Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(4): 1410-5, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23297238

RESUMEN

Appropriate control of immune responses is a critical determinant of health. Here, we show that choline acetyltransferase (ChAT) is expressed and ACh is produced by B cells and other immune cells that have an impact on innate immunity. ChAT expression occurs in mucosal-associated lymph tissue, subsequent to microbial colonization, and is reduced by antibiotic treatment. MyD88-dependent Toll-like receptor up-regulates ChAT in a transient manner. Unlike the previously described CD4(+) T-cell population that is stimulated by norepinephrine to release ACh, ChAT(+) B cells release ACh after stimulation with sulfated cholecystokinin but not norepinephrine. ACh-producing B-cells reduce peritoneal neutrophil recruitment during sterile endotoxemia independent of the vagus nerve, without affecting innate immune cell activation. Endothelial cells treated with ACh in vitro reduced endothelial cell adhesion molecule expression in a muscarinic receptor-dependent manner. Despite this ability, ChAT(+) B cells were unable to suppress effector T-cell function in vivo. Therefore, ACh produced by lymphocytes has specific functions, with ChAT(+) B cells controlling the local recruitment of neutrophils.


Asunto(s)
Acetilcolina/biosíntesis , Inmunidad Adaptativa/fisiología , Inmunidad Innata/fisiología , Linfocitos/inmunología , Linfocitos/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Tejido Linfoide/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Metagenoma/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Neuroinmunomodulación , Embarazo , Receptores de Neurotransmisores/inmunología , Receptores de Neurotransmisores/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo
2.
Immunol Rev ; 248(1): 188-204, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22725962

RESUMEN

Neural reflex circuits regulate cytokine release to prevent potentially damaging inflammation and maintain homeostasis. In the inflammatory reflex, sensory input elicited by infection or injury travels through the afferent vagus nerve to integrative regions in the brainstem, and efferent nerves carry outbound signals that terminate in the spleen and other tissues. Neurotransmitters from peripheral autonomic nerves subsequently promote acetylcholine-release from a subset of CD4(+) T cells that relay the neural signal to other immune cells, e.g. through activation of α7 nicotinic acetylcholine receptors on macrophages. Here, we review recent progress in the understanding of the inflammatory reflex and discuss potential therapeutic implications of current findings in this evolving field.


Asunto(s)
Inflamación/inmunología , Inflamación/metabolismo , Neuroinmunomodulación , Reflejo/inmunología , Acetilcolina/metabolismo , Animales , Colina O-Acetiltransferasa/metabolismo , Humanos , Inflamación/terapia , Reflejo/fisiología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Nervio Vago/inmunología
3.
Brain Behav Immun ; 44: 19-27, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25063706

RESUMEN

Inflammatory conditions characterized by excessive immune cell activation and cytokine release, are associated with bidirectional immune system-brain communication, underlying sickness behavior and other physiological responses. The vagus nerve has an important role in this communication by conveying sensory information to the brain, and brain-derived immunoregulatory signals that suppress peripheral cytokine levels and inflammation. Brain muscarinic acetylcholine receptor (mAChR)-mediated cholinergic signaling has been implicated in this regulation. However, the possibility of controlling inflammation by peripheral administration of centrally-acting mAChR agonists is unexplored. To provide insight we used the centrally-acting M1 mAChR agonist xanomeline, previously developed in the context of Alzheimer's disease and schizophrenia. Intraperitoneal administration of xanomeline significantly suppressed serum and splenic TNF levels, alleviated sickness behavior, and increased survival during lethal murine endotoxemia. The anti-inflammatory effects of xanomeline were brain mAChR-mediated and required intact vagus nerve and splenic nerve signaling. The anti-inflammatory efficacy of xanomeline was retained for at least 20h, associated with alterations in splenic lymphocyte, and dendritic cell proportions, and decreased splenocyte responsiveness to endotoxin. These results highlight an important role of the M1 mAChR in a neural circuitry to spleen in which brain cholinergic activation lowers peripheral pro-inflammatory cytokines to levels favoring survival. The therapeutic efficacy of xanomeline was also manifested by significantly improved survival in preclinical settings of severe sepsis. These findings are of interest for strategizing novel therapeutic approaches in inflammatory diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Agonistas Muscarínicos/farmacología , Piridinas/farmacología , Sepsis/prevención & control , Bazo/fisiología , Tiadiazoles/farmacología , Nervio Vago/fisiología , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/uso terapéutico , Citocinas/fisiología , Conducta de Enfermedad/efectos de los fármacos , Inyecciones Intraperitoneales , Lipopolisacáridos , Masculino , Ratones Endogámicos BALB C , Agonistas Muscarínicos/administración & dosificación , Agonistas Muscarínicos/uso terapéutico , Piridinas/administración & dosificación , Piridinas/uso terapéutico , Ratas Sprague-Dawley , Receptor Muscarínico M1/agonistas , Sepsis/mortalidad , Bazo/efectos de los fármacos , Bazo/inervación , Análisis de Supervivencia , Tiadiazoles/administración & dosificación , Tiadiazoles/uso terapéutico , Factor de Necrosis Tumoral alfa/sangre , Vagotomía
4.
Proc Natl Acad Sci U S A ; 107(26): 11942-7, 2010 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-20547845

RESUMEN

During infection, vertebrates develop "sickness syndrome," characterized by fever, anorexia, behavioral withdrawal, acute-phase protein responses, and inflammation. These pathophysiological responses are mediated by cytokines, including TNF and IL-1, released during the innate immune response to invasion. Even in the absence of infection, qualitatively similar physiological syndromes occur following sterile injury, ischemia reperfusion, crush injury, and autoimmune-mediated tissue damage. Recent advances implicate high-mobility group box 1 (HMGB1), a nuclear protein with inflammatory cytokine activities, in stimulating cytokine release. HMGB1 is passively released during cell injury and necrosis, or actively secreted during immune cell activation, positioning it at the intersection of sterile and infection-associated inflammation. To date, eight candidate receptors have been implicated in mediating the biological responses to HMGB1, but the mechanism of HMGB1-dependent cytokine release is unknown. Here we show that Toll-like receptor 4 (TLR4), a pivotal receptor for activation of innate immunity and cytokine release, is required for HMGB1-dependent activation of macrophage TNF release. Surface plasmon resonance studies indicate that HMGB1 binds specifically to TLR4, and that this binding requires a cysteine in position 106. A wholly synthetic 20-mer peptide containing cysteine 106 from within the cytokine-stimulating B box mediates TLR4-dependent activation of macrophage TNF release. Inhibition of TLR4 binding with neutralizing anti-HMGB1 mAb or by mutating cysteine 106 prevents HMGB1 activation of cytokine release. These results have implications for rationale, design, and development of experimental therapeutics for use in sterile and infectious inflammation.


Asunto(s)
Citocinas/biosíntesis , Proteína HMGB1/química , Proteína HMGB1/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Receptor Toll-Like 4/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Células CHO , Cricetinae , Cricetulus , Cisteína/química , Cartilla de ADN/genética , Proteína HMGB1/genética , Proteína HMGB1/inmunología , Humanos , Inmunidad Innata , Técnicas In Vitro , Antígeno 96 de los Linfocitos/metabolismo , Activación de Macrófagos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Transducción de Señal , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
5.
J Exp Med ; 203(7): 1623-8, 2006 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-16785311

RESUMEN

The innate immune system protects against infection and tissue injury through the specialized organs of the reticuloendothelial system, including the lungs, liver, and spleen. The central nervous system regulates innate immune responses via the vagus nerve, a mechanism termed the cholinergic antiinflammatory pathway. Vagus nerve stimulation inhibits proinflammatory cytokine production by signaling through the alpha7 nicotinic acetylcholine receptor subunit. Previously, the functional relationship between the cholinergic antiinflammatory pathway and the reticuloendothelial system was unknown. Here we show that vagus nerve stimulation fails to inhibit tumor necrosis factor (TNF) production in splenectomized animals during lethal endotoxemia. Selective lesioning of the common celiac nerve abolishes TNF suppression by vagus nerve stimulation, suggesting that the cholinergic pathway is functionally hard wired to the spleen via this branch of the vagus nerve. Administration of nicotine, an alpha7 agonist that mimics vagus nerve stimulation, increases proinflammatory cytokine production and lethality from polymicrobial sepsis in splenectomized mice, indicating that the spleen is critical to the protective response of the cholinergic pathway. These results reveal a specific, physiological connection between the nervous and innate immune systems that may be exploited through either electrical vagus nerve stimulation or administration of alpha7 agonists to inhibit proinflammatory cytokine production during infection and tissue injury.


Asunto(s)
Acetilcolina/antagonistas & inhibidores , Acetilcolina/fisiología , Endotoxemia/inmunología , Sepsis/inmunología , Transducción de Señal/fisiología , Esplenectomía , Animales , Endotoxemia/tratamiento farmacológico , Endotoxemia/mortalidad , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Endogámicas Lew , Sepsis/tratamiento farmacológico , Sepsis/microbiología , Transducción de Señal/efectos de los fármacos
6.
J Exp Med ; 203(7): 1637-42, 2006 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-16818669

RESUMEN

Severe sepsis, a lethal syndrome after infection or injury, is the third leading cause of mortality in the United States. The pathogenesis of severe sepsis is characterized by organ damage and accumulation of apoptotic lymphocytes in the spleen, thymus, and other organs. To examine the potential causal relationships of apoptosis to organ damage, we administered Z-VAD-FMK, a broad-spectrum caspase inhibitor, to mice with sepsis. We found that Z-VAD-FMK-treated septic mice had decreased levels of high mobility group box 1 (HMGB1), a critical cytokine mediator of organ damage in severe sepsis, and suppressed apoptosis in the spleen and thymus. In vitro, apoptotic cells activate macrophages to release HMGB1. Monoclonal antibodies against HMGB1 conferred protection against organ damage but did not prevent the accumulation of apoptotic cells in the spleen. Thus, our data indicate that HMGB1 production is downstream of apoptosis on the final common pathway to organ damage in severe sepsis.


Asunto(s)
Apoptosis/inmunología , Proteína HMGB1/fisiología , Sepsis/mortalidad , Sepsis/patología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Anticuerpos Monoclonales/uso terapéutico , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Femenino , Proteína HMGB1/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Sepsis/inmunología , Sepsis/terapia
7.
Mol Med ; 18: 618-27, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22354214

RESUMEN

Entry of blood-borne pathogens into the spleen elicits a series of changes in cellular architecture that culminates in the systemic release of protective antibodies. Despite an abundance of work that has characterized these processes, the regulatory mechanisms that coordinate cell trafficking and antibody production are still poorly understood. Here, marginal zone (MZ) B cells responding to streptococcus in the blood were observed to migrate along splenic nerves, arriving at the red pulp venous sinuses where they become antibody-secreting cells. Electrical stimulation of the vagus nerve, which in turn regulates the splenic nerve, arrested B-cell migration and decreased antibody secretion. Thus, neural circuits regulate the first wave of antibody production following B-cell exposure to blood-borne antigen.


Asunto(s)
Antígenos/inmunología , Linfocitos B/inmunología , Transducción de Señal , Bazo/inmunología , Bazo/inervación , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos/sangre , Movimiento Celular/inmunología , Fibras Colinérgicas/inmunología , Fibras Colinérgicas/metabolismo , Femenino , Inmunidad Humoral/efectos de los fármacos , Inflamación/inmunología , Cinética , Ratones , Ratones Endogámicos BALB C , Nicotina/farmacología , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Bazo/metabolismo , Sindecano-1/metabolismo , Nervio Vago/efectos de los fármacos , Nervio Vago/inmunología
8.
Mol Med ; 18: 539-43, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22183893

RESUMEN

The immune response to infection or injury coordinates host defense and tissue repair, but also has the capacity to damage host tissues. Recent advances in understanding protective mechanisms have found neural circuits that suppress release of damaging cytokines. Stimulation of the vagus nerve protects from excessive cytokine production and ameliorates experimental inflammatory disease. This mechanism, the inflammatory reflex, requires the α7 nicotinic acetylcholine receptor (α7nAChR), a ligand-gated ion channel expressed on macrophages, lymphocytes, neurons and other cells. To investigate cell-specific function of α7nAChR in the inflammatory reflex, we created chimeric mice by cross-transferring bone marrow between wild-type (WT) and α7nAChR-deficient mice. Deficiency of α7nAChR in bone marrow-derived cells significantly impaired vagus nerve-mediated regulation of tumor necrosis factor (TNF), whereas α7nAChR deficiency in neurons and other cells had no significant effect. In agreement with recent work, the inflammatory reflex was not functional in nude mice, because functional T cells are required for the integrity of the pathway. To investigate the role of T-cell α7nAChR, we adoptively transferred α7nAChR-deficient or WT T cells to nude mice. Transfer of WT and α7nAChR-deficient T cells restored function, indicating that α7nAChR expression on T cells is not necessary for this pathway. Together, these results indicate that α7nAChR expression in bone marrow-derived non-T cells is required for the integrity of the inflammatory reflex.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Inflamación/metabolismo , Receptores Nicotínicos/metabolismo , Traslado Adoptivo , Animales , Ratones , Ratones Noqueados , Ratones Desnudos , Receptores Nicotínicos/genética , Bazo/citología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/sangre , Nervio Vago/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7
9.
Mol Med ; 17(7-8): 599-606, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21738953

RESUMEN

Obesity, a serious and growing health threat, is associated with low-grade inflammation that plays a role in mediating its adverse consequences. Previously, we have discovered a role for neural cholinergic signaling in controlling inflammation, and demonstrated that the cholinergic agent galantamine suppresses excessive proinflammatory cytokine release. The main objective of this study was to examine the efficacy of galantamine, a clinically-approved drug, in alleviating obesity-related inflammation and associated complications. After 8 wks on a high-fat diet, C57BL/6J mice were treated with either galantamine (4 mg/kg, intraperitoneally [i.p.]) or saline for 4 wks in parallel with mice on a low-fat diet and treated with saline. Galantamine treatment of obese mice significantly reduced body weight, food intake, abdominal adiposity, plasma cytokine and adipokine levels, and significantly improved blood glucose, insulin resistance and hepatic steatosis. In addition, galantamine alleviated impaired insulin sensitivity and glucose intolerance significantly. These results indicate a previously unrecognized potential of galantamine in alleviating obesity, inflammation and other obesity-related complications in mice. These findings are of interest for studying the efficacy of this clinically-approved drug in the context of human obesity and metabolic syndrome.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Galantamina/farmacología , Inflamación/prevención & control , Obesidad/prevención & control , Adipoquinas/sangre , Adiposidad/efectos de los fármacos , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Colesterol/sangre , Inhibidores de la Colinesterasa/farmacología , Citocinas/sangre , Ingestión de Alimentos/efectos de los fármacos , Ayuno/sangre , Hígado Graso/etiología , Hígado Graso/prevención & control , Inflamación/sangre , Inflamación/etiología , Insulina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/etiología , Resistina/sangre , Factores de Tiempo , Resultado del Tratamiento , Aumento de Peso/efectos de los fármacos
10.
J Immunol ; 183(1): 552-9, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19542466

RESUMEN

The cholinergic anti-inflammatory pathway is a physiological mechanism that inhibits cytokine production and diminishes tissue injury during inflammation. Recent studies demonstrate that cholinergic signaling reduces adhesion molecule expression and chemokine production by endothelial cells and suppresses leukocyte migration during inflammation. It is unclear how vagus nerve stimulation regulates leukocyte trafficking because the vagus nerve does not innervate endothelial cells. Using mouse models of leukocyte trafficking, we show that the spleen, which is a major point of control for cholinergic modulation of cytokine production, is essential for vagus nerve-mediated regulation of neutrophil activation and migration. Administration of nicotine, a pharmacologic agonist of the cholinergic anti-inflammatory pathway, significantly reduces levels of CD11b, a beta(2)-integrin involved in cell adhesion and leukocyte chemotaxis, on the surface of neutrophils in a dose-dependent manner and this function requires the spleen. Similarly, vagus nerve stimulation significantly attenuates neutrophil surface CD11b levels only in the presence of an intact and innervated spleen. Further mechanistic studies reveal that nicotine suppresses F-actin polymerization, the rate-limiting step for CD11b surface expression. These studies demonstrate that modulation of leukocyte trafficking via cholinergic signaling to the spleen is a specific, centralized neural pathway positioned to suppress the excessive accumulation of neutrophils at inflammatory sites. Activating this mechanism may have important therapeutic potential for preventing tissue injury during inflammation.


Asunto(s)
Antígeno CD11b/fisiología , Inhibición de Migración Celular/inmunología , Agonistas Colinérgicos/administración & dosificación , Regulación hacia Abajo/inmunología , Infiltración Neutrófila/inmunología , Transducción de Señal/inmunología , Bazo/inmunología , Bazo/inervación , Animales , Antígeno CD11b/biosíntesis , Antígeno CD11b/metabolismo , Carragenina/fisiología , Femenino , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología , Nicotina/administración & dosificación , Bazo/citología , Esplenectomía , Nervio Vago/inmunología
11.
Proc Natl Acad Sci U S A ; 105(31): 11008-13, 2008 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-18669662

RESUMEN

The autonomic nervous system maintains homeostasis through its sympathetic and parasympathetic divisions. During infection, cells of the immune system release cytokines and other mediators that cause fever, hypotension, and tissue injury. Although the effect of cytokines on the nervous system has been known for decades, only recently has it become evident that the autonomic nervous system, in turn, regulates cytokine production through neural pathways. We have previously shown that efferent vagus nerve signals regulate cytokine production through the nicotinic acetylcholine receptor subunit alpha7, a mechanism termed "the cholinergic antiinflammatory pathway." Here, we show that vagus nerve stimulation during endotoxemia specifically attenuates TNF production by spleen macrophages in the red pulp and the marginal zone. Administration of nicotine, a pharmacological agonist of alpha7, attenuated TNF immunoreactivity in these specific macrophage subpopulations. Synaptophysin-positive nerve endings were observed in close apposition to red pulp macrophages, but they do not express choline acetyltransferase or vesicular acetylcholine transporter. Surgical ablation of the splenic nerve and catecholamine depletion by reserpine indicate that these nerves are catecholaminergic and are required for functional inhibition of TNF production by vagus nerve stimulation. Thus, the cholinergic antiinflammatory pathway regulates TNF production in discrete macrophage populations via two serially connected neurons: one preganglionic, originating in the dorsal motor nucleus of the vagus nerve, and the second postganglionic, originating in the celiac-superior mesenteric plexus, and projecting in the splenic nerve.


Asunto(s)
Sistema Nervioso Autónomo/inmunología , Endotoxemia/inmunología , Bazo/inervación , Factores de Necrosis Tumoral/inmunología , Nervio Vago/inmunología , Animales , Estimulación Eléctrica , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Nicotina/inmunología , Nicotina/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Nicotínicos/inmunología , Bazo/citología , Receptor Nicotínico de Acetilcolina alfa 7
12.
J Immunol ; 181(5): 3535-9, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18714026

RESUMEN

High mobility group box 1 (HMGB1) is a critical mediator of lethal sepsis. Previously, we showed that apoptotic cells can activate macrophages to release HMGB1. During sepsis, apoptosis occurs primarily in lymphoid organs, including the spleen and thymus. Currently, it is unclear whether this accelerated lymphoid organ apoptosis contributes to systemic release of HMGB1 in sepsis. In this study, we report that splenectomy significantly reduces systemic HMGB1 release and improves survival in mice with polymicrobial sepsis. Treatment with a broad-spectrum caspase inhibitor reduces systemic lymphocyte apoptosis, suppresses circulating HMGB1 concentrations, and improves survival during polymicrobial sepsis, but fails to protect septic mice following splenectomy. These findings indicate that apoptosis in the spleen is essential to the pathogenesis of HMGB1-mediated sepsis lethality.


Asunto(s)
Proteína HMGB1/sangre , Sepsis/terapia , Esplenectomía , Animales , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Linfocitos/citología , Ratones , Sepsis/mortalidad , Bazo/citología , Tasa de Supervivencia
13.
Front Immunol ; 11: 131, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32132994

RESUMEN

Altered lipid metabolism in macrophages is associated with various important inflammatory conditions. Although lipid metabolism is an important target for therapeutic intervention, the metabolic requirement involved in lipid accumulation during pro-inflammatory activation of macrophages remains incompletely characterized. We show here that macrophage activation with IFNγ results in increased aerobic glycolysis, iNOS-dependent inhibition of respiration, and accumulation of triacylglycerol. Surprisingly, metabolite tracing with 13C-labeled glucose revealed that the glucose contributed to the glycerol groups in triacylglycerol (TAG), rather than to de novo synthesis of fatty acids. This is in stark contrast to the otherwise similar metabolism of cancer cells, and previous results obtained in activated macrophages and dendritic cells. Our results establish a novel metabolic pathway whereby glucose provides glycerol to the headgroup of TAG during classical macrophage activation.


Asunto(s)
Ácidos Grasos/metabolismo , Gotas Lipídicas/metabolismo , Activación de Macrófagos/fisiología , Animales , Glucosa/metabolismo , Glucólisis/fisiología , Interferones/farmacología , Metabolismo de los Lípidos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Cultivo Primario de Células , Respiración , Triglicéridos/metabolismo
14.
Mol Med ; 15(7-8): 195-202, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19593403

RESUMEN

The cholinergic antiinflammatory pathway modulates inflammatory cytokine production through a mechanism dependent on the vagus nerve and the alpha7 subunit of the nicotinic acetylcholine receptor. GTS-21 [3-(2,4-dimethoxybenzylidene) anabaseine], a selective alpha7 agonist, inhibits inflammatory cytokine production in murine and human macrophages and in several models of inflammatory disease in vivo, but to date its antiinflammatory efficacy in human monocytes has not been characterized. We report here our findings that GTS-21 attenuates tumor necrosis factor (TNF) and interleukin 1beta levels in human whole blood activated by exposure to endotoxin. GTS-21 inhibited TNF production in endotoxin-stimulated primary human monocytes in vitro at the transcriptional level. The suppressive effect of GTS-21 was more potent than nicotine in whole blood and monocytes. Furthermore, GTS-21 attenuated TNF production in monocytes stimulated with peptidoglycan, polyinosinic-polycytidylic acid, CpG, HMGB1 (high-mobility group box 1 protein), and advanced glycation end product-modified albumin. GTS-21 decreased TNF levels in endotoxin-stimulated whole blood obtained from patients with severe sepsis. These findings establish the immunoregulatory effect of GTS-21 on human monocytes, and indicate the potential benefits of further exploration of GTS-21's therapeutic uses in human inflammatory disease.


Asunto(s)
Compuestos de Bencilideno/farmacología , Agonistas Colinérgicos/farmacología , Citocinas/biosíntesis , Monocitos/efectos de los fármacos , Piridinas/farmacología , Receptores Inmunológicos/agonistas , Receptores Nicotínicos/metabolismo , Receptores Toll-Like/agonistas , Adulto , Anciano , Anciano de 80 o más Años , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/sangre , Citocinas/genética , Endotoxinas/antagonistas & inhibidores , Endotoxinas/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1beta/biosíntesis , Interleucina-1beta/sangre , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Nicotina/farmacología , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/metabolismo , Sepsis/sangre , Sepsis/metabolismo , Receptores Toll-Like/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/genética , Receptor Nicotínico de Acetilcolina alfa 7
15.
Brain Behav Immun ; 23(1): 41-5, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18639629

RESUMEN

The excessive release of cytokines by the immune system contributes importantly to the pathogenesis of inflammatory diseases. Recent advances in understanding the biology of cytokine toxicity led to the discovery of the "cholinergic anti-inflammatory pathway," defined as neural signals transmitted via the vagus nerve that inhibit cytokine release through a mechanism that requires the alpha7 subunit-containing nicotinic acetylcholine receptor (alpha7nAChR). Vagus nerve regulation of peripheral functions is controlled by brain nuclei and neural networks, but despite considerable importance, little is known about the molecular basis for central regulation of the vagus nerve-based cholinergic anti-inflammatory pathway. Here we report that brain acetylcholinesterase activity controls systemic and organ specific TNF production during endotoxemia. Peripheral administration of the acetylcholinesterase inhibitor galantamine significantly reduced serum TNF levels through vagus nerve signaling, and protected against lethality during murine endotoxemia. Administration of a centrally-acting muscarinic receptor antagonist abolished the suppression of TNF by galantamine, indicating that suppressing acetylcholinesterase activity, coupled with central muscarinic receptors, controls peripheral cytokine responses. Administration of galantamine to alpha7nAChR knockout mice failed to suppress TNF levels, indicating that the alpha7nAChR-mediated cholinergic anti-inflammatory pathway is required for the anti-inflammatory effect of galantamine. These findings show that inhibition of brain acetylcholinesterase suppresses systemic inflammation through a central muscarinic receptor-mediated and vagal- and alpha7nAChR-dependent mechanism. Our data also indicate that a clinically used centrally-acting acetylcholinesterase inhibitor can be utilized to suppress abnormal inflammation to therapeutic advantage.


Asunto(s)
Acetilcolinesterasa/metabolismo , Encéfalo/efectos de los fármacos , Inhibidores de la Colinesterasa/farmacología , Citocinas/sangre , Receptores Nicotínicos/fisiología , Alcaloides , Análisis de Varianza , Animales , Derivados de Atropina/administración & dosificación , Derivados de Atropina/farmacología , Encéfalo/enzimología , Inhibidores de la Colinesterasa/administración & dosificación , Endotoxemia/sangre , Endotoxemia/inducido químicamente , Endotoxemia/tratamiento farmacológico , Ensayo de Inmunoadsorción Enzimática , Femenino , Galantamina/administración & dosificación , Galantamina/farmacología , Inyecciones Intraperitoneales , Interleucina-6/sangre , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Parasimpatolíticos/administración & dosificación , Parasimpatolíticos/farmacología , Receptores Nicotínicos/deficiencia , Receptores Nicotínicos/genética , Sesquiterpenos/administración & dosificación , Sesquiterpenos/farmacología , Factor de Necrosis Tumoral alfa/sangre , Vagotomía/métodos , Receptor Nicotínico de Acetilcolina alfa 7
16.
Mol Med ; 14(9-10): 567-74, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18584048

RESUMEN

The alpha7 subunit-containing nicotinic acetylcholine receptor (alpha7nAChR) is an essential component in the vagus nerve-based cholinergic anti-inflammatory pathway that regulates the levels of TNF, high mobility group box 1 (HMGB1), and other cytokines during inflammation. Choline is an essential nutrient, a cell membrane constituent, a precursor in the biosynthesis of acetylcholine, and a selective natural alpha7nAChR agonist. Here, we studied the anti-inflammatory potential of choline in murine endotoxemia and sepsis, and the role of the alpha7nAChR in mediating the suppressive effect of choline on TNF release. Choline (0.1-50 mM) dose-dependently suppressed TNF release from endotoxin-activated RAW macrophage-like cells, and this effect was associated with significant inhibition of NF-kappaB activation. Choline (50 mg/kg, intraperitoneally [i.p.]) treatment prior to endotoxin administration in mice significantly reduced systemic TNF levels. In contrast to its TNF suppressive effect in wild type mice, choline (50 mg/kg, i.p.) failed to inhibit systemic TNF levels in alpha7nAChR knockout mice during endotoxemia. Choline also failed to suppress TNF release from endotoxin-activated peritoneal macrophages isolated from alpha7nAChR knockout mice. Choline treatment prior to endotoxin resulted in a significantly improved survival rate as compared with saline-treated endotoxemic controls. Choline also suppressed HMGB1 release in vitro and in vivo, and choline treatment initiated 24 h after cecal ligation and puncture (CLP)-induced polymicrobial sepsis significantly improved survival in mice. In addition, choline suppressed TNF release from endotoxin-activated human whole blood and macrophages. Collectively, these data characterize the anti-inflammatory efficacy of choline and demonstrate that the modulation of TNF release by choline requires alpha7nAChR-mediated signaling.


Asunto(s)
Antiinflamatorios/farmacología , Colina/farmacología , Colina/fisiología , Macrófagos/metabolismo , Receptores Nicotínicos/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Células Cultivadas , Endotoxemia/tratamiento farmacológico , Endotoxemia/inmunología , Endotoxemia/metabolismo , Endotoxemia/mortalidad , Endotoxinas/inmunología , Femenino , Regulación de la Expresión Génica , Proteína HMGB1/metabolismo , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Nicotínicos/genética , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Sepsis/metabolismo , Sepsis/mortalidad , Receptor Nicotínico de Acetilcolina alfa 7
17.
Front Immunol ; 9: 2648, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30538698

RESUMEN

Macrophage cytokine production is regulated by neural signals, for example in the inflammatory reflex. Signals in the vagus and splenic nerves are relayed by choline acetyltransferase+ T cells that release acetylcholine, the cognate ligand for alpha7 nicotinic acetylcholine subunit-containing receptors (α7nAChR), and suppress TNF release in macrophages. Here, we observed that electrical vagus nerve stimulation with a duration of 0.1-60 s significantly reduced systemic TNF release in experimental endotoxemia. This suppression of TNF was sustained for more than 24 h, but abolished in mice deficient in the α7nAChR subunit. Exposure of primary human macrophages and murine RAW 264.7 macrophage-like cells to selective ligands for α7nAChR for 1 h in vitro attenuated TNF production for up to 24 h in response to endotoxin. Pharmacological inhibition of adenylyl cyclase (AC) and knockdown of adenylyl cyclase 6 (AC6) or c-FOS abolished cholinergic suppression of endotoxin-induced TNF release. These findings indicate that action potentials in the inflammatory reflex trigger a change in macrophage behavior that requires AC and phosphorylation of the cAMP response element binding protein (CREB). These observations further our mechanistic understanding of neural regulation of inflammation and may have implications for development of bioelectronic medicine treatment of inflammatory diseases.


Asunto(s)
Adenilil Ciclasas/metabolismo , Inflamación/metabolismo , Reflejo/fisiología , Factores de Necrosis Tumoral/metabolismo , Animales , Proteína de Unión a CREB/metabolismo , Línea Celular , Endotoxinas/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley , Bazo/metabolismo , Nervio Vago/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
18.
Shock ; 25(6): 571-4, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16721263

RESUMEN

Cerebral and myocardial ischemia, two of the leading causes of morbidity and mortality worldwide, are associated with inflammation that can lead to multiple organ failure and death. High-mobility group box 1(HMGB1), a recently described mediator of lethal systemic inflammation, has been detected in individuals with severe sepsis and hemorrhagic shock, but its role during ischemic injury in humans is unknown. To determine whether systemic HMGB1 levels are elevated after ischemic injury, a prospective observational study was performed in subjects with a diagnosis of either Acute Coronary Syndrome (ACS) or cerebral vascular ischemia (transient ischemic attack or cerebral vascular accident). Subjects (n, 16; age [mean], 67+/-16.3 years) were enrolled in the North Shore-LIJ emergency department within 24 h of symptom onset. Blood samples were collected, and HMGB1 levels analyzed by Western blot analysis using previously described methods (Wang et al. Science. 1999). Control samples were obtained from healthy age- and sex-matched volunteers (n, 16; age [mean], 68+/-15.8 years). Here, we report that serum HMGB1 levels were significantly elevated in both myocardial ischemia subjects (myocardial control serum HMGB1, 1.94+/-2.05 ng/mL, vs. myocardial ischemia serum HMGB1, 159+/-54.3 ng/mL; P<0.001); and in cerebral ischemia subjects (cerebral control serum HMGB1, 16.8+/-10.9 ng/mL, vs. cerebral ischemia serum HMGB1, 218+/-18.8 ng/mL; P<0.001). These results suggest that systemic HMGB1 levels are elevated in human ischemic disease.


Asunto(s)
Isquemia Encefálica/sangre , Isquemia Miocárdica/sangre , Enfermedad Aguda , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Enfermedad Coronaria/sangre , Femenino , Humanos , Inflamación/sangre , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Sepsis/sangre , Sepsis/mortalidad , Choque Hemorrágico/sangre , Choque Hemorrágico/mortalidad , Accidente Cerebrovascular/sangre
19.
Shock ; 40(6): 492-5, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24089009

RESUMEN

Severe sepsis is a life-threatening complication of infection and injury affecting more than 700,000 people in the United States each year. Two thirds of patients with severe sepsis will survive to be discharged. Survivors have high incidence of cognitive impairment, immune dysregulation, functional impairments with marked disability, and 5-year mortality rates of 82%. High-mobility group box 1 (HMGB1) is necessary and sufficient mediator of sepsis pathogenesis in experimental models of this syndrome. The spleen is a crucial organ in the immune response to severe infection, and splenocyte dysfunction occurs in sepsis survivors. We hypothesized that HMGB1 plays a key role in mediating the immune dysfunction of splenocytes in sepsis survivors. Mice that survived cecal ligation and puncture-induced sepsis develop persistent splenomegaly; furthermore, splenocytes derived from sepsis survivors had enhanced responses to lipopolysaccharide ex vivo. Administration of neutralizing anti-HMGB1 antibody to sepsis survivors attenuated development of splenomegaly and reversed splenocyte priming. Splenocytes exposed to HMGB1 and subsequently challenged with cognate ligands to Toll-like receptor 2 (TLR2,) TLR4, TLR9, and RAGE (receptor for advanced glycation end product) receptors had enhanced cytokine release as compared with splenocytes not previously exposed to HMGB1. Exposure of TLR2, TLR9, or RAGE splenocytes to HMGB1 enhanced responses to other TLR receptor ligands; in contrast, HMGB1 failed to prime TLR4 splenocytes. These findings indicate that exposure to HMGB1 enhances splenocyte responses to secondary inflammatory challenges, a priming effect dependent on TLR4, and that anti-HMGB1 monoclonal antibody may be beneficial in sepsis survivors.


Asunto(s)
Proteína HMGB1/inmunología , Sepsis/inmunología , Bazo/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Células Cultivadas , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Proteína HMGB1/antagonistas & inhibidores , Mediadores de Inflamación/inmunología , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/inmunología , Sepsis/complicaciones , Esplenomegalia/etiología , Esplenomegalia/prevención & control , Receptor Toll-Like 4/inmunología
20.
Science ; 334(6052): 98-101, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21921156

RESUMEN

Neural circuits regulate cytokine production to prevent potentially damaging inflammation. A prototypical vagus nerve circuit, the inflammatory reflex, inhibits tumor necrosis factor-α production in spleen by a mechanism requiring acetylcholine signaling through the α7 nicotinic acetylcholine receptor expressed on cytokine-producing macrophages. Nerve fibers in spleen lack the enzymatic machinery necessary for acetylcholine production; therefore, how does this neural circuit terminate in cholinergic signaling? We identified an acetylcholine-producing, memory phenotype T cell population in mice that is integral to the inflammatory reflex. These acetylcholine-producing T cells are required for inhibition of cytokine production by vagus nerve stimulation. Thus, action potentials originating in the vagus nerve regulate T cells, which in turn produce the neurotransmitter, acetylcholine, required to control innate immune responses.


Asunto(s)
Acetilcolina/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Inmunidad Innata , Neuroinmunomodulación , Nervio Vago/fisiología , Potenciales de Acción , Animales , Colina O-Acetiltransferasa/metabolismo , Colinérgicos/metabolismo , Femenino , Memoria Inmunológica , Inflamación , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Norepinefrina/farmacología , Receptores Nicotínicos/metabolismo , Transducción de Señal , Bazo/inmunología , Bazo/inervación , Bazo/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/sangre , Estimulación del Nervio Vago , Receptor Nicotínico de Acetilcolina alfa 7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA