Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 41(24): 5206-5218, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33941650

RESUMO

We examined the signaling route for fever during localized inflammation in male and female mice, elicited by casein injection into a preformed air pouch. The localized inflammation gave rise to high concentrations of prostaglandins of the E species (PGE2) and cytokines in the air pouch and elevated levels of these inflammatory mediators in plasma. There were also elevated levels of PGE2 in the cerebrospinal fluid, although there was little evidence for PGE2 synthesis in the brain. Global deletion of the PGE2 prostaglandin E receptor 3 (EP3) abolished the febrile response as did deletion of the EP3 receptor in neural cells, whereas its deletion on peripheral nerves had no effect, implying that PGE2 action on this receptor in the CNS elicited the fever. Global deletion of the interleukin-1 receptor type 1 (IL-1R1) also abolished the febrile response, whereas its deletion on neural cells or peripheral nerves had no effect. However, deletion of the IL-1R1 on brain endothelial cells, as well as deletion of the interleukin-6 receptor α on these cells, attenuated the febrile response. In contrast, deletion of the PGE2 synthesizing enzymes cyclooxygenase-2 and microsomal prostaglandin synthase-1 in brain endothelial cells, known to attenuate fever evoked by systemic inflammation, had no effect. We conclude that fever during localized inflammation is not mediated by neural signaling from the inflamed site, as previously suggested, but is dependent on humoral signaling that involves interleukin actions on brain endothelial cells, probably facilitating PGE2 entry into the brain from the circulation and hence representing a mechanism distinct from that at work during systemic inflammation.


Assuntos
Encéfalo/metabolismo , Endotélio/metabolismo , Febre/metabolismo , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Animais , Feminino , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
2.
Mol Metab ; 39: 101022, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32446877

RESUMO

OBJECTIVES: Infections, cancer, and systemic inflammation elicit anorexia. Despite the medical significance of this phenomenon, the question of how peripheral inflammatory mediators affect the central regulation of food intake is incompletely understood. Therefore, we have investigated the sickness behavior induced by the prototypical inflammatory mediator IL-1ß. METHODS: IL-1ß was injected intravenously. To interfere with IL-1ß signaling, we deleted the essential modulator of NF-κB signaling (Nemo) in astrocytes and tanycytes. RESULTS: Systemic IL-1ß increased the activity of the transcription factor NF-κB in tanycytes of the mediobasal hypothalamus (MBH). By activating NF-κB signaling, IL-1ß induced the expression of cyclooxygenase-2 (Cox-2) and stimulated the release of the anorexigenic prostaglandin E2 (PGE2) from tanycytes. When we deleted Nemo in astrocytes and tanycytes, the IL-1ß-induced anorexia was alleviated whereas the fever response and lethargy response were unchanged. Similar results were obtained after the selective deletion of Nemo exclusively in tanycytes. CONCLUSIONS: Tanycytes form the brain barrier that mediates the anorexic effect of systemic inflammation in the hypothalamus.


Assuntos
Anorexia/etiologia , Células Ependimogliais/metabolismo , Inflamação/complicações , Inflamação/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Animais , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Hibridização In Situ , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Ratos
3.
J Clin Invest ; 128(7): 3160-3170, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29911992

RESUMO

It is critical for survival to assign positive or negative valence to salient stimuli in a correct manner. Accordingly, harmful stimuli and internal states characterized by perturbed homeostasis are accompanied by discomfort, unease, and aversion. Aversive signaling causes extensive suffering during chronic diseases, including inflammatory conditions, cancer, and depression. Here, we investigated the role of melanocortin 4 receptors (MC4Rs) in aversive processing using genetically modified mice and a behavioral test in which mice avoid an environment that they have learned to associate with aversive stimuli. In normal mice, robust aversions were induced by systemic inflammation, nausea, pain, and κ opioid receptor-induced dysphoria. In sharp contrast, mice lacking MC4Rs displayed preference or indifference toward the aversive stimuli. The unusual flip from aversion to reward in mice lacking MC4Rs was dopamine dependent and associated with a change from decreased to increased activity of the dopamine system. The responses to aversive stimuli were normalized when MC4Rs were reexpressed on dopamine D1 receptor-expressing cells or in the striatum of mice otherwise lacking MC4Rs. Furthermore, activation of arcuate nucleus proopiomelanocortin neurons projecting to the ventral striatum increased the activity of striatal neurons in an MC4R-dependent manner and elicited aversion. Our findings demonstrate that melanocortin signaling through striatal MC4Rs is critical for assigning negative motivational valence to harmful stimuli.


Assuntos
Corpo Estriado/fisiologia , Motivação/fisiologia , Receptor Tipo 4 de Melanocortina/fisiologia , Animais , Aprendizagem da Esquiva/fisiologia , Comportamento Animal/fisiologia , Benzazepinas/administração & dosagem , Corpo Estriado/efeitos dos fármacos , Dopamina/fisiologia , Antagonistas de Dopamina/administração & dosagem , Feminino , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Pró-Opiomelanocortina/fisiologia , Receptor Tipo 4 de Melanocortina/deficiência , Receptor Tipo 4 de Melanocortina/genética , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/fisiologia , Recompensa
4.
FASEB J ; 32(10): 5751-5759, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29738273

RESUMO

The mode of action of paracetamol (acetaminophen), which is widely used for treating pain and fever, has remained obscure, but may involve several distinct mechanisms, including cyclooxygenase inhibition and transient receptor potential ankyrin 1 (TRPA1) channel activation, the latter being recently associated with paracetamol's propensity to elicit hypothermia at higher doses. Here, we examined whether the antipyretic effect of paracetamol was due to TRPA1 activation or cyclooxygenase inhibition. Treatment of wild-type and TRPA1 knockout mice rendered febrile by immune challenge with LPS with a dose of paracetamol that did not produce hypothermia (150 mg/kg) but is known to be analgetic, abolished fever in both genotypes. Paracetamol completely suppressed the LPS-induced elevation of prostaglandin E2 in the brain and also reduced the levels of several other prostanoids. The hypothermia induced by paracetamol was abolished in mice treated with the electrophile-scavenger N-acetyl cysteine. We conclude that paracetamol's antipyretic effect in mice is dependent on inhibition of cyclooxygenase activity, including the formation of pyrogenic prostaglandin E2, whereas paracetamol-induced hypothermia likely is mediated by the activation of TRPA1 by electrophilic metabolites of paracetamol, similar to its analgesic effect in some experimental paradigms.-Mirrasekhian, E., Nilsson, J. L. Å., Shionoya, K., Blomgren, A., Zygmunt, P. M., Engblom, D., Högestätt, E. D., Blomqvist, A. The antipyretic effect of paracetamol occurs independent of transient receptor potential ankyrin 1-mediated hypothermia and is associated with prostaglandin inhibition in the brain.


Assuntos
Acetaminofen/efeitos adversos , Antipiréticos/efeitos adversos , Encéfalo/metabolismo , Dinoprostona/biossíntese , Hipotermia/metabolismo , Canal de Cátion TRPA1/biossíntese , Acetaminofen/farmacologia , Animais , Antipiréticos/farmacologia , Encéfalo/patologia , Hipotermia/induzido quimicamente , Hipotermia/patologia , Camundongos , Camundongos Knockout
5.
Brain Behav Immun ; 66: 165-176, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28655587

RESUMO

Sickness responses to lipopolysaccharide (LPS) were examined in mice with deletion of the interleukin (IL)-1 type 1 receptor (IL-1R1). IL-1R1 knockout (KO) mice displayed intact anorexia and HPA-axis activation to intraperitoneally injected LPS (anorexia: 10 or 120µg/kg; HPA-axis: 120µg/kg), but showed attenuated but not extinguished fever (120µg/kg). Brain PGE2 synthesis was attenuated, but Cox-2 induction remained intact. Neither the tumor necrosis factor-α (TNFα) inhibitor etanercept nor the IL-6 receptor antibody tocilizumab abolished the LPS induced fever in IL-1R1 KO mice. Deletion of IL-1R1 specifically in brain endothelial cells attenuated the LPS induced fever, but only during the late, 3rd phase of fever, whereas deletion of IL-1R1 on neural cells or on peripheral nerves had little or no effect on the febrile response. We conclude that while IL-1 signaling is not critical for LPS induced anorexia or stress hormone release, IL-1R1, expressed on brain endothelial cells, contributes to the febrile response to LPS. However, also in the absence of IL-1R1, LPS evokes a febrile response, although this is attenuated. This remaining fever seems not to be mediated by IL-6 receptors or TNFα, but by some yet unidentified pyrogenic factor.


Assuntos
Anorexia/metabolismo , Febre/metabolismo , Comportamento de Doença , Receptores Tipo I de Interleucina-1/metabolismo , Hormônio Adrenocorticotrópico/sangue , Animais , Anorexia/induzido quimicamente , Encéfalo/metabolismo , Corticosterona/sangue , Ingestão de Alimentos , Células Endoteliais/metabolismo , Feminino , Febre/induzido quimicamente , Hipotálamo/metabolismo , Inflamação/sangue , Inflamação/complicações , Mediadores da Inflamação/sangue , Lipopolissacarídeos/administração & dosagem , Masculino , Camundongos Knockout , Receptores Tipo I de Interleucina-1/genética
6.
J Cachexia Sarcopenia Muscle ; 8(3): 417-427, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28025863

RESUMO

BACKGROUND: The cancer-anorexia-cachexia syndrome (CACS) negatively affects survival and therapy success in cancer patients. Inflammatory mediators and tumour-derived factors are thought to play an important role in the aetiology of CACS. However, the central and peripheral mechanisms contributing to CACS are insufficiently understood. The area postrema (AP) and the nucleus tractus solitarii are two important brainstem centres for the control of eating during acute sickness conditions. Recently, the tumour-derived macrophage inhibitory cytokine-1 (MIC-1) emerged as a possible mediator of cancer anorexia because lesions of these brainstem areas attenuated the anorectic effect of exogenous MIC-1 in mice. METHODS: Using a rat hepatoma tumour model, we examined the roles of the AP and of vagal afferents in the mediation of CACS. Specifically, we investigated whether a lesion of the AP (APX) or subdiaphragmatic vagal deafferentation (SDA) attenuate anorexia, body weight, muscle, and fat loss. Moreover, we analysed MIC-1 levels in this tumour model and their correlation with tumour size and the severity of the anorectic response. RESULTS: In tumour-bearing sham-operated animals mean daily food intake significantly decreased. The anorectic response was paralleled by a significant loss of body weight and muscle mass. APX rats were protected against anorexia, body weight loss, and muscle atrophy after tumour induction. In contrast, subdiaphragmatic vagal deafferentation did not attenuate cancer-induced anorexia or body weight loss. Tumour-bearing rats had substantially increased MIC-1 levels, which positively correlated with tumour size and cancer progression and negatively correlated with food intake. CONCLUSIONS: These findings demonstrate the importance of the AP in the mediation of cancer-dependent anorexia and body weight loss and support a pathological role of MIC-1 as a tumour-derived factor mediating CACS, possibly via an AP-dependent action.


Assuntos
Anorexia/etiologia , Anorexia/metabolismo , Área Postrema/metabolismo , Caquexia/etiologia , Caquexia/metabolismo , Fator 15 de Diferenciação de Crescimento/metabolismo , Neoplasias Hepáticas/complicações , Nervo Vago/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Composição Corporal , Peso Corporal , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Metabolismo Energético , Xenoenxertos , Neoplasias Hepáticas/patologia , Masculino , Atividade Motora , Músculo Esquelético/inervação , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Ratos
7.
J Neurosci ; 34(48): 15957-61, 2014 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-25429137

RESUMO

The cytokine IL-6, which is released upon peripheral immune challenge, is critical for the febrile response, but the mechanism by which IL-6 is pyrogenic has remained obscure. Here we generated mice with deletion of the membrane bound IL-6 receptor α (IL-6Rα) on neural cells, on peripheral nerves, on fine sensory afferent fibers, and on brain endothelial cells, respectively, and examined its role for the febrile response to peripherally injected lipopolysaccharide. We show that IL-6Rα on neural cells, peripheral nerves, and fine sensory afferents are dispensable for the lipopolysaccharide-induced fever, whereas IL-6Rα in the brain endothelium plays an important role. Hence deletion of IL-6Rα on brain endothelial cells strongly attenuated the febrile response, and also led to reduced induction of the prostaglandin synthesizing enzyme Cox-2 in the hypothalamus, the temperature-regulating center in the brain, as well as reduced expression of SOCS3, suggesting involvement of the STAT signaling pathway. Furthermore, deletion of STAT3 in the brain endothelium also resulted in attenuated fever. These data show that IL-6, when endogenously released during systemic inflammation, is pyrogenic by binding to IL-6Rα on brain endothelial cells to induce prostaglandin synthesis in these cells, probably in concerted action with other peripherally released cytokines.


Assuntos
Encéfalo/metabolismo , Células Endoteliais/metabolismo , Febre/metabolismo , Subunidade alfa de Receptor de Interleucina-6/deficiência , Prostaglandinas/biossíntese , Fator de Transcrição STAT3/deficiência , Animais , Encéfalo/efeitos dos fármacos , Feminino , Febre/induzido quimicamente , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/fisiologia , Interleucina-6/metabolismo , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
PLoS One ; 9(7): e103479, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25078278

RESUMO

BACKGROUND: The endocannabinoid ligand anandamide (AEA) is removed from the extracellular space by a process of cellular uptake followed by metabolism. In many cells, such as the RBL-2H3 cell line, inhibition of FAAH activity reduces the observed uptake, indicating that the enzyme regulates uptake by controlling the intra- : extracellular AEA concentration gradient. However, in other FAAH-expressing cells, no such effect is seen. It is not clear, however, whether these differences are methodological in nature or due to properties of the cells themselves. In consequence, we have reinvestigated the role of FAAH in gating the uptake of AEA. METHODOLOGY/PRINCIPAL FINDINGS: The effects of FAAH inhibition upon AEA uptake were investigated in four cell lines: AT1 rat prostate cancer, RBL-2H3 rat basophilic leukaemia, rat C6 glioma and mouse P19 embryonic carcinoma cells. Semi-quantitative PCR for the cells and for a rat brain lysate confirmed the expression of FAAH. No obvious expression of a transcript with the expected molecular weight of FLAT was seen. FAAH expression differed between cells, but all four could accumulate AEA in a manner inhibitable by the selective FAAH inhibitor URB597. However, there was a difference in the sensitivities seen in the reduction of uptake for a given degree of FAAH inhibition produced by a reversible FAAH inhibitor, with C6 cells being more sensitive than RBL-2H3 cells, despite rather similar expression levels and activities of FAAH. The four cell lines all expressed FABP5, and AEA uptake was reduced in the presence of the FABP5 inhibitor SB-FI-26, suggesting that the different sensitivities to FAAH inhibition for C6 and RBL2H3 cells is not due to differences at the level of FABP-5. CONCLUSIONS/SIGNIFICANCE: When assayed using the same methodology, different FAAH-expressing cells display different sensitivities of uptake to FAAH inhibition.


Assuntos
Amidoidrolases/metabolismo , Ácidos Araquidônicos/metabolismo , Endocanabinoides/metabolismo , Proteínas do Olho/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Alcamidas Poli-Insaturadas/metabolismo , Animais , Ácidos Araquidônicos/farmacologia , Linhagem Celular Tumoral , Endocanabinoides/farmacologia , Reação em Cadeia da Polimerase , Alcamidas Poli-Insaturadas/farmacologia , Ratos
9.
Brain Behav Immun ; 40: 166-73, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24681250

RESUMO

The proinflammatory cytokine interleukin-1ß (IL-1ß) plays a major role in the signal transduction of immune stimuli from the periphery to the central nervous system, and has been shown to be an important mediator of the immune-induced stress hormone release. The signaling pathway by which IL-1ß exerts this function involves the blood-brain-barrier and induced central prostaglandin synthesis, but the identity of the blood-brain-barrier cells responsible for this signal transduction has been unclear, with both endothelial cells and perivascular macrophages suggested as critical components. Here, using an irradiation and transplantation strategy, we generated mice expressing IL-1 type 1 receptors (IL-1R1) either in hematopoietic or non-hematopoietic cells and subjected these mice to peripheral immune challenge with IL-1ß. Following both intraperitoneal and intravenous administration of IL-1ß, mice lacking IL-1R1 in hematopoietic cells showed induced expression of the activity marker c-Fos in the paraventricular hypothalamic nucleus, and increased plasma levels of ACTH and corticosterone. In contrast, these responses were not observed in mice with IL-1R1 expression only in hematopoietic cells. Immunoreactivity for IL-1R1 was detected in brain vascular cells that displayed induced expression of the prostaglandin synthesizing enzyme cyclooxygenase-2 and that were immunoreactive for the endothelial cell marker CD31, but was not seen in cells positive for the brain macrophage marker CD206. These results imply that activation of the HPA-axis by IL-1ß is dependent on IL-1R1s on non-hematopoietic cells, such as brain endothelial cells, and that IL-1R1 on perivascular macrophages are not involved.


Assuntos
Células da Medula Óssea/imunologia , Sistema Hipotálamo-Hipofisário/imunologia , Interleucina-1beta/farmacologia , Sistema Hipófise-Suprarrenal/imunologia , Receptores de Interleucina-1/metabolismo , Hormônio Adrenocorticotrópico/sangue , Animais , Células da Medula Óssea/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Corticosterona/sangue , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Receptores de Interleucina-1/genética
10.
Front Neuroendocrinol ; 34(4): 329-49, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23817054

RESUMO

Gonadal hormones not only play a pivotal role in reproductive behavior and sexual differentiation, they also contribute to thermoregulation, feeding, memory, neuronal survival, and the perception of somatosensory stimuli. Numerous studies on both animals and human subjects have also demonstrated the potential effects of gonadal hormones, such as estrogens, on pain transmission. These effects most likely involve multiple neuroanatomical circuits as well as diverse neurochemical systems and they therefore need to be evaluated specifically to determine the localization and intrinsic characteristics of the neurons engaged. The aim of this review is to summarize the morphological as well as biochemical evidence in support for gonadal hormone modulation of nociceptive processing, with particular focus on estrogens and spinal cord mechanisms.


Assuntos
Encéfalo/metabolismo , Estrogênios/metabolismo , Dor/metabolismo , Medula Espinal/metabolismo , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Humanos , Neurônios/metabolismo , Dor/fisiopatologia , Receptores de Estrogênio/metabolismo , Medula Espinal/patologia
11.
Brain Behav Immun ; 33: 123-30, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23827828

RESUMO

Interleukin-6 (IL-6) is critical for the lipopolysaccharide (LPS)-induced febrile response. However, the exact source(s) of IL-6 involved in regulating the LPS-elicited fever is still to be identified. One known source of IL-6 is hematopoietic cells, such as monocytes. To clarify the contribution of hematopoietically derived IL-6 to fever, we created chimeric mice expressing IL-6 selectively either in cells of hematopoietic or, conversely, in cells of non-hematopoietic origin. This was performed by extinguishing hematopoietic cells in wild-type (WT) or IL-6 knockout (IL-6 KO) mice by whole-body irradiation and transplanting them with new stem cells. Mice on a WT background but lacking IL-6 in hematopoietic cells displayed normal fever to LPS and were found to have similar levels of IL-6 protein in the cerebrospinal fluid (CSF) and in plasma and of IL-6 mRNA in the brain as WT mice. In contrast, mice on an IL-6 KO background, but with intact IL-6 production in cells of hematopoietic origin, only showed a minor elevation of the body temperature after peripheral LPS injection. While they displayed significantly elevated levels of IL-6 both in plasma and CSF compared with control mice, the increase was modest compared with that seen in LPS injected mice on a WT background, the latter being approximately 20 times larger in magnitude. These results suggest that IL-6 of non-hematopoietic origin is the main source of IL-6 in LPS-induced fever, and that IL-6 produced by hematopoietic cells only plays a minor role.


Assuntos
Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Febre/imunologia , Hematopoese/imunologia , Interleucina-6/biossíntese , Interleucina-6/fisiologia , Lipopolissacarídeos/farmacologia , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea/métodos , Feminino , Febre/genética , Febre/patologia , Raios gama , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/sangue , Proteínas de Fluorescência Verde/genética , Hematopoese/genética , Interleucina-6/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Quimera por Radiação , Distribuição Aleatória
12.
FASEB J ; 27(5): 1973-80, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23395911

RESUMO

Loss of appetite is a hallmark of inflammatory diseases. The underlying mechanisms remain undefined, but it is known that myeloid differentiation primary response gene 88 (MyD88), an adaptor protein critical for Toll-like and IL-1 receptor family signaling, is involved. Here we addressed the question of determining in which cells the MyD88 signaling that results in anorexia development occurs by using chimeric mice and animals with cell-specific deletions. We found that MyD88-knockout mice, which are resistant to bacterial lipopolysaccharide (LPS)-induced anorexia, displayed anorexia when transplanted with wild-type bone marrow cells. Furthermore, mice with a targeted deletion of MyD88 in hematopoietic or myeloid cells were largely protected against LPS-induced anorexia and displayed attenuated weight loss, whereas mice with MyD88 deletion in hepatocytes or in neural cells or the cerebrovascular endothelium developed anorexia and weight loss of similar magnitude as wild-type mice. Furthermore, in a model for cancer-induced anorexia-cachexia, deletion of MyD88 in hematopoietic cells attenuated the anorexia and protected against body weight loss. These findings demonstrate that MyD88-dependent signaling within the brain is not required for eliciting inflammation-induced anorexia. Instead, we identify MyD88 signaling in hematopoietic/myeloid cells as a critical component for acute inflammatory-driven anorexia, as well as for chronic anorexia and weight loss associated with malignant disease.


Assuntos
Anorexia/fisiopatologia , Encéfalo/citologia , Caquexia/fisiopatologia , Células Endoteliais/fisiologia , Inflamação/fisiopatologia , Células Mieloides/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Sarcoma Experimental/fisiopatologia , Animais , Quimera/fisiologia , Metilcolantreno , Camundongos , Camundongos Knockout , Neurônios/citologia , Sarcoma Experimental/induzido quimicamente , Transdução de Sinais/fisiologia , Redução de Peso/fisiologia
13.
Brain Behav Immun ; 29: 124-135, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23305935

RESUMO

It is well-established that prostaglandins (PGs) affect tumorigenesis, and evidence indicates that PGs also are important for the reduced food intake and body weight loss, the anorexia-cachexia syndrome, in malignant cancer. However, the identity of the PGs and the PG producing cyclooxygenase (COX) species responsible for cancer anorexia-cachexia is unknown. Here, we addressed this issue by transplanting mice with a tumor that elicits anorexia. Meal pattern analysis revealed that the anorexia in the tumor-bearing mice was due to decreased meal frequency. Treatment with a non-selective COX inhibitor attenuated the anorexia, and also tumor growth. When given at manifest anorexia, non-selective COX-inhibitors restored appetite and prevented body weight loss without affecting tumor size. Despite COX-2 induction in the cerebral blood vessels of tumor-bearing mice, a selective COX-2 inhibitor had no effect on the anorexia, whereas selective COX-1 inhibition delayed its onset. Tumor growth was associated with robust increase of PGE(2) levels in plasma - a response blocked both by non-selective COX-inhibition and by selective COX-1 inhibition, but not by COX-2 inhibition. However, there was no increase in PGE(2)-levels in the cerebrospinal fluid. Neutralization of plasma PGE(2) with specific antibodies did not ameliorate the anorexia, and genetic deletion of microsomal PGE synthase-1 (mPGES-1) affected neither anorexia nor tumor growth. Furthermore, tumor-bearing mice lacking EP(4) receptors selectively in the nervous system developed anorexia. These observations suggest that COX-enzymes, most likely COX-1, are involved in cancer-elicited anorexia and weight loss, but that these phenomena occur independently of host mPGES-1, PGE(2) and neuronal EP(4) signaling.


Assuntos
Anorexia/enzimologia , Anorexia/etiologia , Ciclo-Oxigenase 1/genética , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/psicologia , Animais , Anorexia/tratamento farmacológico , Temperatura Corporal/fisiologia , Ciclo-Oxigenase 1/biossíntese , Ciclo-Oxigenase 2/fisiologia , Inibidores de Ciclo-Oxigenase/farmacologia , DNA Complementar/biossíntese , DNA Complementar/genética , Dinoprostona/sangue , Dinoprostona/líquido cefalorraquidiano , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Feminino , Imuno-Histoquímica , Oxirredutases Intramoleculares/biossíntese , Masculino , Camundongos , Neoplasias Experimentais/complicações , Prostaglandina-E Sintases , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Prostaglandina E Subtipo EP4/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
14.
Endocrinology ; 153(10): 4849-61, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22872578

RESUMO

Immune-induced prostaglandin E2 (PGE2) synthesis is critical for fever and other centrally elicited disease symptoms. The production of PGE2 depends on cyclooxygenase-2 and microsomal prostaglandin E synthase-1 (mPGES-1), but the identity of the cells involved has been a matter of controversy. We generated mice expressing mPGES-1 either in cells of hematopoietic or nonhematopoietic origin. Mice lacking mPGES-1 in hematopoietic cells displayed an intact febrile response to lipopolysaccharide, associated with elevated levels of PGE2 in the cerebrospinal fluid. In contrast, mice that expressed mPGES-1 only in hematopoietic cells, although displaying elevated PGE2 levels in plasma but not in the cerebrospinal fluid, showed no febrile response to lipopolysaccharide, thus pointing to the critical role of brain-derived PGE2 for fever. Immunohistochemical stainings showed that induced cyclooxygenase-2 expression in the brain exclusively occurred in endothelial cells, and quantitative PCR analysis on brain cells isolated by flow cytometry demonstrated that mPGES-1 is induced in endothelial cells and not in vascular wall macrophages. Similar analysis on liver cells showed induced expression in macrophages and not in endothelial cells, pointing at the distinct role for brain endothelial cells in PGE2 synthesis. These results identify the brain endothelial cells as the PGE2-producing cells critical for immune-induced fever.


Assuntos
Encéfalo/metabolismo , Dinoprostona/biossíntese , Células Endoteliais/metabolismo , Febre/metabolismo , Lipopolissacarídeos/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Febre/induzido quimicamente , Febre/imunologia , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prostaglandina-E Sintases
15.
J Pain ; 13(5): 459-66, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22480441

RESUMO

UNLABELLED: We examined the relationship between estrogen and pain in women undergoing in vitro fertilization (IVF). Quantitative sensory tests (QST) were performed twice during the IVF-regimen: once during hormonal down-regulation and once during hormonal up-regulation. A group of healthy men and a group of women using monophasic contraceptives were also examined, to control for session-to-session effects. Among the women undergoing IVF, serum 17ß-estradiol levels differed strongly between treatments as expected, and increased from 65.7 (SD = 26) pmol/L during the down-regulation phase, to 5,188 (SD = 2,524) pmol/L during the up-regulation phase. Significant outcomes in the QST were only seen for temperature perception thresholds (1.7 °C versus 2.2 °C; P = .003) and cold pain threshold (11.5 °C versus 14.5 °C; P = .04). A similar change in cold pain threshold was also seen in the 2 control groups, however, and statistical analysis suggested that this change was due to a session-to-session effect rather than being the result of hormonal modulation. Heat pain thresholds, heat tolerance, pressure pain thresholds, and the cold pressor test showed no significant differences between sessions. These data demonstrate that pain perception and pain thresholds in healthy women show little, if any, changes even with major variations in serum estradiol levels. PERSPECTIVE: This study shows that pain perception and tolerance in women undergoing in vitro fertilization do not vary, despite the dramatic changes in 17ß-estradiol levels induced by the treatment regimen. The result thus suggests that in humans, contrary to experimental animals, changes in estrogen levels have little influence on pain sensitivity.


Assuntos
Estradiol/sangue , Fertilização in vitro/efeitos adversos , Percepção da Dor/fisiologia , Limiar da Dor/fisiologia , Dor/sangue , Dor/etiologia , Adulto , Análise de Variância , Temperatura Baixa/efeitos adversos , Regulação para Baixo , Feminino , Humanos , Masculino , Dor/psicologia , Medição da Dor , Fatores Sexuais , Regulação para Cima
16.
Rheumatology (Oxford) ; 50(3): 544-51, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21078629

RESUMO

OBJECTIVES: FM is a condition that preferentially affects women. Sex hormones, and in particular oestrogens, have been shown to affect pain processing and pain sensitivity, and oestrogen deficit has been considered a potentially promoting factor for FM. However, the effects of oestrogen treatment in patients suffering from FM have not been studied. Here, we examined the effect of transdermal oestrogen substitution treatment on experimental as well as self-estimated pain in women suffering from FM. METHODS: Twenty-nine post-menopausal women were randomized to either 8 weeks of treatment with transdermal 17ß-oestradiol (50 µg/day) or placebo according to a double-blind protocol. A self-estimation of pain, a set of quantitative sensory tests measuring thresholds to temperature, thermal pain, cold pain and pressure pain, and a cold pressor test were performed on three occasions: before treatment, after 8 weeks of treatment and 20 weeks after cessation of treatment. RESULTS: Hormonal replacement treatment significantly increased serum oestradiol levels as expected (P < 0.01). However, no differences in self-estimated pain were seen between treatment and placebo groups, nor were there any differences between the two groups regarding the results of the quantitative sensory tests or the cold pressor test at any of the examined time points. CONCLUSION: Eight weeks of transdermal oestradiol treatment does not influence perceived pain, pain thresholds or pain tolerance as compared with placebo treatment in post-menopausal women suffering from FM. TRIAL REGISTRATION: ClinicalTrials.gov Registration; http://www.clinicaltrials.gov; NCT01087593.


Assuntos
Terapia de Reposição de Estrogênios , Estrogênios/administração & dosagem , Fibromialgia/tratamento farmacológico , Dor/tratamento farmacológico , Administração Cutânea , Método Duplo-Cego , Estradiol/administração & dosagem , Feminino , Fibromialgia/fisiopatologia , Humanos , Pessoa de Meia-Idade , Dor/diagnóstico , Medição da Dor , Limiar da Dor , Pós-Menopausa
17.
Brain Behav Immun ; 24(4): 554-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20093176

RESUMO

The anorexia-cachexia syndrome, characterized by a rise in energy expenditure and loss of body weight that paradoxically are associated with loss of appetite and decreased food intake, contributes significantly to the morbidity and mortality in cancer. While the pathophysiology of cancer anorexia-cachexia is poorly understood, evidence indicates that pro-inflammatory cytokines are key mediators of this response. Although inflammation hence is recognized as an important component of cancer anorexia-cachexia, the molecular pathways involved are largely unknown. We addressed this issue in mice carrying a deletion of the gene encoding MyD88, the key intracellular adaptor molecule in Toll-like and interleukin-1 family receptor signaling. Wild-type and MyD88-deficient mice were transplanted subcutaneously with a syngenic methylcholanthrene-induced tumor (MCG 101) and daily food intake and body weight were recorded. Wild-type mice showed progressively reduced food intake from about 5days after tumor transplantation and displayed a slight body weight loss after 10days when the experiment was terminated. In contrast, MyD88-deficient mice did not develop anorexia, and displayed a positive body weight development during the observation period. While the MyD88-deficient mice on average developed somewhat smaller tumors than wild-type mice, this did not explain the absence of anorexia, because anorexia was seen in wild-type mice with similar tumor mass as non-anorexic knock-out mice. These data suggest that MyD88-dependent mechanisms are involved in the metabolic derangement during cancer anorexia-cachexia and that innate immune signaling is important for the development of this syndrome.


Assuntos
Anorexia/imunologia , Anorexia/fisiopatologia , Peso Corporal/imunologia , Ingestão de Alimentos/imunologia , Fator 88 de Diferenciação Mieloide/genética , Neoplasias/fisiopatologia , Deleção de Sequência , Animais , Apetite/imunologia , Caquexia/imunologia , Caquexia/fisiopatologia , Modelos Animais de Doenças , Feminino , Metilcolantreno , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Síndrome , Transplante Isogênico , Redução de Peso/imunologia
18.
Eur J Pain ; 14(3): 245-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19525133

RESUMO

Estrogens exert a substantial influence on the transmission of nociceptive stimuli and the susceptibility to pain disorders as made evident by studies in both animals and human subjects. The estrogen receptor (ER) seems to be of crucial importance to the cellular mechanisms underlying such an influence. However, it has not been clarified whether nociceptive neurons activated by pain express ERs. In this study, a noxious injection of formalin was given into the lower lip of female rats, thereby activating nociceptive neurons in the trigeminal subnucleus caudalis as demonstrated by immunohistochemical labeling of Fos. Using a dual-label immunohistochemistry protocol ERalpha-containing cells were visualized in the same sections. In the superficial layers of the medullary dorsal horn, 12% of ERalpha-labeled cells, mainly located in lamina II, also expressed noxious-induced Fos. These findings show that nociceptive-responsive neurons in the medullary dorsal horn express ERalpha, thus providing a possible morphological basis for the hypothesis that estrogens directly regulate pain transmission at this level.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Nociceptores/metabolismo , Células do Corno Posterior/metabolismo , Núcleo Inferior Caudal do Nervo Trigêmeo/metabolismo , Animais , Feminino , Imuno-Histoquímica , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley
19.
Endocrinology ; 150(4): 1850-60, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19022895

RESUMO

Fever has been shown to be elicited by prostaglandin E(2) (PGE(2)) binding to its receptors on thermoregulatory neurons in the anterior hypothalamus. The signals that trigger PGE(2) production are thought to include proinflammatory cytokines, such as IL-6. However, although the presence of IL-6 is critical for fever, IL-6 by itself is not or only weakly pyrogenic. Here we examined the relationship between IL-6 and PGE(2) in lipopolysaccharide (LPS)-induced fever. Immune-challenged IL-6 knockout mice did not produce fever, in contrast to wild-type mice, but the expression of the inducible PGE(2)-synthesizing enzymes, cyclooxygenase-2 and microsomal prostaglandin E synthase-1, was similarly up-regulated in the hypothalamus of both genotypes, which also displayed similarly elevated PGE(2) levels in the cerebrospinal fluid. Nevertheless, both wild-type and knockout mice displayed a febrile response to graded concentrations of PGE(2) injected into the lateral ventricle. There was no major genotype difference in the expression of IL-1beta and TNFalpha or their receptors, and pretreatment of IL-6 knockout mice with soluble TNFalpha receptor ip or intracerebroventricularly or a cyclooxygenase-2 inhibitor ip did not abolish the LPS unresponsiveness. Hence, although IL-6 knockout mice have both an intact PGE(2) synthesis and an intact fever-generating pathway downstream of PGE(2), endogenously produced PGE(2) is not sufficient to produce fever in the absence of IL-6. The findings suggest that IL-6 controls some factor(s) in the inflammatory cascade, which render(s) IL-6 knockout mice refractory to the pyrogenic action of PGE(2), or that it is involved in the mechanisms that govern release of synthesized PGE(2) onto its target neurons.


Assuntos
Dinoprostona/metabolismo , Dinoprostona/farmacologia , Febre/induzido quimicamente , Interleucina-6/farmacologia , Interleucina-6/fisiologia , Lipopolissacarídeos/farmacologia , Animais , Temperatura Corporal/efeitos dos fármacos , Encéfalo/metabolismo , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Genótipo , Imunoensaio , Imuno-Histoquímica , Interleucina-6/genética , Isoxazóis/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/farmacologia , Vísceras/metabolismo
20.
Endocrinology ; 149(4): 1436-50, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18174279

RESUMO

There is evidence from in vitro studies that inflammatory messengers influence the release of stress hormone via direct effects on the adrenal gland; however, the mechanisms underlying these effects in the intact organism are unknown. Here we demonstrate that systemic inflammation in rats elicited by iv injection of lipopolysaccharide results in dynamic changes in the adrenal immune cell population, implying a rapid depletion of dendritic cells in the inner cortical layer and the recruitment of immature cells to the outer layers. These changes are accompanied by an induced production of IL-1beta and IL-1 receptor type 1 as well as cyclooxygenase-2 and microsomal prostaglandin E synthase-1 in these cells, implying local cytokine-mediated prostaglandin E(2) production in the adrenals, which also displayed prostaglandin E(2) receptors of subtypes 1 and 3 in the cortex and medulla. The IL-1beta expression was also induced by systemically administrated IL-1beta and was in both cases attenuated by IL-1 receptor antagonist, consistent with an autocrine signaling loop. IL-1beta similarly induced expression of cyclooxygenase-2, but the cyclooxygenase-2 expression was, in contrast, further enhanced by IL-1 receptor antagonist. These data demonstrate a mechanism by which systemic inflammatory agents activate an intrinsically regulated local signaling circuit that may influence the adrenals' response to immune stress and may help explain the dissociation between plasma levels of ACTH and corticosteroids during chronic immune perturbations.


Assuntos
Glândulas Suprarrenais/fisiologia , Sistema Imunitário/fisiologia , Inflamação/imunologia , Animais , Ciclo-Oxigenase 2/genética , Células Dendríticas/fisiologia , Regulação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Interleucina-1beta/genética , Oxirredutases Intramoleculares/genética , Lipopolissacarídeos/farmacologia , Macrófagos/fisiologia , Masculino , Prostaglandina-E Sintases , Ratos , Ratos Sprague-Dawley , Receptores de Interleucina-1/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA