Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Pediatr Res ; 85(5): 711-718, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30759452

RESUMO

BACKGROUND: Congenital diaphragmatic hernia (CDH) is a complex birth anomaly with significant mortality and morbidity. Lung hypoplasia and persistent pulmonary hypertension (PPHN) limit survival in CDH. Macrophage migration inhibitory factor (MIF), a key regulator of innate immunity, is involved in hypoxia-induced vascular remodeling and PPHN. We hypothesized that antenatal inhibition of MIF in CDH fetuses, would reduce vascular remodeling, and improve angiogenesis and lung development. METHODS: Pregnant rats were randomized into three groups: Control, nitrofen, and nitrofen + ISO-92. Lung volumes of pups were measured by CT scanning. Right ventricular systolic pressure (RVSP) and vascular wall thickness (VWT) were measured together with MIF concentration, angiogenesis markers, lung morphometry, and histology. RESULTS: Prenatal treatment with ISO-92, an MIF inhibitor, improved normalization of static lung volume, lung volume-to-body weight ratio, decreased alveolar septal thickness, RVSP and VWT and improved radial alveolar count as compared to the non-treated group. Expression of MIF was unaffected by ISO-92; however, ISO-92 increased p-eNOS and VEGF activities and reduced arginase 1, 2 and Sflt-1. CONCLUSION: Prenatal inhibition of MIF activity in CDH rat model improves angiogenesis and lung development. This selective intervention may be a future therapeutic strategy to reduce the morbidity and mortality of this devastating condition.


Assuntos
Hérnias Diafragmáticas Congênitas/terapia , Oxirredutases Intramoleculares/antagonistas & inibidores , Pulmão/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Peso Corporal , Modelos Animais de Doenças , Feminino , Hemodinâmica , Hérnias Diafragmáticas Congênitas/induzido quimicamente , Hérnias Diafragmáticas Congênitas/patologia , Hipertensão Pulmonar/etiologia , Imunidade Inata , Inflamação , Pulmão/crescimento & desenvolvimento , Exposição Materna , Éteres Fenílicos , Gravidez , Prenhez , Ratos , Sístole , Tomografia Computadorizada por Raios X , Remodelação Vascular , Função Ventricular Direita
2.
Exp Lung Res ; 42(7): 335-345, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27585839

RESUMO

INTRODUCTION: Sepsis is characterized by dysregulated systemic inflammation and cytokine storm. Angiopoietin-2 (Ang-2) is known to closely correlate with severity of sepsis-related acute lung injury and mortality. The aim of this study was to clarify the mechanisms involved in Ang-2 secretion to better understand the pathophysiology of sepsis. MATERIALS AND METHODS: The concentration of Ang-2 was assessed in culture medium of pulmonary microvascular endothelial cells in the presence or absence of Gram-positive bacteria cell wall components [lipoteichoic acid (LTA) and peptidoglycan (PGN)] stimulation at different time points ranging from 15 minutes to 24 hours. Constitutive and LTA-PGN-stimulated Ang-2 level changes were also assessed after cells were pretreated with different pathway inhibitors for 1 hour. RESULTS: Two distinctive mechanisms of Ang-2 secretion, constitutive and stimulated secretion, were identified. Constitutive secretion resulted in slow but continuous increase in Ang-2 in culture medium over time. It was regulated by 3'5'-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA)-Ca2+ and nitric oxide (NO)-3'5'-cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG)-Ca2+ pathways and partially regulated by N-ethyl-maleimide-sensitive factor-Ca2+ pathways. LTA-PGN stimulation caused rapid and potent increase followed by gradual decrease of Ang-2. It was partially regulated by both Ral A-phospholipase D and NSF-Ca2+ pathways. CONCLUSIONS: We demonstrated characteristics and involved pathways for two distinctive secretory mechanisms, constitutive and stimulated, of Ang-2 in pulmonary microvascular endothelial cells. Considering the close correlation of Ang-2 with sepsis outcomes, our findings provide a better understanding of an important mechanism associated with sepsis pathophysiology and identify possible therapeutic targets to improve outcomes in the potentially lethal disease.


Assuntos
Angiopoietina-2/metabolismo , Células Endoteliais/metabolismo , Pulmão/irrigação sanguínea , Parede Celular/química , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Bactérias Gram-Positivas/química , Humanos , Lipopolissacarídeos/farmacologia , Pulmão/citologia , Peptidoglicano/farmacologia , Sepse/fisiopatologia , Ácidos Teicoicos/farmacologia
3.
J Immunol ; 183(1): 552-9, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19542466

RESUMO

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.


Assuntos
Antígeno CD11b/fisiologia , Inibição de Migração Celular/imunologia , Agonistas Colinérgicos/administração & dosagem , Regulação para Baixo/imunologia , Infiltração de Neutrófilos/imunologia , Transdução de Sinais/imunologia , Baço/imunologia , Baço/inervação , Animais , Antígeno CD11b/biossíntese , Antígeno CD11b/metabolismo , Carragenina/fisiologia , Feminino , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Nicotina/administração & dosagem , Baço/citologia , Esplenectomia , Nervo Vago/imunologia
4.
Brain Behav Immun ; 23(1): 41-5, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18639629

RESUMO

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.


Assuntos
Acetilcolinesterase/metabolismo , Encéfalo/efeitos dos fármacos , Inibidores da Colinesterase/farmacologia , Citocinas/sangue , Receptores Nicotínicos/fisiologia , Alcaloides , Análise de Variância , Animais , Derivados da Atropina/administração & dosagem , Derivados da Atropina/farmacologia , Encéfalo/enzimologia , Inibidores da Colinesterase/administração & dosagem , Endotoxemia/sangue , Endotoxemia/induzido quimicamente , Endotoxemia/tratamento farmacológico , Ensaio de Imunoadsorção Enzimática , Feminino , Galantamina/administração & dosagem , Galantamina/farmacologia , Injeções Intraperitoneais , Interleucina-6/sangue , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Parassimpatolíticos/administração & dosagem , Parassimpatolíticos/farmacologia , Receptores Nicotínicos/deficiência , Receptores Nicotínicos/genética , Sesquiterpenos/administração & dosagem , Sesquiterpenos/farmacologia , Fator de Necrose Tumoral alfa/sangue , Vagotomia/métodos , Receptor Nicotínico de Acetilcolina alfa7
5.
Proc Natl Acad Sci U S A ; 105(31): 11008-13, 2008 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-18669662

RESUMO

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.


Assuntos
Sistema Nervoso Autônomo/imunologia , Endotoxemia/imunologia , Baço/inervação , Fatores de Necrose Tumoral/imunologia , Nervo Vago/imunologia , Animais , Estimulação Elétrica , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nicotina/imunologia , Nicotina/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Nicotínicos/imunologia , Baço/citologia , Receptor Nicotínico de Acetilcolina alfa7
6.
Mol Med ; 14(9-10): 567-74, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18584048

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Colina/farmacologia , Colina/fisiologia , Macrófagos/metabolismo , Receptores Nicotínicos/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Endotoxemia/tratamento farmacológico , Endotoxemia/imunologia , Endotoxemia/metabolismo , Endotoxemia/mortalidade , Endotoxinas/imunologia , Feminino , Regulação da Expressão Gênica , Proteína HMGB1/metabolismo , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nicotínicos/genética , Sepse/tratamento farmacológico , Sepse/imunologia , Sepse/metabolismo , Sepse/mortalidade , Receptor Nicotínico de Acetilcolina alfa7
7.
Crit Care Med ; 35(12): 2762-8, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17901837

RESUMO

OBJECTIVE: Electrical vagus nerve stimulation inhibits proinflammatory cytokine production and prevents shock during lethal systemic inflammation through an alpha7 nicotinic acetylcholine receptor (alpha7nAChR)-dependent pathway to the spleen, termed the cholinergic anti-inflammatory pathway. Pharmacologic alpha7nAChR agonists inhibit production of the critical proinflammatory mediator high mobility group box 1 (HMGB1) and rescue mice from lethal polymicrobial sepsis. Here we developed a method of transcutaneous mechanical vagus nerve stimulation and then investigated whether this therapy can protect mice against sepsis lethality. DESIGN: Prospective, randomized study. SETTING: Institute-based research laboratory. SUBJECTS: Male BALB/c mice. INTERVENTIONS: Mice received lipopolysaccharide to induce lethal endotoxemia or underwent cecal ligation and puncture to induce polymicrobial sepsis. Mice were then randomized to receive electrical, transcutaneous, or sham vagus nerve stimulation and were followed for survival or euthanized at predetermined time points for cytokine analysis. MEASUREMENTS AND MAIN RESULTS: Transcutaneous vagus nerve stimulation dose-dependently reduced systemic tumor necrosis factor levels during lethal endotoxemia. Treatment with transcutaneous vagus nerve stimulation inhibited HMGB1 levels and improved survival in mice with polymicrobial sepsis, even when administered 24 hrs after the onset of disease. CONCLUSIONS: Transcutaneous vagus nerve stimulation is an efficacious treatment for mice with lethal endotoxemia or polymicrobial sepsis.


Assuntos
Proteína HMGB1/sangue , Sepse/terapia , Estimulação Elétrica Nervosa Transcutânea/métodos , Nervo Vago , Animais , Citocinas/sangue , Endotoxemia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neuroimunomodulação , Estudos Prospectivos , Distribuição Aleatória , Sepse/imunologia , Análise de Sobrevida
8.
Crit Care Med ; 35(4): 1139-44, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17334244

RESUMO

OBJECTIVE: Tumor necrosis factor and high mobility group box 1 are critical cytokine mediators of inflammation. The efferent vagus nerve inhibits cytokine release through alpha7-nicotinic acetylcholine receptor-mediated cholinergic signaling. Here we studied whether GTS-21, a selective alpha7-nicotinic acetylcholine receptor agonist, inhibits proinflammatory cytokines in vitro and in vivo and improves survival in murine endotoxemia and severe sepsis. DESIGN: Randomized and controlled in vitro and in vivo study. SETTINGS: Research laboratory and animal facility rooms. SUBJECTS: RAW 264.7 cells and BALB/c mice treated with endotoxin or subjected to cecal ligation and puncture (CLP). INTERVENTIONS: RAW 264.7 cells were exposed to endotoxin (4 ng/mL or 10 ng/mL) in the presence or absence of GTS-21 (1-100 muM), and tumor necrosis factor and high mobility group box 1 release and nuclear factor-kappaB activation were analyzed. Mice were treated with GTS-21 (0.4 mg/kg or 4 mg/kg, intraperitoneally) or saline 30 mins before endotoxin (6 mg/kg, intraperitoneally), and serum tumor necrosis factor was analyzed 1.5 hrs after the onset of endotoxemia. In survival experiments, mice were treated with GTS-21 (0.4 or 4.0 mg/kg, intraperitoneally) or saline 30 mins before and 6 hrs after endotoxin and then twice daily for 3 days. Severe sepsis was induced by CLP. Mice were treated with GTS-21 (4 mg/kg) or saline immediately and 6 hrs and 24 hrs after CLP, and serum high mobility group box 1 was analyzed 30 hrs after CLP. In survival experiments, GTS-21 (0.4 or 4 mg/kg) treatment was initiated 24 hrs after CLP and continued twice daily for 3 days. MEASUREMENTS AND MAIN RESULTS: GTS-21 dose-dependently inhibited tumor necrosis factor and high mobility group box 1 release and nuclear factor-kappaB activation in vitro. GTS-21 (4 mg/kg) significantly inhibited serum tumor necrosis factor during endotoxemia and improved survival (p < .0001). GTS-21 (4 mg/kg) significantly inhibited serum high mobility group box 1 levels in CLP mice and improved survival (p < .0006). CONCLUSION: These findings are of interest for the development of alpha7-nicotinic acetylcholine receptor agonists as a new class of anti-inflammatory therapeutics.


Assuntos
Compostos de Benzilideno/farmacologia , Endotoxemia/tratamento farmacológico , Agonistas Nicotínicos/farmacologia , Piridinas/farmacologia , Receptores Nicotínicos/metabolismo , Sepse/tratamento farmacológico , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Endotoxemia/mortalidade , Proteína HMGB1/antagonistas & inibidores , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/antagonistas & inibidores , Sepse/mortalidade , Índice de Gravidade de Doença , Taxa de Sobrevida , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Receptor Nicotínico de Acetilcolina alfa7
9.
J Exp Med ; 203(7): 1623-8, 2006 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-16785311

RESUMO

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.


Assuntos
Acetilcolina/antagonistas & inibidores , Acetilcolina/fisiologia , Endotoxemia/imunologia , Sepse/imunologia , Transdução de Sinais/fisiologia , Esplenectomia , Animais , Endotoxemia/tratamento farmacológico , Endotoxemia/mortalidade , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Endogâmicos Lew , Sepse/tratamento farmacológico , Sepse/microbiologia , Transdução de Sinais/efeitos dos fármacos
10.
Proc Natl Acad Sci U S A ; 103(13): 5219-23, 2006 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-16549778

RESUMO

TNF has a critical mediator role in inflammation and is an important therapeutic target. We recently discovered that TNF production is regulated by neural signals through the vagus nerve. Activation of this "cholinergic antiinflammatory pathway" inhibits the production of TNF and other cytokines and protects animals from the inflammatory damage caused by endotoxemia and severe sepsis. Here, we describe a role for central muscarinic acetylcholine receptors in the activation of the cholinergic antiinflammatory pathway. Central muscarinic cholinergic activation by muscarine, the M1 receptor agonist McN-A-343, and the M2 receptor antagonist methoctramine inhibited serum TNF levels significantly during endotoxemia. Centrally administered methoctramine stimulated vagus-nerve activity measured by changes in instantaneous heart-rate variability. Blockade of peripheral muscarinic receptors did not abolish antiinflammatory signaling through the vagus nerve, indicating that peripheral muscarinic receptors on immune cells are not required for the cytokine-regulating activities of the cholinergic antiinflammatory pathway. The role of central muscarinic receptors in activating the cholinergic antiinflammatory pathway is of interest for the use of centrally acting muscarinic cholinergic enhancers as antiinflammatory agents.


Assuntos
Encéfalo/metabolismo , Endotoxemia/metabolismo , Inflamação/metabolismo , Receptores Muscarínicos/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Diaminas/administração & dosagem , Diaminas/farmacologia , Endotoxemia/induzido quimicamente , Frequência Cardíaca/efeitos dos fármacos , Masculino , Agonistas Muscarínicos/farmacologia , Ratos , Síndrome de Resposta Inflamatória Sistêmica/induzido quimicamente , Fator de Necrose Tumoral alfa/metabolismo
11.
J Biol Chem ; 280(44): 36541-4, 2005 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-16115897

RESUMO

MIF is a proinflammatory cytokine that has been implicated in the pathogenesis of sepsis, arthritis, and other inflammatory diseases. Antibodies against MIF are effective in experimental models of inflammation, and there is interest in strategies to inhibit its deleterious cytokine activities. Here we identify a mechanism of inhibiting MIF pro-inflammatory activities by targeting MIF tautomerase activity. We designed small molecules to inhibit this tautomerase activity; a lead molecule, "ISO-1 ((S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid methyl ester)," significantly inhibits the cytokine activity in vitro. Moreover, ISO-1 inhibits tumor necrosis factor release from macrophages isolated from LPStreated wild type mice but has no effect on cytokine release from MIFdeficient macrophages. The therapeutic importance of the MIF inhibition by ISO-1 is demonstrated by the significant protection from sepsis, induced by cecal ligation and puncture in a clinically relevant time frame. These results identify ISO-1 as the first small molecule inhibitor of MIF proinflammatory activities with therapeutic implications and indicate the potential of the MIF active site as a novel target for therapeutic interventions in human sepsis.


Assuntos
Anti-Inflamatórios/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Oxirredutases Intramoleculares/química , Isoxazóis/farmacologia , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Sepse/prevenção & controle , Animais , Sítios de Ligação , Ceco/efeitos dos fármacos , Ceco/metabolismo , Sobrevivência Celular/fisiologia , Ensaio de Desvio de Mobilidade Eletroforética , Endotoxemia/tratamento farmacológico , Endotoxemia/etiologia , Isoxazóis/química , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Punções , Sepse/tratamento farmacológico , Sepse/patologia , Taxa de Sobrevida , Fatores de Necrose Tumoral/metabolismo
12.
J Lipid Res ; 46(4): 623-7, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15687351

RESUMO

Stearoyl lysophosphatidylcholine (LPC) has recently been proven protective against lethal sepsis by stimulating neutrophils to eliminate invading pathogens through an H2O2-dependent mechanism. Here, we demonstrate that stearoyl LPC, but not caproyl LPC, significantly attenuates circulating high-mobility group box 1 (HMGB1) levels in endotoxemia and sepsis by suppressing endotoxin-induced HMGB1 release from macrophages/monocytes. Neutralizing antibodies against G2A, a potential cell surface receptor for LPC, partially abrogated stearoyl LPC-mediated suppression of HMGB1 release. Thus, stearoyl LPC confers protection against lethal experimental sepsis partly by facilitating the elimination of the invading pathogens and partly by inhibiting endotoxin-induced release of a late proinflammatory cytokine, HMGB1.


Assuntos
Proteína HMGB1/metabolismo , Lisofosfatidilcolinas/farmacologia , Lisofosfatidilcolinas/uso terapêutico , Sepse/tratamento farmacológico , Sepse/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Endotoxinas/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Transporte Proteico , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA