Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Int J Mol Sci ; 19(4)2018 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-29642561

RESUMO

While interleukin-1ß (IL-1ß) is a potent pro-inflammatory cytokine essential for host defense, high systemic levels cause life-threatening inflammatory syndromes. ATP, a stimulus of IL-1ß maturation, is released from damaged cells along with ß-nicotinamide adenine dinucleotide (ß-NAD). Here, we tested the hypothesis that ß-NAD controls ATP-signaling and, hence, IL-1ß release. Lipopolysaccharide-primed monocytic U937 cells and primary human mononuclear leukocytes were stimulated with 2'(3')-O-(4-benzoyl-benzoyl)ATP trieethylammonium salt (BzATP), a P2X7 receptor agonist, in the presence or absence of ß-NAD. IL-1ß was measured in cell culture supernatants. The roles of P2Y receptors, nicotinic acetylcholine receptors (nAChRs), and Ca2+-independent phospholipase A2 (iPLA2ß, PLA2G6) were investigated using specific inhibitors and gene-silencing. Exogenous ß-NAD signaled via P2Y receptors and dose-dependently (IC50 = 15 µM) suppressed the BzATP-induced IL-1ß release. Signaling involved iPLA2ß, release of a soluble mediator, and nAChR subunit α9. Patch-clamp experiments revealed that ß-NAD inhibited BzATP-induced ion currents. In conclusion, we describe a novel triple membrane-passing signaling cascade triggered by extracellular ß-NAD that suppresses ATP-induced release of IL-1ß by monocytic cells. This cascade links activation of P2Y receptors to non-canonical metabotropic functions of nAChRs that inhibit P2X7 receptor function. The biomedical relevance of this mechanism might be the control of trauma-associated systemic inflammation.


Assuntos
Interleucina-1beta/metabolismo , Monócitos/metabolismo , NAD/farmacologia , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Lipopolissacarídeos/farmacologia , Antagonistas Nicotínicos/farmacologia , Inibidores de Fosfolipase A2/farmacologia , Fosfolipases A2/genética , Fosfolipases A2/metabolismo , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Receptores Purinérgicos P2Y/genética , Receptores Purinérgicos P2Y/metabolismo
2.
J Lipid Res ; 58(6): 1055-1066, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28404637

RESUMO

Interleukin (IL)-1ß is a potent pro-inflammatory cytokine of innate immunity involved in host defense. High systemic IL-1ß levels, however, cause life-threatening inflammatory diseases, including systemic inflammatory response syndrome. In response to various danger signals, the pro-form of IL-1ß is synthesized and stays in the cytoplasm unless a second signal, such as extracellular ATP, activates the inflammasome, which enables processing and release of mature IL-1ß. As pulmonary surfactant is known for its anti-inflammatory properties, we hypothesize that surfactant inhibits ATP-induced release of IL-1ß. Lipopolysaccharide-primed monocytic U937 cells were stimulated with an ATP analog in the presence of natural or synthetic surfactant composed of recombinant surfactant protein (rSP)-C, palmitoylphosphatidylglycerol, and dipalmitoylphosphatidylcholine (DPPC). Both surfactant preparations dose-dependently inhibited IL-1ß release from U937 cells. DPPC was the active constituent of surfactant, whereas rSP-C and palmitoylphosphatidylglycerol were inactive. DPPC was also effective in primary mononuclear leukocytes isolated from human blood. Experiments with nicotinic antagonists, siRNA technology, and patch-clamp experiments suggested that stimulation of nicotinic acetylcholine receptors (nAChRs) containing subunit α9 results in a complete inhibition of the ion channel function of ATP receptor, P2X7. In conclusion, the surfactant constituent, DPPC, efficiently inhibits ATP-induced inflammasome activation and maturation of IL-1ß in human monocytes by a mechanism involving nAChRs.


Assuntos
Trifosfato de Adenosina/farmacologia , Interleucina-1beta/metabolismo , Surfactantes Pulmonares/farmacologia , Receptores Nicotínicos/metabolismo , 1,2-Dipalmitoilfosfatidilcolina/metabolismo , Trifosfato de Adenosina/química , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Subunidades Proteicas/metabolismo , Células U937
3.
Physiology (Bethesda) ; 31(2): 117-30, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26889017

RESUMO

Obesity is reaching dramatic proportions in humans and is associated with a higher risk for cardiovascular disease, diabetes, and cognitive alterations, and a higher mortality during infection and inflammation. The focus of the present review is on the influence of obesity on the presentation of fever, sickness behavior, and inflammatory responses during acute systemic inflammation.


Assuntos
Comportamento Animal/fisiologia , Encéfalo/fisiopatologia , Comportamento de Doença/fisiologia , Inflamação/fisiopatologia , Obesidade/complicações , Obesidade/fisiopatologia , Doença Aguda , Animais , Humanos , Inflamação/complicações
4.
Brain Behav Immun ; 48: 147-64, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25813145

RESUMO

The transcription factor nuclear factor interleukin 6 (NF-IL6) plays a pivotal role in neuroinflammation and, as we previously suggested, hypothalamus-pituitary-adrenal-axis-activation. Here, we investigated its contribution to immune-to-brain communication and brain controlled sickness symptoms during lipopolysaccharide (LPS)-induced (50 or 2500 µg/kg i.p.) systemic inflammation in NF-IL6-deficient (KO) or wildtype mice (WT). In WT LPS induced a dose-dependent febrile response and reduction of locomotor activity. While KO developed a normal fever after low-dose LPS-injection the febrile response was almost abolished 3-7 h after a high LPS-dose. High-dose LPS-stimulation was accompanied by decreased (8 h) followed by enhanced (24 h) inflammation in KO compared to WT e.g. hypothalamic mRNA-expression including microsomal prostaglandin E synthase, inducible nitric oxide synthase and further inflammatory mediators, neutrophil recruitment to the brain as well as plasma levels of inflammatory markers such as IL-6 and IL-10. Interestingly, KO showed reduced locomotor activity even under basal conditions, but enhanced locomotor activity to novel environment stress. Hypothalamic-pituitary-adrenal-axis-activity of KO was intact, but tryptophan-metabolizing enzymes were shifted to enhanced serotonin production and reuptake. Overall, we showed for the first time that NF-IL6 plays a dual role for sickness response and immune-to-brain communication: acting pro-inflammatory at 8h but anti-inflammatory at 24 h after onset of the inflammatory response reflecting active natural programming of inflammation. Moreover, reduced locomotor activity observed in KO might be due to altered tryptophan metabolism and serotonin reuptake suggesting some role for NF-IL6 as therapeutic target for depressive disorders.


Assuntos
Encéfalo/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Comportamento de Doença/efeitos dos fármacos , Inflamação/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Proteína delta de Ligação ao Facilitador CCAAT/genética , Relação Dose-Resposta a Droga , Feminino , Sistema Hipotálamo-Hipofisário/metabolismo , Inflamação/induzido quimicamente , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Infiltração de Neutrófilos/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo
5.
Pflugers Arch ; 466(7): 1451-66, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24114176

RESUMO

Although peripherally released interleukin (IL)-10 has a critical regulatory role in limiting fever in mild-to-moderate forms of inflammation, its role in regulating the more complex thermoregulatory manifestations of hypothermia and fever noted during severe inflammation is less clear. Using cytokine antagonism, we therefore investigated the involvement of peripherally released IL-10 in mediating hypothermia, fever and inflammation induced by intraperitoneal (IP) administration of a large dose of lipopolysaccharide (LPS). Male Wistar rats (200-250 g) were anaesthetized and implanted intra-abdominally with temperature-sensitive radiotelemeters. Rats were randomly assigned to receive IL-10 antiserum (IL-10AS) or normal sheep serum IP, 4 h before receiving an IP injection of LPS (10 mg/kg) or phosphate-buffered saline (PBS). Inflammatory responses were measured in plasma and tissue samples (spleen, liver and brain) at 90 min and 6 h after the IP injection of LPS or PBS. Administration of LPS induced an initial period of hypothermia (~90 min) after which fever developed. Pre-treating rats with IL-10AS abolished the LPS-induced increase in plasma IL-10 levels, attenuated the hypothermia and increased the amplitude of the fever. Moreover, IL-10AS pre-treatment augmented the LPS-induced increase in plasma levels of tumor necrosis factor-alpha (90 min and 6 h), IL-1ß (90 min), prostaglandin E2 (90 min) and IL-6 (6 h), in the periphery, but not the hypothalamus, over the duration of hypothermia and fever. Via its action on the synthesis of inflammatory mediators in the spleen and liver, endogenous IL-10 plays a crucial regulatory role in mediating hypothermia and fever during severe aspectic (LPS-induced) systemic inflammation.


Assuntos
Regulação da Temperatura Corporal , Febre/metabolismo , Hipotermia/metabolismo , Interleucina-10/metabolismo , Animais , Encéfalo/metabolismo , Febre/fisiopatologia , Hipotermia/fisiopatologia , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Interleucina-10/sangue , Interleucina-10/genética , Lipopolissacarídeos/toxicidade , Fígado/metabolismo , Masculino , Ratos , Ratos Wistar , Baço/metabolismo , Fator de Necrose Tumoral alfa/sangue
6.
J Neuroinflammation ; 10: 140, 2013 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-24279606

RESUMO

BACKGROUND: The transcription factor nuclear factor interleukin 6 (NF-IL6) is known to be activated by various inflammatory stimuli in the brain. Interestingly, we recently detected NF-IL6-activation within the hypothalamus-pituitary-adrenal (HPA)-axis of rats after systemic lipopolysaccharide (LPS)-injection. Thus, the aim of the present study was to investigate whether NF-IL6 is activated during either, inflammatory, or psychological stress in the rat brain. METHODS: Rats were challenged with either the inflammatory stimulus LPS (100 µg/kg, i.p.) or exposed to a novel environment. Core body temperature (Tb) and motor activity were monitored using telemetry, animals were killed at different time points, brains and blood removed, and primary cell cultures of the anterior pituitary lobe (AL) were investigated. Analyses were performed using immunohistochemistry, RT-PCR, and cytokine-specific bioassays. RESULTS: Stress stimulation by a novel environment increased NF-IL6-immunoreactivity (IR) in the pituitary's perivascular macrophages and hypothalamic paraventricular cells and a rise in Tb lasting approximately 2 h. LPS stimulation lead to NF-IL6-IR in several additional cell types including ACTH-IR-positive corticotrope cells in vivo and in vitro. Two other proinflammatory transcription factors, namely signal transducer and activator of transcription (STAT)3 and NFκB, were significantly activated and partially colocalized with NF-IL6-IR in cells of the AL only after LPS-stimulation, but not following psychological stress. In vitro NF-IL6-activation was associated with induction and secretion of TNFα in folliculostellate cells, which could be antagonized by the JAK-STAT-inhibitor AG490. CONCLUSIONS: We revealed, for the first time, that NF-IL6 activation occurs not only during inflammatory LPS stimulation, but also during psychological stress, that is, a novel environment. Both stressors were associated with time-dependent activation of NF-IL6 in different cell types of the brain and the pituitary. Moreover, while NF-IL6-IR was partially linked to STAT3 and NFκB activation, TNFα production, and ACTH-IR after LPS stimulation; this was not the case after exposure to a novel environment, suggesting distinct underlying signaling pathways. Overall, NF-IL6 can be used as a broad activation marker in the brain and might be of interest for therapeutic approaches not only during inflammatory but also psychological stress.


Assuntos
Encéfalo/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Inflamação/metabolismo , Estresse Psicológico/metabolismo , Animais , Imuno-Histoquímica , Masculino , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Pharmaceuticals (Basel) ; 14(6)2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-34208101

RESUMO

High mobility group box (HMGB)1 action contributes to late phases of sepsis, but the effects of increased endogenous plasma HMGB1 levels on brain cells during inflammation are unclear. Here, we aimed to further investigate the role of HMGB1 in the brain during septic-like lipopolysaccharide-induced inflammation in rats (LPS, 10 mg/kg, i.p.). HMGB-1 mRNA expression and release were measured in the periphery/brain by RT-PCR, immunohistochemistry and ELISA. In vitro experiments with disulfide-HMGB1 in primary neuro-glial cell cultures of the area postrema (AP), a circumventricular organ with a leaky blood-brain barrier and direct access to circulating mediators like HMGB1 and LPS, were performed to determine the direct influence of HMGB1 on this pivotal brain structure for immune-to-brain communication. Indeed, HMGB1 plasma levels stayed elevated after LPS injection. Immunohistochemistry of brains and AP cultures confirmed LPS-stimulated cytoplasmatic translocation of HMGB1 indicative of local HMGB1 release. Moreover, disulfide-HMGB1 stimulation induced nuclear factor (NF)-κB activation and a significant release of interleukin-6, but not tumor necrosis factor α, into AP culture supernatants. However, only a few AP cells directly responded to HMGB1 with increased intracellular calcium concentration. Interestingly, priming with LPS induced a seven-fold higher percentage of responsive cells to HMGB1. We conclude that, as a humoral and local mediator, HMGB1 enhances brain inflammatory responses, after LPS priming, linked to sustained sepsis symptoms.

8.
Front Immunol ; 10: 664, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31019507

RESUMO

Interleukin-1ß (IL-1ß) is a potent, pro-inflammatory cytokine of the innate immune system that plays an essential role in host defense against infection. However, elevated circulating levels of IL-1ß can cause life-threatening systemic inflammation. Hence, mechanisms controlling IL-1ß maturation and release are of outstanding clinical interest. Secretory leukocyte protease inhibitor (SLPI), in addition to its well-described anti-protease function, controls the expression of several pro-inflammatory cytokines on the transcriptional level. In the present study, we tested the potential involvement of SLPI in the control of ATP-induced, inflammasome-dependent IL-1ß maturation and release. We demonstrated that SLPI dose-dependently inhibits the ATP-mediated inflammasome activation and IL-1ß release in human monocytic cells, without affecting the induction of pro-IL-1ß mRNA by LPS. In contrast, the ATP-independent IL-1ß release induced by the pore forming bacterial toxin nigericin is not impaired, and SLPI does not directly modulate the ion channel function of the human P2X7 receptor heterologously expressed in Xenopus laevis oocytes. In human monocytic U937 cells, however, SLPI efficiently inhibits ATP-induced ion-currents. Using specific inhibitors and siRNA, we demonstrate that SLPI activates the calcium-independent phospholipase A2ß (iPLA2ß) and leads to the release of a low molecular mass factor that mediates the inhibition of IL-1ß release. Signaling involves nicotinic acetylcholine receptor subunits α7, α9, α10, and Src kinase activation and results in an inhibition of ATP-induced caspase-1 activation. In conclusion, we propose a novel anti-inflammatory mechanism induced by SLPI, which inhibits the ATP-dependent maturation and secretion of IL-1ß. This novel signaling pathway might lead to development of therapies that are urgently needed for the prevention and treatment of systemic inflammation.


Assuntos
Trifosfato de Adenosina/metabolismo , Interleucina-1beta/metabolismo , Monócitos/citologia , Monócitos/metabolismo , Inibidor Secretado de Peptidases Leucocitárias/genética , Animais , Linhagem Celular , Células Cultivadas , Citocinas/biossíntese , Expressão Gênica , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Inibidor Secretado de Peptidases Leucocitárias/metabolismo , Transdução de Sinais , Quinases da Família src/metabolismo
9.
Front Immunol ; 9: 1604, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30105015

RESUMO

Blood levels of the acute phase reactant C-reactive protein (CRP) are frequently measured as a clinical marker for inflammation, but the biological functions of CRP are still controversial. CRP is a phosphocholine (PC)-binding pentraxin, mainly produced in the liver in response to elevated levels of interleukin-1ß (IL-1ß) and of the IL-1ß-dependent cytokine IL-6. While both cytokines play important roles in host defense, excessive systemic IL-1ß levels can cause life-threatening diseases such as trauma-associated systemic inflammation. We hypothesized that CRP acts as a negative feedback regulator of monocytic IL-1ß maturation and secretion. Here, we demonstrate that CRP, in association with PC, efficiently reduces ATP-induced inflammasome activation and IL-1ß release from human peripheral blood mononuclear leukocytes and monocytic U937 cells. Effective concentrations are in the range of marginally pathologic CRP levels (IC50 = 4.9 µg/ml). CRP elicits metabotropic functions at nicotinic acetylcholine (ACh) receptors (nAChRs) containing subunits α7, α9, and α10 and suppresses the function of ATP-sensitive P2X7 receptors in monocytic cells. Of note, CRP does not induce ion currents at conventional nAChRs, suggesting that CRP is a potent nicotinic agonist controlling innate immunity without entailing the risk of adverse effects in the nervous system. In a prospective study on multiple trauma patients, IL-1ß plasma concentrations negatively correlated with preceding CRP levels, whereas inflammasome-independent cytokines IL-6, IL-18, and TNF-α positively correlated. In conclusion, PC-laden CRP is an unconventional nicotinic agonist that potently inhibits ATP-induced inflammasome activation and might protect against trauma-associated sterile inflammation.


Assuntos
Proteína C-Reativa/imunologia , Inflamassomos/imunologia , Inflamação , Adulto , Idoso , Biomarcadores , Proteína C-Reativa/farmacologia , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Inflamassomos/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Masculino , Pessoa de Meia-Idade , Receptores Nicotínicos/imunologia , Receptores Nicotínicos/metabolismo , Receptores Purinérgicos P2X7/imunologia , Receptores Purinérgicos P2X7/metabolismo
10.
J Basic Clin Physiol Pharmacol ; 28(6): 563-571, 2017 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-28820735

RESUMO

BACKGROUND: Studies with NF-IL6-deficient mice indicate that this transcription factor plays a dual role during systemic inflammation with pro- and anti-inflammatory capacities. Here, we aimed to characterize the role of NF-IL6 specifically within the brain. METHODS: In this study, we tested the capacity of short interfering (si) RNA to silence the inflammatory transcription factor nuclear factor-interleukin 6 (NF-IL6) in brain cells under in vitro and in vivo conditions. RESULTS: In cells of a mixed neuronal and glial primary culture from the rat area postrema (AP), short interfering RNA (siRNA) directed against NF-IL6 strongly reduced basal and lipopolysaccharide (LPS)-induced nuclear immunoreactivity of this transcription factor, with the strongest effect on astrocytes. The siRNA did not exert inflammatory effects in the primary culture as confirmed by unaltered levels of IL-6 in supernatants. In vivo, intracerebroventricular (i.c.v.) injections of fluorochrome labelled siRNA caused its appearance in relevant brain structures for fever induction pathways such as the vascular organ of lamina terminalis, the subfornical organ, the median preoptic nucleus (MnPO) and the AP in several cell types, including microglial cells. However, i.c.v. injections of siRNA per se caused signs of fever, anorexia and reduced locomotor activity, i.e. sickness behavior. CONCLUSIONS: This approach was, thus, not suitable to characterize the role NF-IL6 in the brain in vivo, namely during experimentally induced systemic inflammation.


Assuntos
Encéfalo/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/deficiência , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos , RNA Interferente Pequeno/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Proteína beta Intensificadora de Ligação a CCAAT/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Comportamento de Doença/efeitos dos fármacos , Infusões Intraventriculares , Interleucina-6/metabolismo , Masculino , Neuroglia/efeitos dos fármacos , Neurônios , Cultura Primária de Células , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/metabolismo , Ratos
11.
Neuropharmacology ; 71: 98-111, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23548702

RESUMO

The functional significance for activation of inflammatory transcription factors, such as signal transducer and activator of transcription (STAT3), nuclear factor (NF)κB or NF-interleukin (IL)6 and their contribution to the induction of brain controlled sickness responses, such as fever, during infection and inflammation is unknown. Using AG490, previously shown to inhibit the STAT3- and NF-IL6-signaling pathway, we therefore investigated the central involvement of these two signaling pathways in mediating sickness behavior, fever and accompanying brain inflammation induced by peripheral lipopolysaccharide (LPS)-stimulation. Rats pre-treated i.c.v. with AG490 1 h before the i.p. LPS-challenge (100 µg/kg) showed a modestly exaggerated fever, attenuated adipsia and almost unimpaired locomotor activity compared to LPS-controls receiving vehicle (i.c.v.). The LPS-induced anorexia was not altered and AG490 did not have any effect on rats receiving PBS (i.p.). We did observe effects of AG490 on STAT3-signaling at 4 h, while AG490-mediated changes in brain activity of inflammatory transcription factors at 8 h were not significant. Increased NF-IL6 and suppressor of cytokines 3 mRNA-expression in AG490/LPS-treated rats were indicative of a compensative activation at 24 h. Moreover, a significant decrease in hypothalamic anti-inflammatory IL-10-expression and an increase in inflammatory microsomal prostaglandin E synthase (mPGES) mRNA-expression 8 h after LPS-injection was revealed in AG490 pre-treated animals compared to solvent-treated LPS-controls. In summary, we have shown a dissociation between the effects of central AG490 treatment on fever and components of sickness behavior, which appears to be related to reduced IL-10 and increased mPGES-expression in the brain. Thus, AG490 might have therapeutic potential to reduce sickness behavior.


Assuntos
Encéfalo/efeitos dos fármacos , Encefalite/prevenção & controle , Inibidores Enzimáticos/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Janus Quinases/antagonistas & inibidores , Fatores de Transcrição STAT/antagonistas & inibidores , Tirfostinas/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Regulação da Temperatura Corporal/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Encefalite/imunologia , Encefalite/metabolismo , Encefalite/patologia , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/efeitos adversos , Mediadores da Inflamação/agonistas , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Injeções Intraventriculares , Janus Quinases/metabolismo , Lipopolissacarídeos , Masculino , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Fatores de Transcrição STAT/metabolismo , Tirfostinas/administração & dosagem , Tirfostinas/efeitos adversos
12.
J Comp Neurol ; 519(3): 480-505, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21192080

RESUMO

Rats injected with lipopolysaccharide (LPS) show brain-controlled sickness symptoms, including fever. In these animals, early genomic activation of brain cells was previously monitored by immunohistochemical detection of transcription factors such as nuclear factor (NF)-κB or signal transducer and activator of transcription (STAT)3 and was linked to the initiation or maintenance of the febrile response. To investigate whether NF-IL6 might be another important transcription factor implicated in this kind of immune-to-brain signaling, rats were injected with LPS (100 µg/kg, intraperitoneally) or phosphate-buffered saline, and brains were analyzed by immunohistochemistry, real-time PCR, or Western blot 4, 6, 8, and 10 hours later. Moderate to strong LPS-induced nuclear NF-IL6 immunoreactivity (IR) occurred in a time-dependent manner within circumventricular organs, namely, the vascular organ of the lamina terminalis, the subfornical organ, the area postrema, and the median eminence, brain structures with a leaky blood-brain barrier. Furthermore, nuclear NF-IL6-IR was observed in the pituitary gland, the choroid plexus, and the meninges as well as blood vessels throughout the entire brain. Endothelial, microglial, and ependymal cells, astrocytes, perivascular macrophages, and neurons exhibited LPS-induced nuclear NF-IL6-IR; mRNA levels of NF-IL6, responsive inflammatory genes, and NF-IL6 protein levels were significantly elevated. As opposed to observations on STAT3 or NFκB, the percentage of NF-IL6-reactive cells increased in parallel to late phases of the febrile response. In conclusion, these results suggest a potential role for NF-IL6 in the maintenance or possibly the termination of LPS-induced fever. Moreover, we propose NF-IL6 to be a delayed brain cell activation marker.


Assuntos
Febre/induzido quimicamente , Hipotálamo/fisiologia , Inflamação/imunologia , Interleucina-6/sangue , Lipopolissacarídeos/farmacologia , Animais , Biomarcadores/metabolismo , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Hipotálamo/anatomia & histologia , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/imunologia , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Masculino , Fenótipo , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA