Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Biotechnol Bioeng ; 117(6): 1904-1908, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32068245

RESUMEN

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.


Asunto(s)
Antiinflamatorios/química , Antiinflamatorios/farmacología , Complemento C3a/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Leucina/análogos & derivados , Leucina/farmacología , Células Cultivadas , Activación de Complemento/efectos de los fármacos , Complemento C3a/inmunología , Humanos , Inflamación/inmunología , Modelos Moleculares
2.
J Immunol ; 199(3): 1069-1085, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28687659

RESUMEN

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.


Asunto(s)
Astrocitos/fisiología , Diferenciación Celular/fisiología , Complemento C1q/biosíntesis , Complemento C3a/biosíntesis , Células-Madre Neurales/fisiología , Neutrófilos/metabolismo , Animales , Astrocitos/efectos de los fármacos , Movimiento Celular , Células Cultivadas , Complemento C1q/antagonistas & inhibidores , Complemento C1q/genética , Complemento C1q/inmunología , Complemento C3a/antagonistas & inhibidores , Complemento C3a/genética , Complemento C3a/inmunología , Medios de Cultivo Condicionados , Humanos , Inmunidad Innata , Ratones , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/inmunología , Neutrófilos/inmunología , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/fisiopatología
3.
Wound Repair Regen ; 25(1): 41-50, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28019718

RESUMEN

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.


Asunto(s)
Complemento C3a/antagonistas & inhibidores , Complemento C5a/antagonistas & inhibidores , Dípteros/metabolismo , Inflamación/fisiopatología , Larva/fisiología , Cicatrización de Heridas/fisiología , Animales , Secreciones Corporales/metabolismo , Activación de Complemento/fisiología , Complemento C3a/metabolismo , Complemento C5a/metabolismo , Desbridamiento/métodos , Regulación hacia Abajo , Inflamación/metabolismo , Activación Neutrófila/fisiología
4.
J Pharmacol Exp Ther ; 351(3): 527-37, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25232192

RESUMEN

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.


Asunto(s)
Complemento C3a/antagonistas & inhibidores , Factor D del Complemento/antagonistas & inhibidores , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Degeneración Macular/tratamiento farmacológico , Animales , Bovinos , Complemento C3a/inmunología , Factor D del Complemento/inmunología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Inyecciones Intravítreas , Macaca fascicularis , Degeneración Macular/sangre , Degeneración Macular/inmunología , Masculino , Ratones , Resultado del Tratamiento
5.
J Pharmacol Exp Ther ; 351(2): 344-51, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25150279

RESUMEN

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.


Asunto(s)
Sistema Cardiovascular/inmunología , Sistema Cardiovascular/fisiopatología , Activación de Complemento/inmunología , Complemento C3a/inmunología , Complemento C5a/inmunología , Hipertensión/inmunología , Hipertensión/fisiopatología , Animales , Complemento C3a/antagonistas & inhibidores , Complemento C5a/antagonistas & inhibidores , Modelos Animales de Enfermedad , Endotelio Vascular/inmunología , Endotelio Vascular/fisiopatología , Femenino , Frecuencia Cardíaca/inmunología , Isquemia/inmunología , Isquemia/fisiopatología , Placenta/inmunología , Embarazo , Ratas , Receptores de Complemento/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología
6.
J Immunol ; 188(11): 5694-705, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22539791

RESUMEN

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.


Asunto(s)
Asma/inmunología , Asma/patología , Hiperreactividad Bronquial/inmunología , Complemento C3a/fisiología , Inmunoglobulina E/fisiología , Interleucina-17/fisiología , Neutrófilos/inmunología , Neutrófilos/patología , Animales , Anticuerpos Monoclonales/fisiología , Asma/metabolismo , Hiperreactividad Bronquial/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Complemento C3a/antagonistas & inhibidores , Complemento C3a/inmunología , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/fisiología , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Receptores de Complemento/antagonistas & inhibidores , Factores de Tiempo
7.
J Immunol ; 186(5): 3120-9, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21270401

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/fisiología , Antígenos de Superficie/fisiología , Proteínas de la Membrana Bacteriana Externa/fisiología , Complemento C3d/metabolismo , Evasión Inmune/inmunología , Moraxella catarrhalis/inmunología , Adulto , Animales , Antígenos Bacterianos/metabolismo , Antígenos de Superficie/metabolismo , Proteínas de la Membrana Bacteriana Externa/biosíntesis , Proteínas de la Membrana Bacteriana Externa/metabolismo , Niño , Activación de Complemento/inmunología , Complemento C3a/antagonistas & inhibidores , Complemento C3a/fisiología , Proteínas Inactivadoras de Complemento/fisiología , Vía Alternativa del Complemento/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Ratones , Moraxella catarrhalis/aislamiento & purificación , Moraxella catarrhalis/patogenicidad , Infecciones por Moraxellaceae/inmunología , Infecciones por Moraxellaceae/microbiología , Infecciones por Moraxellaceae/patología , Unión Proteica/inmunología , Conejos , Ovinos
8.
Circ Res ; 105(11): 1094-101, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19815824

RESUMEN

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.


Asunto(s)
Muerte Encefálica/inmunología , Complemento C3a/inmunología , Trasplante de Corazón/inmunología , Daño por Reperfusión Miocárdica/inmunología , Donantes de Tejidos , Animales , Muerte Encefálica/patología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Activación de Complemento/inmunología , Complemento C3a/antagonistas & inhibidores , Modelos Animales de Enfermedad , Expresión Génica/inmunología , Precondicionamiento Isquémico/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocarditis/inmunología , Miocarditis/patología , Miocarditis/prevención & control
9.
Nat Med ; 9(4): 431-8, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12612546

RESUMEN

High-dose intravenous immunoglobulin (IVIG) prevents immune damage by scavenging complement fragments C3b and C4b. We tested the hypothesis that exogenous immunoglobulin molecules also bind anaphylatoxins C3a and C5a, thereby neutralizing their pro-inflammatory effects. Single-cell calcium measurements in HMC-1 human mast cells showed that a rise in intracellular calcium caused by C3a and C5a was inhibited in a concentration-dependent manner by IVIG, F(ab)2-IVIG and irrelevant human monoclonal antibody. C3a- and C5a-induced thromboxane (TXB2) generation and histamine release from HMC-1 cells and whole-blood basophils were also suppressed by exogenous immunoglobulins. In a mouse model of asthma, immunoglobulin treatment reduced cellular migration to the lung. Lethal C5a-mediated circulatory collapse in pigs was prevented by pretreatment with F(ab)2-IVIG. Molecular modeling, surface plasmon resonance (SPR) and western blot analyses suggested a physical association between anaphylatoxins and the constant region of F(ab)2. This binding could interfere with the role of C3a and C5a in inflammation.


Asunto(s)
Complemento C3a/antagonistas & inhibidores , Complemento C5a/antagonistas & inhibidores , Inmunoglobulinas Intravenosas/farmacología , gammaglobulinas/farmacología , Animales , Asma/metabolismo , Presión Sanguínea/efectos de los fármacos , Calcio , Línea Celular , Inhibición de Migración Celular , Complemento C3a/metabolismo , Complemento C5a/metabolismo , Relación Dosis-Respuesta a Droga , Liberación de Histamina/efectos de los fármacos , Humanos , Mastocitos/metabolismo , Ratones , Síndrome de Dificultad Respiratoria/prevención & control , Porcinos , Tromboxano B2/metabolismo
10.
Hum Immunol ; 82(4): 264-269, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33632561

RESUMEN

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.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Activación de Complemento/efectos de los fármacos , Complemento C3a/antagonistas & inhibidores , Complemento C5a/antagonistas & inhibidores , Inactivadores del Complemento/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , COVID-19/patología , COVID-19/fisiopatología , Activación de Complemento/inmunología , Complemento C3a/inmunología , Complemento C5a/inmunología , Humanos , SARS-CoV-2/inmunología
11.
J Pharmacol Sci ; 112(1): 56-63, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20051658

RESUMEN

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.


Asunto(s)
Anticuerpos/toxicidad , Arginina/análogos & derivados , Artritis Experimental/prevención & control , Compuestos de Bencidrilo/farmacología , Compuestos de Bencidrilo/uso terapéutico , Complemento C3a/antagonistas & inhibidores , Óvulo/inmunología , Animales , Arginina/farmacología , Arginina/uso terapéutico , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Complemento C3a/metabolismo , Masculino , Ratones , Ratones Endogámicos DBA , Ratas , Ratas Endogámicas Lew , Receptores de Complemento/antagonistas & inhibidores , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo
12.
J Am Soc Nephrol ; 20(3): 593-603, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19158354

RESUMEN

Tubulointerstitial inflammation and progressive fibrosis are common pathways that lead to kidney failure in proteinuric nephropathies. Activation of the complement system has been implicated in the development of tubulointerstitial injury in clinical and animal studies, but the mechanism by which complement induces kidney injury is not fully understood. Here, we studied the effect of complement on the phenotype of tubular epithelial cells. Tubular epithelial cells exposed to serum proteins adopted phenotypic and functional characteristics of mesenchymal cells. Expression of E-cadherin protein decreased and expression of both alpha-smooth muscle actin protein and collagen I mRNA increased. Exposure of the cells to the complement anaphylotoxin C3a induced similar features. Treating with a C3a receptor (C3aR) antagonist prevented both C3a- and serum-induced epithelial-to-mesenchymal transition. In the adriamycin-induced proteinuria model, C3aR-deficient mice demonstrated less injury, preserved renal function, and improved survival compared with wild-type mice. Furthermore, the kidneys of C3aR-deficient mice had significantly less interstitial collagen I and alpha-smooth muscle actin. In summary, the complement anaphylotoxin C3a is an important mediator of glomerular and tubulointerstitial injury and can induce tubular epithelial-to-mesenchymal transition.


Asunto(s)
Complemento C3a/metabolismo , Enfermedades Renales/inmunología , Proteinuria/inmunología , Animales , Arginina/análogos & derivados , Arginina/farmacología , Compuestos de Bencidrilo/farmacología , Línea Celular , Colágeno Tipo I/metabolismo , Activación de Complemento , Complemento C3a/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Epitelio/efectos de los fármacos , Epitelio/inmunología , Epitelio/patología , Femenino , Fibroblastos/patología , Humanos , Enfermedades Renales/patología , Túbulos Renales Proximales/inmunología , Túbulos Renales Proximales/lesiones , Túbulos Renales Proximales/patología , Antígeno de Macrófago-1/genética , Antígeno de Macrófago-1/metabolismo , Macrófagos/patología , Mesodermo/efectos de los fármacos , Mesodermo/inmunología , Mesodermo/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteinuria/patología , Transducción de Señal
13.
Inflammation ; 43(6): 2128-2136, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32617860

RESUMEN

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.


Asunto(s)
Caspasas/metabolismo , Colitis Ulcerosa/diagnóstico , Complemento C3a/antagonistas & inhibidores , Regulación de la Expresión Génica , Piroptosis , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Colitis Ulcerosa/metabolismo , Inflamación , Interleucina-6/metabolismo , Intestinos/patología , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Proteínas de Unión a Fosfato/biosíntesis , Pronóstico , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Factor de Necrosis Tumoral alfa/biosíntesis , Regulación hacia Arriba
14.
J Cereb Blood Flow Metab ; 28(5): 1048-58, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18197178

RESUMEN

The complement anaphylatoxin C3a contributes to injury after cerebral ischemia in mice. This study assesses the effect of C3a receptor antagonist (C3aRA) on leukocyte infiltration into the ischemic zone. Transient or permanent middle cerebral artery occlusion (MCAO) was induced in wild-type C57Bl/6 mice. Intraperitoneal C3aRA or vehicle was administered 45 mins before or 1 h after occlusion. Twenty-four hours after occlusion, we harvested brain tissue and purified inflammatory cells using flow cytometry. Soluble intercellular adhesion molecule (ICAM)-1 protein levels were assessed using enzyme-linked immunosorbent assays, and ICAM-1 and C3a receptor (C3aR) expression was confirmed via immunohistochemistry. In the transient MCAO model, animals receiving C3aRA showed smaller strokes, less upregulation of C3aR-positive granulocytes, and less ICAM-1 protein on endothelial cells than vehicle-treated animals; no significant differences in other inflammatory cell populations were observed. C3a receptor antagonist-treated and vehicle-treated animals showed no differences in stroke volume or inflammatory cell populations after permanent MCAO. These data suggest that blocking the binding of C3a to C3aR modulates tissue injury in reperfused stroke by inhibiting the recruitment of neutrophils to the ischemic zone. It further establishes antagonism of the C3a anaphylatoxin as a promising strategy for ameliorating injury after ischemia/reperfusion.


Asunto(s)
Arginina/análogos & derivados , Compuestos de Bencidrilo/farmacología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Complemento C3a/antagonistas & inhibidores , Granulocitos/patología , Receptores de Complemento/antagonistas & inhibidores , Anafilatoxinas/metabolismo , Animales , Arginina/farmacología , Encéfalo/patología , Isquemia Encefálica/patología , Complemento C3a/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Receptores de Complemento/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
15.
PLoS One ; 13(8): e0202594, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30133517

RESUMEN

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.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Traumatismos por Explosión/tratamiento farmacológico , Antígenos CD55/administración & dosificación , Proteína HMGB1/genética , Inflamación/tratamiento farmacológico , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/fisiopatología , Animales , Traumatismos por Explosión/genética , Traumatismos por Explosión/patología , Activación de Complemento/efectos de los fármacos , Complemento C3a/antagonistas & inhibidores , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/fisiopatología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/fisiopatología , FN-kappa B/genética , Presión/efectos adversos , Ratas , Ratas Sprague-Dawley
16.
Sci Rep ; 8(1): 9679, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29946065

RESUMEN

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.


Asunto(s)
Complemento C3a/metabolismo , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Anafilatoxinas/antagonistas & inhibidores , Anafilatoxinas/metabolismo , Arginina/análogos & derivados , Arginina/farmacología , Compuestos de Bencidrilo/farmacología , Células Cultivadas , Activación de Complemento/efectos de los fármacos , Complemento C3a/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Ensayo de Inmunoadsorción Enzimática , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos , Leupeptinas/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Epitelio Pigmentado de la Retina/efectos de los fármacos
17.
Sci Rep ; 8(1): 5165, 2018 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-29581525

RESUMEN

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.


Asunto(s)
Anemia de Diamond-Blackfan/inmunología , Anemia de Diamond-Blackfan/metabolismo , Inmunidad Innata/fisiología , Pez Cebra/inmunología , Pez Cebra/metabolismo , Receptores de Activinas/antagonistas & inhibidores , Activinas/metabolismo , Animales , Arginina/análogos & derivados , Arginina/farmacología , Benzamidas/farmacología , Compuestos de Bencidrilo/farmacología , Complemento C3a/antagonistas & inhibidores , Complemento C3a/metabolismo , Dioxoles/farmacología , Modelos Animales de Enfermedad , Humanos , Interferones/metabolismo , Células K562 , ARN Interferente Pequeño/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Proteínas Ribosómicas/metabolismo , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Pez Cebra/embriología , Proteínas de Pez Cebra/metabolismo
18.
Mol Immunol ; 93: 266-277, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28860090

RESUMEN

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.


Asunto(s)
Candida albicans/enzimología , Complemento C3/antagonistas & inhibidores , Proteínas Fúngicas/fisiología , Secuencia de Aminoácidos , Unión Competitiva , Señalización del Calcio/efectos de los fármacos , Candida albicans/efectos de los fármacos , Candida albicans/inmunología , Línea Celular , Complemento C3/inmunología , Complemento C3/metabolismo , Complemento C3/farmacología , Complemento C3a/antagonistas & inhibidores , Complemento C3a/farmacología , Complemento C3b/antagonistas & inhibidores , Complemento C3b/farmacología , Proteínas Fúngicas/antagonistas & inhibidores , Proteínas Fúngicas/farmacología , Células HEK293 , Humanos , Interleucina-8/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Proteínas Opsoninas/inmunología , Fragmentos de Péptidos/metabolismo , Fagocitosis/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Proteolisis , Receptores de Complemento/antagonistas & inhibidores , Receptores de Complemento/metabolismo , Virulencia/inmunología
19.
Nat Commun ; 8(1): 2078, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29233958

RESUMEN

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.


Asunto(s)
Complemento C3a/fisiología , Inflamación/inmunología , Monocitos/fisiología , Músculo Esquelético/fisiología , Receptores de Complemento/fisiología , Regeneración/inmunología , Animales , Trasplante de Médula Ósea , Cardiotoxinas/toxicidad , Movimiento Celular/fisiología , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimera/fisiología , Complemento C3a/antagonistas & inhibidores , Vía Alternativa del Complemento/fisiología , Modelos Animales de Enfermedad , Venenos Elapídicos/farmacología , Humanos , Macrófagos/fisiología , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/lesiones , Receptores de Complemento/deficiencia , Regeneración/efectos de los fármacos , Transducción de Señal/fisiología , Cicatrización de Heridas/fisiología
20.
Mol Immunol ; 42(1): 31-8, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15488941

RESUMEN

The feeding success of a tick upon a host depends on its ability to suppress host anti-tick responses which include activation of the complement system. We investigated the mechanism of inhibition of the alternative pathway of complement by salivary gland extract (SGE) of the ixodid tick species, Ixodes ricinus. SGE treatment strongly inhibited C3a generation and factor B cleavage in serum when rabbit erythrocytes were used as complement activator, but not when cobra venom factor (CVF) was used as an activator. SGE treatment strongly inhibited C3b deposition on rabbit erythrocytes, and the turnover of C3 (to C3b/iC3b) in serum. However, there was no significant effect upon the formation, stability or activity of C3 convertase (C3bBb) when formed from purified C3b, factor B and factor D. SGE treatment of isolated C3 resulted in a shift in mobility of the alpha-chain (by about 5 kDa). N-terminal sequencing of this species suggests that cleavage occurs at the C-terminus of the alpha-chain of C3. Consistent with this hypothesis, the modified alpha-chain was still a substrate for pre-formed convertase. The activity was specific for the alpha-chain of C3 but not of C3(H2O) nor the alpha'-chain of C3b. It is proposed that SGE-modified C3 does not participate in convertase formation, probably having a reduced affinity for factor B.


Asunto(s)
Extractos Celulares/farmacología , Activación de Complemento/efectos de los fármacos , Ixodes/inmunología , Glándulas Salivales/química , Animales , C3 Convertasa de la Vía Alternativa del Complemento , Complemento C3a/antagonistas & inhibidores , Complemento C3a/metabolismo , Complemento C3b/efectos de los fármacos , Complemento C3b/metabolismo , Factor B del Complemento/antagonistas & inhibidores , Factor B del Complemento/metabolismo , Eritrocitos/inmunología , Eritrocitos/parasitología , Humanos , Ixodes/patogenicidad , Fragmentos de Péptidos/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Conejos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda