 3b/administração & dosagem , Traumatismo por Reperfusão/prevenção & controle , Anticorpos de Cadeia Única/administração & dosagem , Alotransplante de Tecidos Compostos Vascularizados , Animais , Inativadores do Complemento/farmacocinética , Modelos Animais de Doenças , Fibrinolíticos/administração & dosagem , Sobrevivência de Enxerto/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Selectina-P/metabolismo , Fluxo Sanguíneo Regional , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Transdução de Sinais , Anticorpos de Cadeia Única/farmacocinética
7.
Front Immunol ; 12: 791429, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899761

RESUMO

The complex pathophysiologic interplay between SARS-CoV-2 infection and complement activation is the subject of active investigation. It is clinically mirrored by the occurrence of exacerbations of complement mediated diseases during COVID-19 infection. These include complement-mediated hemolytic anemias such as paroxysmal nocturnal hemoglobinuria (PNH), autoimmune hemolytic anemia (AIHA), particularly cold agglutinin disease (CAD), and hemolytic uremic syndrome (HUS). All these conditions may benefit from complement inhibitors that are also under study for COVID-19 disease. Hemolytic exacerbations in these conditions may occur upon several triggers including infections and vaccines and may require transfusions, treatment with complement inhibitors and/or immunosuppressors (i.e., steroids and rituximab for AIHA), and result in thrombotic complications. In this manuscript we describe four patients (2 with PNH and 2 with CAD) who experienced hemolytic flares after either COVID-19 infection or SARS-Cov2 vaccine and provide a review of the most recent literature. We report that most episodes occurred within the first 10 days after COVID-19 infection/vaccination and suggest laboratory monitoring (Hb and LDH levels) in that period. Moreover, in our experience and in the literature, hemolytic exacerbations occurring during COVID-19 infection were more severe, required greater therapeutic intervention, and carried more complications including fatalities, as compared to those developing after SARS-CoV-2 vaccine, suggesting the importance of vaccinating this patient population. Patient education remains pivotal to promptly recognize signs/symptoms of hemolytic flares and to refer to medical attention. Treatment choice should be based on the severity of the hemolytic exacerbation as well as of that of COVID-19 infection. Therapies include transfusions, complement inhibitor initiation/additional dose in the case of PNH, steroids/rituximab in patients with CAD and warm type AIHA, plasma exchange, hemodialysis and complement inhibitor in the case of atypical HUS. Finally, anti-thrombotic prophylaxis should be always considered in these settings, provided safe platelet counts.


Assuntos
Anemia Hemolítica Autoimune/imunologia , Vacinas contra COVID-19/imunologia , COVID-19/imunologia , Proteínas do Sistema Complemento/imunologia , Hemoglobinúria Paroxística/imunologia , SARS-CoV-2/imunologia , Adolescente , Adulto , Idoso , Anemia Hemolítica Autoimune/terapia , COVID-19/prevenção & controle , COVID-19/virologia , Vacinas contra COVID-19/administração & dosagem , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Inativadores do Complemento/uso terapêutico , Feminino , Hemoglobinúria Paroxística/terapia , Humanos , Masculino , Pessoa de Meia-Idade , SARS-CoV-2/fisiologia
8.
Sci Rep ; 11(1): 21873, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34750424

RESUMO

The complement system is a potent mediator of ischemia-reperfusion injury (IRI), which detrimentally affects the function and survival of transplanted kidneys. Human complement receptor 1 (HuCR1) is an integral membrane protein that inhibits complement activation by blocking the convertases that activate C3 and C5. We have previously reported that CSL040, a truncated form of recombinant soluble HuCR1 (sHuCR1), has enhanced complement inhibitory activity and improved pharmacokinetic properties compared to the parent molecule. Here, we compared the capacity of CSL040 and full-length sHuCR1 to suppress complement-mediated organ damage in a mouse model of warm renal IRI. Mice were treated with two doses of CSL040 or sHuCR1, given 1 h prior to 22 min unilateral renal ischemia and again 3 h later. 24 h after reperfusion, mice treated with CSL040 were protected against warm renal IRI in a dose-dependent manner, with the highest dose of 60 mg/kg significantly reducing renal dysfunction, tubular injury, complement activation, endothelial damage, and leukocyte infiltration. In contrast, treatment with sHuCR1 at a molar equivalent dose to 60 mg/kg CSL040 did not confer significant protection. Our results identify CSL040 as a promising therapeutic candidate to attenuate renal IRI and demonstrate its superior efficacy over full-length sHuCR1 in vivo.


Assuntos
Rim/lesões , Receptores de Complemento 3b/administração & dosagem , Traumatismo por Reperfusão/prevenção & controle , Animais , Ativação do Complemento/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Rim/efeitos dos fármacos , Rim/imunologia , Transplante de Rim/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Receptores de Complemento 3b/química , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/imunologia , Solubilidade
9.
PLoS One ; 16(10): e0259133, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34710157

RESUMO

Acute lung injury (ALI) often causes severe trauma that may progress to significant morbidity and mortality. ALI results from a combination of the underlying clinical condition of the patient (e.g., inflammation) with a secondary insult such as viral pneumonia or a blood transfusion. While the secondary insult may be variable, the rapidly progressive disease process leading to pulmonary failure is typically mediated by an overwhelming innate immunological or inflammatory reaction driven by excessive complement and neutrophil-mediated inflammatory responses. We recently developed a 'two-hit' ALI rat model mediated by lipopolysaccharide followed by transfusion of incompatible human erythrocytes resulting in complement activation, neutrophil-mediated ALI and free DNA in the blood indicative of neutrophil extracellular trap formation. The objective of this study was to evaluate the role of peptide inhibitor of complement C1 (RLS-0071), a classical complement pathway inhibitor and neutrophil modulator in this animal model. Adolescent male Wistar rats were infused with lipopolysaccharide followed by transfusion of incompatible erythrocytes in the presence or absence of RLS-0071. Blood was collected at various time points to assess complement C5a levels, free DNA and cytokines in isolated plasma. Four hours following erythrocyte transfusion, lung tissue was recovered and assayed for ALI by histology. Compared to animals not receiving RLS-0071, lungs of animals treated with a single dose of RLS-0071 showed significant reduction in ALI as well as reduced levels of C5a, free DNA and inflammatory cytokines in the blood. These results demonstrate that RLS-0071 can modulate neutrophil-mediated ALI in this novel rat model.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Anti-Inflamatórios/uso terapêutico , Ativação do Complemento/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Infiltração de Neutrófilos/efeitos dos fármacos , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/patologia , Animais , Anti-Inflamatórios/administração & dosagem , Citocinas/metabolismo , Modelos Animais de Doenças , Transfusão de Eritrócitos , Humanos , Lipopolissacarídeos , Pulmão/patologia , Masculino , Ratos , Ratos Wistar
10.
J Gen Virol ; 102(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34704923

RESUMO

The highly pathogenic Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is a severe respiratory virus. Recent reports indicate additional central nervous system (CNS) involvement. In this study, human DPP4 transgenic mice were infected with MERS-CoV, and viral antigens were first detected in the midbrain-hindbrain 4 days post-infection, suggesting the virus may enter the brainstem via peripheral nerves. Neurons and astrocytes throughout the brain were infected, followed by damage of the blood brain barrier (BBB), as well as microglial activation and inflammatory cell infiltration, which may be caused by complement activation based on the observation of deposition of complement activation product C3 and high expression of C3a receptor (C3aR) and C5a receptor (C5aR1) in neurons and glial cells. It may be concluded that these effects were mediated by complement activation in the brain, because of their reduction resulted from the treatment with mouse C5aR1-specific mAb. Such mAb significantly reduced nucleoprotein expression, suppressed microglial activation and decreased activation of caspase-3 in neurons and p38 phosphorylation in the brain. Collectively, these results suggest that MERS-CoV infection of CNS triggers complement activation, leading to inflammation-mediated damage of brain tissue, and regulating of complement activation could be a promising intervention and adjunctive treatment for CNS injury by MERS-CoV and other coronaviruses.


Assuntos
Encéfalo/patologia , Proteínas do Sistema Complemento/imunologia , Infecções por Coronavirus/patologia , Dipeptidil Peptidase 4/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Encéfalo/irrigação sanguínea , Encéfalo/imunologia , Encéfalo/virologia , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/uso terapêutico , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Modelos Animais de Doenças , Humanos , Inflamação , Camundongos , Camundongos Transgênicos , Microglia/imunologia , Microglia/patologia
11.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34502392

RESUMO

Tick saliva is a rich source of antihemostatic, anti-inflammatory, and immunomodulatory molecules that actively help the tick to finish its blood meal. Moreover, these molecules facilitate the transmission of tick-borne pathogens. Here we present the functional and structural characterization of Iripin-8, a salivary serpin from the tick Ixodes ricinus, a European vector of tick-borne encephalitis and Lyme disease. Iripin-8 displayed blood-meal-induced mRNA expression that peaked in nymphs and the salivary glands of adult females. Iripin-8 inhibited multiple proteases involved in blood coagulation and blocked the intrinsic and common pathways of the coagulation cascade in vitro. Moreover, Iripin-8 inhibited erythrocyte lysis by complement, and Iripin-8 knockdown by RNA interference in tick nymphs delayed the feeding time. Finally, we resolved the crystal structure of Iripin-8 at 1.89 Å resolution to reveal an unusually long and rigid reactive center loop that is conserved in several tick species. The P1 Arg residue is held in place distant from the serpin body by a conserved poly-Pro element on the P' side. Several PEG molecules bind to Iripin-8, including one in a deep cavity, perhaps indicating the presence of a small-molecule binding site. This is the first crystal structure of a tick serpin in the native state, and Iripin-8 is a tick serpin with a conserved reactive center loop that possesses antihemostatic activity that may mediate interference with host innate immunity.


Assuntos
Coagulação Sanguínea/fisiologia , Ativação do Complemento/fisiologia , Ixodes/metabolismo , Serpinas/metabolismo , Animais , Proteínas de Artrópodes/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Eritrócitos/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Ixodes/enzimologia , Ixodes/genética , Doença de Lyme , Ninfa , Saliva/química , Glândulas Salivares/metabolismo , Serpinas/ultraestrutura
12.
Adv Biol Regul ; 81: 100822, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34454870

RESUMO

Severe COVID-19 is characterized by lung and multiorgan inflammation and coagulation in the presence of overactivation of the complement system. Complement is a double edged-sward in SARS-Cov-2 infection. On one hand, it can control the viral infection in milder cases, on the other hand in cases with severe and prolonged infection massive complement activation occurs, which can intensify lung and systemic inflammation and promote a procoagulant and prothrombotic state. Several uncontrolled studies and controlled clinical trials with different complement inhibitors have been performed and others are ongoing. Results are promising in some but negative in others. Further studies are required to elucidate the benefit to risk profile of complement inhibitors in COVID-19 patients at different stages of the disease and to clarify the best targets in the complement cascade.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19/sangue , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/uso terapêutico , Proteínas do Sistema Complemento/metabolismo , SARS-CoV-2/metabolismo , Humanos , Pulmão/metabolismo , Pulmão/patologia , Índice de Gravidade de Doença
13.
J Immunol Res ; 2021: 9945725, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34368372

RESUMO

Age-related macular degeneration (AMD) is a multifactorial disease, which can culminate in irreversible vision loss and blindness in elderly. Nowadays, there is a big gap between dry AMD and wet AMD on treatment. Accounting for nearly 90% of AMD, dry AMD still lacks effective treatment. Numerous genetic and molecular researches have confirmed the significant role of the complement system in the pathogenesis of AMD, leading to a deeper exploration of complement inhibitors in the treatment of AMD. To date, at least 14 different complement inhibitors have been or are being explored in AMD in almost 40 clinical trials. While most complement inhibitors fail to treat AMD successfully, two of them are effective in inhibiting the rate of GA progression in phase II clinical trials, and both of them successfully entered phase III trials. Furthermore, recently emerging complement gene therapy and combination therapy also offer new opportunities to treat AMD in the future. In this review, we aim to introduce genetic and molecular associations between the complement system and AMD, provide the updated progress in complement inhibitors in AMD on clinical trials, and discuss the challenges and prospects of complement therapeutic strategies in AMD.


Assuntos
Inativadores do Complemento/uso terapêutico , Proteínas do Sistema Complemento/imunologia , Degeneração Macular/tratamento farmacológico , Degeneração Macular/etiologia , Animais , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Inativadores do Complemento/farmacologia , Gerenciamento Clínico , Suscetibilidade a Doenças/imunologia , Predisposição Genética para Doença , Humanos , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Terapia de Alvo Molecular , Fatores de Risco
14.
Nat Commun ; 12(1): 4858, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34381048

RESUMO

Complement is an enzymatic humoral pattern-recognition defence system of the body. Non-specific deposition of blood biomolecules on nanomedicines triggers complement activation through the alternative pathway, but complement-triggering mechanisms of nanomaterials with dimensions comparable to or smaller than many globular blood proteins are unknown. Here we study this using a library of <6 nm poly(amido amine) dendrimers bearing different end-terminal functional groups. Dendrimers are not sensed by C1q and mannan-binding lectin, and hence do not trigger complement activation through these pattern-recognition molecules. While, pyrrolidone- and carboxylic acid-terminated dendrimers fully evade complement response, and independent of factor H modulation, binding of amine-terminated dendrimers to a subset of natural IgM glycoforms triggers complement activation through lectin pathway-IgM axis. These findings contribute to mechanistic understanding of complement surveillance of dendrimeric materials, and provide opportunities for dendrimer-driven engineering of complement-safe nanomedicines and medical devices.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento/metabolismo , Dendrímeros/metabolismo , Imunoglobulina M/metabolismo , Ativação do Complemento/efeitos dos fármacos , Complemento C1q/metabolismo , Dendrímeros/química , Dendrímeros/farmacologia , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacologia , Humanos , Lectina de Ligação a Manose/metabolismo , Serina Proteases Associadas a Proteína de Ligação a Manose , Poliaminas/química , Poliaminas/metabolismo , Poliaminas/farmacologia
15.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34202223

RESUMO

Age-related macular degeneration (AMD) is a complex multifactorial disease characterized in its late form by neovascularization (wet type) or geographic atrophy of the retinal pigment epithelium cell layer (dry type). The complement system is an intrinsic component of innate immunity. There has been growing evidence that the complement system plays an integral role in maintaining immune surveillance and homeostasis in AMD. Based on the association between the genotypes of complement variants and AMD occurrence and the presence of complement in drusen from AMD patients, the complement system has become a therapeutic target for AMD. However, the mechanism of complement disease propagation in AMD has not been fully understood. This concise review focuses on an overall understanding of the role of the complement system in AMD and its ongoing clinical trials. It provides further insights into a strategy for the treatment of AMD targeting the complement system.


Assuntos
Proteínas do Sistema Complemento/imunologia , Degeneração Macular/etiologia , Ensaios Clínicos como Assunto , Ativação do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/metabolismo , Diagnóstico por Imagem , Gerenciamento Clínico , Suscetibilidade a Doenças , Humanos , Degeneração Macular/diagnóstico , Degeneração Macular/metabolismo , Degeneração Macular/terapia , Terapia de Alvo Molecular , Fatores de Risco , Resultado do Tratamento
16.
Front Immunol ; 12: 682334, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34276669

RESUMO

Both acute and chronic antibody-mediated allograft rejection (AMR), which are directly mediated by B cells, remain difficult to treat. Long-lived plasma cells (LLPCs) in bone marrow (BM) play a crucial role in the production of the antibodies that induce AMR. However, LLPCs survive through a T cell-independent mechanism and resist conventional immunosuppressive therapy. Desensitization therapy is therefore performed, although it is accompanied by severe side effects and the pathological condition may be at an irreversible stage when these antibodies, which induce AMR development, are detected in the serum. In other words, AMR control requires the development of a diagnostic method that predicts its onset before LLPC differentiation and enables therapeutic intervention and the establishment of humoral immune monitoring methods providing more detailed information, including individual differences in the susceptibility to immunosuppressive agents and the pathological conditions. In this study, we reviewed recent studies related to the direct or indirect involvement of immunocompetent cells in the differentiation of naïve-B cells into LLPCs, the limitations of conventional methods, and the possible development of novel control methods in the context of AMR. This information will significantly contribute to the development of clinical applications for AMR and improve the prognosis of patients who undergo organ transplantation.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Rejeição de Enxerto/tratamento farmacológico , Rejeição de Enxerto/etiologia , Imunossupressores/uso terapêutico , Aloenxertos/imunologia , Animais , Linfócitos B/metabolismo , Biomarcadores , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/metabolismo , Humanos , Memória Imunológica/efeitos dos fármacos , Imunossupressores/farmacologia , Imunoterapia , Técnicas de Diagnóstico Molecular/métodos , Terapia de Alvo Molecular/métodos , Plasmócitos/efeitos dos fármacos , Plasmócitos/imunologia , Plasmócitos/metabolismo , Transdução de Sinais
17.
Fish Shellfish Immunol ; 117: 1-16, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34274424

RESUMO

The effect of cinnamaldehyde (CM) enriched diet on immunity and cytokine gene expression in Channa striatus against Aphanomyces invadans is reported. C. striatus was uniformly divided into eight groups (n = 25 fish each) and fed with formulated diets with 0, 5, 10, and 15 mg kg-1 CM enriched diet. In healthy and infected groups fed with 5 mg kg-1 diet the leukocytes count increased significantly after 4th week; with 10 mg kg-1 CM diet the increase manifested after 6th week, but with 15 mg kg-1 not even after 8th week. In both groups, 5 mg kg-1 CM diet resulted in a significant increase in the serum total protein, albumin, and globulin levels after 4th week, whereas with other diets this effect was observed only after 6th week. Similarly, with any enriched diet the lysozyme activity increased significantly, but with 15 mg kg-1 CM diet only after 6th week. In both groups the complement activity and lymphocyte production increased significantly when fed with 5 mg kg-1 CM diet after 4th week while with other enriched diets only after 6th week. The phagocytic activity increased significantly in both groups fed with 5 mg kg-1 CM diet after 6th week, whereas the SOD activity increased after 4th week. The IgM production increased significantly in both groups fed with 5 mg kg-1 CM diet after 2nd week, while with 5 and 10 mg kg-1 CM diet after 4th week. In both groups, the expression of CXCR3α was significant on 4th week when fed with 10 mg kg-1 CM diet, while in the healthy group fed with 15 mg kg-1 CM diet the expression manifested earlier than 4th week. However, when fed with 10 and 15 mg kg-1 CM diets the increase was observed on 6th week; whereas, the expression of MHC-I reached the maximum on 6th week with any enriched diet. The results indicate that in C. striatus the innate immunity and expression of cytokine and immune related genes were significantly modulated when fed with 5 mg kg-1 CM diet on 4th week against A. invadans.


Assuntos
Acroleína/análogos & derivados , Aphanomyces , Doenças dos Peixes , Peixes/genética , Peixes/imunologia , Expressão Gênica/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Infecções , Acroleína/administração & dosagem , Animais , Ativação do Complemento/efeitos dos fármacos , Dieta , Doenças dos Peixes/genética , Doenças dos Peixes/imunologia , Imunoglobulina M/imunologia , Infecções/genética , Infecções/imunologia , Infecções/veterinária , Contagem de Leucócitos , Muramidase/imunologia , Fagocitose/efeitos dos fármacos , Espécies Reativas de Oxigênio/imunologia
18.
BMC Nephrol ; 22(1): 252, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34229609

RESUMO

BACKGROUND: Thrombotic microangiopathy (TMA)-mediated acute kidney injury (AKI) following massive haemorrhage is a rare but severe complication of the post-partum period. It is associated with a poor renal prognosis and a high risk of end-stage kidney disease. Complement activation may occur in this picture. However, whether complement activation, and thus complement blockade, may be critically relevant in this setting is unknown. CASE PRESENTATION: A 50 year-old woman presented with massive delayed post-partum haemorrhage (PPH). Despite bleeding control and normalization of coagulation parameters, she rapidly developed AKI stage 3 associated with dysmorphic microhematuria and proteinuria up to 2 g/day with the need of renal replacement therapy. Blood tests showed signs of TMA associated with markedly increased sC5b-9 and factor Bb plasma levels, respectively markers of terminal and alternative complement pathway over-activation. This clinical picture prompted us to initiate anti-C5 therapy. sC5b-9 normalized within 12 h after the first dose of eculizumab, factor Bb and C3 after seven days, platelet count after nine days and haptoglobin after 3 weeks. The clinical picture improved rapidly with blood pressure control within 48 h. Diuresis resumed after three days, kidney function rapidly improved and haemodialysis could be discontinued after the sixth and last dose. Serum creatinine returned to normal two years after presentation. CONCLUSIONS: We suggest that massive PPH induced major activation of complement pathways, which ultimately lead to TMA-induced AKI. Various causes, such as oocyte-donation, the potential retention of placental material and the use of tranexamic acid may have contributed to complement activation due to PPH. The prompt administration of anti-C5 therapy may have rapidly restored kidney microcirculation patency, thus reversing signs of TMA and AKI. We propose that complement activation may represent a major pathophysiological player of this complication and may provide a novel therapeutic avenue to improve renal prognosis in TMA-induced AKI following massive PPH.


Assuntos
Injúria Renal Aguda/etiologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Ativação do Complemento , Inativadores do Complemento/uso terapêutico , Hemorragia Pós-Parto/tratamento farmacológico , Hemorragia Pós-Parto/imunologia , Microangiopatias Trombóticas/etiologia , Injúria Renal Aguda/terapia , Biomarcadores/sangue , Ativação do Complemento/efeitos dos fármacos , Complemento C3/metabolismo , Fator B do Complemento/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Hemorragia Pós-Parto/sangue , Gravidez , Diálise Renal , Microangiopatias Trombóticas/terapia
19.
J Biomed Mater Res A ; 109(10): 1955-1966, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34085403

RESUMO

A main problem in the design of blood-contacting biomaterials has been the deficiency of a systematic understanding of blood-biomaterial interactions and the strategy to modulate blood responses. In this work, different functional groups including carboxyl (COOH), hydroxyl (OH) and zwitterionic sulfobetaine group (⊕N((CH3 )2 )(CH2 )3 SO3-○- , SMDB) were grafted on the poly (butylene terephthalate) (PBT) film to study how the functional groups modulate blood responses and in terms of interaction with the coagulation system, the complement system, and platelets. The results showed protein absorption and platelet adhesion was stronger on the PBT bearing COOH group than PBT films bearing OH and zwitterionic sulfobetaine groups (total protein (µg/cm2 ): 32.92 ± 5.89 vs. 22.02 ± 1.44 vs. 19.09 ± 1.59; platelet adhesion (/mm2 ): 1,626.7 ± 120.1 vs. 1,395.6 ± 363.3 vs. 1,102.2 ± 373.7), which had a rougher and negatively charged surface, and the coagulation system was inhibited by binding fibrinogen (Fg) and coagulation factors. Meanwhile, PBT-PSMDB showed anticoagulant property and induced platelet activation. As a result, complement formation on these two films were less than PBT bearing OH groups by inhibiting the coagulation system (C3a (ng/ml): 3,745.4 ± 143.9 vs. 3,290.9 ± 249.7 vs. 4,887.9 ± 88.9; C5a (ng/ml): 22.1 ± 2.6 vs. 22.3 ± 1.8 vs. 27.9 ± 2.0). On the other hand, PBT bearing OH groups did not facilitate remarkable platelet adhesion and activation, and had no influence on platelet aggregation, hypotonic shock response, and coagulation system. The above results showed that the blood responses were highly interlinked, and could be modulated by grafting with different functional groups on the biomaterial surfaces. These findings may help identify a strategy to design materials with better hemocompatibility for blood contact, filtration, and purification applications.


Assuntos
Eritrócitos/efeitos dos fármacos , Poliésteres/química , Poliésteres/farmacologia , Adsorção , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Ativação do Complemento/efeitos dos fármacos , Fibrinogênio/metabolismo , Hemólise/efeitos dos fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Teste de Materiais , Espectroscopia Fotoeletrônica , Adesividade Plaquetária/efeitos dos fármacos , Espectroscopia de Infravermelho com Transformada de Fourier , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...