Hypothalamic AMPK and fatty acid metabolism mediate thyroid regulation of energy balance

Miguel López, Luis Varela, María J Vázquez, Sergio Rodríguez-Cuenca, Carmen R González, Vidya R Velagapudi, Donald A Morgan, Erik Schoenmakers, Khristofor Agassandian, Ricardo Lage, Pablo Blanco Martínez de Morentin, Sulay Tovar, Rubén Nogueiras, David Carling, Christopher Lelliott, Rosalía Gallego, Matej Oresic, Krishna Chatterjee, Asish K Saha, Kamal Rahmouni, Carlos Diéguez, Antonio Vidal-Puig, Miguel López, Luis Varela, María J Vázquez, Sergio Rodríguez-Cuenca, Carmen R González, Vidya R Velagapudi, Donald A Morgan, Erik Schoenmakers, Khristofor Agassandian, Ricardo Lage, Pablo Blanco Martínez de Morentin, Sulay Tovar, Rubén Nogueiras, David Carling, Christopher Lelliott, Rosalía Gallego, Matej Oresic, Krishna Chatterjee, Asish K Saha, Kamal Rahmouni, Carlos Diéguez, Antonio Vidal-Puig

Abstract

Thyroid hormones have widespread cellular effects; however it is unclear whether their effects on the central nervous system (CNS) contribute to global energy balance. Here we demonstrate that either whole-body hyperthyroidism or central administration of triiodothyronine (T3) decreases the activity of hypothalamic AMP-activated protein kinase (AMPK), increases sympathetic nervous system (SNS) activity and upregulates thermogenic markers in brown adipose tissue (BAT). Inhibition of the lipogenic pathway in the ventromedial nucleus of the hypothalamus (VMH) prevents CNS-mediated activation of BAT by thyroid hormone and reverses the weight loss associated with hyperthyroidism. Similarly, inhibition of thyroid hormone receptors in the VMH reverses the weight loss associated with hyperthyroidism. This regulatory mechanism depends on AMPK inactivation, as genetic inhibition of this enzyme in the VMH of euthyroid rats induces feeding-independent weight loss and increases expression of thermogenic markers in BAT. These effects are reversed by pharmacological blockade of the SNS. Thus, thyroid hormone-induced modulation of AMPK activity and lipid metabolism in the hypothalamus is a major regulator of whole-body energy homeostasis.

Figures

Figure 1. Energy balance, AMPK pathway and…
Figure 1. Energy balance, AMPK pathway and POMC expression
(a) Western blots (left panel) for hypothalamic protein levels (middle panels) of pAMPKα, AMPKα1, AMPKα2, tAMPKα, pACCα, ACCα (lower band in the tAMPK gel), ACCβ (upper band in the tAMPK gel) and FAS and hypothalamic AMPKα1 and AMPKα2 activities (right panel) in euthyroid and hyperthyroid rats. (b–d) Hypothalamic levels of Fasn (b), malonyl-CoA content (c) and CPT1 activity (d) in euthyroid and hyperthyroid rats. (e–i) Body weight change (e), daily food intake (f), hypothalamic malonyl-CoA levels (g), Pomc mRNA levels in the ARC (h) and western blots (left panel) for hypothalamic protein levels (right panel) of pFoxO1 and pSTAT3 (i) of euthyroid and hyperthyroid rats treated ICV with vehicle or cerulenin for 4 d. ¶P = 0.1, *P < 0.05, **P < 0.01, ***P < 0.001 vs. vehicle or euthyroid vehicle; #P < 0.05 euthyroid cerulenin vs. hyperthyroid cerulenin; ###P < 0.001 hyperthyroid vehicle vs. hyperthyroid cerulenin; all data are expressed as mean ± SEM.
Figure 2. Effects of chronic central T3…
Figure 2. Effects of chronic central T3 administration
(a–d) Body weight change (a, b), daily food intake (c, d) of euthyroid and hypothyroid rats ICV-treated with T3 for 4 d. (e–f) Western blots (left panel) for hypothalamic protein levels (right panel) of pAMPKα, AMPKα1, AMPKα2, pACCα and ACCα (e) and mRNA expression profiles in BAT (f) of euthyroid rats ICV-treated with T3 for 4 d. (g–i) Body weight change (g), daily food intake (h) and mRNA expression profiles in BAT (i) of euthyroid rats ICV-treated with T3 and subcutaneously (SC) -treated with the β3-AR specific antagonist SR59230A for 4 d.!P = 0.09, ¡P = 0.08, +P = 0.06, *P < 0.05, **P < 0.01, ***P < 0.001 vs. vehicle; #P < 0.05, ##P < 0.01 T3 ICV vs. T3 ICV SR59230A; all data are expressed as mean ± SEM.
Figure 3. Effects of central T3 on…
Figure 3. Effects of central T3 on BAT activation via the SNS
(a) Double immunohistochemistry (upper: 40 ×, scale bar, 200 μm; lower: 200 ×; scale bar, 20 μm) showing pAMPKα and TRα coexpression in the VMH. (b–e) Western blots (left panel) for hypothalamic protein levels (right panel) of pAMPKα, AMPKα1, AMPKα2, pACCα and ACCα (b), immunohistochemistry showing c-FOS immunoreactivity (IR) in the DMV (upper images 40 ×, scale bar, 200 μm) and in the RPa and the IO (lower images 100 ×, scale bar 100 μm) and c-FOS-IR cells in those nuclei (c) and BAT SNA (d, e) of euthyroid rats 1–3 h (protein and c-FOS) or 6 h (SNA) after ICV treatment with T3. (f, g) Western blots (left panel) for hypothalamic protein levels (right panel) of pAMPKα, AMPKα1, AMPKα2, pACCα and ACCα (f) and BAT SNA (g) of euthyroid rats 1 h after VMH microinjection of T3. P = 0.1, *P < 0.05, **P < 0.01, ***P < 0.001 vs. vehicle; #P < 0.05 T3 ICV 2 ng vs. T3 ICV 4 ng; all data are expressed as mean ± SEM. 3V: third ventricle; CC: central canal; HN: hypoglossal nucleus.
Figure 4. Effects of genetic ablation of…
Figure 4. Effects of genetic ablation of thyroid hormone receptor in the VMH
(a–e) Body weight change (a), daily food intake (b), plasma T3 (c) and T4 (d) levels and mRNA expression profiles in BAT (e) of hyperthyroid (and euthyroid when indicated) rats stereotaxically treated with GFP-expressing adenoviruses or GFP plus TR-DN adenoviruses into the VMH. *P < 0.05, **P < 0.01, ***P < 0.001 vs. euthyroid GFP or hyperthyroid GFP; all data are expressed as mean ± SEM.
Figure 5. Effects of inactivation of hypothalamic…
Figure 5. Effects of inactivation of hypothalamic de novo lipogenesis
(a–d) Body weight change (left panel) and daily food intake (right panel) (a), hypothalamic malonyl-CoA levels (b) and Ucp1 and Ucp3 mRNA in the BAT (c) of hyperthyroid (and euthyroid when indicated) rats treated with vehicle or TOFA. (d–f) Body weight change (left panel) daily food intake (right panel) (d), hypothalamic malonyl-CoA levels (e) and Ucp1 and Ucp3 mRNA and in the BAT (f) of hyperthyroid (and euthyroid when indicated) rats treated with vehicle or AICAR. (g–i) Malonyl-CoA levels in the ventral hypothalamus (g) body weight change (left panel), food intake (right panel) (h) and mRNA expression profiles in BAT (i) of hyperthyroid (or euthyroid when indicated) rats stereotaxically treated with a GFP-expressing adenoviruses or GFP plus AMPK constitutively active (AMPKα-CA) adenoviruses into the VMH. *P < 0.05, **P < 0.01, ***P < 0.001 vs. vehicle or GFP; ###P < 0.01 hyperthyroid vehicle vs. hyperthyroid TOFA or AICAR and hyperthyroid GFP vs. hyperthyroid AMPKα-CA; all data are expressed as mean ± SEM.
Figure 6. Effects of selective inactivation of…
Figure 6. Effects of selective inactivation of AMPK in the VMH
(a–d) Malonyl-CoA levels in the ventral hypothalamus (a), body weight change (b), food intake (c), and mRNA expression profiles in BAT (d) of euthyroid rats stereotaxically treated with a GFP-expressing adenoviruses or GFP plus AMPK dominant negative (AMPKα-DN) into the VMH. (e–g) Body weight change (e), food intake (f), and mRNA expression profiles in BAT (g) of rats stereotaxically treated into the VMH with GFP-expressing adenoviruses SC-treated with vehicle, GFP plus AMPKα-DN SC-treated with vehicle and GFP plus AMPKα-DN SC-treated with the β3-AR specific antagonist SR59230A. (h) Proposed model of the effect of thyroid hormones excess on hypothalamic fatty acid metabolism. Hyperthyroidism and T3 upregulate de novo lipogenesis in the hypothalamus which results from decreased activity of AMPK, activation of ACC and increased expression of Fasn. Thyroid hormone-induced changes in hypothalamic lipid biosynthetic pathway increases levels of hypothalamic malonyl-CoA and complex lipids. These changes are associated with the activation of the SNS through the RPa and the IO, resulting in increased expression of BAT markers, such as Ucp1, Upc3, Ppargc1a (which encodes PGC1α) and Ppargc1b (which encodes PGC1β), promoting negative energy balance and weight loss. *: P < 0.05, **P < 0.01, ***P < 0.001 vs. GFP; #P < 0.05, ##P < 0.01 AMPKα-DN vehicle vs. AMPKα-DN SR59230A; all data are expressed as mean ± SEM.

References

    1. Silva JE. Thyroid hormone control of thermogenesis and energy balance. Thyroid. 1995;5:481–492.
    1. Coppola A, et al. A central thermogenic-like mechanism in feeding regulation: an interplay between arcuate nucleus T3 and UCP2. Cell Metab. 2007;5:21–33.
    1. Herwing A, Ross AW, Nilaweera KN, Morgan PJ, Barrett P. Hypothalamic thyroid hormone in energy balance regulation. Obes. Facts. 2008;1:71–79.
    1. Pijl H, et al. Food choice in hyperthyroidism: potential influence of the autonomic nervous system and brain serotonin precursor availability. J. Clin. Endocrinol. Metab. 2001;86:5848–5853.
    1. Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84:277–359.
    1. Volpe JJ, Kishimoto Y. Fatty acid synthetase of brain: development, influence of nutritional and hormonal factors and comparison with liver enzyme. J Neurochem. 1972;19:737–753.
    1. Gnoni GV, Landriscina C, Ruggiero FM, Quagliariello E. Effect of hyperthyroidism on lipogenesis in brown adipose tissue of young rats. Biochim. Biophys. Acta. 1983;751:271–279.
    1. Blennemann B, Leahy P, Kim TS, Freake HC. Tissue-specific regulation of lipogenic mRNAs by thyroid hormone. Mol. Cell Endocrinol. 1995;110:1–8.
    1. Cachefo A, et al. Hepatic lipogenesis and cholesterol synthesis in hyperthyroid patients. J. Clin. Endocrinol. Metab. 2001;86:5353–5357.
    1. Park SH, et al. Effects of thyroid state on AMP-activated protein kinase and acetyl-CoA carboxylase expression in muscle. J Appl. Physiol. 2002;93:2081–2088.
    1. Winder WW, et al. Long-term regulation of AMP-activated protein kinase and acetyl-CoA carboxylase in skeletal muscle. Biochem. Soc. Trans. 2003;31:182–185.
    1. Branvold DJ, et al. Thyroid Hormone effects on LKB1, MO25, phospho-AMPK, phospho-CREB, and PGC-1{alpha} in Rat Muscle. J. Appl. Physiol. 2008;105:1218–1227.
    1. Irrcher I, Walkinshaw DR, Sheehan TE, Hood DA. Thyroid hormone (T3) rapidly activates p38 and AMPK in skeletal muscle in vivo. J. Appl. Physiol. 2008;104:178–185.
    1. Morini P, Conserva AR, Lippolis R, Casalino E, Landriscina C. Differential action of thyroid hormones on the activity of certain enzymes in rat kidney and brain. Biochem. Med. Metab Biol. 1991;46:169–176.
    1. Blennemann B, Moon YK, Freake HC. Tissue-specific regulation of fatty acid synthesis by thyroid hormone. Endocrinology. 1992;130:637–643.
    1. Minokoshi Y, et al. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature. 2004;428:569–574.
    1. Gao S, et al. Leptin activates hypothalamic acetyl-CoA carboxylase to inhibit food intake. Proc. Natl. Acad. Sci. U. S. A. 2007;104:17358–17363.
    1. Kola B, et al. The orexigenic effect of ghrelin is mediated through central activation of the endogenous cannabinoid system. PLoS. ONE. 2008;3:e1797.
    1. López M, et al. Hypothalamic fatty acid metabolism mediates the orexigenic action of ghrelin. Cell Metab. 2008;7:389–399.
    1. Andrews ZB, et al. UCP2 mediates ghrelin’s action on NPY/AgRP neurons by lowering free radicals. Nature. 2008;454:846–851.
    1. Loftus TM, et al. Reduced food intake and body weight in mice treated with fatty acid synthase inhibitors. Science. 2000;288:2379–2381.
    1. Hu Z, Cha SH, Chohnan S, Lane MD. Hypothalamic malonyl-CoA as a mediator of feeding behavior. Proc. Natl. Acad. Sci. U. S. A. 2003;100:12624–12629.
    1. Obici S, Feng Z, Arduini A, Conti R, Rossetti L. Inhibition of hypothalamic carnitine palmitoyltransferase-1 decreases food intake and glucose production. Nat. Med. 2003;9:756–761.
    1. Lam TK, Schwartz GJ, Rossetti L. Hypothalamic sensing of fatty acids. Nat. Neurosci. 2005;8:579–584.
    1. Wolfgang MJ, et al. The brain-specific carnitine palmitoyltransferase-1c regulates energy homeostasis. Proc. Natl. Acad. Sci. U. S. A. 2006;103:7282–7287.
    1. López M, et al. Tamoxifen-induced anorexia is associated with fatty acid synthase inhibition in the ventromedial nucleus of the hypothalamus and accumulation of malonyl-CoA. Diabetes. 2006;55:1327–1336.
    1. Chakravarthy MV, et al. Brain fatty acid synthase activates PPAR-alpha to maintain energy homeostasis. J. Clin. Invest. 2007;117:2539–2552.
    1. Lam TK. Neuronal regulation of homeostasis by nutrient sensing. Nat. Med. 2010;16:392–395.
    1. Dulloo AG. Biomedicine. A sympathetic defense against obesity. Science. 2002;297:780–781.
    1. Commins SP, Watson PM, Levin N, Beiler RJ, Gettys TW. Central leptin regulates the UCP1 and ob genes in brown and white adipose tissue via different beta-adrenoceptor subtypes. J. Biol. Chem. 2000;275:33059–33067.
    1. Tong Q, et al. Synaptic glutamate release by ventromedial hypothalamic neurons is part of the neurocircuitry that prevents hypoglycemia. Cell Metab. 2007;5:383–393.
    1. Chatterjee VK, et al. Thyroid hormone resistance syndrome. Inhibition of normal receptor function by mutant thyroid hormone receptors. J. Clin. Invest. 1991;87:1977–1984.
    1. Lage R, et al. Ghrelin effects on neuropeptides in the rat hypothalamus depend on fatty acid metabolism actions on BSX but not on gender. FASEB J. 2010;24:2670–2679.
    1. Hagenfeldt L, Wennlung A, Felig P, Wahren J. Turnover and splanchnic metabolism of free fatty acids in hyperthyroid patients. J. Clin. Invest. 1981;67:1672–1677.
    1. Beylot M, et al. Lipolytic and ketogenic fluxes in human hyperthyroidism. J. Clin. Endocrinol. Metab. 1991;73:42–49.
    1. Riis AL, et al. Elevated regional lipolysis in hyperthyroidism. J. Clin. Endocrinol. Metab. 2002;87:4747–4753.
    1. Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated protein kinase: Ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 2005;1:15–25.
    1. Lage R, Diéguez C, Vidal-Puig A, López M. AMPK: a metabolic gauge regulating whole-body energy homeostasis. Trends Mol. Med. 2008;14:539–549.
    1. Plum L, et al. The obesity susceptibility gene Cpe links FoxO1 signaling in hypothalamic proopiomelanocortin neurons with regulation of food intake. Nat. Med. 2009;15:1195–1201.
    1. Belgardt BF, et al. PDK1 deficiency in POMC-expressing cells reveals FOXO1-dependent and - independent pathways in control of energy homeostasis and stress response. Cell Metab. 2008;7:291–301.
    1. Pocai A, et al. Restoration of hypothalamic lipid sensing normalizes energy and glucose homeostasis in overfed rats. J. Clin. Invest. 2006;116:1081–1091.
    1. He W, Lam TK, Obici S, Rossetti L. Molecular disruption of hypothalamic nutrient sensing induces obesity. Nat. Neurosci. 2006;9:227–233.
    1. Sangiao-Alvarellos S, et al. Influence of ghrelin and GH deficiency on AMPK and hypothalamic lipid metabolism. J. Neuroendocrinol. 2010;22:543–556.
    1. Niijima A, Rohner-Jeanrenaud F, Jeanrenaud B. Role of ventromedial hypothalamus on sympathetic efferents of brown adipose tissue. Am. J. Physiol. 1984;247:R650–R654.
    1. Holt SJ, Wheal HV, York DA. Hypothalamic control of brown adipose tissue in Zucker lean and obese rats. Effect of electrical stimulation of the ventromedial nucleus and other hypothalamic centres. Brain Res. 1987;405:227–233.
    1. Halvorson I, Gregor L, Thornhill JA. Brown adipose tissue thermogenesis is activated by electrical and chemical (L-glutamate) stimulation of the ventromedial hypothalamic nucleus in cold-acclimated rats. Brain Res. 1990;522:76–82.
    1. McCrimmon RJ, et al. Key role for AMP-activated protein kinase in the ventromedial hypothalamus in regulating counterregulatory hormone responses to acute hypoglycemia. Diabetes. 2008;57:444–450.
    1. Nedergaard J, Bengtsson T, Cannon B. Unexpected evidence for active brown adipose tissue in adult humans. Am. J. Physiol Endocrinol. Metab. 2007;293:E444–E452.
    1. Marken Lichtenbelt WD, et al. Cold-activated brown adipose tissue in healthy men. N. Engl. J. Med. 2009;360:1500–1508.
    1. Cypess AM, et al. Identification and importance of brown adipose tissue in adult humans. N. Engl. J. Med. 2009;360:1509–1517.
    1. Virtanen KA, et al. Functional brown adipose tissue in healthy adults. N. Engl. J. Med. 2009;360:1518–1525.
    1. Skarulis MC, et al. Thyroid hormone induced brown adipose tissue and amelioration of diabetes in a patient with extreme insulin resistance. J. Clin. Endocrinol. Metab. 2010;95:256–262.
    1. Rahmouni K, et al. Hypothalamic PI3K and MAPK differentially mediate regional sympathetic activation to insulin. J. Clin. Invest. 2004;114:652–658.
    1. Nogueiras R, et al. Direct control of peripheral lipid deposition by CNS GLP-1 receptor signaling is mediated by the sympathetic nervous system and blunted in diet induced obesity. J Neurosci. 2009;29:5916–5925.

Source: PubMed

Подписаться