1,256
Views
14
CrossRef citations to date
0
Altmetric
Research Paper

MicroRNAs are involved in the hypothalamic leptin sensitivity

, ORCID Icon, , , ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 1127-1140 | Received 31 Jul 2018, Accepted 22 Oct 2018, Published online: 11 Nov 2018

References

  • Derghal A, Djelloul M, Trouslard J, et al. The role of MicroRNA in the modulation of the melanocortinergic system. Front Neurosci. 2017;11:181.
  • Cowley MA, Smart JL, Rubinstein M, et al. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature. 2001;411:480–484.
  • Ibrahim N, Bosch MA, Smart JL, et al. Hypothalamic proopiomelanocortin neurons are glucose responsive and express K(ATP) channels. Endocrinology. 2003;144:1331–1340.
  • Plum L, Belgardt BF, Brüning JC. Central insulin action in energy and glucose homeostasis. J Clin Invest. 2006;116:1761–1766.
  • Parton LE, Ye CP, Coppari R, et al. Glucose sensing by POMC neurons regulates glucose homeostasis and is impaired in obesity. Nature. 2007;449:228–232.
  • Mounien L, Do Rego J-C, Bizet P, et al. Pituitary adenylate cyclase-activating polypeptide inhibits food intake in mice through activation of the hypothalamic melanocortin system. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2009;34:424–435.
  • Mounien L, Marty N, Tarussio D, et al. Glut2-dependent glucose-sensing controls thermoregulation by enhancing the leptin sensitivity of NPY and POMC neurons. FASEB J Off Publ Fed Am Soc Exp Biol. 2010;24:1747–1758.
  • Hill JW, Elias CF, Fukuda M, Williams KW, Berglund ED, Holland WL, Cho Y-R, Chuang J-C, Xu Y, Choi M, et al. Direct insulin and leptin action on pro-opiomelanocortin neurons is required for normal glucose homeostasis and fertility. Cell Metab. 2010;11:286–297.
  • Huszar D, Lynch CA, Fairchild-Huntress V, et al. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell. 1997;88:131–141.
  • Yaswen L, Diehl N, Brennan MB, et al. Obesity in the mouse model of pro-opiomelanocortin deficiency responds to peripheral melanocortin. Nat Med. 1999;5:1066–1070.
  • Lee YS. The role of leptin-melanocortin system and human weight regulation: lessons from experiments of nature. Ann Acad Med Singapore. 2009;38:34.
  • Bouret SG, Draper SJ, Simerly RB. Trophic action of leptin on hypothalamic neurons that regulate feeding. Science. 2004;304:108–110.
  • Gao Q, Wolfgang MJ, Neschen S, et al. Disruption of neural signal transducer and activator of transcription 3 causes obesity, diabetes, infertility, and thermal dysregulation. Proc Natl Acad Sci U S A. 2004;101:4661–4666.
  • Pinto S, Roseberry AG, Liu H, et al. Rapid rewiring of arcuate nucleus feeding circuits by leptin. Science. 2004;304:110–115.
  • Coupé B, Ishii Y, Dietrich MO, et al. Loss of autophagy in pro-opiomelanocortin neurons perturbs axon growth and causes metabolic dysregulation. Cell Metab. 2012;15:247–255.
  • Stevens A, Begum G, Cook A, et al. Epigenetic changes in the hypothalamic proopiomelanocortin and glucocorticoid receptor genes in the ovine fetus after periconceptional undernutrition. Endocrinology. 2010;151:3652–3664.
  • Gali Ramamoorthy T, Allen T-J, Davies A, et al. Maternal overnutrition programs epigenetic changes in the regulatory regions of hypothalamic Pomc in the offspring of rats. Int J Obes. 2018;42:1431–1444.
  • Vogt MC, Paeger L, Hess S, et al. Neonatal insulin action impairs hypothalamic neurocircuit formation in response to maternal high-fat feeding. Cell. 2014;156:495–509.
  • Marco A, Kisliouk T, Tabachnik T, et al. Overweight and CpG methylation of the Pomc promoter in offspring of high-fat-diet-fed dams are not “reprogrammed” by regular chow diet in rats. FASEB J Off Publ Fed Am Soc Exp Biol. 2014;28:4148–4157.
  • Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297.
  • Derghal A, Djelloul M, Trouslard J, et al. An emerging role of micro-RNA in the effect of the endocrine disruptors. Front Neurosci. 2016;10:318.
  • Esau C, Davis S, Murray SF, et al. miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab. 2006;3:87–98.
  • Frost RJA, Olson EN. Control of glucose homeostasis and insulin sensitivity by the Let-7 family of microRNAs. Proc Natl Acad Sci U S A. 2011;108:21075–21080.
  • Grueter CE, van Rooij E, Johnson BA, et al. A cardiac microRNA governs systemic energy homeostasis by regulation of MED13. Cell. 2012;149:671–683.
  • Mang GM, Pradervand S, Du N-H, et al. A neuron-specific deletion of the microRNA-processing enzyme DICER induces severe but transient obesity in mice. PloS One. 2015;10:e0116760.
  • Vinnikov IA, Hajdukiewicz K, Reymann J, et al. Hypothalamic miR-103 protects from hyperphagic obesity in mice. J Neurosci Off J Soc Neurosci. 2014;34:10659–10674.
  • Schneeberger M, Altirriba J, García A, et al. Deletion of miRNA processing enzyme Dicer in POMC-expressing cells leads to pituitary dysfunction, neurodegeneration and development of obesity. Mol Metab. 2012;2:74–85.
  • Greenman Y, Kuperman Y, Drori Y, et al. Postnatal ablation of POMC neurons induces an obese phenotype characterized by decreased food intake and enhanced anxiety-like behavior. Mol Endocrinol Baltim Md. 2013;27:1091–1102.
  • Crépin D, Benomar Y, Riffault L, et al. The over-expression of miR-200a in the hypothalamus of ob/ob mice is linked to leptin and insulin signaling impairment. Mol Cell Endocrinol. 2014;384:1–11.
  • Derghal A, Djelloul M, Airault C, et al. Leptin is required for hypothalamic regulation of miRNAs targeting POMC 3ʹUTR. Front Cell Neurosci. 2015;9:172.
  • Harfe BD, McManus MT, Mansfield JH, et al. The RNaseIII enzyme Dicer is required for morphogenesis but not patterning of the vertebrate limb. Proc Natl Acad Sci U S A. 2005;102:10898–10903.
  • Balthasar N, Coppari R, McMinn J, et al. Leptin receptor signaling in POMC neurons is required for normal body weight homeostasis. Neuron. 2004;42:983–991.
  • Mercader JM, González JR, Lozano JJ, et al. Aberrant brain microRNA target and miRISC gene expression in the anx/anx anorexia mouse model. Gene. 2012;497:181–190.
  • Meyer CW, Reitmeir P, Tschöp MH. Exploration of energy metabolism in the mouse using indirect calorimetry: measurement of daily energy expenditure (DEE) and basal metabolic rate (BMR). Curr Protoc Mouse Biol. 2015;5:205–222.
  • Tschöp MH, Speakman JR, Arch JRS, et al. A guide to analysis of mouse energy metabolism. Nat Methods. 2011;9:57–63.
  • Do Carmo JM, Da Silva AA, Dubinion J, et al. Control of metabolic and cardiovascular function by the leptin-brain melanocortin pathway. IUBMB Life. 2013;65:692–698.
  • Fan W, Boston BA, Kesterson RA, et al. Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature. 1997;385:165–168.
  • Butler AA, Kesterson RA, Khong K, et al. A unique metabolic syndrome causes obesity in the melanocortin-3 receptor-deficient mouse. Endocrinology. 2000;141:3518–3521.
  • Chen AS, Marsh DJ, Trumbauer ME, et al. Inactivation of the mouse melanocortin-3 receptor results in increased fat mass and reduced lean body mass. Nat Genet. 2000;26:97–102.
  • Pritchard LE, Turnbull AV, White A. Pro-opiomelanocortin processing in the hypothalamus: impact on melanocortin signalling and obesity. J Endocrinol. 2002;172:411–421.
  • Flynn MC, Scott TR, Pritchard TC, et al. Mode of action of OB protein (leptin) on feeding. Am J Physiol. 1998;275:R174–179.
  • Smith GP. The controls of eating: a shift from nutritional homeostasis to behavioral neuroscience. Nutr Burbank Los Angel Cty Calif. 2000;16:814–820.
  • Baran K, Preston E, Wilks D, et al. Chronic central melanocortin-4 receptor antagonism and central neuropeptide-Y infusion in rats produce increased adiposity by divergent pathways. Diabetes. 2002;51:152–158.
  • Small CJ, Liu YL, Stanley SA, Connoley IP, Kennedy A, Stock MJ, Bloom SR. Chronic CNS administration of Agouti-related protein (Agrp) reduces energy expenditure. Int J Obes Relat Metab Disord J Int Assoc Study Obes. 2003;27:530–533.
  • Yasuda T, Masaki T, Kakuma T, et al. Hypothalamic melanocortin system regulates sympathetic nerve activity in brown adipose tissue. Exp Biol Med Maywood NJ. 2004;229:235–239.
  • Cano G, Passerin AM, Schiltz JC, et al. Anatomical substrates for the central control of sympathetic outflow to interscapular adipose tissue during cold exposure. J Comp Neurol. 2003;460:303–326.
  • Voss-Andreae A, Murphy JG, Ellacott KLJ, et al. Role of the central melanocortin circuitry in adaptive thermogenesis of brown adipose tissue. Endocrinology. 2007;148:1550–1560.
  • Williams DL, Bowers RR, Bartness TJ, et al. Brainstem melanocortin 3/4 receptor stimulation increases uncoupling protein gene expression in brown fat. Endocrinology. 2003;144:4692–4697.
  • Song CK, Vaughan CH, Keen-Rhinehart E, et al. Melanocortin-4 receptor mRNA expressed in sympathetic outflow neurons to brown adipose tissue: neuroanatomical and functional evidence. Am J Physiol Regul Integr Comp Physiol. 2008;295:R417–428.
  • Martins FF, Bargut TCL, Aguila MB, et al. Thermogenesis, fatty acid synthesis with oxidation, and inflammation in the brown adipose tissue of ob/ob (-/-) mice. Ann Anat Anat Anz Off Organ Anat Ges. 2017;210:44–51.
  • Herzer S, Silahtaroglu A, Meister B. Locked nucleic acid-based in situ hybridisation reveals miR-7a as a hypothalamus-enriched microRNA with a distinct expression pattern. J Neuroendocrinol. 2012;24:1492–1504.
  • Sangiao-Alvarellos S, Pena-Bello L, Manfredi-Lozano M, et al. Perturbation of hypothalamic microRNA expression patterns in male rats after metabolic distress: impact of obesity and conditions of negative energy balance. Endocrinology. 2014;155:1838–1850.
  • Novak A, Guo C, Yang W, et al. Z/EG, a double reporter mouse line that expresses enhanced green fluorescent protein upon Cre-mediated excision. Genes N Y N. 2000;28:147–155.
  • Dominguez JF, Guo L, Carrasco Molnar MA, et al. Novel indirect calorimetry technology to analyze metabolism in individual neonatal rodent pups. PloS One. 2009;4:e6790.
  • Marcotorchino J, Tourniaire F, Astier J, et al. Vitamin D protects against diet-induced obesity by enhancing fatty acid oxidation. J Nutr Biochem. 2014;25:1077–1083.
  • Karkeni E, Bonnet L, Astier J, et al. All-trans-retinoic acid represses chemokine expression in adipocytes and adipose tissue by inhibiting NF-κB signaling. J Nutr Biochem. 2017;42:101–107.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.