Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
J Immunol ; 205(1): 251-260, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32444389

RESUMEN

Over the first days of polymicrobial sepsis, there is robust activation of the innate immune system, causing the appearance of proinflammatory cytokines and chemokines, along with the appearance of extracellular histones, which are highly proinflammatory and prothrombotic. In the current study, we studied different innate immune responses in mice with knockout (KO) of complement protein 6 (C6). Polymorphonuclear neutrophils (PMNs) from these KO mice had defective innate immune responses, including defective expression of surface adhesion molecules, generation of superoxide anion, and appearance of reactive oxygen species and histone release after activation of PMNs, along with defective phagocytosis. In addition, in C6-/- mice, the NLRP3 inflammasome was defective both in PMNs and in macrophages. When these KO mice were subjected to polymicrobial sepsis, their survival was improved, associated with reduced levels in the plasma of proinflammatory cytokines and chemokines and lower levels of histones in plasma. In addition, sepsis-induced cardiac dysfunction was attenuated in these KO mice. In a model of acute lung injury induced by LPS, C6-/- mice showed reduced PMN buildup and less lung epithelial/endothelial cell dysfunction (edema and hemorrhage). These data indicate that C6-/- mice have reduced innate immune responses that result in less organ injury and improved survival after polymicrobial sepsis.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Cardiomiopatías/inmunología , Coinfección/inmunología , Complemento C6/metabolismo , Inmunidad Innata , Sepsis/inmunología , Lesión Pulmonar Aguda/diagnóstico , Lesión Pulmonar Aguda/patología , Animales , Cardiomiopatías/diagnóstico , Cardiomiopatías/patología , Coinfección/complicaciones , Coinfección/diagnóstico , Coinfección/patología , Complemento C6/genética , Modelos Animales de Enfermedad , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Sepsis/complicaciones , Sepsis/diagnóstico , Sepsis/genética , Índice de Severidad de la Enfermedad
2.
FASEB J ; 31(9): 4129-4139, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28572445

RESUMEN

Polymicrobial sepsis in mice causes myocardial dysfunction after generation of the complement anaphylatoxin, complement component 5a (C5a). C5a interacts with its receptors on cardiomyocytes (CMs), resulting in redox imbalance and cardiac dysfunction that can be functionally measured and quantitated using Doppler echocardiography. In this report we have evaluated activation of MAPKs and Akt in CMs exposed to C5a in vitro and after cecal ligation and puncture (CLP) in vivo In both cases, C5a in vitro caused activation (phosphorylation) of MAPKs and Akt in CMs, which required availability of both C5a receptors. Using immunofluorescence technology, activation of MAPKs and Akt occurred in left ventricular (LV) CMs, requiring both C5a receptors, C5aR1 and -2. Use of a water-soluble p38 inhibitor curtailed activation in vivo of MAPKs and Akt in LV CMs as well as the appearance of cytokines and histones in plasma from CLP mice. When mouse macrophages were exposed in vitro to LPS, activation of MAPKs and Akt also occurred. The copresence of the p38 inhibitor blocked these activation responses. Finally, the presence of the p38 inhibitor in CLP mice reduced the development of cardiac dysfunction. These data suggest that polymicrobial sepsis causes cardiac dysfunction that appears to be linked to activation of MAPKs and Akt in heart.-Fattahi, F., Kalbitz, M., Malan, E. A., Abe, E., Jajou, L., Huber-Lang, M. S., Bosmann, M., Russell, M. W., Zetoune, F. S., Ward, P. A. Complement-induced activation of MAPKs and Akt during sepsis: role in cardiac dysfunction.


Asunto(s)
Complemento C5a/metabolismo , Regulación de la Expresión Génica/fisiología , Cardiopatías/etiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sepsis/metabolismo , Animales , Complemento C5a/genética , Cardiopatías/metabolismo , Interleucinas , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Ratas Sprague-Dawley , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo
3.
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
4.
FASEB J ; 30(12): 3997-4006, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27543123

RESUMEN

Cardiac dysfunction develops during sepsis in humans and rodents. In the model of polymicrobial sepsis induced by cecal ligation and puncture (CLP), we investigated the role of the NLRP3 inflammasome in the heart. Mouse heart homogenates from sham-procedure mice contained high mRNA levels of NLRP3 and IL-1ß. Using the inflammasome protocol, exposure of cardiomyocytes (CMs) to LPS followed by ATP or nigericin caused release of mature IL-1ß. Immunostaining of left ventricular frozen sections before and 8 h after CLP revealed the presence of NLRP3 and IL-1ß proteins in CMs. CLP caused substantial increases in mRNAs for IL-1ß and NLRP3 in CMs which are reduced in the absence of either C5aR1 or C5aR2. After CLP, NLRP3-/- mice showed reduced plasma levels of IL-1ß and IL-6. In vitro exposure of wild-type CMs to recombinant C5a (rC5a) caused elevations in both cytosolic and nuclear/mitochondrial reactive oxygen species (ROS), which were C5a-receptor dependent. Use of a selective NOX2 inhibitor prevented increased cytosolic and nuclear/mitochondrial ROS levels and release of IL-1ß. Finally, NLRP3-/- mice had reduced defects in echo/Doppler parameters in heart after CLP. These studies establish that the NLRP3 inflammasome contributes to the cardiomyopathy of polymicrobial sepsis.-Kalbitz, M., Fattahi, F., Grailer, J. J., Jajou, L., Malan, E. A., Zetoune, F. S., Huber-Lang, M., Russell, M. W., Ward, P. A. Complement-induced activation of the cardiac NLRP3 inflammasome in sepsis.


Asunto(s)
Complemento C5a/metabolismo , Inflamasomas/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Sepsis/metabolismo , Animales , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Ratones , Ratas , Especies Reactivas de Oxígeno/metabolismo
5.
J Immunol ; 194(3): 868-72, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25539817

RESUMEN

In the early stages of sepsis, lymphocytes undergo apoptosis, resulting in lymphopenia and immunosuppression. The trigger for septic lymphopenia is unknown. Using the polymicrobial model of murine sepsis, we investigated the role of C5a receptors in septic lymphopenia. In wild-type mice, cecal ligation and puncture resulted in splenocyte apoptosis and significant lymphopenia after 3 d, which was not observed in C5aR1(-/-) or C5aR2(-/-) mice. Our data show that mouse neutrophils exposed to recombinant mouse C5a cause release of histones in a dose-dependent and time-dependent manner. Histone levels in spleen were significantly elevated following cecal ligation and puncture but were reduced by the absence of C5aR1. Histones induced significant lymphocyte apoptosis in vitro. Ab-mediated neutralization of histones prevented the development of lymphopenia in sepsis. Together, these results describe a new pathway of septic lymphopenia involving complement and extracellular histones. Targeting of this pathway may have therapeutic benefit for patients with sepsis or other serious illness.


Asunto(s)
Linfopenia/etiología , Linfopenia/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Sepsis/complicaciones , Animales , Apoptosis , Complemento C5a/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Espacio Extracelular , Histonas/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Ratones , Ratones Noqueados , Receptor de Anafilatoxina C5a/genética
6.
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
7.
J Immunol ; 192(12): 5974-83, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24795455

RESUMEN

The inflammasome is a key factor in innate immunity and senses soluble pathogen and danger-associated molecular patterns as well as biological crystals (urate, cholesterol, etc.), resulting in expression of IL-1ß and IL-18. Using a standard model of acute lung injury (ALI) in mice featuring airway instillation of LPS, ALI was dependent on availability of NLRP3 as well as caspase-1, which are known features of the NLRP3 inflammasome. The appearance of IL-1ß, a product of NLRP3 inflammasome activation, was detected in bronchoalveolar lavage fluids (BALF) in a macrophage- and neutrophil-dependent manner. Neutrophil-derived extracellular histones appeared in the BALF during ALI and directly activated the NLRP3 inflammasome. Ab-mediated neutralization of histones significantly reduced IL-1ß levels in BALF during ALI. Inflammasome activation by extracellular histones in LPS-primed macrophages required NLRP3 and caspase-1 as well as extrusion of K(+), increased intracellular Ca(2+) concentration, and generation of reactive oxygen species. NLRP3 and caspase-1 were also required for full extracellular histone presence during ALI, suggesting a positive feedback mechanism. Extracellular histone and IL-1ß levels in BALF were also elevated in C5a-induced and IgG immune complex ALI models, suggesting a common inflammatory mechanism. These data indicate an interaction between extracellular histones and the NLRP3 inflammasome, resulting in ALI. Such findings suggest novel targets for treatment of ALI, for which there is currently no known efficacious drug.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Proteínas Portadoras/inmunología , Inflamasomas/inmunología , Macrófagos Alveolares/inmunología , Neutrófilos/inmunología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Proteínas Portadoras/genética , Caspasa 1/genética , Caspasa 1/inmunología , Modelos Animales de Enfermedad , Histonas/genética , Histonas/inmunología , Inflamasomas/genética , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Lipopolisacáridos/farmacología , Macrófagos Alveolares/patología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Neutrófilos/patología
8.
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
9.
Mediators Inflamm ; 2016: 1340156, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27382187

RESUMEN

C5a is an inflammatory mediator generated by complement activation that positively regulates various arms of immune defense, including Toll-like receptor 4 (TLR4) signaling. The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome is activated by pathogen products and cellular/tissue damage products and is a major contributor of IL-1ß. In this study, we investigate whether C5a modulates lipopolysaccharide- (LPS-) induced NLRP3 inflammasome activation in myeloid cells. Appearance of plasma IL-1ß during endotoxemia was reduced in C5aR1(-/-) mice when compared to wild-type mice. In vitro, C5a significantly enhanced LPS-induced production of IL-1ß in bone marrow Ly6C-high inflammatory monocytes, accompanied by augmented intracellular pro-IL-1ß expression. This effect was abolished during p38 blockade by SB 203580 and in the absence of C5aR1. Conversely, C5a suppressed LPS-induced macrophage production of IL-1ß, which was accompanied by attenuated levels of pro-IL-1ß, NLRP3, and caspase-1 expression. C5a's suppressive effects were negated during phosphoinositide 3-kinase (PI3K) inhibition by wortmannin but were largely preserved in the absence of C5aR1. Thus, C5a bidirectionally amplifies TLR4-mediated NLRP3 inflammasome activation in monocytes while suppressing this pathway in macrophages. However, as C5aR1 deficiency attenuates the IL-1ß response to LPS challenge in vivo, our results suggest overall that C5a augments physiologic inflammasome responses.


Asunto(s)
Inflamasomas/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Animales , Western Blotting , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Complemento C5a/metabolismo , Complemento C5a/farmacología , Modelos Animales de Enfermedad , Endotoxemia/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inflamasomas/efectos de los fármacos , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
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
11.
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
12.
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
13.
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
14.
Am J Physiol Lung Cell Mol Physiol ; 304(12): L863-72, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23564505

RESUMEN

Zonulin is a protein involved in the regulation of tight junctions (TJ) in epithelial or endothelial cells. Zonulin is known to affect TJ in gut epithelial cells, but little is known about its influences in other organs. Prehaptoglobin2 has been identified as zonulin and is related to serine proteases (MASPs, C1qrs) that activate the complement system. The current study focused on the role of zonulin in development of acute lung injury (ALI) in C57BL/6 male mice following intrapulmonary deposition of IgG immune complexes. A zonulin antagonist (AT-1001) and a related peptide with permeability agonist activities (AT-1002) were employed and given intratracheally or intravenously. Also, zonulin was blocked in lung with a neutralizing antibody. In a dose-dependent manner, AT-1001 or zonulin neutralizing antibody attenuated the intensity of ALI (as quantitated by albumin leak, neutrophil accumulation, and proinflammatory cytokines). A similar pattern was found using the bacterial lipopolysaccharide model of ALI. Using confocal microscopy on sections of injured lungs, staining patterns for TJ proteins were discontinuous, reduced, and fragmented. As expected, the leak of blood products into the alveolar space confirmed the passage of 3 and 20 kDa dextran, and albumin. In contrast to AT-1001, application of the zonulin agonist AT-1002 intensified ALI. Zonulin both in vitro and in vivo induced generation of complement C3a and C5a. Collectively, these data suggest that zonulin facilitates development of ALI both by enhancing albumin leak and complement activation as well as increased buildup of neutrophils and cytokines during development of ALI.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Toxina del Cólera/genética , Proteínas del Sistema Complemento/agonistas , Precursores de Proteínas/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Complejo Antígeno-Anticuerpo/farmacología , Toxina del Cólera/agonistas , Toxina del Cólera/antagonistas & inhibidores , Toxina del Cólera/inmunología , Activación de Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/inmunología , Citocinas/biosíntesis , Citocinas/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Haptoglobinas , Inmunoglobulina G/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/patología , Oligopéptidos/farmacología , Péptidos/farmacología , Permeabilidad/efectos de los fármacos , Precursores de Proteínas/agonistas , Precursores de Proteínas/antagonistas & inhibidores , Precursores de Proteínas/inmunología , Transducción de Señal/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/inmunología , Uniones Estrechas/patología , Tráquea/efectos de los fármacos , Tráquea/inmunología , Tráquea/patología
15.
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
16.
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
17.
Nat Med ; 12(6): 682-7, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16715088

RESUMEN

Complement-mediated tissue injury in humans occurs upon deposition of immune complexes, such as in autoimmune diseases and acute respiratory distress syndrome. Acute lung inflammatory injury in wild-type and C3-/- mice after deposition of IgG immune complexes was of equivalent intensity and was C5a dependent, but injury was greatly attenuated in Hc-/- mice (Hc encodes C5). Injury in lungs of C3-/- mice and C5a levels in bronchoalveolar lavage (BAL) fluids from these mice were greatly reduced in the presence of antithrombin III (ATIII) or hirudin but were not reduced in similarly treated C3+/+ mice. Plasma from C3-/- mice contained threefold higher levels of thrombin activity compared to plasma from C3+/+ mice. There were higher levels of F2 mRNA (encoding prothrombin) as well as prothrombin and thrombin protein in liver of C3-/- mice compared to C3+/+ mice. A potent solid-phase C5 convertase was generated using plasma from either C3+/+ or C3-/- mice. Human C5 incubated with thrombin generated C5a that was biologically active. These data suggest that, in the genetic absence of C3, thrombin substitutes for the C3-dependent C5 convertase. This linkage between the complement and coagulation pathways may represent a new pathway of complement activation.


Asunto(s)
Activación de Complemento/fisiología , Complemento C3/inmunología , Complemento C5a/inmunología , Animales , Complejo Antígeno-Anticuerpo , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Complemento C3/genética , Complemento C5a/genética , Humanos , Inmunoglobulina G/inmunología , Hígado/citología , Hígado/metabolismo , Pulmón/inmunología , Pulmón/patología , Lesión Pulmonar , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Protrombina/genética , Protrombina/metabolismo , Trombina/metabolismo
18.
Nature ; 449(7163): 721-5, 2007 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-17914358

RESUMEN

It is becoming increasingly clear that the autonomic nervous system and the immune system demonstrate cross-talk during inflammation by means of sympathetic and parasympathetic pathways. We investigated whether phagocytes are capable of de novo production of catecholamines, suggesting an autocrine/paracrine self-regulatory mechanism by catecholamines during inflammation, as has been described for lymphocytes. Here we show that exposure of phagocytes to lipopolysaccharide led to a release of catecholamines and an induction of catecholamine-generating and degrading enzymes, indicating the presence of the complete intracellular machinery for the generation, release and inactivation of catecholamines. To assess the importance of these findings in vivo, we chose two models of acute lung injury. Blockade of alpha2-adrenoreceptors or catecholamine-generating enzymes greatly suppressed lung inflammation, whereas the opposite was the case either for an alpha2-adrenoreceptor agonist or for inhibition of catecholamine-degrading enzymes. We were able to exclude T cells or sympathetic nerve endings as sources of the injury-modulating catecholamines. Our studies identify phagocytes as a new source of catecholamines, which enhance the inflammatory response.


Asunto(s)
Catecolaminas/metabolismo , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Fagocitos/metabolismo , Enfermedad Aguda , Antagonistas de Receptores Adrenérgicos alfa 2 , Animales , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fagocitos/efectos de los fármacos , Fagocitos/enzimología , Ratas , Ratas Long-Evans , Receptores Adrenérgicos alfa 2/metabolismo , Linfocitos T/metabolismo
19.
Shock ; 59(2): 311-317, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36377404

RESUMEN

ABSTRACT: This report deals with the advances made in the areas of complement and its role in sepsis, both in mice and in humans. The study relates to work over the past 25 years (late 1990s to October 2022). During this period, there has been very rapid progress in understanding the activation pathways of complement and the activation products of complement, especially the anaphylatoxin C5a and its receptors, C5aR1 and C5aR2. Much has also been learned about these pathways of activation that trigger activation of the innate immune system and the array of strong proinflammatory cytokines that can cause cell and organ dysfunction, as well as complement products that cause immunosuppression. The work in septic humans and mice, along with patients who develop lung dysfunction caused by COVID-19, has taught us that there are many strategies for treatment of humans who are septic or develop COVID-19-related lung dysfunction. To date, treatments in humans with these disorders suggest that we are in the midst of a new and exciting area related to the complement system.


Asunto(s)
COVID-19 , Sepsis , Humanos , Ratones , Animales , Citocinas , Proteínas del Sistema Complemento , Complemento C5a/metabolismo , Receptor de Anafilatoxina C5a
20.
J Exp Med ; 203(1): 53-61, 2006 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-16380509

RESUMEN

Defective cardiac function during sepsis has been referred to as "cardiomyopathy of sepsis." It is known that sepsis leads to intensive activation of the complement system. In the current study, cardiac function and cardiomyocyte contractility have been evaluated in rats after cecal ligation and puncture (CLP). Significant reductions in left ventricular pressures occurred in vivo and in cardiomyocyte contractility in vitro. These defects were prevented in CLP rats given blocking antibody to C5a. Both mRNA and protein for the C5a receptor (C5aR) were constitutively expressed on cardiomyocytes; both increased as a function of time after CLP. In vitro addition of recombinant rat C5a induced dramatic contractile dysfunction in both sham and CLP cardiomyocytes, but to a consistently greater degree in cells from CLP animals. These data suggest that CLP induces C5aR on cardiomyocytes and that in vivo generation of C5a causes C5a-C5aR interaction, causing dysfunction of cardiomyocytes, resulting in compromise of cardiac performance.


Asunto(s)
Cardiomiopatías/etiología , Complemento C5a/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Sepsis/complicaciones , Animales , Anticuerpos/farmacología , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica , Técnicas In Vitro , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , ARN Mensajero/metabolismo , Ratas , Receptor de Anafilatoxina C5a/genética , Sarcómeros/fisiología , Sepsis/metabolismo , Sepsis/fisiopatología , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA