Canna~Fangled Abstracts

Hypothalamic POMC neurons promote cannabinoid-induced feeding

By February 18, 2015December 17th, 2020No Comments
ARTICLE PREVIEW view full access options

Marco KochLuis Varela, Jae Geun Kim, Jung Dae Kim, Francisco Hernández-Nuño, Stephanie E. Simonds, Carlos M. Castorena, Claudia R. Vianna, Joel K. Elmquist, Yury M. Morozov, Pasko Rakic, Ingo Bechmann, Michael A. Cowley, Klara Szigeti-Buck, Marcelo O. Dietrich, Xiao-Bing Gao, Sabrina Diano Tamas L. Horvath

Affiliations

Nature (2015) doi:10.1038/nature14260
Received
Accepted
Published online
18 February 2015

Nature MedicineHypothalamic pro-opiomelanocortin (POMC) neurons promote satiety. Cannabinoid receptor 1 (CB1R) is critical for the central regulation of food intake. Here we test whether CB1R-controlled feeding in sated mice is paralleled by decreased activity of POMC neurons. We show that chemical promotion of CB1R activity increases feeding, and notably, CB1R activation also promotes neuronal activity of POMC cells. This paradoxical increase in POMC activity was crucial for CB1R-induced feeding, because designer-receptors-exclusively-activated-by-designer-drugs (DREADD)-mediated inhibition of POMC neurons diminishes, whereas DREADD-mediated activation of POMC neurons enhances CB1R-driven feeding. The Pomc gene encodes both the anorexigenic peptide α-melanocyte-stimulating hormone, and the opioid peptide β-endorphin. CB1R activation selectively increases β-endorphin but not α-melanocyte-stimulating hormone release in the hypothalamus, and systemic or hypothalamic administration of the opioid receptor antagonist naloxone blocks acute CB1R-induced feeding. These processes involve mitochondrial adaptations that, when blocked, abolish CB1R-induced cellular responses and feeding. Together, these results uncover a previously unsuspected role of POMC neurons in the promotion of feeding by cannabinoids.

References

  1. Dietrich, M. O. & Horvath, T. L. Hypothalamic control of energy balance: insights into the role of synaptic plasticityTrends Neurosci. 366573 (2013)
  2. Aponte, Y.Atasoy, D. & Sternson, S. M. AGRP neurons are sufficient to orchestrate feeding behavior rapidly and without trainingNature Neurosci. 14351355 (2011)
  3. Krashes, M. J. et al. Rapid, reversible activation of AgRP neurons drives feeding behavior in miceJ. Clin. Invest. 12114241428 (2011)
  4. Yang, Y.Atasoy, D.Su, H. H. & Sternson, S. M. Hunger states switch a flip-flop memory circuit via a synaptic AMPK-dependent positive feedback loopCell 1469921003 (2011)
  5. DiPatrizio, N. V. & Piomelli, D. The thrifty lipids: endocannabinoids and the neural control of energy conservationTrends Neurosci. 35403411 (2012)
  6. Bermudez-Silva, F. J.Cardinal, P. & Cota, D. The role of the endocannabinoid system in the neuroendocrine regulation of energy balanceJ. Psychopharmacol. 26114124 (2012)
  7. Sinnayah, P. et al. Feeding induced by cannabinoids is mediated independently of the melanocortin systemPLoS ONE 3e2202 (2008)
  8. Bakkali-Kassemi, L. et al. Effects of cannabinoids on neuropeptide Y and β-endorphin expression in the rat hypothalamic arcuate nucleusBr. J. Nutr. 105654660 (2011)
  9. Ho, J.Cox, J. M. & Wagner, E. J. Cannabinoid-induced hyperphagia: correlation with inhibition of proopiomelanocortin neurons? Physiol. Behav. 92507519 (2007)
  10. Hentges, S. T.Low, M. J. & Williams, J. T. Differential regulation of synaptic inputs by constitutively released endocannabinoids and exogenous cannabinoidsJ. Neurosci. 2597469751 (2005)
  11. Bellocchio, L. et al. Bimodal control of stimulated food intake by the endocannabinoid systemNature Neurosci. 13281283 (2010)
  12. Hentges, S. T. Synaptic regulation of proopiomelanocortin neurons can occur distal to the arcuate nucleusJ. Neurophysiol. 9732983304 (2007)
  13. Zhan, C. et al. Acute and long-term suppression of feeding behavior by POMC neurons in the brainstem and hypothalamus, respectivelyJ. Neurosci. 3336243632 (2013)
  14. Dores, R. M. & Baron, A. J. Evolution of POMC: origin, phylogeny, posttranslational processing, and the melanocortinsAnn. NY Acad. Sci. 12203448 (2011)
  15. Dube, M. G.Horvath, T. L.Leranth, C.Kalra, P. S. & Kalra, S. P. Naloxone reduces the feeding evoked by intracerebroventricular galanin injectionPhysiol. Behav. 56811813(1994)
  16. Kalra, S. P. & Horvath, T. L. Neuroendocrine interactions between galanin, opioids, and neuropeptide Y in the control of reproduction and appetiteAnn. NY Acad. Sci. 863236240(1998)
  17. Mountjoy, K. G.Mortrud, M. T.Low, M. J.Simerly, R. B. & Cone, R. D. Localization of the melanocortin-4 receptor (MC4-R) in neuroendocrine and autonomic control circuits in the brainMol. Endocrinol. 812981308 (1994)
  18. Balthasar, N. et al. Divergence of melanocortin pathways in the control of food intake and energy expenditureCell 123493505 (2005)
  19. Leibowitz, S. F. & Hor, L. Endorphinergic and alpha-noradrenergic systems in the paraventricular nucleus: effects on eating behaviorPeptides 3421428 (1982)
  20. Leibowitz, S. F. Brain neurotransmitters and appetite regulationPsychopharmacol. Bull. 21412418 (1985)
  21. Morozov, Y. M. et al. Antibodies to cannabinoid type 1 receptor co-react with stomatin-like protein 2 in mouse brain mitochondriaEur. J. Neurosci. 3823412348 (2013)
  22. Morozov, Y. M.Horvath, T. L. & Rakic, P. A tale of two methods: Identifying neuronal CB1 receptorsMol. Metabol. 3338 (2014)
  23. Bénard, G. et al. Mitochondrial CB(1) receptors regulate neuronal energy metabolismNature Neurosci. 15558564 (2012)
  24. Hebert-Chatelain, E. et al. Studying mitochondrial CB1 receptors: Yes we canMolecular Metabol. 3339 (2014)
  25. Hebert-Chatelain, E. et al. Cannabinoid control of brain bioenergetics: Exploring the subcellular localization of the CB1 receptorMol. Metabol. 3495504 (2014)
  26. Schneeberger, M. et al. Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalanceCell 155172187 (2013)
  27. Nasrallah, C. M. & Horvath, T. L. Mitochondrial dynamics in the central regulation of metabolismNature Rev. Endocrinol. 10650658 (2014)
  28. Diano, S. et al. Peroxisome proliferation-associated control of reactive oxygen species sets melanocortin tone and feeding in diet-induced obesityNature Med. 1711211127 (2011)
  29. Negre-Salvayre, A. et al. A role for uncoupling protein-2 as a regulator of mitochondrial hydrogen peroxide generationFASEB 11809815 (1997)
  30. Echtay, K. S. et al. Superoxide activates mitochondrial uncoupling proteinsNature 4159699(2002)
  31. Andrews, Z. B. et al. UCP2 mediates ghrelin’s action on NPY/AgRP neurons by lowering free radicalsNature 454846851 (2008)
  32. Diano, S. & Horvath, T. L. Mitochondrial uncoupling protein 2 (UCP2) in glucose and lipid metabolismTrends Mol. Med. 185258 (2012)
  33. Huszar, D. et al. Targeted disruption of the melanocortin-4 receptor results in obesity in miceCell 88131141 (1997)
  34. Fan, W.Boston, B. A.Kesterson, R. A.Hruby, V. J. & Cone, R. D. Role of melanocortinergic neurons in feeding and the agouti obesity syndromeNature 385165168 (1997)
  35. Cone, R. D. Anatomy and regulation of the central melanocortin systemNature Neurosci. 8571578 (2005)
  36. Elias, C. F. et al. Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic areaNeuron 23775786 (1999)
  37. Cowley, M. A. et al. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleusNature 411480484 (2001)
  38. Horvath, T. L.Naftolin, F.Kalra, S. P. & Leranth, C. Neuropeptide-Y innervation of beta-endorphin-containing cells in the rat mediobasal hypothalamus: a light and electron microscopic double immunostaining analysisEndocrinology 13124612467 (1992)
  39. Soria-Gómez, E. et al. The endocannabinoid system controls food intake via olfactory processesNature Neurosci. 17407415 (2014)
  40. Greenberg, I.Kuehnle, J.Mendelson, J. H. & Bernstein, J. G. Effects of marihuana use on body weight and caloric intake in humansPsychopharmacology 497984 (1976)
  41. Foltin, R. W.Brady, J. V. & Fischman, M. W. Behavioral analysis of marijuana effects on food intake in humansPharmacol. Biochem. Behav. 25577582 (1986)
  42. Coppola, A. et al. A central thermogenic-like mechanism in feeding regulation: an interplay between arcuate nucleus T3 and UCP2Cell Metab. 52133 (2007)
  43. Parton, L. E. et al. Glucose sensing by POMC neurons regulates glucose homeostasis and is impaired in obesityNature 449228232 (2007)
  44. Horvath, T. L. et al. Brain uncoupling protein 2: uncoupled neuronal mitochondria predict thermal synapses in homeostatic centersJ. Neurosci. 191041710427 (1999)
  45. Andrews, Z. B. et al. Ghrelin promotes and protects nigrostriatal dopamine function via a UCP2-dependent mitochondrial mechanismJ. Neurosci. 291405714065 (2009)
  46. Landry, M.Vila-Porcile, E.Hokfelt, T. & Calas, A. Differential routing of coexisting neuropeptides in vasopressin neuronsEur. J. Neurosci. 17579589 (2003)
  47. van den Pol, A. N. Neuropeptide transmission in brain circuitsNeuron 7698115 (2012)
  48. Pinto, S. et al. Rapid rewiring of arcuate nucleus feeding circuits by leptinScience 304110115 (2004)
  49. Zimmer, A.Zimmer, A. M.Hohmann, A. G.Herkenham, M. & Bonner, T. I. Increased mortality, hypoactivity, and hypoalgesia in cannabinoid CB1 receptor knockout miceProc. Natl Acad. Sci. USA 9657805785 (1999)
  50. Zhang, C. Y. et al. Uncoupling protein-2 negatively regulates insulin secretion and is a major link between obesity, beta cell dysfunction, and type 2 diabetesCell 105745755 (2001)
  51. Dietrich, M. O.Liu, Z. W. & Horvath, T. L. Mitochondrial dynamics controlled by mitofusins regulate Agrp neuronal activity and diet-induced obesityCell 155188199 (2013)
  52. Dietrich, M. O. et al. AgRP neurons regulate development of dopamine neuronal plasticity and nonfood-associated behaviorsNature Neurosci. 1511081110 (2012)
  53. Cetin, A.Komai, S.Eliava, M.Seeburg, P. H. & Osten, P. Stereotaxic gene delivery in the rodent brainNature Protocols 131663173 (2006)
  54. Moreira, P. I. et al. Mitochondria from distinct tissues are differently affected by 17β-estradiol and tamoxifenJ. Steroid Biochem. Mol. Biol. 123816 (2011)

Download references

Author information

  1. Present address: Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 406-772, South Korea.

    • Jae Geun Kim

Affiliations

  1. Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06520, USA

    • Marco Koch,
    • Luis Varela,
    • Jae Geun Kim,
    • Jung Dae Kim,
    • Francisco Hernández-Nuño,
    • Klara Szigeti-Buck,
    • Marcelo O. Dietrich,
    • Xiao-Bing Gao,
    • Sabrina Diano &
    • Tamas L. Horvath
  2. Institute of Anatomy, University of Leipzig, 04103 Leipzig, Germany

    • Marco Koch &
    • Ingo Bechmann
  3. Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520, USA

    • Jung Dae Kim,
    • Sabrina Diano &
    • Tamas L. Horvath
  4. Obesity & Diabetes Institute, Department of Physiology, Monash University, Clayton, Victoria 3800, Australia

    • Stephanie E. Simonds &
    • Michael A. Cowley
  5. Division of Endocrinology & Metabolism, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA

    • Carlos M. Castorena,
    • Claudia R. Vianna &
    • Joel K. Elmquist
  6. Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA

    • Yury M. Morozov,
    • Pasko Rakic,
    • Marcelo O. Dietrich,
    • Sabrina Diano &
    • Tamas L. Horvath
  7. Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520, USA

    • Pasko Rakic &
    • Tamas L. Horvath

Contributions

M.K., S.D. and T.L.H. developed the conceptual framework of this study. M.K., M.O.D., X.-B.G., S.D. and T.L.H. interpreted results. M.K. performed experiments and analysed results. Experimental contributions: L.V. contributed to Figs 4h–j, 5d and Extended Data Figs 1b, 5e and 6a, b; J.G.K. contributed to Figs 2e, f, 3i, 5a, b and Extended Data Fig. 2g; J.D.K. contributed to Figs 3b–d, 5e–g and Extended Data Figs 5c and 6c; F.H. contributed to Figs 4a, 5c and Extended Data Fig. 5a, b, d; S.E.S. contributed to Fig. 3a; C.M.C., C.R.V. and J.K.E. provided key animal models; Y.M.M. and P.R. contributed to Fig. 3b and Extended Data Fig. 1c; P.R., I.B. and M.A.C. provided materials, animals and equipment; K.S.-B. contributed to Figs 3f and 4d–g; X.-B.G. contributed to Figs 1C, Da–c and 3j. M.K. and T.L.H. wrote the paper.

Competing financial interests

The authors declare no competing financial interests.

Corresponding author

Correspondence to:

Extended data figures and tables

Extended Data Figures

  1. Extended Data Figure 1: Characterization of CB1R-dependent food intake. (449 KB)
    a, Bimodal effects of different ACEA doses on food intake in fed mice (vehicle, n = 23 mice, 100 ± 16.3%; ACEA (in mg kg−1 body weight, intraperitoneal): 0.1, n = 8, 104.5 ± 46.6%; 0.5, n = 3, 190.8 ± 40.4%; 1.0, n = 19, 196.7 ± 30%; 2.5, n = 16, 87.1 ± 18%; 5.0, n = 11, 59.2 ± 15.5%; P < 0.01 versus vehicle, one-way ANOVA, followed by Dunnett’s multiple comparisons test; six independent experiments with litters from different parents). b, Neutral dose of ACEA on feeding (5 mg kg−1 body weight, intraperitoneal) did not alter locomotor activity of fed mice (n = 3 mice/group; P > 0.05). c, Impaired feeding response to ACEA (1 mg kg−1 body weight, intraperitoneal) in CB1R-heterozygote mice (Cnr1+/−n = 6 mice, 1 h: 0.04 ± 0.01 g, 2 h: 0.07 ± 0.01 g) and CB1R-deficient mice (Cnr1−/−, 1 h: n = 6, 0.02 ± 0.01 g, 2 h: n = 4, 0.03 ± 0.01 g) mice, when compared to CB1R wild-type mice (Cnr1+/+, 1 h: n = 12, 0.13 ± 0.01 g, 2 h: n = 4, 0.18 ± 0.04 g; P < 0.01, P < 0.001 versus wild-type; two independent experiments). d, Central, local ACEA injection into the ARC induced food intake (vehicle, n = 4 mice, 1 h: 0.05 ± 0.03 g, 2 h: 0.12 ± 0.01 g; ACEA, n = 4, 1 h: 0.25 ± 0.03 g; 2 h: 0.43 ± 0.05 g; P < 0.01, P < 0.001). e, Verification of correct ARC cannula placement by HOECHST (blue) injection (representative image (two different magnifications) of four independent experiments). f, Hyperphagic CB1R activation (1 mg kg−1 body weight ACEA, intraperitoneal) was abolished by central, local ARC RIMO-mediated CB1R blockade (vehicle plus vehicle, n = 8 mice, 0.05 ± 0.01 g; vehicle plus ACEA, n = 8, 0.15 ± 0.02 g; RIMO plus vehicle, n = 8, 0.09 ± 0.02 g; RIMO plus ACEA, n = 8, 0.09 ± 0.02 g; P < 0.05, #P < 0.05 for interaction between RIMO and ACEA, two-way ANOVA, followed by Šidák’s multiple comparisons test; two independent experiments). g, Hyperphagic CB1R activation (1 mg kg−1 body weight WIN, intraperitoneal) was reduced by local ARC RIMO-mediated CB1R blockade (vehicle plus WIN, n = 8 mice, 0.21 ± 0.03 g; RIMO+WIN, n = 8, 0.1 ± 0.02 g; P < 0.01). h, RIMO-induced hypophagic blockade of CB1R in fasted mice (vehicle, n = 10 mice, 1 h: 0.76 ± 0.07 g, 2 h: 1.18 ± 0.07 g; RIMO, n = 11 mice, 1 h: 0.42 ± 0.05 g, 2 h: 0.75 ± 0.08 g; P < 0.01, P < 0.001; two independent experiments). Values (biological replicates) denote mean ± s.e.m. If not otherwise stated, Pvalues (unpaired comparisons) by two-tailed Student’s t-test. Scale bars, 25 μm.
  2. Extended Data Figure 2: DREADD-mediated regulation of POMC neurons. (622 KB)
    a, Selective DREADD expression specified by local ARC mCherry fluorescence. b, POMC neurons (green) contain mCherry-labelled DREADD (red, arrowheads). c, CNO-activated inhibitory DREADD reduced ARC cFOS immunolabelled neurons in fed mice (arrowheads). Representative images of four independent experiments (ac). de, CNO-activated inhibitory DREADD blocked ACEA-induced POMC activation (cFOS; vehicle plus ACEA, n= 6 mice, 60.4 ± 3.6%; CNO plus ACEA, n = 5, 32.3 ± 2.5%; P < 0.001). f, CNO-activated POMC-specific inhibitory DREADD did not acutely affect feeding but enhanced it after 8 h (vehicle, n = 17 mice, 0.42 ± 0.04 g; CNO, n = 16, 0.58 ± 0.04 g; 24 h after injection: vehicle, n = 5 mice, 2.57 ± 0.07 g; CNO, n = 5, 3.37 ± 0.18 g; P < 0.01 versus vehicle; three independent experiments). g, CNO-activated POMC-specific stimulating DREADD did not acutely affect feeding but reduced it after 8 h (vehicle, n = 6 mice, 0.58 ± 0.05 g; CNO, n = 6, 0.34 ± 0.05 g; P < 0.01 versus vehicle; 24 h after injection: vehicle, 3.96 ± 0.15 g; CNO, 3.65 ± 0.21 g; P > 0.05 versus vehicle). Values (biological replicates) denote mean ± s.e.m. If not otherwise stated, P values (unpaired comparisons) by two-tailed Student’s t-test. Scale bars, 100 μm (a), 25 μm (b) and 50 μm (cd).
  3. Extended Data Figure 3: Hyperphagic CB1R activation selectively increased PVN β-endorphin. (735 KB)
    adi, PVN α-MSH remained unchanged after hyperphagic CB1R activation (PVN unilateral analysis; vehicle, n = 6 values (technical replicates)/6 sections/3 mice (biological replicates); 60 min ACEA, n = 10/10/5; 90 min ACEA, n = 6/6/3; values, see Extended Data Table 1a). ehj, In contrast, hyperphagic ACEA increased PVN β-endorphin 60 and 90 min after application (PVN unilateral analysis; vehicle, n = 13 values/13 sections/6 mice; 60 min ACEA, n = 4/4/4; 90 min ACEA, n = 14/14/7; values, see Extended Data Table 1bP < 0.001, P < 0.05 versus vehicle, one-way ANOVA, followed by Dunnett’s multiple comparisons test, two independent experiments using litters from different parents). Error bars indicate mean ± s.e.m. Scale bars, 25 μm.
  4. Extended Data Figure 4: Bimodal character of ARC CB1R-driven β-endorphin increase. (239 KB)
    a, Compared to vehicle (bilateral PVN analysis; n = 22 values (technical replicates)/11 sections/4 mice (biological replicates), hyperphagic doses (1 mg kg−1 body weight, respectively) of WIN (n = 24/12/4) or ACEA (n = 18/9/3) induced PVN β-endorphin immunoreactivity. Neutral dose (5 mg kg−1 BW) of ACEA (n = 18/9/3) on feeding showed no effects (see Extended Data Table 2 for all values). P < 0.05, P < 0.01, P < 0.001 versus vehicle, one-way ANOVA, followed by Dunnett’s multiple comparisons test. b, Representative binary images of four independent experiments showing β-endorphin immunoreactivity after thresholding (image segmentation) using ImageJ software (see Methods). c, Compared to vehicle (unilateral PVN analysis; n = 4 mice (biological replicates), 2–3 sections (technical replicates) per mouse), central, hyperphagic local ARC injection of ACEA (n = 5 mice, 3 sections per mouse) increased PVN β-endorphin immunoreactivity (see Extended Data Table 3 for all values; P < 0.05, P < 0.01). Error bars indicate mean ± s.e.m. If not otherwise stated, P values (unpaired comparisons) by two-tailed Student’s t-test. Scale bars, 100 μm.
  5. Extended Data Figure 5: Post-transcriptional regulation of hypothalamic pro-protein convertases, normal Cnr1 expression in Ucp2−/− mice and presence of CB1R in POMC neurons. (289 KB)
    ab, ACEA did not affect transcripts of pro-protein convertases 1 (Pcsk1) and 2 (Pcsk2) (in fold change; Pcsk1: vehicle, n = 11 mice, 1.00 ± 0.07; ACEA, n = 10 mice, 1.17 ± 0.09; Pcsk2: vehicle, n = 11 mice, 1.00 ± 0.13; ACEA, n = 11 mice, 1.14 ± 0.19; P > 0.05; two independent experiments). c, Representative western blot membranes for PC-1 (~80 kilodaltons (kDa)) and PC-2 (~72 kDa) immunolabelling. d, Equal Cnr1 expression in wild-type and Ucp2−/− mice (in fold change: all groups n = 6 mice; wild type, 1.00 ± 0.1; Ucp2−/−, 0.98 ± 0.12; P > 0.05). e, We have previously shown that antibodies raised against CB1R also recognized the mitochondrial protein, stomatin-like protein 2 (ref. 21). In line with this, mitochondrial labelling of CB1R was found substantially diminished but not completely eliminated in CB1R-KO (Cnr1−/−) mice232425. We observed that in contrast to wild-type animals (Cnr1+/+ mice), which showed ~80% (77 out of 97, 79.5 ± 3.9%) of POMC neurons (red fluorescence) to contain labelling with the CB1R antisera (green fluorescence), in CB1R knockout (KO; Cnr1−/−) mice, less than 30% (37 out of 128, 29.2 ± 3.3%) of POMC neurons retained immunolabelling. Thus, we concluded that a large population of POMC neurons contains CB1R (P < 0.001). All values (biological replicates: acd; biological replicates including technical replicates: e) denote mean ± s.e.m. If not otherwise stated, P values (unpaired comparisons) by two-tailed Student’s t-test. Scale bar, 25 μm.
  6. Extended Data Figure 6: Bimodal CB1R-dependent regulation of mitochondrial respiration and UCP2-dependent control of POMC. (511 KB)
    ab, Bimodal CB1R-controlled mitochondrial respiration in hippocampus. a, Hyperphagic (1 mg kg−1 body weight ACEA, intraperitoneal) CB1R activation increased ex vivomitochondrial respiration (in nmol O2 min−1 mg−1 protein; state 3: vehicle, n = 6 mice, 170.7 ± 12; ACEA, n = 8, 252.7 ± 17.2; state 4: vehicle, 92.7 ± 5.4; ACEA, 139.7 ± 6; P < 0.01, P < 0.001). b, Neutral dose of ACEA on feeding (5 mg kg−1 body weight, intraperitoneal) reduced mitochondrial respiration (state 3: vehicle, n = 7 mice, 178.2 ± 12.2; ACEA, n = 5, 118.9 ± 9.4; state 4: vehicle, 100 ± 5.1; ACEA, 64.3 ± 6.3; two independent experiments). c, Representative western blot membranes for POMC (pro-POMC, ~31 kDa; POMC, ~27 kDa). d, The 24-h food intake did not differ between wild-type (n = 28 mice, 100 ± 3.2%) and Ucp2−/− (n = 29, 98.9 ± 4.7%; P > 0.05) mice after ACEA (1 mg kg−1 body weight, intraperitoneal) treatment (six independent experiments using litters from different parents). All values (biological replicates) denote ± s.e.m. If not otherwise stated, P values (unpaired comparisons) by two-tailed Student’s t-test.

Extended Data Tables

  1. Extended Data Table 1: Semi-quantitative measurements of α-MSH and β-endorphin immunoreactivity (144 KB)
  2. Extended Data Table 2: Semi-quantitative measurements of β-endorphin immunoreactivity (111 KB)
  3. Extended Data Table 3: Semi-quantitative measurements of β-endorphin immunoreactivity (58 KB)
  4. Extended Data Table 4: Semi-quantitative measurements of β-endorphin immunoreactivity (101 KB)

Supplementary information

PDF files

  1. Supplementary Table (103 KB)
    This file contains Supplementary Table 1.twin memes II