23 research outputs found

    Role of Hypothalamic Melanocortin System in Adaptation of Food Intake to Food Protein Increase in Mice

    Get PDF
    The hypothalamic melanocortin system—the melanocortin receptor of type 4 (MC4R) and its ligands: α-melanin-stimulating hormone (α-MSH, agonist, inducing hypophagia), and agouti-related protein (AgRP, antagonist, inducing hyperphagia)—is considered to play a central role in the control of food intake. We tested its implication in the mediation of the hunger-curbing effects of protein-enriched diets (PED) in mice. Whereas there was a 20% decrease in food intake in mice fed on the PED, compared to mice fed on an isocaloric starch-enriched diet, there was a paradoxical decrease in expression of the hypothalamic proopiomelanocortin gene, precursor of α-MSH, and increase in expression of the gene encoding AgRP. The hypophagia effect of PED took place in mice with invalidation of either MC4R or POMC, and was even strengthened in mice with ablation of the AgRP-expressing neurons. These data strongly suggest that the hypothalamic melanocortin system does not mediate the hunger-curbing effects induced by changes in the macronutrient composition of food. Rather, the role of this system might be to defend the body against the variations in food intake generated by the nutritional environment

    Quand les syndicats présentent leurs solutions

    No full text
    "Plans sociaux, unité de site, charte, telles sont les trois "propositions de solutions" qui seront présentées à la direction du CERN par les syndicats..." (1/2 page)

    Dégeler des crédits pour congeler une nappe d'eau

    No full text
    The LHC budget has been exceeded, partially due to serious engineering difficulties such as a large body of water at point 5 on the construction site (1 page)

    Un convoi exceptionnel perturbe la circulation

    No full text
    "Le passage d'un convoi exceptionnel hier après-midi dans le secteur de Bellegarde en direction de Cessy aura causé d'importantes perturbations. Entre la deux fois deux voies obturée et les routes secondaires bouclées, les automobilistes ont dû prendre leur mal en patience" (1 page

    Opioid-induced mitogen-activated protein kinase signaling in rat enteric neurons following chronic morphine treatment.

    No full text
    Opioids, acting at ÎĽ opioid receptors, are commonly used for pain management. Chronic opioid treatment induces cellular adaptations, which trigger long-term side effects, including constipation mediated by enteric neurons. We tested the hypothesis that chronic opioid treatment induces alterations of ÎĽ opioid receptor signaling in enteric neurons, which are likely to serve as mechanisms underlying opioid-induced constipation. In cultured rat enteric neurons, either untreated (naĂŻve) or exposed to morphine for 4 days (chronic), we compared the effect of morphine and DAMGO (D-Ala2,MePhe4,Gly-ol5 enkephalin) on ÎĽ opioid receptor internalization and downstream signaling by examining the activation of the mitogen-activated protein kinase/extracellular signal-regulated kinases 1 and 2 (MAPK/ERK) pathway, cAMP accumulation and transcription factor cAMP Response Element-Binding protein (CREB) expression. ÎĽ opioid receptor internalization and MAPK/ERK phosphorylation were induced by DAMGO, but not morphine in naĂŻve neurons, and by both opioids in chronic neurons. MAPK/ERK activation was prevented by the receptor antagonist naloxone, by blocking receptor trafficking with hypertonic sucrose, dynamin inhibitor, or neuronal transfection with mutated dynamin, and by MAPK inhibitor. Morphine and DAMGO inhibited cAMP in naĂŻve and chronic enteric neurons, and induced desensitization of cAMP signaling. Chronic morphine treatment suppressed desensitization of cAMP and MAPK signaling, increased CREB phosphorylation through a MAPK/ERK pathway and induced delays of gastrointestinal transit, which was prevented by MAPK/ERK blockade. This study showed that opioids induce endocytosis- and dynamin-dependent MAPK/ERK activation in enteric neurons and that chronic morphine treatment triggers changes at the receptor level and downstream signaling resulting in MAPK/ERK-dependent CREB activation. Blockade of this signaling pathway prevents the development of gastrointestinal motility impairment induced by chronic morphine treatment. These findings suggest that alterations in ÎĽ opioid receptor downstream signaling including MAPK/ERK pathway in enteric neurons chronically treated with morphine contribute to the development of opioid-induced constipation

    Desensitization of ÎĽOR signaling in enteric neurons.

    No full text
    <p>A: Single exposure to DAMGO (1 µM, 5 min) induced significant MAPK activation in naïve enteric neurons, whereas a second exposure to the same DAMGO dose following 2 hours DAMGO pretreatment abolished DAMGO-mediated MAPK response, indicating desensitization. B: Single exposure to DAMGO (1 µM) or morphine (10 µM) activated MAPK in chronic neurons. A second exposure to the same dose of DAMGO or morphine following 2 hours DAMGO or morphine pretreatment induced the same effect in chronic neurons as single exposures, indicating suppression of desensitization. (** p<0.01 vs. control in A and B). C and D: DAMGO and morphine inhibit forskolin-stimulated cAMP in naïve (C) and chronic (D) enteric neurons. This effect was not observed in naïve enteric neurons (C) with a second opioid stimulation following a prior 2 hour exposure, indicative of desensitization. D: Note the over 2 fold increase in cAMP in unstimulated chronic neurons (cAMP superactivation or “overshooting”) vs. naïve control; DAMGO and morphine inhibition of cAMP was not prevented by 2 hours DAMGO or morphine pretreatment in chronic neurons, indicating suppression of desensitization. **p<0.01 vs. controls. N = 5–7 experiments performed in duplicate per group.</p

    Opioid-induced MAPK activation in naĂŻve (A) and chronically treated (B) enteric neurons.

    No full text
    <p>DAMGO (1 µM, black bars) induced a transient MAPK/ERK1/2 activation in naïve (A) and chronic (B) neurons at 5 and 10 minutes, whereas morphine (grey bars) induced MAPK/ERK1/2 activation only in chronic (B) neurons. **p<0.01 compared to controls (white bars). N = 4–7 experiments in triplicate. Representative gels of pERK1/2 and tERK are shown at the bottom of each graph. tERK was used to verify that the treatment did not affect the total level of this protein and to confirm equal gel loading.</p

    Activation of \u3bc opioid receptors modulates inflammation in acute experimental colitis

    No full text
    Background: \u3bc opioid receptors (\u3bcORs) are expressed by neurons and inflammatory cells, and mediate immune response. We tested whether activation of peripheral \u3bcORs ameliorates the acute and delayed phase of colitis. Methods: C57BL/6J mice were treated with 3% dextran sodium sulfate (DSS) in water, 5 days with or without the peripherally acting \u3bcOR agonist, [D-Ala2, N-Me-Phe4, Gly5-ol]-Enkephalin (DAMGO) or with DAMGO+\u3bcOR antagonist at day 2-5, then euthanized. Other mice received DSS followed by water for 4 weeks, or DSS with DAMGO starting at day 2 of DSS for 2 or 3 weeks followed by water, then euthanized at 4 weeks. Disease activity index (DAI), histological damage, and myeloperoxidase assay (MPO), as index of neutrophil infiltration, were evaluated. Cytokines and \u3bcOR mRNAs were measured with RT-PCR, and nuclear factor-kB (NF-kB), the antiapoptotic factor Bcl-xL, and caspase 3 and 7 with Western blot. Key Results: DSS induced acute colitis with elevated DAI, tissue damage, apoptosis and increased MPO, cytokines, \u3bcOR mRNA, and NF-kB. DAMGO significantly reduced DAI, inflammatory indexes, cytokines, caspases, and NF-kB, and upregulated Bcl-xL, effects prevented by \u3bcOR antagonist. In DSS mice plus 4 weeks of water, DAI, NF-kB, and \u3bcOR were normal, whereas MPO, histological damage, and cytokines were still elevated; DAMGO did not reduce inflammation, and did not upregulate Bcl-xL. Conclusions & Inferences: \u3bcOR activation ameliorated the acute but not the delayed phase of DSS colitis by reducing cytokines, likely through activation of the antiapoptotic factor, Bcl-xL, and suppression of NF-kB, a potentiator of inflammation. \u3bc opioid receptors (\u3bcORs) mediate a variety of biological processes, including gastrointestinal functions and immune response. This study showed that activation of peripheral \u3bcOR with DAMGO, a selective \u3bcOR agonist, reduces the inflammatory response and disease activity index in acute experimental colitis through a mechanism involving the activation of the antiapoptotic factor, Bcl-xL, a member of the Bcl-2 family of apoptosis regulator proteins, and the suppression of the transcription nuclear factor-kB (shown here), which in turns reduces the release of pro-inflammatory cytokines and immune cells infiltration. By contrast, the delayed phase of colitis, which is characterized by lower level of inflammation, does not appear to benefit for additional treatment with peripherally acting \u3bcOR agonists as indicated by the lack of effect of DAMGO on the expression of Bcl-xL and NF-kB
    corecore