25 research outputs found

    Resolvin rvd2 reduces hypothalamic inflammation and rescues mice from diet-induced obesity

    Get PDF
    Diet-induced hypothalamic inflammation is an important mechanism leading to dysfunction of neurons involved in controlling body mass. Studies have shown that polyunsaturated fats can reduce hypothalamic inflammation. Here, we evaluated the presence and function of RvD2, a resolvin produced from docosahexaenoic acid, in the hypothalamus of mice. Methods: Male Swiss mice were fed either chow or a high-fat diet. RvD2 receptor and synthetic enzymes were evaluated by real-time PCR and immunofluorescence. RvD2 was determined by mass spectrometry. Dietary and pharmacological approaches were used to modulate the RvD2 system in the hypothalamus, and metabolic phenotype consequences were determined. Results: All enzymes involved in the synthesis of RvD2 were detected in the hypothalamus and were modulated in response to the consumption of dietary saturated fats, leading to a reduction of hypothalamic RvD2. GPR18, the receptor for RvD2, which was detected in POMC and NPY neurons, was also modulated by dietary fats. The substitution of saturated by polyunsaturated fats in the diet resulted in increased hypothalamic RvD2, which was accompanied by reduced body mass and improved glucose tolerance. The intracerebroventricular treatment with docosahexaenoic acid resulted in increased expression of the RvD2 synthetic enzymes, increased expression of anti-inflammatory cytokines and improved metabolic phenotype. Finally, intracerebroventricular treatment with RvD2 resulted in reduced adiposity, improved glucose tolerance and increased hypothalamic expression of anti-inflammatory cytokines. Conclusions: Thus, RvD2 is produced in the hypothalamus, and its receptor and synthetic enzymes are modulated by dietary fats. The improved metabolic outcomes of RvD2 make this substance an attractive approach to treat obesity14511

    Hypothalamic Inhibition Of Acetyl-coa Carboxylase Stimulates Hepatic Counter-regulatory Response Independent Of Ampk Activation In Rats.

    Get PDF
    Hypothalamic AMPK acts as a cell energy sensor and can modulate food intake, glucose homeostasis, and fatty acid biosynthesis. Intrahypothalamic fatty acid injection is known to suppress liver glucose production, mainly by activation of hypothalamic ATP-sensitive potassium (K(ATP)) channels. Since all models employed seem to involve malonyl-CoA biosynthesis, we hypothesized that acetyl-CoA carboxylase can modulate the counter-regulatory response independent of nutrient availability. In this study employing immunoblot, real-time PCR, ELISA, and biochemical measurements, we showed that reduction of the hypothalamic expression of acetyl-CoA carboxylase by antisense oligonucleotide after intraventricular injection increased food intake and NPY mRNA, and diminished the expression of CART, CRH, and TRH mRNA. Additionally, as in fasted rats, in antisense oligonucleotide-treated rats, serum glucagon and ketone bodies increased, while the levels of serum insulin and hepatic glycogen diminished. The reduction of hypothalamic acetyl-CoA carboxylase also increased PEPCK expression, AMPK phosphorylation, and glucose production in the liver. Interestingly, these effects were observed without modification of hypothalamic AMPK phosphorylation. Hypothalamic ACC inhibition can activate hepatic counter-regulatory response independent of hypothalamic AMPK activation.8e6266

    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

    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

    Loss of Toll-Like Receptor 4 Function Partially Protects against Peripheral and Cardiac Glucose Metabolic Derangements During a Long-Term High-Fat Diet

    Get PDF
    We would like to acknowledge Matt Priest for excellent technical assistance.Diabetes is a chronic inflammatory disease that carries a high risk of cardiovascular disease. However, the pathophysiological link between these disorders is not well known. We hypothesize that TLR4 signaling mediates high fat diet (HFD)-induced peripheral and cardiac glucose metabolic derangements. Mice with a loss-of-function mutation in TLR4 (C3H/HeJ) and age-matched control (C57BL/6) mice were fed either a high-fat diet or normal diet for 16 weeks. Glucose tolerance and plasma insulin were measured. Protein expression of glucose transporters (GLUT), AKT (phosphorylated and total), and proinflammatory cytokines (IL-6, TNF-α and SOCS-3) were quantified in the heart using Western Blotting. Both groups fed a long-term HFD had increased body weight, blood glucose and insulin levels, as well as impaired glucose tolerance compared to mice fed a normal diet. TLR4-mutant mice were partially protected against long-term HFD-induced insulin resistance. In control mice, feeding a HFD decreased cardiac crude membrane GLUT4 protein content, which was partially rescued in TLR4-mutant mice. TLR4-mutant mice fed a HFD also had increased expression of GLUT8, a novel isoform, compared to mice fed a normal diet. GLUT8 content was positively correlated with SOCS-3 and IL-6 expression in the heart. No significant differences in cytokine expression were observed between groups, suggesting a lack of inflammation in the heart following a HFD. Loss of TLR4 function partially restored a healthy metabolic phenotype, suggesting that TLR4 signaling is a key mechanism in HFD-induced peripheral and cardiac insulin resistance. Our data further suggest that TLR4 exerts its detrimental metabolic effects in the myocardium through a cytokine-independent pathway.Yeshttp://www.plosone.org/static/editorial#pee

    Topiramate Treatment Improves Hypothalamic Insulin and Leptin Signaling and Action and Reduces Obesity in Mice

    No full text
    Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP)Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq)Topiramate (TPM) treatment has been shown to reduce adiposity in humans and rodents. The reduction in adiposity is related to decreased food intake and increased energy expenditure. However, the molecular mechanisms through which TPM induces weight loss are contradictory and remain to be clarified. Whether TPM treatment alters hypothalamic insulin, or leptin signaling and action, is not well established. Thus, we investigate herein whether short-term TPM treatment alters energy balance by affecting insulin and leptin signaling, action, or neuropeptide expression in the hypothalamus of mice fed with a high-fat diet. As expected, short-term treatment with TPM diminished adiposity in obese mice mainly due to reduced food intake. TPM increased anorexigenic signaling by enhancing the leptin-induced leptin receptor/Janus kinase 2/signal transducer and activator of transcription 3 pathway and the insulin-induced insulin receptor substrate/Akt/forkhead box O1 pathway in parallel to reduced phosphatase protein expression in the hypothalamus of obese mice. These effects were independent of body weight. TPM also raised anorexigenic neuropeptides such as POMC, TRH, and CRH mRNA levels in obese mice. In addition, TPM increased the activation of the hypothalamic MAPK/ERK pathway induced by leptin, accompanied by an increase in peroxisome proliferator-activated receptor-coactivator alpha and uncoupling protein 1 protein levels in brown adipose tissue. Furthermore, TPM increased AMP-activated protein kinase and acetyl-coenzyme A carboxylase phosphorylation in peripheral tissues, which may help improve energy metabolism in these tissues. Together, these results provide novel insights into the molecular mechanisms through which TPM treatment reduces adiposity. (Endocrinology 153: 4401-4411, 2012)o TEXTO COMPLETO DESTE ARTIGO, ESTARÁ DISPONÍVEL À PARTIR DE AGOSTO DE 2015.153944014411Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP)Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq)Instituto Nacional Ciencia e Tecnologia de Obesidade e Diabetes [573856/2008-7]Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP)Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq)FAPESP [2008/55674-8]CNPq [480131/2009-0]Instituto Nacional Ciencia e Tecnologia de Obesidade e Diabetes [573856/2008-7

    Fractalkine (cx3cl1) is involved in the early activation of hypothalamic inflammation in experimental obesity

    No full text
    Hypothalamic inflammation is a common feature of experimental obesity. Dietary fats are important triggers of this process, inducing the activation of toll-like receptor-4 (TLR4) signaling and endoplasmic reticulum stress. Microglia cells, which are the cellular components of the innate immune system in the brain, are expected to play a role in the early activation of diet-induced hypothalamic inflammation. Here, we use bone marrow transplants to generate mice chimeras that express a functional TLR4 in the entire body except in bone marrow-derived cells or only in bone marrow-derived cells. We show that a functional TLR4 in bone marrow-derived cells is required for the complete expression of the diet-induced obese phenotype and for the perpetuation of inflammation in the hypothalamus. In an obesity-prone mouse strain, the chemokine CX3CL1 (fractalkine) is rapidly induced in the neurons of the hypothalamus after the introduction of a high-fat diet. The inhibition of hypothalamic fractalkine reduces diet-induced hypothalamic inflammation and the recruitment of bone marrow-derived monocytic cells to the hypothalamus; in addition, this inhibition reduces obesity and protects against diet-induced glucose intolerance. Thus, fractalkine is an important player in the early induction of diet-induced hypothalamic inflammation, and its inhibition impairs the induction of the obese and glucose intolerance phenotypes.Hypothalamic inflammation is a common feature of experimental obesity. Dietary fats are important triggers of this process, inducing the activation of toll-like receptor-4 (TLR4) signaling and endoplasmic reticulum stress. Microglia cells, which are the cellular components of the innate immune system in the brain, are expected to play a role in the early activation of diet-induced hypothalamic inflammation. Here, we use bone marrow transplants to generate mice chimeras that express a functional TLR4 in the entire body except in bone marrow-derived cells or only in bone marrow-derived cells. We show that a functional TLR4 in bone marrow-derived cells is required for the complete expression of the diet-induced obese phenotype and for the perpetuation of inflammation in the hypothalamus. In an obesity-prone mouse strain, the chemokine CX3CL1 (fractalkine) is rapidly induced in the neurons of the hypothalamus after the introduction of a high-fat diet. The inhibition of hypothalamic fractalkine reduces diet-induced hypothalamic inflammation and the recruitment of bone marrow-derived monocytic cells to the hypothalamus; in addition, this inhibition reduces obesity and protects against diet-induced glucose intolerance. Thus, fractalkine is an important player in the early induction of diet-induced hypothalamic inflammation, and its inhibition impairs the induction of the obese and glucose intolerance phenotypes63113770378

    Hypothalamic stearoyl-CoA desaturase-2 (SCD2) controls whole-body energy expenditure

    No full text
    BACKGROUND/OBJECTIVES: Stearoyl-CoA desaturase-2 (SCD2) is the main delta 9 desaturase expressed in the central nervous system. Because of its potential involvement in controlling whole-body adiposity, we evaluated the expression and function of SCD2 in the hypothalami of mice. SUBJECTS/METHODS: Male mice of different strains were used in real-time PCR, immunoblot and metabolic experiments. In addition, antisense oligonucleotides and lentiviral vectors were used to reduce and increase the expression of SCD2 in the hypothalamus. RESULTS: The level of SCD2 in the hypothalamus is similar to other regions of the central nervous system and is similar to 10-fold higher than in any other region of the body. In the arcuate nucleus, SCD2 is expressed in proopiomelanocortin and neuropeptide-Y neurons. Upon high fat feeding, the level of hypothalamic SCD2 increases. Inhibition of hypothalamic SCD2 as accomplished by two distinct approaches, an antisense oligonucleotide or a short-hairpin RNA delivered by a lentivirus, resulted in reduced body mass gain mostly due to increased energy expenditure and increased spontaneous activity. Increasing hypothalamic SCD2 by a lentivirus approach resulted in no change in body mass and food intake. CONCLUSIONS: Thus, SCD2 is highly expressed in the hypothalami of rodents and its knockdown reduces body mass due to increased whole-body energy expenditure403471478CONSELHO NACIONAL DE DESENVOLVIMENTO CIENTÍFICO E TECNOLÓGICO - CNPQFUNDAÇÃO DE AMPARO À PESQUISA DO ESTADO DE SÃO PAULO - FAPESPsem informaçã
    corecore