7 research outputs found

    Diet-induced obesity induces endoplasmic reticulum stress and insulin resistance in the amygdala of rats

    Get PDF
    FAPESP - FUNDAÇÃO DE AMPARO À PESQUISA DO ESTADO DE SÃO PAULOInsulin acts in the hypothalamus, decreasing food intake (FI) by the IR/PI3K/Akt pathway. This pathway is impaired in obese animals and endoplasmic reticulum (ER) stress and low-grade inflammation are possible mechanisms involved in this impairment. Here, we highlighted the amygdala as an important brain region for FI regulation in response to insulin. This regulation was dependent on PI3K/AKT pathway similar to the hypothalamus. Insulin was able to decrease neuropeptide Y (NPY) and increase oxytocin mRNA levels in the amygdala via PI3K, which may contribute to hypophagia. Additionally, obese rats did not reduce FI in response to insulin and AKT phosphorylation was decreased in the amygdala, suggesting insulin resistance. Insulin resistance was associated with ER stress and low-grade inflammation in this brain region. The inhibition of ER stress with PBA reverses insulin action/signaling, decreases NPY and increases oxytocin mRNA levels in the amygdala from obese rats, suggesting that ER stress is probably one of the mechanisms that induce insulin resistance in the amygdala. © 2013 The Authors.Insulin acts in the hypothalamus, decreasing food intake (FI) by the IR/PI3K/Akt pathway. This pathway is impaired in obese animals and endoplasmic reticulum (ER) stress and low-grade inflammation are possible mechanisms involved in this impairment. Here, we highlighted the amygdala as an important brain region for FI regulation in response to insulin. This regulation was dependent on PI3K/AKT pathway similar to the hypothalamus. Insulin was able to decrease neuropeptide Y (NPY) and increase oxytocin mRNA levels in the amygdala via PI3K, which may contribute to hypophagia. Additionally, obese rats did not reduce FI in response to insulin and AKT phosphorylation was decreased in the amygdala, suggesting insulin resistance. Insulin resistance was associated with ER stress and low-grade inflammation in this brain region. The inhibition of ER stress with PBA reverses insulin action/signaling, decreases NPY and increases oxytocin mRNA levels in the amygdala from obese rats, suggesting that ER stress is probably one of the mechanisms that induce insulin resistance in the amygdala31443449FAPESP - FUNDAÇÃO DE AMPARO À PESQUISA DO ESTADO DE SÃO PAULOFAPESP - FUNDAÇÃO DE AMPARO À PESQUISA DO ESTADO DE SÃO PAULOsem informaçã

    Hypothalamic S1p/s1pr1 axis controls energy homeostasis

    Get PDF
    Sphingosine 1-phosphate receptor 1 (S1PR1) is a G-protein-coupled receptor for sphingosine-1-phosphate (S1P) that has a role in many physiological and pathophysiological processes. Here we show that the S1P/S1PR1 signalling pathway in hypothalamic neurons regulates energy homeostasis in rodents. We demonstrate that S1PR1 protein is highly enriched in hypothalamic POMC neurons of rats. Intracerebroventricular injections of the bioactive lipid, S1P, reduce food consumption and increase rat energy expenditure through persistent activation of STAT3 and the melanocortin system. Similarly, the selective disruption of hypothalamic S1PR1 increases food intake and reduces the respiratory exchange ratio. We further show that STAT3 controls S1PR1 expression in neurons via a positive feedback mechanism. Interestingly, several models of obesity and cancer anorexia display an imbalance of hypothalamic S1P/S1PR1/STAT3 axis, whereas pharmacological intervention ameliorates these phenotypes. Taken together, our data demonstrate that the neuronal S1P/S1PR1/STAT3 signalling axis plays a critical role in the control of energy homeostasis in rats.Sphingosine 1-phosphate receptor 1 (S1PR1) is a G-protein-coupled receptor for sphingosine-1-phosphate (S1P) that has a role in many physiological and pathophysiological processes. Here we show that the S1P/S1PR1 signalling pathway in hypothalamic neurons regulates energy homeostasis in rodents. We demonstrate that S1PR1 protein is highly enriched in hypothalamic POMC neurons of rats. Intracerebroventricular injections of the bioactive lipid, S1P, reduce food consumption and increase rat energy expenditure through persistent activation of STAT3 and the melanocortin system. Similarly, the selective disruption of hypothalamic S1PR1 increases food intake and reduces the respiratory exchange ratio. We further show that STAT3 controls S1PR1 expression in neurons via a positive feedback mechanism. Interestingly, several models of obesity and cancer anorexia display an imbalance of hypothalamic S1P/S1PR1/STAT3 axis, whereas pharmacological intervention ameliorates these phenotypes. Taken together, our data demonstrate that the neuronal S1P/S1PR1/STAT3 signalling axis plays a critical role in the control of energy homeostasis in rats5485

    Hypothalamic S1p/s1pr1 Axis Controls Energy Homeostasis

    Get PDF
    Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES)Sphingosine 1-phosphate receptor 1 (S1PR1) is a G-protein-coupled receptor for sphingosine-1-phosphate (S1P) that has a role in many physiological and pathophysiological processes. Here we show that the S1P/S1PR1 signalling pathway in hypothalamic neurons regulates energy homeostasis in rodents. We demonstrate that S1PR1 protein is highly enriched in hypothalamic POMC neurons of rats. Intracerebroventricular injections of the bioactive lipid, S1P, reduce food consumption and increase rat energy expenditure through persistent activation of STAT3 and the melanocortin system. Similarly, the selective disruption of hypothalamic S1PR1 increases food intake and reduces the respiratory exchange ratio. We further show that STAT3 controls S1PR1 expression in neurons via a positive feedback mechanism. Interestingly, several models of obesity and cancer anorexia display an imbalance of hypothalamic S1P/S1PR1/STAT3 axis, whereas pharmacological intervention ameliorates these phenotypes. Taken together, our data demonstrate that the neuronal S1P/S1PR1/STAT3 signalling axis plays a critical role in the control of energy homeostasis in rats.5Capes-12900-13-3; CAPES; Coordenação de Aperfeiçoamento de Pessoal de Nível SuperiorCoordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES)Schwartz, M.W., Woods, S.C., Porte, D., Jr., Seeley, R.J., Baskin, D.G., Central nervous system control of food intake (2000) Nature, 404, pp. 661-671Munzberg, H., Huo, L., Nillni, E.A., Hollenberg, A.N., Bjorbaek, C., Role of signal transducer and activator of transcription 3 in regulation of hypothalamic proopiomelanocortin gene expression by leptin (2003) Endocrinology, 144, pp. 2121-2131Myers, M.G., Cowley, M.A., Munzberg, H., Mechanisms of leptin action and leptin resistance (2008) Annu. Rev. Physiol., 70, pp. 537-556Janoschek, R., Gp130 signaling in proopiomelanocortin neurons mediates the acute anorectic response to centrally applied ciliary neurotrophic factor (2006) Proc. Natl Acad. Sci. USA, 103, pp. 10707-10712Johnen, H., Tumor-induced anorexia and weight loss are mediated by the TGF-beta superfamily cytokine MIC-1 (2007) Nat. Med., 13, pp. 1333-1340Ropelle, E.R., IL-6 and IL-10 anti-inflammatory activity links exercise to hypothalamic insulin and leptin sensitivity through IKKbeta and ER stress inhibition (2010) PLoS Biol., 8El-Haschimi, K., Pierroz, D.D., Hileman, S.M., Bjorbaek, C., Flier, J.S., Two defects contribute to hypothalamic leptin resistance in mice with diet-induced obesity (2000) J. Clin. Invest., 105, pp. 1827-1832Ernst, M.B., Enhanced Stat3 activation in POMC neurons provokes negative feedback inhibition of leptin and insulin signaling in obesity (2009) J. Neurosci., 29, pp. 11582-11593Gao, Q., Anorectic estrogen mimics leptin's effect on the rewiring of melanocortin cells and Stat3 signaling in obese animals (2007) Nat. Med., 13, pp. 89-94Ghilardi, N., Defective STAT signaling by the leptin receptor in diabetic mice (1996) Proc. Natl Acad. Sci. USA, 93, pp. 6231-6235Frias, M.A., James, R.W., Gerber-Wicht, C., Lang, U., Native and reconstituted HDL activate Stat3 in ventricular cardiomyocytes via ERK1/2: Role of sphingosine-1-phosphate (2009) Cardiovasc. Res., 82, pp. 313-323Gurgui, M., Broere, R., Kalff, J.C., Van Echten-Deckert, G., Dual action of sphingosine 1-phosphate in eliciting proinflammatory responses in primary cultured rat intestinal smooth muscle cells (2010) Cell Signal., 22, pp. 1727-1733Lee, H., STAT3-induced S1PR1 expression is crucial for persistent STAT3 activation in tumors (2010) Nat. Med., 16, pp. 1421-1428Liang, J., Sphingosine-1-phosphate links persistent STAT3 activation, chronic intestinal inflammation, and development of colitis-associated cancer (2013) Cancer Cell, 23, pp. 107-120Spiegel, S., Milstien, S., The outs and the ins of sphingosine-1-phosphate in immunity (2011) Nat. Rev. Immunol., 11, pp. 403-415Spiegel, S., Milstien, S., Functions of the multifaceted family of sphingosine kinases and some close relatives (2007) J. Biol. Chem., 282, pp. 2125-2129Loh, K.C., Sphingosine-1-phosphate enhances satellite cell activation in dystrophic muscles through a S1PR2/STAT3 signaling pathway (2012) PLoS One, 7Lopez, M., Hypothalamic fatty acid metabolism mediates the orexigenic action of ghrelin (2008) Cell. Metab., 7, pp. 389-399Oo, M.L., Engagement of S1P(1)-degradative mechanisms leads to vascular leak in mice (2011) J. Clin. Invest., 121, pp. 2290-2300Means, C.K., Brown, J.H., Sphingosine-1-phosphate receptor signalling in the heart (2009) Cardiovasc. Res., 82, pp. 193-200Schwartz, M.W., Leptin increases hypothalamic pro-opiomelanocortin mRNA expression in the rostral arcuate nucleus (1997) Diabetes, 46, pp. 2119-2123Cowley, M.A., Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus (2001) Nature, 411, pp. 480-484Bates, S.H., STAT3 signalling is required for leptin regulation of energy balance but not reproduction (2003) Nature, 421, pp. 856-859Mynard, V., Guignat, L., Devin-Leclerc, J., Bertagna, X., Catelli, M.G., Different mechanisms for leukemia inhibitory factor-dependent activation of two proopiomelanocortin promoter regions (2002) Endocrinology, 143, pp. 3916-3924Bousquet, C., Zatelli, M.C., Melmed, S., Direct regulation of pituitary proopiomelanocortin by STAT3 provides a novel mechanism for immunoneuroendocrine interfacing (2000) J. Clin. Invest., 106, pp. 1417-1425Bousquet, C., Melmed, S., Critical role for STAT3 in murine pituitary adrenocorticotropin hormone leukemia inhibitory factor signaling (1999) J. Biol. Chem., 274, pp. 10723-10730Andreux, P.A., Systems genetics of metabolism: The use of the BXD murine reference panel for multiscalar integration of traits (2012) Cell, 150, pp. 1287-1299Guissouma, H., Froidevaux, M.S., Hassani, Z., Demeneix, B.A., In vivo siRNA delivery to the mouse hypothalamus confirms distinct roles of TR beta isoforms in regulating TRH transcription (2006) Neurosci. Lett., 406, pp. 240-243Froidevaux, M.S., The co-chaperone XAP2 is required for activation of hypothalamic thyrotropin-releasing hormone transcription in vivo (2006) EMBO Rep., 7, pp. 1035-1039Nagahashi, M., Sphingosine-1-phosphate produced by sphingosine kinase 1 promotes breast cancer progression by stimulating angiogenesis and lymphangiogenesis (2012) Cancer Res., 72, pp. 726-735Brinkmann, V., Fingolimod (FTY720): Discovery and development of an oral drug to treat multiple sclerosis (2010) Nat. Rev. Drug Discov., 9, pp. 883-897Graler, M.H., Goetzl, E.J., The immunosuppressant FTY720 down-regulates sphingosine 1-phosphate G-protein-coupled receptors (2004) FASEB J., 18, pp. 551-553Guo, H., An activated protein C analog stimulates neuronal production by human neural progenitor cells via a PAR1-PAR3-S1PR1-Akt pathway (2013) J. Neurosci., 33, pp. 6181-6190Ishii, I., Fukushima, N., Ye, X., Chun, J., Lysophospholipid receptors: Signaling and biology (2004) Annu. Rev. Biochem., 73, pp. 321-354Walter, D.H., Sphingosine-1-phosphate stimulates the functional capacity of progenitor cells by activation of the CXCR4-dependent signaling pathway via the S1P3 receptor (2007) Arterioscler. Thromb. Vasc. Biol., 27, pp. 275-282Grossberg, A.J., Arcuate nucleus proopiomelanocortin neurons mediate the acute anorectic actions of leukemia inhibitory factor via gp130 (2010) Endocrinology, 151, pp. 606-616Febbraio, M.A., Gp130 receptor ligands as potential therapeutic targets for obesity (2007) J. Clin. Invest., 117, pp. 841-849Samad, F., Hester, K.D., Yang, G., Hannun, Y.A., Bielawski, J., Altered adipose and plasma sphingolipid metabolism in obesity: A potential mechanism for cardiovascular and metabolic risk (2006) Diabetes, 55, pp. 2579-2587Bence, K.K., Neuronal PTP1B regulates body weight, adiposity and leptin action (2006) Nat. Med., 12, pp. 917-924Chiarreotto-Ropelle, E.C., Acute exercise suppresses hypothalamic PTP1B protein level and improves insulin and leptin signaling in obese rats (2013) Am. J. Physiol. Endocrinol. Metab., 305, pp. E649-E659Picardi, P.K., Reduction of hypothalamic protein tyrosine phosphatase improves insulin and leptin resistance in diet-induced obese rats (2008) Endocrinology, 149, pp. 3870-3880Milanski, M., Saturated fatty acids produce an inflammatory response predominantly through the activation of TLR4 signaling in hypothalamus: Implications for the pathogenesis of obesity (2009) J. Neurosci., 29, pp. 359-370Purkayastha, S., Zhang, G., Cai, D., Uncoupling the mechanisms of obesity and hypertension by targeting hypothalamic IKK-beta and NF-kappaB (2011) Nat. Med., 17, pp. 883-887Zhang, X., Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity (2008) Cell, 135, pp. 61-73Laviano, A., Meguid, M.M., Rossi-Fanelli, F., Cancer anorexia: Clinical implications, pathogenesis, and therapeutic strategies (2003) Lancet Oncol., 4, pp. 686-694Tisdale, M.J., Biology of cachexia (1997) J. Natl Cancer Inst., 89, pp. 1763-1773Pchejetski, D., Circulating sphingosine-1-phosphate inversely correlates with chemotherapy-induced weight gain during early breast cancer (2010) Breast Cancer Res. Treat., 124, pp. 543-549Ponnusamy, S., Communication between host organism and cancer cells is transduced by systemic sphingosine kinase 1/sphingosine 1-phosphate signalling to regulate tumour metastasis (2012) EMBO Mol. Med., 4, pp. 761-775Mellon, P.L., Immortalization of hypothalamic GnRH neurons by genetically targeted tumorigenesis (1990) Neuron, 5, pp. 1-1

    Analysis Of The Physical Activity Effects And Measurement Of Pro-inflammatory Cytokines In Irradiated Lungs In Rats [análise Dos Efeitos Da Atividade Física E Mensuração De Citocinas Pró-inflamatórias Em Pulmões Irradiados Em Ratos]

    No full text
    PURPOSE: To study if the pre-radiotherapy physical activity has radio-protective elements, by measuring the radio-induced activation of pro-inflammatory cytokines as interleukin-6 (il-6), transforming growth factor -β (tgf -β), tumor necrosis factor -α (tnf-α) and protein beta kinase β (ikkβ), through western blotting analysis. METHODS: A randomized study with 28 Wistar hannover rats, males, with a mean age of 90 days and weighing about 200 grams. The animals were divided into three groups: (GI, GII and GIII). GIII group were submitted to swimming for eight weeks (zero load, three times a week, about 30 minutes). Then, the groups (except the control group) were submitted to irradiation by cobalt therapy, single dose of 3.5 gray in the whole body. All animals were sacrificed by overdose of pentobarbital, according to the time for analysis of cytokines, and then a fragment of the lower lobe of the right lung went to western blotting analysis. RESULTS: The cytokines IKK β, TNF-α and IL-6 induced by radiation in the lung were lower in the exercised animals. However, exercise did not alter the radiation-induced increase in tgf-β. CONCLUSION: The results show a lower response in relation to inflammatory cytokines in the group that practiced the exercise pre-radiotherapy, showing that exercise can protect tissues from tissue damage due to irradiation.273223230Liao, Z., Cox, J.D., Komaki, R., Radiochemotherapy of esofaphageal cancer (2007) J Thorac Oncol, 2 (6), pp. 553-568D'amico, T.A., Outcomes after surgery for esophageal cancer (2007) Gastrointest Cancer Res, 1 (5), pp. 188-196Tercioti Jr., V., Lopes, L.R., Coelho, N.J.D., Carvalheira, J.B., Andreollo, N.A., Local effectiveness and complications of neoadjuvant therapy in esophageal squamous cell carcinoma: Radiotherapy versus chemoradiotherapy (2011) Rev Col Bras Cir, 38 (4), pp. 227-234Miyata, H., Yamasaki, M., Takiguchi, S., Nakajima, K., Fujiwara, Y., Nishida, T., Mori, M., Doki, Y., Salvage esophagectomy after definitive chemoradiotherapy for thoracic esophageal cancer (2009) J Surg Oncol, 100, pp. 442-446Asakura, H., Hashimoto, T., Zenda, S., Harada, H., Hirakawa, K., Mizumoto, M., Furutani, K., Nishimura, T., Analysis of dose-volume histogram parameters for radiation pneumonitis after definitive concurrent chemoradiotherapy for esophageal cancer (2010) Radiother Oncol, 95, pp. 240-244Yu, H.M., Liu, Y.F., Cheng, Y.F., Hu, L.K., Hou, M., Effects of rhubarb extract on radiation induced lung toxicity via decreasing transforming growth factor-beta-1 and interleukin-6 in lung cancer patients treated with radiotherapy (2008) Lung Cancer, 59, pp. 219-226Sakai, M., Iwakawa, M., Iwakura, Y., Ohta, T., Tsujii, H., Imai, T., CD44 and bak expression in IL-6 or TNF-alpha gene knockout mice after whole lung irradiation (2008) J Radiat Res, 49, pp. 409-416Novakova-Jiresova, A., Luijk, P.V., Goor, H.V., Kampinga, H.H., Coppes, R.P., Pulmonary radiation injury: Identification of risk factors associated with regional hypersensitivity (2005) Cancer Res, 65, pp. 3568-3576Bao, P., Gao, W., Li, S., Zhang, L., Qu, S., Wu, C., Qi, H., Effect of pretreatment with high-dose ulinastatin in preventing radiation-induced pulmonary injury in rats (2009) Eur J Pharmacol, 603, pp. 114-119Schmid, J.A., Birbach, A., IkB kinase β (Ikkβ/Ikk2/IkBkB)- a key molecule in signaling to the transcription factor NF-kB (2008) Cytokine Growth Factor Rev, 19, pp. 157-165Mussi, R.K., Camargo, E.A., Ferreira, T., de Moraes, C., Delbin, M.A., Toro, I.F., Brancher, S., Antunes, E., Exercise training reduces pulmonary ischaemia-reperfusion-induced inflammatory responses (2008) Eur Respir J, 31 (3), pp. 645-649Gannon, G.A., Rhind, S.G., Suzui, M., Shek, P.N., Shephard, R.J., Circulating levels of peripheral blood leucocytes and cytokines following competitive cycling (1997) Can J Appl Physiol, 22, pp. 133-147Nieman, D.C., Special feature for the Olympics: Exercise effects on systemic immunity (2000) Immunol Cell Biol, 78, pp. 496-501Triboulet, J.P., Mariette, C., Esophageal squamous cell carcinoma stage III: State of surgery after radiochemotherapy (2006) Cancer Radiother, 10, pp. 456-461Hermann, R.M., Horstmann, O., Haller, F., Perske, C., Christiansen, H., Hille, A., Schimidberger, H., Füzesi, L., Histomorphological tumor regression grading esophageal carcinoma after neoadjuvante radiochemotherapy: With score to use? (2006) Dis Esophagus, 5, pp. 329-334Cagol, M., Ruol, A., Sileni, V.C., de Salvo, G.L., Corti, L., Alfieri, R., Innocente, R., Ancona, E., Multinodal treatment in locally advanced esophageal cancer: A multicenter phase II study with neoadjuvant oxaliplatin, 5-fluorouracil, leucovorin and neoadjuvante radiotherapy: Prelimirary results (2006) Chir Ital, 4, pp. 433-439Rübe, C.E., Wilfert, F., Uthe, D., Schmid, K.W., Knoop, R., Willich, N., Schuck, A., Rübe, C., Modulation of radiation-induced tumor necrosis factor α (TNF-α) expression in the lung tissue by pentoxifyline (2002) Radiother Oncol, 64, pp. 177-187Arnold, R., Humbert, B., Werchau, H., Gallati, H., Königg, W., Interleukin-8, interleukin-6, and soluble tumor necrosis factor receptor type I release from a human pulmonary epithelial cell line (A549) exposed to respiratory syncytial virus (1994) Immunology, 82, pp. 126-133Chen, Y., Rubin, P., Williams, J., Hernady, E., Smudzin, T., Okunieff, P., (2001) Int J Radiat Oncol Biol Phys, 49 (3), pp. 641-648Sriwijitkamol, A., Coletta, D.K., Wajcberg, E., Balbontin, G.B., Reyna, S.M., Barrientes, J., Eagan, P.A., Musi, N., Effect of acute exercise on AMPK signiling in skeletal muscle of subjects with type 2 diabetes: A time-course and dose-response study (2007) Diabetes, 56 (3), pp. 836-848Anscher, M.S., Peters, W.P., Reisenbichler, H., Petros, W.P., Jirtle, R.L., Transforming growth factor beta as predictor of liver and lung fibrosis after autologous bone marrow transplantation for advanced breast cancer (1993) N Engl J Med, 328, pp. 1592-1598Fu, X.L., Huang, H., Bentel, G., Clough, R., Jirtle, R.L., Kong, F.M., Marks, L.B., Anscher, M.S., Predicting the risk of symptomatic radiation-induced lung injury using both the physical and biologic parameters V30 and transforming growth factor β (2001) Int J Radiat Oncol Biol Phys, 50 (4), pp. 899-908Mattos, M.D., Kimura, E.T., Silva, M.R.R., Egami, M.I., Segreto, R.A., Segreto, H.R.C., Ativação da proteína TGFβ1 latente em pulmão irradiado in vivo (2002) Rev Assoc Med Bras, 48 (4), pp. 329-334Lixuan, Z., Jingcheng, D., Wenqin, Y., Jianhua, H., Baojun, L., Xiaotao, F., Baicalin attenuates inflammation by inhibiting NF-kappaβ activation in cigarette smoke induced inflammatory models (2010) Pulm Pharmacol Ther, 23 (5), pp. 411-419Colbert, L.H., Davis, J.M., Essig, D.A., Ghaffar, A., Mayer, E.P., Tissue expression and plasma concentrations of TNF-α, IL-1β, and IL-6 following treadmill exercise in mice (2001) Int J Sports Med, 22, pp. 261-267Ostrowski, K., Rohde, T., Zacho, M., Asp, S., Pedersen, B.K., Evidence that interleukin-6 is produced in human skeletal muscle during prolonged running (1998) J Physiol, 508, pp. 949-953Ullum, H., Haahr, P.M., Diamant, M., Palmo, J., Halkjaer-Kristensen, J., Pedersen, B.K., Bicycle exercise enhances plasma IL-6 but does not change IL-1α, IL-1β, IL-6 or TNFα pre-mRNA in BMNC (1994) J Appl Physiol, 77, pp. 93-97Oliveira, A.G., Carvalho, B.M., Tobar, N., Ropelle, E.R., Pauli, J.R., Bagarolli, R.A., Guadagnini, D., Saad, M.J., Physical exercise reduces circulating lipopolysaccharide and TLR4 activation and improves insulin signaling in tissues of DIO rats (2011) Diabetes, 60, pp. 784-796Lira, F.S., Rosa, J.C., Pimentel, G.D., Tarini, V.A.F., Arida, R.M., Faloppa, F., Alves, E.S., Santos, R.V., Inflammation and adipose tissue: Effects of progressive load training in rats (2010) Lipids Health Dis, 9, p. 109Ropelle, E.R., Flores, M.B., Cintra, D.E., Rocha, G.Z., Pauli, J.R., Morari, J., de Souza, C.T., Carvalheira, J.B., IL-6 and IL-10 anti-inflammatory activity links exercise to hypothalamic insulin and leptin sensitivity through IKKbeta and ER stress inhibition (2010) PLoS Biol, 8 (8), pp. e1000465Chen, L., Wang, T., Wang, X., Sun, B.B., Li, J.Q., Liu, D.S., Zhang, S.F., Wen, F.Q., Blockade of advanced glycation end product formation attenuates bleomycin-induced pulmonary fibrosis in rats (2009) Respir Res, 10, p. 55Kim, J.Y., Kim, Y.S., Kim, Y.K., Park, H.J., Kim, S.J., Kang, J.H., Wang, Y.P., Yoon, S.C., TGF-β1 dynamics during radiation therapy and its correlation to symptomatic radiation pneumonitis in lung cancer patients (2009) Radiat Oncol, 4, p. 59Moon, C., Lee, Y.J., Park, H.J., Chong, Y.H., Kang, J.L., N-acetylcysteine inhibits RhoA and promotes apoptotic cell clearance during intense lung inflammation (2010) Am J Respir Crit Care Med, 181, pp. 374-387Morita, M., Yoshida, R., Ikeda, K., Egashira, A., Oki, E., Sadanaga, N., Kakeji, Y., Maehara, Y., Acute lung injury following an esophagectomy for esophageal cancer, with special reference to the clinical factors and cytokine levels of peripheral blood and pleural drainage fluid (2008) Dis Esophagus, 21 (1), pp. 30-36Sakamoto, K., Arakawa, H., Mita, S., Ishiko, T., Ikei, S., Egami, H., Hisano, S., Ogawa, M., Elevation of circulating interleukin-6 after surgery: Factors influencing serum level (1994) Cytokine, 6, pp. 181-186Adams, E.J., Warrington, A.P., A comparison between cobalt and linear accelerator-based treatment plans for conformal and intensity-modulated radiotherapy (2008) Br J Radiol, 81, pp. 304-310Jin, H., Tucker, S.L., Liu, H.H., Wei, X., Yom, S.S., Wang, S., Komaki, R., Liao, Z., Dose-volume thresholds and smoking status for the risk of treatment-related pneumonitis in inoperable non-small cell lung cancer treated with definitive radiotherapy (2009) Radiother Oncol, 91, pp. 427-432Werner-Wasik, M., Yorke, E., Deasy, J., Nam, J., Marks, L.B., Radiation dose-volume effects in the esophagus (2010) Int J Radiat Oncol Biol Phys, 76 (3), pp. S86-S93Kocak, Z., Borst, G.R., Zeng, J., Zhou, S., Hollis, R., Zhang, J., Evans, E.S., Marks, L.B., Prospective assessment of dosimetric/physiologic-based models for predicting radiation pneumonitis (2007) Int J Radiat Oncol Biol Phys, 67 (1), pp. 178-186Yorke, E.D., Jackson, A., Rosenzweig, K.E., Braban, L., Leibel, S.A., Ling, C.C., Correlation of dosimetric factors and radiation pneumonitis for non-small-cell lung cancer patients in a recently completed dose escalation study (2005) Int J Radiat Oncol Biol Phys, 63 (3), pp. 672-682Hope, A.J., Lindsay, P.E., El-Naga, I., Alaly, J.R., Vicic, M., Bradley, J.D., Deasy, G.O., Modeling radiation pneumonitis risk with clinical, dosimetric, and spatial parameters (2006) Int J Radiat Oncol Biol Phys, 65 (1), pp. 112-124Tsujino, K., Hirota, S., Endo, M., Obayashi, K., Kotani, Y., Satouchi, M., Kado, T., Takada, Y., Predictive value of dose-volume histogram parameters for predicting radiation pneumonitis after concurrent chemoradiation for lung cancer (2003) Int J Radiat Oncol Biol Phys, 55 (1), pp. 110-115Fay, M., Tan, A., Fisher, R., Mac, M.M., Wirth, A., Ball, D., Dose-volume histogram analysis as predictor of radiation pneumonitis in primary lung cancer patients treated with radiotherapy (2005) Int J Radiat Oncol Biol Phys, 61 (5), pp. 1355-1363Szabo, S., Ghosh, S.N., Fish, B.L., Bodiga, S., Tomic, R., Kumar, G., Morrow, N.V., Medhora, M., Cellular inflammatory infiltrate in pneumonitis induced by a single moderate dose of thoracic versus radiation in rats (2010) Radiat Res, 173 (4), pp. 545-556Dähn, D., Martell, J., Vorwerk, H., Hess, C.F., Becker, H., Jung, K., Hilgers, R., Christiansen, H., Influence of irradiated lung volumes on perioperative morbidity and mortality in patients after neoadjuvante radiochemotherapy for esophageal cancer (2010) Int J Radiat Oncol Biol Phys, 77 (1), pp. 44-52Ysayama, L., Lopes, L.R., Silva, A.M.O., Andreollo, N.A., The influence of the respiratory muscular training in the recovery of esophagectomies (2008) ABCD Arq Bras Cir Dig, 21 (2), pp. 61-64Nakamura, M., Iwahashi, M., Nakamori, M., Ishida, K., Naka, T., Iida, T., Katsuda, M., Yamaue, H., An analysis of the factors contributing to a reduction in the incidence of pulmonary complications following an esophagectomy for esophageal cancer (2008) Langenbecks Arch Surg, 393, pp. 127-13

    Exercise Training Decreases Mitogen-activated Protein Kinase Phosphatase-3 Expression And Suppresses Hepatic Gluconeogenesis In Obese Mice

    No full text
    Insulin plays an important role in the control of hepatic glucose production. Insulin resistant states are commonly associated with excessive hepatic glucose production, which contributes to both fasting hyperglycaemia and exaggerated postprandial hyperglycaemia. In this regard, increased activity of phosphatases may contribute to the dysregulation of gluconeogenesis. Mitogen-activated protein kinase phosphatase-3 (MKP-3) is a key protein involved in the control of gluconeogenesis. MKP-3-mediated dephosphorylation activates FoxO1 (a member of the forkhead family of transcription factors) and subsequently promotes its nuclear translocation and binding to the promoters of gluconeogenic genes such as phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase). In this study, we investigated the effects of exercise training on the expression of MKP-3 and its interaction with FoxO1 in the livers of obese animals. We found that exercised obese mice had a lower expression of MKP-3 and FoxO1/MKP-3 association in the liver. Further, the exercise training decreased FoxO1 phosphorylation and protein levels of Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and gluconeogenic enzymes (PEPCK and G6Pase). These molecular results were accompanied by physiological changes, including increased insulin sensitivity and reduced hyperglycaemia, which were not caused by reductions in total body mass. Similar results were also observed with oligonucleotide antisense (ASO) treatment. However, our results showed that only exercise training could reduce an obesity-induced increase in HNF-4α protein levels while ASO treatment alone had no effect. These findings could explain, at least in part, why additive effects of exercise training treatment and ASO treatment were not observed. Finally, the suppressive effects of exercise training on MKP-3 protein levels appear to be related, at least in part, to the reduced phosphorylation of Extracellular signal-regulated kinases (ERK) in the livers of obese mice. © 2014 The Authors. The Journal of Physiology © 2014 The Physiological Society.592613251340Band, C.J., Posner, B.I., Phosphatidylinositol 3′-kinase and p70s6k are required for insulin but not bisperoxovanadium 1,10-phenanthroline (bpV(phen)) inhibition of insulin-like growth factor binding protein gene expression. Evidence for MEK-independent activation of mitogen-activated protein kinase by bpV(phen) (1997) J Biol Chem, 272, pp. 138-145Barthel, A., Schmoll, D., Krüger, K.D., Bahrenberg, G., Walther, R., Roth, R.A., Joost, H.G., Differential regulation of endogenous glucose-6-phosphatase and phosphoenol pyruvate carboxykinase gene expression by the forkhead transcription factor FKHR in H4IIE hepatoma cells (2001) Biochem Biophys Res Commun, 285, pp. 897-902Bonora, E., Moghetti, P., Zancanaro, C., Cigolini, M., Querena, M., Cacciatori, V., Corgnati, A., Muggeo, M., Estimates of in vivo insulin action in man: comparison of insulin tolerance tests with euglycemic and hyperglycemic glucose clamp studies (1989) J Clin Endocrinol Metab, 68, pp. 374-378Bradford, M.M., A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding (1976) Anal Biochem, 72, pp. 248-254Burgess, S.C., He, T., Yan, Z., Lindner, J., Sherry, A.D., Malloy, C.R., Browning, J.D., Magnuson, M.A., Cytosolic phosphoenolpyruvate carboxykinase does not solely control the rate of hepatic gluconeogenesis in the intact mouse liver (2007) Cell Metab, 5, pp. 313-320Camps, M., Chabert, C., Muda, M., Boschert, U., Gillieron, C., Arkinstall, S., Induction of the mitogen-activated protein kinase phosphatase MKP3 by nerve growth factor in differentiating PC12 (1998) FEBS Lett, 425, pp. 271-276Chambers, M.A., Moylan, J.S., Smith, J.D., Goodyear, L.J., Reid, M.B., Stretch-stimulated glucose uptake in skeletal muscle is mediated by reactive oxygen species and p38 MAP-kinase (2009) J Physiol, 587, pp. 3363-3373Cintra, D.E., Pauli, J.R., Araújo, E.P., Moraes, J.C., de Souza, C.T., Milanski, M., Morari, J., Velloso, L.A., Interleukin-10 is a protective factor against diet-induced insulin resistance in liver (2008) J Hepatol, 48, pp. 628-637Daitoku, H., Yamagata, K., Matsuzaki, H., Hatta, M., Fukamizu, A., Regulation of PGC-1 promoter activity by protein kinase B and the forkhead transcription factor FKHR (2003) Diabetes, 52, pp. 642-649da Luz, G., Frederico, M.J., da Silva, S., Vitto, M.F., Cesconetto, P.A., de Pinho, R.A., Pauli, J.R., De Souza, C.T., Endurance exercise training ameliorates insulin resistance and reticulum stress in adipose and hepatic tissue in obese rats (2011) Eur J Appl Physiol, 111, pp. 2015-2023de Moura, L.P., Souza Pauli, L.S., Cintra, D.E., de Souza, C.T., da Silva, A.S., Marinho, R., de Melo, M.A., Pauli, J.R., Acute exercise decreases PTP-1B protein level and improves insulin signaling in the liver of old rats (2013) Immun Ageing, 10, p. 8De Souza, C.T., Frederico, M.J., da Luz, G., Cintra, D.E., Ropelle, E.R., Pauli, J.R., Velloso, L.A., Acute exercise reduces hepatic glucose production through inhibition of the Foxo1/HNF-4α pathway in insulin resistant mice (2010) J Physiol, 588, pp. 2239-2253Feng, B., Jiao, P., Yang, Z., Xu, H., MEK/ERK pathway mediates insulin-promoted degradation of MKP-3 protein in liver cells (2012) Mol Cell Endocrinol, 361, pp. 116-123Fjeld, C.C., Rice, A.E., Kim, Y., Gee, K.R., Denu, J.M., Mechanistic basis for catalytic activation of mitogen-activated protein kinase phosphatase 3 by extracellular signal-regulated kinase (2000) J Biol Chem, 275, pp. 6749-6757Gonzalez, F.J., Regulation of hepatocyte nuclear factor 4α-mediated transcription (2008) Drug Metab Pharmacokinet, 23, pp. 2-7Heled, Y., Shapiro, Y., Shani, Y., Moran, D.S., Langzam, L., Barash, V., Sampson, S.R., Meyerovitch, J., Physical exercise enhances hepatic insulin signaling and inhibits phosphoenolpyruvate carboxykinase activity in diabetes-prone Psammomys obesus (2004) Metabolism, 53, pp. 836-841Hirota, K., Sakamaki, J., Ishida, J., Shimamoto, Y., Nishihara, S., Kodama, N., Ohta, K., Fukamizu, A., A combination of HNF-4 and Foxo1 is required for reciprocal transcriptional regulation of glucokinase and glucose-6-phosphatase genes in response to fasting and feeding (2008) J Biol Chem, 283, pp. 32432-32441Jiao, P., Feng, B., Xu, H., Mapping MKP-3/FOXO1 interaction and evaluating the effect on gluconeogenesis (2012) PLoS One, 7, pp. e41168Jurek, A., Amagasaki, K., Gembarska, A., Heldin, C.H., Lennartsson, J., Negative and positive regulation of MAPK phosphatase 3 controls platelet-derived growth factor-induced Erk activation (2009) J Biol Chem, 284, pp. 4626-4634Liao, J., Barthel, A., Nakatani, K., Roth, R.A., Activation of protein kinase B/Akt is sufficient to repress the glucocorticoid and cAMP induction of phosphoenolpyruvate carboxykinase gene (1998) J Biol Chem, 273, pp. 27320-27324Lima, A.F., Ropelle, E.R., Pauli, J.R., Cintra, D.E., Frederico, M.J., Pinho, R.A., Velloso, L.A., De Souza, C.T., Acute exercise reduces insulin resistance-induced TRB3 expression and amelioration of the hepatic production of glucose in the liver of diabetic mice (2009) J Cell Physiol, 221, pp. 92-97Lin, J., Tarr, P.T., Yang, R., Rhee, J., Puigserver, P., Newgard, C.B., Spiegelman, B.M., PGC-1β in the regulation of hepatic glucose and energy metabolism (2003) J Biol Chem, 278, pp. 30843-30848Maillet, M., Purcell, N.H., Sargent, M.A., York, A.J., Bueno, O.F., Molkentin, J.D., DUSP6 (MKP3) null mice show enhanced ERK1/2 phosphorylation at baseline and increased myocyte proliferation in the heart affecting disease susceptibility (2008) J Biol Chem, 283, pp. 31246-31255Marchetti, S., Gimond, C., Chambard, J.C., Touboul, T., Roux, D., Pouysségur, J., Pagès, G., Extracellular signal-regulated kinases phosphorylate mitogen-activated protein kinase phosphatase 3/DUSP6 at serines 159 and 197, two sites critical for its proteasomal degradation (2005) Mol Cell Biol, 25, pp. 854-864Marinho, R., Ropelle, E.R., Cintra, D.E., De Souza, C.T., Da Silva, A.S., Bertoli, F.C., Colantonio, E., Pauli, J.R., Endurance exercise training increases APPL1 expression and improves insulin signaling in the hepatic tissue of diet-induced obese mice, independently of weight loss (2012) J Cell Physiol, pp. 2917-2926Miyake, K., Ogawa, W., Matsumoto, M., Nakamura, T., Sakaue, H., Kasuga, M., Hyperinsulinemia, glucose intolerance, and dyslipidemia induced by acute inhibition of phosphoinositide 3-kinase signaling in the liver (2002) J Clin Invest, 110, pp. 1483-1491Oliveira, A.G., Carvalho, B.M., Tobar, N., Ropelle, E.R., Pauli, J.R., Bagarolli, R.A., Guadagnini, D., Saad, M.J., Physical exercise reduces circulating lipopolysaccharide and TLR4 activation and improves insulin signaling in tissues of DIO rats (2011) Diabetes, 60, pp. 784-796Pagliassotti, M.J., Kang, J., Thresher, J.S., Sung, C.K., Bizeau, M.E., Elevated basal PI 3-kinase activity and reduced insulin signaling in sucrose-induced hepatic insulin resistance (2002) Am J Physiol Endocrinol Metab, 282, pp. E170-E176Pauli, J.R., Ropelle, E.R., Cintra, D.E., Carvalho-Filho, M.A., Moraes, J.C., De Souza, C.T., Velloso, L.A., Saad, M.J., Acute physical exercise reverses S-nitrosation of the insulin receptor, insulin receptor substrate 1 and protein kinase B/Akt in diet-induced obese Wistar rats (2008) J Physiol, 586, pp. 659-671Pilkis, S.J., Granner, D.K., Molecular physiology of the regulation of hepatic gluconeogenesis and glycolysis (1992) Annu Rev Physiol, 54, pp. 885-909Prada, P.O., Pauli, J.R., Ropelle, E.R., Zecchin, H.G., Carvalheira, J.B., Velloso, L.A., Saad, M.J., Selective modulation of the CAP/Cbl pathway in the adipose tissue of high fat diet treated rats (2006) FEBS Lett, 580, pp. 4889-4894Puigserver, P., Rhee, J., Donovan, J., Walkey, C.J., Yoon, J.C., Oriente, F., Kitamura, Y., Spiegelman, B.M., Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1α interaction (2003) Nature, 423, pp. 550-555Puigserver, P., Spiegelman, B.M., Peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α): transcriptional coactivator and metabolic regulator (2003) Endocr Rev, 24, pp. 78-90Rhee, J., Inoue, Y., Yoon, J.C., Puigserver, P., Fan, M., Gonzalez, F.J., Spiegelman, B.M., Regulation of hepatic fasting response by PPARγ coactivator-1α (PGC-1): requirement for hepatocyte nuclear factor 4α in gluconeogenesis (2003) Proc Natl Acad Sci U S A, 100, pp. 4012-4017Ropelle, E.R., Pauli, J.R., Cintra, D.E., Frederico, M.J., de Pinho, R.A., Velloso, L.A., De Souza, C.T., Acute exercise modulates the Foxo1/PGC-1α pathway in the liver of diet-induced obesity rats (2009) J Physiol, 587, pp. 2069-2076Ryder, J.W., Fahlman, R., Wallberg-Henriksson, H., Alessi, D.R., Krook, A., Zierath, J.R., Effect of contraction on mitogen-activated protein kinase signal transduction in skeletal muscle. Involvement of the mitogen- and stress-activated protein kinase 1 (2000) J Biol Chem, 275, pp. 1457-1462Schmoll, D., Walker, K.S., Alessi, D.R., Grempler, R., Burchell, A., Guo, S., Walther, R., Unterman, T.G., Regulation of glucose-6-phosphatase gene expression by protein kinase Bα and the forkhead transcription factor FKHR. Evidence for insulin response unit-dependent and -independent effects of insulin on promoter activity (2000) J Biol Chem, 275, pp. 36324-36333Scott, A.M., Atwater, I., Rojas, E., A method for the simultaneous measurement of insulin release and B cell membrane potential in single mouse islets of Langerhans (1981) Diabetologia, 21, pp. 470-475Sutherland, C., O'Brien, R.M., Granner, D.K., Phosphatidylinositol 3-kinase, but not p70/p85 ribosomal S6 protein kinase, is required for the regulation of phosphoenolpyruvate carboxykinase (PEPCK) gene expression by insulin. Dissociation of signaling pathways for insulin and phorbol ester regulation of PEPCK gene expression (1995) J Biol Chem, 270, pp. 15501-15506Sutherland, C., O'Brien, R.M., Granner, D.K., New connections in the regulation of PEPCK gene expression by insulin (1996) Philos Trans R Soc Lond B Biol Sci, 351, pp. 191-199Vidal-Puig, A., O'Rahilly, S., Metabolism. Controlling the glucose factory (2001) Nature, 413, pp. 125-126Widegren, U., Jiang, X.J., Krook, A., Chibalin, A.V., Björnholm, M., Tally, M., Roth, R.A., Zierath, J.R., Divergent effects of exercise on metabolic and mitogenic signaling pathways in human skeletal muscle (1998) FASEB J, 12, pp. 1379-1389Wu, Z., Jiao, P., Huang, X., Feng, B., Feng, Y., Yang, S., Hwang, P., Xu, H., MAPK phosphatase-3 promotes hepatic gluconeogenesis through dephosphorylation of forkhead box O1 in mice (2010) J Clin Invest, 120, pp. 3901-3911Yoon, J.C., Puigserver, P., Chen, G., Donovan, J., Wu, Z., Rhee, J., Adelmant, G., Spiegelman, B.M., Control of hepatic gluconeogenesis through the transcriptional coactivator PGC-1 (2001) Nature, 413, pp. 131-13
    corecore