DOI QR코드

DOI QR Code

Metformin ameliorates olanzapine-induced disturbances in POMC neuron number, axonal projection, and hypothalamic leptin resistance

  • Kim, Jaedeok (Department of Biomedical Science, Hallym University) ;
  • Lee, Nayoung (Department of Biomedical Science, Hallym University) ;
  • Suh, Sang Bum (University of Ulsan College of Medicine) ;
  • Jang, Sooyeon (Department of Biomedical Science, Hallym University) ;
  • Kim, Saeha (Department of Biomedical Science, Hallym University) ;
  • Kim, Dong-Gyu (Department of Biomedical Science, Hallym University) ;
  • Park, Jong Kook (Department of Biomedical Science, Hallym University) ;
  • Lee, Keun-Wook (Department of Biomedical Science, Hallym University) ;
  • Choi, Soo Young (Department of Biomedical Science, Hallym University) ;
  • Lee, Chan Hee (Department of Biomedical Science, Hallym University)
  • Received : 2022.02.15
  • Accepted : 2022.04.22
  • Published : 2022.06.30

Abstract

Antipsychotics have been widely accepted as a treatment of choice for psychiatric illnesses such as schizophrenia. While atypical antipsychotics such as aripiprazole are not associated with obesity and diabetes, olanzapine is still widely used based on the anticipation that it is more effective in treating severe schizophrenia than aripiprazole, despite its metabolic side effects. To address metabolic problems, metformin is widely prescribed. Hypothalamic proopiomelanocortin (POMC) neurons have been identified as the main regulator of metabolism and energy expenditure. Although the relation between POMC neurons and metabolic disorders is well established, little is known about the effects of olanzapine and metformin on hypothalamic POMC neurons. In the present study, we investigated the effect of olanzapine and metformin on the hypothalamic POMC neurons in female mice. Olanzapine administration for 5 days significantly decreased Pomc mRNA expression, POMC neuron numbers, POMC projections, and induced leptin resistance before the onset of obesity. It was also observed that coadministration of metformin with olanzapine not only increased POMC neuron numbers and projections but also improved the leptin response of POMC neurons in the olanzapine-treated female mice. These findings suggest that olanzapine-induced hypothalamic POMC neuron abnormality and leptin resistance, which can be ameliorated by metformin administration, are the possible causes of subsequent hyperphagia.

Keywords

Acknowledgement

This research was supported by Hallym University Research Fund, 202110-005 (HRF-202110-005).

References

  1. Palmer BA, Pankratz VS and Bostwick JM (2005) The lifetime risk of suicide in schizophrenia: a reexamination. Arch Gen Psychiatry 62, 247-253 https://doi.org/10.1001/archpsyc.62.3.247
  2. Meltzer HY (2013) Update on typical and atypical antipsychotic drugs. Annu Rev Med 64, 393-406 https://doi.org/10.1146/annurev-med-050911-161504
  3. Bever KA and Perry PJ (1998) Olanzapine: a serotonin-dopamine-receptor antagonist for antipsychotic therapy. Am J Health Syst Pharm 55, 1003-1016 https://doi.org/10.1093/ajhp/55.10.1003
  4. Bak M, Fransen A, Janssen J, van Os J and Drukker M (2014) Almost all antipsychotics result in weight gain: a meta-analysis. PLoS One 9, e94112 https://doi.org/10.1371/journal.pone.0094112
  5. Pillinger T, McCutcheon RA, Vano L et al (2020) Comparative effects of 18 antipsychotics on metabolic function in patients with schizophrenia, predictors of metabolic dysregulation, and association with psychopathology: a systematic review and network meta-analysis. Lancet Psychiatry 7, 64-77 https://doi.org/10.1016/s2215-0366(19)30416-x
  6. Huang J, Hei GR, Yang Y et al (2020) Increased appetite plays a key role in olanzapine-induced weight gain in first-episode schizophrenia patients. Front Pharmacol 11, 739 https://doi.org/10.3389/fphar.2020.00739
  7. Schwartz MW, Woods SC, Porte D Jr, Seeley RJ and Baskin DG (2000) Central nervous system control of food intake. Nature 404, 661-671 https://doi.org/10.1038/35007534
  8. Cone RD (2005) Anatomy and regulation of the central melanocortin system. Nat Neurosci 8, 571-578 https://doi.org/10.1038/nn1455
  9. Williams DL and Schwartz MW (2005) The melanocortin system as a central integrator of direct and indirect controls of food intake. Am J Physiol Regul Integr Comp Physiol 289, R2-3 https://doi.org/10.1152/ajpregu.00226.2005
  10. Quarta C, Claret M, Zeltser LM et al (2021) POMC neuronal heterogeneity in energy balance and beyond: an integrated view. Nat Metab 3, 299-308 https://doi.org/10.1038/s42255-021-00345-3
  11. Timper K and Bruning JC (2017) Hypothalamic circuits regulating appetite and energy homeostasis: pathways to obesity. Dis Model Mech 10, 679-689 https://doi.org/10.1242/dmm.026609
  12. Ferno J, Varela L, Skrede S et al (2011) Olanzapine-induced hyperphagia and weight gain associate with orexigenic hypothalamic neuropeptide signaling without concomitant AMPK phosphorylation. PLoS One 6, e20571 https://doi.org/10.1371/journal.pone.0020571
  13. Weston-Green K, Huang XF and Deng C (2012) Alterations to melanocortinergic, GABAergic and cannabinoid neurotransmission associated with olanzapine-induced weight gain. PLoS One 7, e33548 https://doi.org/10.1371/journal.pone.0033548
  14. Zhang Q, He M, Deng C, Wang H, Lian J and Huang XF (2014) Hypothalamic ghrelin signalling mediates olanzapine-induced hyperphagia and weight gain in female rats. Int J Neuropsychopharmacol 17, 807-818 https://doi.org/10.1017/S1461145713001697
  15. Kim K, Yang WH, Jung YS and Cha JH (2020) A new aspect of an old friend: the beneficial effect of metformin on anti-tumor immunity. BMB Rep 53, 512-520 https://doi.org/10.5483/BMBRep.2020.53.10.149
  16. Lee CH, Han JH, Kim S et al (2020) Metformin ameliorates bile duct ligation-induced acute hepatic injury via regulation of ER stress. BMB Rep 53, 311-316 https://doi.org/10.5483/BMBRep.2020.53.6.169
  17. Lee A and Morley JE (1998) Metformin decreases food consumption and induces weight loss in subjects with obesity with type II non-insulin-dependent diabetes. Obes Res 6, 47-53 https://doi.org/10.1002/j.1550-8528.1998.tb00314.x
  18. Inzucchi SE, Maggs DG, Spollett GR et al (1998) Efficacy and metabolic effects of metformin and troglitazone in type II diabetes mellitus. N Engl J Med 338, 867-872 https://doi.org/10.1056/NEJM199803263381303
  19. Baptista T, Martinez J, Lacruz A et al (2006) Metformin for prevention of weight gain and insulin resistance with olanzapine: a double-blind placebo-controlled trial. Can J Psychiatry 51, 192-196 https://doi.org/10.1177/070674370605100310
  20. Wu RR, Zhao JP, Jin H et al (2008) Lifestyle intervention and metformin for treatment of antipsychotic-induced weight gain: a randomized controlled trial. JAMA 299, 185-193 https://doi.org/10.1001/jama.2007.56-b
  21. Wu RR, Zhao JP, Guo XF et al (2008) Metformin addition attenuates olanzapine-induced weight gain in drug-naive first-episode schizophrenia patients: a double-blind, placebo-controlled study. Am J Psychiatry 165, 352-358 https://doi.org/10.1176/appi.ajp.2007.07010079
  22. Cui D, Peng Y, Zhang C et al (2018) Macrophage migration inhibitory factor mediates metabolic dysfunction induced by atypical antipsychotic therapy. J Clin Invest 128, 4997-5007 https://doi.org/10.1172/jci93090
  23. Lord CC, Wyler SC, Wan R et al (2017) The atypical antipsychotic olanzapine causes weight gain by targeting serotonin receptor 2C. J Clin Invest 127, 3402-3406 https://doi.org/10.1172/JCI93362
  24. German AJ (2006) The growing problem of obesity in dogs and cats. J Nutr 136, 1940s-1946s https://doi.org/10.1093/jn/136.7.1940S
  25. Guo C, Liu J and Li H (2021) Metformin ameliorates olanzapine-induced insulin resistance via suppressing macrophage infiltration and inflammatory responses in rats. Biomed Pharmacother 133, 110912 https://doi.org/10.1016/j.biopha.2020.110912
  26. Praharaj SK, Jana AK, Goyal N and Sinha VK (2011) Metformin for olanzapine-induced weight gain: a systematic review and meta-analysis. Br J Clin Pharmacol 71, 377-382 https://doi.org/10.1111/j.1365-2125.2010.03783.x
  27. Bouret SG, Draper SJ and Simerly RB (2004) Formation of projection pathways from the arcuate nucleus of the hypothalamus to hypothalamic regions implicated in the neural control of feeding behavior in mice. J Neurosci 24, 2797-2805 https://doi.org/10.1523/JNEUROSCI.5369-03.2004
  28. Balthasar N, Coppari R, McMinn J et al (2004) Leptin receptor signaling in POMC neurons is required for normal body weight homeostasis. Neuron 42, 983-991 https://doi.org/10.1016/j.neuron.2004.06.004
  29. Myers MG, Cowley MA and Munzberg H (2008) Mechanisms of leptin action and leptin resistance. Annu Rev Physiol 70, 537-556 https://doi.org/10.1146/annurev.physiol.70.113006.100707
  30. Berglund ED, Vianna CR, Donato J Jr et al (2012) Direct leptin action on POMC neurons regulates glucose homeostasis and hepatic insulin sensitivity in mice. J Clin Invest 122, 1000-1009 https://doi.org/10.1172/JCI59816
  31. Elmquist JK, Elias CF and Saper CB (1999) From lesions to leptin: hypothalamic control of food intake and body weight. Neuron 22, 221-232 https://doi.org/10.1016/S0896-6273(00)81084-3
  32. Myers MG Jr (2004) Leptin receptor signaling and the regulation of mammalian physiology. Recent Prog Horm Res 59, 287-304 https://doi.org/10.1210/rp.59.1.287
  33. Elias CF, Aschkenasi C, Lee C et al (1999) Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area. Neuron 23, 775-786 https://doi.org/10.1016/S0896-6273(01)80035-0
  34. Munzberg H, Jobst EE, Bates SH et al (2007) Appropriate inhibition of orexigenic hypothalamic arcuate nucleus neurons independently of leptin receptor/STAT3 signaling. J Neurosci 27, 69-74 https://doi.org/10.1523/JNEUROSCI.3168-06.2007
  35. Tam J, Szanda G, Drori A et al (2017) Peripheral cannabinoid-1 receptor blockade restores hypothalamic leptin signaling. Mol Metab 6, 1113-1125 https://doi.org/10.1016/j.molmet.2017.06.010
  36. Lee CH, Kim HJ, Lee YS et al (2018) Hypothalamic macrophage inducible nitric oxide synthase mediates obesity-associated hypothalamic inflammation. Cell Rep 25, 934-946.e935 https://doi.org/10.1016/j.celrep.2018.09.070
  37. Lee CH, Suk K, Yu R and Kim MS (2020) Cellular contributors to hypothalamic inflammation in obesity. Mol Cells 43, 431-437 https://doi.org/10.14348/molcells.2020.0055
  38. Li H, Peng S, Li S et al (2019) Chronic olanzapine administration causes metabolic syndrome through inflammatory cytokines in rodent models of insulin resistance. Sci Rep 9, 1582 https://doi.org/10.1038/s41598-018-36930-y
  39. He M, Qian K, Zhang Y et al (2021) Olanzapine-induced activation of hypothalamic astrocytes and toll-like receptor-4 signaling via endoplasmic reticulum stress were related to olanzapine-induced weight gain. Front Neurosci 14, 589650 https://doi.org/10.3389/fnins.2020.589650
  40. Pedroso JAB, Buonfiglio DC, Cardinali LI et al (2014) Inactivation of SOCS3 in leptin receptor-expressing cells protects mice from diet-induced insulin resistance but does not prevent obesity. Mol Metab 3, 608-618 https://doi.org/10.1016/j.molmet.2014.06.001
  41. Picardi PK, Calegari VC, de Oliveira Prada Pc et al (2008) Reduction of hypothalamic protein tyrosine phosphatase improves insulin and leptin resistance in diet-induced obese rats. Endocrinology 149, 3870-3880 https://doi.org/10.1210/en.2007-1506
  42. Giri S, Nath N, Smith B, Viollet B, Singh AK and Singh I (2004) 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside inhibits proinflammatory response in glial cells: a possible role of AMP-activated protein kinase. J Neurosci 24, 479-487 https://doi.org/10.1523/jneurosci.4288-03.2004
  43. Prasad R, Giri S, Nath N, Singh I and Singh AK (2006) 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside attenuates experimental autoimmune encephalomyelitis via modulation of endothelial-monocyte interaction. J Neurosci Res 84, 614-625 https://doi.org/10.1002/jnr.20953