Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Immunol ; 197(6): 2353-61, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27521340

RESUMEN

There is accumulating evidence during sepsis that cardiomyocyte (CM) homeostasis is compromised, resulting in cardiac dysfunction. An important role for complement in these outcomes is now demonstrated. Addition of C5a to electrically paced CMs caused prolonged elevations of intracellular Ca(2+) concentrations during diastole, together with the appearance of spontaneous Ca(2+) transients. In polymicrobial sepsis in mice, we found that three key homeostasis-regulating proteins in CMs were reduced: Na(+)/K(+)-ATPase, which is vital for effective action potentials in CMs, and two intracellular Ca(2+) concentration regulatory proteins, that is, sarcoplasmic/endoplasmic reticulum calcium ATPase 2 and the Na(+)/Ca(2+) exchanger. Sepsis caused reduced mRNA levels and reductions in protein concentrations in CMs for all three proteins. The absence of either C5a receptor mitigated sepsis-induced reductions in the three regulatory proteins. Absence of either C5a receptor (C5aR1 or C5aR2) diminished development of defective systolic and diastolic echocardiographic/Doppler parameters developing in the heart (cardiac output, left ventricular stroke volume, isovolumic relaxation, E' septal annulus, E/E' septal annulus, left ventricular diastolic volume). We also found in CMs from septic mice the presence of defective current densities for Ik1, l-type calcium channel, and Na(+)/Ca(2+) exchanger. These defects were accentuated in the copresence of C5a. These data suggest complement-related mechanisms responsible for development of cardiac dysfunction during sepsis.


Asunto(s)
Coinfección/inmunología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Sepsis/inmunología , Sepsis/fisiopatología , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/inmunología , Coinfección/microbiología , Coinfección/fisiopatología , Complemento C5a/inmunología , Citoplasma/química , Citoplasma/metabolismo , Corazón/fisiopatología , Ratones , Miocitos Cardíacos/microbiología , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/inmunología , Receptor de Anafilatoxina C5a/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/inmunología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Sepsis/complicaciones
2.
FASEB J ; 29(5): 2185-93, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25681459

RESUMEN

The purpose of this study was to define the relationship in polymicrobial sepsis (in adult male C57BL/6 mice) between heart dysfunction and the appearance in plasma of extracellular histones. Procedures included induction of sepsis by cecal ligation and puncture and measurement of heart function using echocardiogram/Doppler parameters. We assessed the ability of histones to cause disequilibrium in the redox status and intracellular [Ca(2+)]i levels in cardiomyocytes (CMs) (from mice and rats). We also studied the ability of histones to disturb both functional and electrical responses of hearts perfused with histones. Main findings revealed that extracellular histones appearing in septic plasma required C5a receptors, polymorphonuclear leukocytes (PMNs), and the Nacht-, LRR-, and PYD-domains-containing protein 3 (NLRP3) inflammasome. In vitro exposure of CMs to histones caused loss of homeostasis of the redox system and in [Ca(2+)]i, as well as defects in mitochondrial function. Perfusion of hearts with histones caused electrical and functional dysfunction. Finally, in vivo neutralization of histones in septic mice markedly reduced the parameters of heart dysfunction. Histones caused dysfunction in hearts during polymicrobial sepsis. These events could be attenuated by histone neutralization, suggesting that histones may be targets in the setting of sepsis to reduce cardiac dysfunction.


Asunto(s)
Cardiomiopatías/etiología , Modelos Animales de Enfermedad , Histonas/efectos adversos , Mitocondrias/patología , Sepsis/complicaciones , Animales , Calcio/metabolismo , Cardiomiopatías/sangre , Cardiomiopatías/diagnóstico , Proteínas Portadoras/fisiología , Caspasa 1/fisiología , Células Cultivadas , Histonas/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Sepsis/sangre , Sepsis/patología , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/fisiología
3.
J Immunol ; 193(11): 5668-77, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25348624

RESUMEN

Severe sepsis and septic shock are leading causes of morbidity and mortality worldwide. Infection-associated inflammation promotes the development and progression of adverse outcomes in sepsis. The effects of heterodimeric IL-27 (p28/EBI3) have been implicated in the natural course of sepsis, whereas the molecular mechanisms underlying the regulation of gene expression and release of IL-27 in sepsis are poorly understood. We studied the events regulating the p28 subunit of IL-27 in endotoxic shock and polymicrobial sepsis following cecal ligation and puncture. Neutralizing Abs to IL-27(p28) improved survival rates, restricted cytokine release, and reduced bacterial burden in C57BL/6 mice during sepsis. Genetic disruption of IL-27 signaling enhanced the respiratory burst of macrophages. Experiments using splenectomized mice or treatment with clodronate liposomes suggested that macrophages in the spleen may be a significant source of IL-27(p28) during sepsis. In cultures of TLR4-activated macrophages, the frequency of F4/80(+)CD11b(+)IL-27(p28)(+) cells was reduced by the addition of IL-10. IL-10 antagonized both MyD88-dependent and TRIF-dependent release of IL-27(p28). Genetic deletion of STAT3 in Tie2-Cre/STAT3flox macrophages completely interrupted the inhibition of IL-27(p28) by IL-10 after TLR4 activation. In contrast, IL-10 remained fully active to suppress IL-27(p28) with deletion of SOCS3 in Tie2-Cre/SOCS3flox macrophages. Blockade of IL-10R by Ab or genetic deficiency of IL-10 resulted in 3-5-fold higher concentrations of IL-27(p28) in endotoxic shock and polymicrobial sepsis. Our studies identify IL-10 as a critical suppressing factor for IL-27(p28) production during infection-associated inflammation. These findings may be helpful for a beneficial manipulation of adverse IL-27(p28) release during sepsis.


Asunto(s)
Interleucina-10/metabolismo , Interleucinas/metabolismo , Macrófagos/fisiología , Factor de Transcripción STAT3/metabolismo , Sepsis/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Anticuerpos Bloqueadores/administración & dosificación , Carga Bacteriana , Ciego/cirugía , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Interleucina-10/genética , Interleucinas/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Receptores de Citocinas/genética , Receptores de Interleucina , Factor de Transcripción STAT3/genética , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Receptor Toll-Like 4/inmunología
4.
Neurobiol Dis ; 67: 57-70, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24657919

RESUMEN

Proinflammatory mediators trigger intensive postischemic inflammatory remodeling of the blood-brain barrier (BBB) including extensive brain endothelial cell surface and junctional complex changes. Junctional adhesion molecule-A (JAM-A) is a component of the brain endothelial junctional complex with dual roles: paracellular route occlusion and regulating leukocyte docking and migration. The current study examined the contribution of JAM-A to the regulation of leukocyte (neutrophils and monocytes/macrophages) infiltration and the postischemic inflammatory response in brain ischemia/reperfusion (I/R injury). Brain I/R injury was induced by transient middle cerebral artery occlusion (MCAO) for 30min in mice followed by reperfusion for 0-5days, during which time JAM-A antagonist peptide (JAM-Ap) was administered. The peptide, which inhibits JAM-A/leukocyte interaction by blocking the interaction of the C2 domain of JAM-A with LFA on neutrophils and monocytes/macrophages, attenuated I/R-induced neutrophil and monocyte infiltration into brain parenchyma. Consequently, mice treated with JAM-A peptide during reperfusion had reduced expression (~3-fold) of inflammatory mediators in the ischemic penumbra, reduced infarct size (94±39 vs 211±38mm3) and significantly improved neurological score. BBB hyperpermeability was also reduced. Collectively, these results indicate that JAM-A has a prominent role in regulating leukocyte infiltration after brain I/R injury and could be a new target in limiting post-ischemic inflammation.


Asunto(s)
Isquemia Encefálica/metabolismo , Movimiento Celular , Encefalitis/metabolismo , Molécula A de Adhesión de Unión/antagonistas & inhibidores , Leucocitos/fisiología , Daño por Reperfusión/metabolismo , Animales , Encéfalo/metabolismo , Isquemia Encefálica/fisiopatología , Encefalitis/fisiopatología , Infarto de la Arteria Cerebral Media/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Daño por Reperfusión/fisiopatología
5.
J Innate Immun ; 6(5): 607-18, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24642449

RESUMEN

The main drivers of acute inflammation are macrophages, which are known to have receptors for catecholamines. Based on their function, macrophages are broadly categorized as having either M1 (proinflammatory) or M2 phenotypes (anti-inflammatory). In this study, we investigated catecholamine-induced alterations in the phenotype of activated macrophages. In the presence of lipopolysaccharide (LPS), mouse peritoneal macrophages acquired an M1 phenotype. However, the copresence of LPS and either epinephrine or norepinephrine resulted in a strong M2 phenotype including high levels of arginase-1 and interleukin-10, and a reduced expression of M1 markers. Furthermore, epinephrine enhanced macrophage phagocytosis and promoted type 2 T-cell responses in vitro, which are known features of M2 macrophages. Analysis of M2 subtype-specific markers indicated that LPS and catecholamine-cotreated macrophages were not alternatively activated but were rather of the regulatory macrophage subtype. Interestingly, catecholamines signaled through the ß2-adrenergic receptor but not the canonical cAMP/protein kinase A signaling pathway. Instead, the M2 pathway required an intact phosphoinositol 3-kinase pathway. Blockade of the ß2-adrenergic receptor reduced survival and enhanced injury in mouse models of endotoxemia and LPS-induced acute lung injury, respectively. These results demonstrate a role for the ß2-adrenergic receptor in promoting the M2 macrophage phenotype.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Endotoxemia/inmunología , Macrófagos Peritoneales/inmunología , Receptores Adrenérgicos beta 2/metabolismo , Células Th2/inmunología , Animales , Catecolaminas/metabolismo , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Epinefrina/metabolismo , Humanos , Lipopolisacáridos/inmunología , Activación de Linfocitos , Activación de Macrófagos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Norepinefrina/metabolismo , Fagocitosis , Fenotipo , Transducción de Señal
6.
FASEB J ; 27(12): 5010-21, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23982144

RESUMEN

We investigated how complement activation promotes tissue injury and organ dysfunction during acute inflammation. Three models of acute lung injury (ALI) induced by LPS, IgG immune complexes, or C5a were used in C57BL/6 mice, all models requiring availability of both C5a receptors (C5aR and C5L2) for full development of ALI. Ligation of C5aR and C5L2 with C5a triggered the appearance of histones (H3 and H4) in bronchoalveolar lavage fluid (BALF). BALF from humans with ALI contained H4 histone. Histones were absent in control BALF from healthy volunteers. In mice with ALI, in vivo neutralization of H4 with IgG antibody reduced the intensity of ALI. Neutrophil depletion in mice with ALI markedly reduced H4 presence in BALF and was highly protective. The direct lung damaging effects of extracellular histones were demonstrated by airway administration of histones into mice and rats (Sprague-Dawley), which resulted in ALI that was C5a receptor-independent, and associated with intense inflammation, PMN accumulation, damage/destruction of alveolar epithelial cells, together with release into lung of cytokines/chemokines. High-resolution magnetic resonance imaging demonstrated lung damage, edema and consolidation in histone-injured lungs. These studies confirm the destructive C5a-dependent effects in lung linked to appearance of extracellular histones.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Activación de Complemento , Complemento C5a/inmunología , Espacio Extracelular/metabolismo , Histonas/inmunología , Lesión Pulmonar Aguda/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/genética , Citocinas/metabolismo , Espacio Extracelular/inmunología , Histonas/metabolismo , Humanos , Inflamación/metabolismo , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Ratas , Ratas Sprague-Dawley , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo
7.
Eur J Immunol ; 43(7): 1907-13, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23575697

RESUMEN

The complement activation product, C5a, is a key factor for regulation of inflammatory responses. C5a and C5adesArg bind to their receptors, C5aR and C5L2, but the functional roles of C5L2 remain controversial. We screened the patterns of 23 inflammatory mediators in cultures of LPS-activated mouse peritoneal elicited macrophages (PEMs) in the presence or absence of recombinant mouse C5a. Production of most mediators studied was suppressed by C5a, whereas G-CSF production was enhanced. G-CSF gene expression and secretion from PEMs was amplified two- to threefold by C5a in a dose- and time-dependent fashion. The degradation product C5adesArg promoted lower levels of G-CSF. The effects of C5a on G-CSF were associated with activation of PI3K/Akt and MEK1/2 signaling pathways. C5a did not enhance G-CSF production in cultures of PEMs from either C5aR- or C5L2-deficient mice, indicating that both C5a receptors are indispensable for mediating the effects of C5a in the production of G-CSF. Finally, G-CSF levels in plasma during polymicrobial sepsis after cecal ligation and puncture were substantially lower in C5aR- or C5L2-deficient mice as compared with that in C57BL/6J WT mice. These findings elucidate the functional characteristics of the C5L2 receptor during the acute inflammatory response.


Asunto(s)
Complemento C5a/inmunología , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Inflamación/inmunología , Receptores de Quimiocina/inmunología , Receptores de Complemento/inmunología , Enfermedad Aguda , Animales , Complemento C5a/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Factor Estimulante de Colonias de Granulocitos/inmunología , Inflamación/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Anafilatoxina C5a , Receptores de Quimiocina/metabolismo , Receptores de Complemento/metabolismo , Sepsis/inmunología , Sepsis/metabolismo , Transducción de Señal/inmunología
8.
Shock ; 39(5): 447-52, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23481504

RESUMEN

Acute lung injury (ALI) is a severe pulmonary disease causing high numbers of fatalities worldwide. Innate immune responses are an integral part of the pathophysiologic events during ALI. Interleukin 23 (IL-23) is a proinflammatory mediator known to direct the inflammatory responses in various settings of infection, autoimmunity, and cancer. Interleukin 23 has been associated with proliferation and effector functions in T(H)17 cells. Surprisingly, little is known about production of IL-23 during ALI. In this study, we found expression of mRNA for IL-23p19 to be 10-fold elevated in lung homogenates of C57BL/6 mice after lipopolysaccharide (LPS)-induced ALI. Likewise, concentrations of IL-23 significantly increased in bronchoalveolar lavage fluids. Experiments with IL-23-deficient mice showed that endogenous IL-23 was required for production of IL-17A during LPS-ALI. CD11c-diphtheria toxin receptor transgenic mice were used to selectively deplete CD11c cells, the data suggesting that IL-23 production is dependent at least in part on CD11c cells during ALI. No alterations of IL-23 levels were observed in Rag-1-deficient mice as compared with wild-type C57BL/6 mice following ALI. The mouse alveolar macrophage cell line, MH-S, as well as primary alveolar macrophages displayed abundant surface expression of CD11c. Activation of these macrophages by LPS resulted in release of IL-23 in vitro. Our findings identify CD11c macrophages in the lung are likely an important source of IL-23 during ALI, which may be helpful for better understanding of this disease.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Antígeno CD11c/metabolismo , Interleucina-23/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos Alveolares/metabolismo , Animales , Masculino , Ratones
9.
J Immunol ; 190(8): 4215-25, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23479227

RESUMEN

During experimental sepsis, excessive generation of the anaphylatoxin C5a results in reduction of the C5a receptor (C5aR) on neutrophils. These events have been shown to result in impaired innate immunity. However, the regulation and fate of C5aR on neutrophils during sepsis are largely unknown. In contrast to 30 healthy volunteers, 60 patients in septic shock presented evidence of complement activation with significantly increased serum levels of C3a, C5a, and C5b-9. In the septic shock group, the corresponding decrease in complement hemolytic activity distinguished survivors from nonsurvivors. Neutrophils from patients in septic shock exhibited decreased C5aR expression, which inversely correlated with serum concentrations of C-reactive protein (CRP) and clinical outcome. In vitro exposure of normal neutrophils to native pentameric CRP led to a dose- and time-dependent loss of C5aR expression on neutrophils, whereas the monomeric form of CRP, as well as various other inflammatory mediators, failed to significantly alter C5aR levels on neutrophils. A circulating form of C5aR (cC5aR) was detected in serum by immunoblotting and a flow-based capture assay, suggestive of an intact C5aR molecule. Levels of cC5aR were significantly enhanced during septic shock, with serum levels directly correlating with lethality. The data suggest that septic shock in humans is associated with extensive complement activation, CRP-dependent loss of C5aR on neutrophils, and appearance of cC5aR in serum, which correlated with a poor outcome. Therefore, cC5aR may represent a new sepsis marker to be considered in tailoring individualized immune-modulating therapy.


Asunto(s)
Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de Complemento/sangre , Choque Séptico/sangre , Choque Séptico/inmunología , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Receptor de Anafilatoxina C5a , Receptores de Complemento/antagonistas & inhibidores , Choque Séptico/mortalidad , Sobrevida
10.
Am J Pathol ; 182(4): 1124-30, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23499051

RESUMEN

Severe sepsis is a life-threatening disease that causes major morbidity and mortality. Catecholamines and glucocorticoids often have been used for the treatment of sepsis. Several recent studies have suggested a potential role of IL-17 during the development and progression of sepsis in small animal models. In this study, the cross-talk of catecholamines and glucocorticoids with members of the IL-17 family was investigated during sepsis in C57BL/6 mice. The concentrations in plasma of IL-17A, IL-17F, and the IL-17AF heterodimer all were increased greatly in mice after endotoxemia or cecal ligation and puncture as compared with sham mice. Surprisingly, when compared with IL-17A (487 pg/mL), the concentrations of IL-17F (2361 pg/mL) and the heterodimer, IL-17AF (5116 pg/mL), were much higher 12 hours after endotoxemia. After surgical removal of the adrenal glands, mice had much higher mortality after endotoxemia or cecal ligation and puncture. The absence of endogenous adrenal gland hormones (cortical and medullary) was associated with 3- to 10-fold higher concentrations of IL-17A, IL-17F, IL-17AF, and IL-23. The addition of adrenaline, noradrenaline, hydrocortisone, or dexamethasone to lipopolysaccharide-activated peritoneal macrophages dose-dependently suppressed the expression and release of IL-17s. The production of IL-17s required activation of c-Jun-N-terminal kinase, which was antagonized by both catecholamines and glucocorticoids. These data provide novel insights into the molecular mechanisms of immune modulation by catecholamines and glucocorticoids during acute inflammation.


Asunto(s)
Catecolaminas/farmacología , Glucocorticoides/farmacología , Interleucina-17/metabolismo , Sepsis/metabolismo , Adrenalectomía , Animales , Endotoxemia/complicaciones , Endotoxemia/genética , Endotoxemia/metabolismo , Endotoxemia/patología , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-17/biosíntesis , Interleucina-17/sangre , Interleucina-17/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Sepsis/complicaciones , Sepsis/genética , Sepsis/patología
11.
Cell Host Microbe ; 13(2): 215-26, 2013 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-23414761

RESUMEN

Placental malaria (PM) is a major cause of fetal growth restriction, yet the underlying mechanism is unclear. Complement C5a and C5a receptor levels are increased with PM. C5a is implicated in fetal growth restriction in non-infection-based animal models. In a case-control study of 492 pregnant Malawian women, we find that elevated C5a levels are associated with an increased risk of delivering a small-for-gestational-age infant. C5a was significantly increased in PM and was negatively correlated with the angiogenic factor angiopoietin-1 and positively correlated with angiopoietin-2, soluble endoglin, and vascular endothelial growth factor. Genetic or pharmacological blockade of C5a or its receptor in a mouse model of PM resulted in greater fetoplacental vessel development, reduced placental vascular resistance, and improved fetal growth and survival. These data suggest that C5a drives fetal growth restriction in PM through dysregulation of angiogenic factors essential for placental vascular remodeling resulting in placental vascular insufficiency.


Asunto(s)
Activación de Complemento , Complemento C5a/metabolismo , Retardo del Crecimiento Fetal/patología , Regulación del Desarrollo de la Expresión Génica , Malaria/patología , Insuficiencia Placentaria/patología , Complicaciones Parasitarias del Embarazo/patología , Adolescente , Animales , Biomarcadores/metabolismo , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/metabolismo , Retardo del Crecimiento Fetal/parasitología , Edad Gestacional , Humanos , Malaria/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/metabolismo , Placenta/irrigación sanguínea , Placenta/parasitología , Insuficiencia Placentaria/metabolismo , Insuficiencia Placentaria/parasitología , Plasmodium berghei/patogenicidad , Embarazo , Receptor de Anafilatoxina C5a , Receptores de Complemento/metabolismo , Transducción de Señal , Transcripción Genética , Adulto Joven
12.
J Immunol ; 188(10): 5086-93, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22491257

RESUMEN

There is accumulating evidence that the complement activation product, C5a, can orchestrate cellular immune functions. IL-27(p28/EBI3) is an emerging key player essential for regulating inflammatory responses and T cells. In this article, we report that C5a robustly suppressed IL-27(p28) gene expression and release in peritoneal macrophages. These cells from C57BL/6J mice abundantly produced IL-27(p28) after engagement of either the TLR3 (polyinosinic-polycytidylic acid) or TLR4 (LPS) receptor. Genetic deficiency of either TLR4 or LBP completely incapacitated the ability of macrophages to secrete IL-27(p28) in response to LPS. IL-27(p28)-producing macrophages also expressed the C5aR receptor, thus displaying an IL-27(p28)(+)F4/80(+)C5aR(+) phenotype. C5a suppressed IL-27(p28) in LPS-stimulated macrophages via interactions with the C5aR receptor rather than the C5L2 receptor. After endotoxemia, C5aR(-/-) mice displayed higher plasma levels of IL-27(p28) compared with C57BL/6J mice. C5a did not affect the release of IL-27(p28) or the frequency of IL-27(p28)(+)F4/80(+) macrophages after engagement of TLR3. Mechanistically, LPS activated both the NF-κB and the PI3K/Akt pathways, whereas C5a activated only the PI3K/Akt pathway. Engagement of PI3K/Akt was inhibitory for IL-27(p28) production, because PI3K/Akt pharmacologic blockade resulted in increased amounts of IL-27(p28) and reversed the suppressive effects of C5a. Blockade of PI3K/Akt in endotoxemic C57BL/6J mice resulted in higher generation of IL-27(p28). In contrast, the PI3K/Akt pathway was not involved in TLR3-mediated release of IL-27(p28). These data provide new evidence about how complement activation may selectively interfere with production of T cell regulatory cytokines by APCs in the varying contexts of either bacterial (TLR4 pathway) or viral (TLR3 pathway) infection.


Asunto(s)
Activación de Complemento/inmunología , Complemento C5a/fisiología , Regulación hacia Abajo/inmunología , Interleucinas/antagonistas & inhibidores , Interleucinas/biosíntesis , Macrófagos Peritoneales/inmunología , Receptor Toll-Like 3/fisiología , Receptor Toll-Like 4/fisiología , Animales , Células Cultivadas , Interleucinas/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Anafilatoxina C5a , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/fisiología , Receptor Toll-Like 3/biosíntesis , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/deficiencia
13.
FASEB J ; 26(5): 2137-44, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22318967

RESUMEN

These studies were undertaken to extend emerging evidence that ß(2) adrenergic receptor (ß(2)AR) agonists, in addition to their bronchorelaxing effects, may have broad anti-inflammatory effects in the lung following onset of experimental acute lung injury (ALI). Young male C57BL/6 mice (25 g) developed ALI following airway deposition of bacterial LPS or IgG immune complexes in the absence or presence of appropriate stereoisomers (enantiomers) of ß(2)AR agonists, albuterol or formoterol. Endpoints included albumin leak into lung and buildup of polymorphonuclear neutrophils and cytokines/chemokines in bronchoalveolar fluids. Both ß(2)AR agonists suppressed lung inflammatory parameters (IC(50)=10(-7) M). Similar effects of ß(2)AR agonists on mediator release were found when mouse macrophages were stimulated in vitro with LPS. The protective effects were associated with reduced activation (phosphorylation) of JNK but not of other signaling proteins. Collectively, these data suggest that ß(2)AR agonists have broad anti-inflammatory effects in the setting of ALI. While ß(2)AR agonists suppress JNK activation, the extent to which this can explain the blunted lung inflammatory responses in the ALI models remains to be determined.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Agonistas Adrenérgicos beta/uso terapéutico , Antiinflamatorios/uso terapéutico , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Líquido del Lavado Bronquioalveolar , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
14.
FASEB J ; 26(4): 1640-51, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22202675

RESUMEN

There is growing evidence that the complement activation product C5a positively or negatively regulates inflammatory functions. The studies presented here report that C5a exerts anti-inflammatory effects by altering production of the cytokines IL-17A and IL-23 during endotoxic shock in young adult male C57BL/6J mice and has similar effects on macrophages from the same mice. IL-17A and IL-23 both appeared in plasma during endotoxemia, and their neutralization improved survival. The relevant sources of IL-17A during endotoxemia were not CD4(+) cells, γδ T cells, or NK cells but CD11b(+)F4/80(+) macrophages. The addition in vitro of C5a to lipopolysaccharide-activated peritoneal macrophages dose dependently antagonized the production of IL-17A (IC(50), 50-100 nM C5a) and IL-23 (IC(50), 10 nM C5a). This suppression required the receptor C5aR, but was independent of the second C5a receptor, C5L2. Genetic absence of C5aR was associated with much higher levels of IL-17A and IL-23 during endotoxic shock. Mechanistically, C5a mediated its effects on the IL-17A/IL-23 axis in a 2-step process. C5a caused activation of the PI3K-Akt and MEK1/2-ERK1/2 pathways, resulting in induction of IL-10, which powerfully inhibited production of IL-17A and IL-23. These data identify previously unknown mechanisms by which the anaphylatoxin C5a limits acute inflammation and antagonizes the IL-17A/IL-23 axis.


Asunto(s)
Antiinflamatorios/inmunología , Complemento C5a/inmunología , Inmunidad Innata/inmunología , Interleucina-17/inmunología , Interleucina-23/inmunología , Animales , Antiinflamatorios/farmacología , Antígenos de Diferenciación/inmunología , Antígeno CD11b/inmunología , Complemento C5a/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Interleucina-10/inmunología , Subunidad p19 de la Interleucina-23/inmunología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , MAP Quinasa Quinasa 1/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología
15.
Cell Health Cytoskelet ; 4: 73-82, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23576881

RESUMEN

Complement activation usually results in the formation of complement fragment 5a (C5a) that interacts with its two receptors, C5aR and C5L2. These receptors belong to the rhodopsin family of G protein-coupled seven transmembrane-containing receptors. C5aR and C5L2 are expressed on/in a wide variety of cells and tissues. Interaction of C5a with C5aR leads to many pleiotropic effects, including the release of cytokines and chemokines and recruitment of inflammatory cells. In certain circumstances, C5a-C5aR interactions can also result in pathophysiological changes as seen in sepsis, rheumatoid arthritis, asthma, acute lung injury and ischemia-reperfusion injury. This overview of the C5a-C5aR interactions describes how such interactions facilitate the pivotal role the complement system plays in the host's innate and adaptive responses.

16.
FASEB J ; 25(12): 4292-300, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21859893

RESUMEN

Growing evidence suggests that transcription factor signal transducer and activator of transcription (Stat) 3 may play an important regulatory role during inflammation. However, the function of Stat3 in acute lung injury (ALI) is largely unknown. In the current study, by using an adenoviral vector expressing a dominant-negative Stat3 isoform (Ad-Stat3-EVA), we determined the role of Stat3 in IgG immune complex (IC)-induced inflammatory responses and injury in the lung from C57BL/6J mice. We show that IgG IC-induced DNA binding activity of Stat3 in the lung was significantly inhibited by Stat3-EVA. We demonstrate that both lung vascular permeability (albumin leak) and lung myeloperoxidase accumulation in the Ad-Stat-EVA treated mice were substantially reduced when compared with values in mice receiving control virus (Ad-GFP) during the injury. Furthermore, intratracheal administration of Ad-Stat3-EVA caused significant decreases in the contents of neutrophils, inflammatory cytokines (TNF-α and IL-6), chemokines [keratinocyte cell-derived chemokine, macrophage inflammatory protein (MIP)-1α, and MIP-1ß], and complement component C5a in bronchoalveolar lavage fluids. Using Stat3-specific small interfering RNA, we show that knocking down Stat3 expression in alveolar macrophages (MH-S cells) significantly reduced the production of proinflammatory mediators on IgG IC stimulation. These data suggest that Stat3 plays an essential role in the pathogenesis of IgG IC-induced ALI by mediating the acute inflammatory responses in the lung and alveolar macrophages.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Inmunoglobulina G/metabolismo , Neumonía/inmunología , Neumonía/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Secuencia de Bases , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Quimiocinas/metabolismo , Citocinas/metabolismo , ADN/metabolismo , Técnicas de Silenciamiento del Gen , Mediadores de Inflamación/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética
17.
FASEB J ; 25(12): 4222-32, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21859896

RESUMEN

The interleukin-17 (IL-17) family of cytokines plays important roles in innate immune defenses against bacterial and fungal pathogens. While much is known about IL-17A, much less information is available about the IL-17F isoform. Here, we investigated gene expression and release of IL-17F and its regulation by the complement system. IL-17F was produced in mouse peritoneal elicited macrophages after TLR4 activation by LPS, peaking after 12 h. This effect was completely dependent on the presence of the adaptor protein MyD88. The copresence of the complement activation product, C5a (EC(50)=10 nM), amplified IL-17F production via the receptor C5aR. In vitro signaling studies indicated that LPS or C5a, or the combination, caused phosphorylation of Akt occurring at threonine 308 but not at serine 473. Treatment of macrophages with pharmacologic inhibitors of PI3K-Akt greatly reduced production of IL-17F as well as mRNA for IL-17F. In endotoxemia, C5a levels peaked at 6 h, while IL-17F levels peaked between 6-12 h. Full in vivo production of IL-17F during endotoxemia required C5a. A similar result was found in the cecal ligation and puncture sepsis model. These data suggest that maximal production of IL-17F requires complement activation and presence of C5a.


Asunto(s)
Complemento C5a/metabolismo , Interleucina-17/biosíntesis , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Activación de Complemento , Endotoxemia/inmunología , Endotoxemia/metabolismo , Inmunidad Innata , Interleucina-17/genética , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo
18.
Shock ; 36(4): 396-401, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21701414

RESUMEN

The contribution of the adaptive and innate immune systems to the pathogenesis and outcome of sepsis remains a fundamental yet controversial question. Here, we use mice lacking the recombination activating gene 1 (Rag-1) to study the role of T and B cells in sepsis after cecal ligation and puncture (CLP). Spleens of Rag-1 mice were atrophic and completely devoid of CD3 T cells and CD19 B cells. Wild-type mice and Rag-1 mice (both on a C57BL/6J background) underwent CLP or sham surgery. Both wild-type and Rag-1 mice developed clinical signs of sepsis within the first day after CLP. This included severe hypothermia as measured by a decrease in body surface temperature and organ dysfunction as detected by plasma increases in blood urea nitrogen and lactate dehydrogenase levels. Survival curves of wild-type and Rag-1 mice after CLP were superimposable, with 35% survival in the wild-type group and 27% survival in the Rag-1 group, respectively (not significant, P = 0.875). Using multiplex bead-based assays, the mediator concentrations for 23 cytokines and chemokines were measured in plasma of wild-type and Rag-1 mice 8 h after CLP or sham surgery. Compared with sham surgery mice, the highest mediator levels were observed for granulocyte colony-stimulating factor, keratinocyte chemoattractant, IL-6, monocyte chemotactic protein 1, and IL-10. Levels for most mediators were unaffected by the absence of T and B lymphocytes. Only the concentrations of IL-6 and IL-17 were found to be significantly lower in Rag-1 mice compared with wild-type mice. In conclusion, the absence of T and B cells in the CLP model used does not appear to affect the acute outcome of severe sepsis.


Asunto(s)
Linfocitos B/inmunología , Sepsis/inmunología , Sepsis/microbiología , Linfocitos T/inmunología , Animales , Antígenos CD19/metabolismo , Complejo CD3/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Sepsis/metabolismo
19.
FASEB J ; 25(7): 2500-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21478262

RESUMEN

We have recently shown that antibody-induced blockade of C5a, C5a receptors, or IL-17A greatly reduced the harmful outcomes of sepsis. In the current study, normal cardiomyocytes from young (300 g) male Sprague-Dawley rats responded in vitro to C5a (ED(50)=55 nM) with release of IL-6 and TNFα, peaking between 2 to 8 h. Neutralizing antibodies to mouse C5a or IL-17A (ED(50)=40 µg for each, based on improved survival) reduced spontaneous in vitro release of cardiosuppressive cytokines and chemokines in cardiomyocytes obtained from mice with polymicrobial sepsis. A non-neutralizing C5a antibody had no such effects. Cardiomyocytes from septic mice (C57Bl/6) showed increased mRNA for TNFR1, IL-6 (gp80), and C5aR at 6 h after sepsis. Cardiomyocytes from septic C5aR(-/-) or C5L2(-/-) mice did not show spontaneous in vitro release of cytokines and chemokines. These data suggest that cardiomyocytes from septic mice release suppressive cytokines in a C5a-, C5aR-, and IL-17A-dependent manner, followed by mediator reactivity with receptors on cardiomyocytes, resulting in defective contractility and relaxation. These data may be relevant to a strategy for the treatment of heart dysfunction developing during sepsis.


Asunto(s)
Complemento C5a/metabolismo , Citocinas/metabolismo , Miocitos Cardíacos/metabolismo , Sepsis/metabolismo , Animales , Quimiocina CCL3/sangre , Quimiocina CCL3/metabolismo , Quimiocina CXCL2/sangre , Quimiocina CXCL2/metabolismo , Quimiocinas/sangre , Quimiocinas/metabolismo , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Interleucina-17/metabolismo , Interleucina-6/sangre , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sepsis/genética , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/metabolismo
20.
Cell Tissue Res ; 343(1): 227-35, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20838815

RESUMEN

The complement system consists of a tightly regulated network of proteins that play an important role in host defense and inflammation. Complement activation results in opsonization of pathogens and their removal by phagocytes, as well as cell lysis. Inappropriate complement activation and complement deficiencies are the underlying cause of the pathophysiology of many diseases such as systemic lupus erythematosus and asthma. This review represents an overview of the complement system in an effort to understand the beneficial as well as harmful roles it plays during inflammatory responses.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Inmunidad Adaptativa/inmunología , Anafilatoxinas/inmunología , Animales , Activación de Complemento/inmunología , Humanos , Evasión Inmune/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA