641
Views
46
CrossRef citations to date
0
Altmetric
Review

Atypical antipsychotics-induced metabolic syndrome and nonalcoholic fatty liver disease: a critical review

&
Pages 2087-2099 | Published online: 22 Jul 2019

References

  • Turner T. Chlorpromazine: unlocking psychosis. BMJ. 2007;334(Suppl 1):S7. doi:10.1136/bmj.39034.609074.9417204765
  • Maric NP, Jovicic MJ, Mihaljevic M, Miljevic C. Improving current treatments for schizophrenia. Drug Dev Res. 2016;77:357–367. doi:10.1002/ddr.2133727633376
  • Warnez S, Alessi-Severini S. Clozapine: a review of clinical practice guidelines and prescribing trends. BMC Psychiatry. 2014;14:102. doi:10.1186/1471-244X-14-10224708834
  • McDonagh M, Peterson K, Carson S, Fu R, Thakurta S. Drug Class Review. A typical antipsychotic drugs: final update 3 report. Drug Class Rev. 2010.
  • Hermann RC, Yang D, Ettner SL, Marcus SC, Yoon C, Abraham M. Prescription of antipsychotic drugs by office-based physicians in the United States, 1989–1997. Psychiatr Serv. 2002;53:425–430. doi:10.1176/appi.ps.53.4.42511919355
  • Carbon M, Hsieh C-H, Kane JM, Correll CU. Tardive dyskinesia prevalence in the period of second-generation antipsychotic use: a meta-analysis. J Clin Psychiatry. 2017;78:e264–e278. doi:10.4088/JCP.16r1083228146614
  • Zhang JP, Gallego JA, Robinson DG, Malhotra AK, Kane JM, Correll CU. Efficacy and safety of individual second-generation vs. first-generation antipsychotics in first-episode psychosis: a systematic review and meta-analysis. Int J Neuropsychopharmacol. 2013;16:1205–1218. doi:10.1017/S146114571200127723199972
  • Mitchell AJ, Vancampfort D, Sweers K, van Winkel R, Yu W, De Hert M. Prevalence of metabolic syndrome and metabolic abnormalities in schizophrenia and related disorders–a systematic review and meta-analysis. Schizophr Bull. 2013;39:306–318. doi:10.1093/schbul/sbr14822207632
  • De Hert M, Yu W, Detraux J, Sweers K, van Winkel R, Correll CU. Body weight and metabolic adverse effects of asenapine, iloperidone, lurasidone and paliperidone in the treatment of schizophrenia and bipolar disorder: a systematic review and exploratory meta-analysis. CNS Drugs. 2012;26:733–759. doi:10.2165/11634500-000000000-0000022900950
  • Hasnain M, Fredrickson SK, Vieweg WV, Pandurangi AK. Metabolic syndrome associated with schizophrenia and atypical antipsychotics. Curr Diab Rep. 2010;10:209–216. doi:10.1007/s11892-010-0112-820425584
  • Newcomer JW. Second-generation (atypical) antipsychotics and metabolic effects: a comprehensive literature review. CNS Drugs. 2005;19(Suppl 1):1–93. doi:10.2165/00023210-200519001-00001
  • Newcomer JW. Antipsychotic medications: metabolic and cardiovascular risk. J Clin Psychiatry. 2007;68(Suppl 4):8–13.
  • Rummel-Kluge C, Komossa K, Schwarz S, et al. Head-to-head comparisons of metabolic side effects of second generation antipsychotics in the treatment of schizophrenia: a systematic review and meta-analysis. Schizophr Res. 2010;123:225–233. doi:10.1016/j.schres.2010.07.01220692814
  • Galassi A, Reynolds K, He J. Metabolic syndrome and risk of cardiovascular disease: a meta-analysis. Am J Med. 2006;119:812–819. doi:10.1016/j.amjmed.2006.02.03117000207
  • Gami AS, Witt BJ, Howard DE, et al. Metabolic syndrome and risk of incident cardiovascular events and death: a systematic review and meta-analysis of longitudinal studies. J Am Coll Cardiol. 2007;49:403–414. doi:10.1016/j.jacc.2006.09.03217258085
  • Mottillo S, Filion KB, Genest J, et al. The metabolic syndrome and cardiovascular risk a systematic review and meta-analysis. J Am Coll Cardiol. 2010;56:1113–1132. doi:10.1016/j.jacc.2010.05.03420863953
  • Eckel RH, Alberti KG, Grundy SM, Zimmet PZ. The metabolic syndrome. Lancet. 2010;375:181–183. doi:10.1016/S0140-6736(09)61794-320109902
  • Massart J, Begriche K, Moreau C, Fromenty B. Role of nonalcoholic fatty liver disease as risk factor for drug-induced hepatotoxicity. J Clin Transl Res. 2017;3:212–232. doi:10.18053/jctres.03.2017S1.00628691103
  • Tarantino G, Conca P, Basile V, et al. A prospective study of acute drug-induced liver injury in patients suffering from non-alcoholic fatty liver disease. Hepatol Res. 2007;37:410–415. doi:10.1111/j.1872-034X.2007.00072.x17539815
  • Slim M, Medina-Caliz I, Gonzalez-Jimenez A, et al. Hepatic safety of atypical antipsychotics: current evidence and future directions. Drug Saf. 2016;39:925–943. doi:10.1007/s40264-016-0436-727449495
  • Said MA, Sulaiman AH, Habil MH, et al. Metabolic syndrome and cardiovascular risk among patients with schizophrenia receiving antipsychotics in Malaysia. Singapore Med J. 2012;53:801–807.23268153
  • Bou KR. Atypical antipsychotic drugs, schizophrenia, and metabolic syndrome in non-Euro- American societies. Clin Neuropharmacol. 2012;35:141–147. doi:10.1097/WNF.0b013e31824d528822592510
  • McEvoy JP, Meyer JM, Goff DC, et al. Prevalence of the metabolic syndrome in patients with schizophrenia: baseline results from the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) schizophrenia trial and comparison with national estimates from NHANES III. Schizophr Res. 2005;80:19–32. doi:10.1016/j.schres.2005.07.01416137860
  • Vanuzzo D, Lo NC, Pilotto L, et al. Cardiovascular epidemiologic observatory 2008–2011: preliminary results. G Ital Cardiol (Rome). 2010;11:25S–30S.20879475
  • Zhang Y, Chen M, Chen J, et al. Metabolic syndrome in patients taking clozapine: prevalence and influence of catechol-O-methyltransferase genotype. Psycho- Pharmacology (berl). 2014;231:2211–2218. doi:10.1007/s00213-013-3410-4
  • Baptista T. Body weight gain induced by antipsychotic drugs: mechanisms and management. Acta Psychiatr Scand. 1999;100:3–16.10442434
  • Leadbetter R, Shutty M, Pavalonis D, Vieweg V, Higgins P, Downs M. Clozapine-induced weight gain: prevalence and clinical relevance. Am J Psychiatry. 1992;149:68–72. doi:10.1176/ajp.149.1.681728188
  • Kumra S, Herion D, Jacobsen LK, Briguglia C, Grothe D. Case study: risperidone-induced hepatotoxicity in pediatric patients. J Am Acad Child Adolesc Psychiatry. 1997;36:701–705. doi:10.1097/00004583-199705000-000229136506
  • Kemner C, Willemsen-Swinkels SH, de Jonge M, Tuynman-Qua H, van Engeland H. Open-label study of olanzapine in children with pervasive developmental disorder. J Clin Psychopharmacol. 2002;22:455–460.12352267
  • Gagliano A, Germanò E, Pustorino G, et al. Risperidone treatment of children with autistic disorder: effectiveness, tolerability, and pharmacokinetic implications. J Child Adolesc Psychopharmacol. 2004;14:39–47. doi:10.1089/10445460477384047215142390
  • Madhusoodanan S, Suresh P, Brenner R, Pillai R. Experience with the atypical antipsychotics risperidone and olanzapine in the elderly. Ann Clin Psychiatry. 1999;11:113–118.10482120
  • Street JS, Clark WS, Gannon KS, et al. Olanzapine treatment of psychotic and behavioral symptoms in patients with Alzheimer disease in nursing care facilities: a double-blind, randomized, placebo-controlled trial. The HGEU study group. Arch Gen Psychiatry. 2000;57:968–976.11015815
  • Barahal HS, Freeman N. Sudden graying of hair, alopecia, and diabetes mellitus of psychogenic origin. Psychiatr Q. 1946;20:31–38.21018012
  • Gury C. Schizophrenia, diabetes mellitus and antipsychotics. Encephale. 2004;30:382–391.15538314
  • Haupt DW, Newcomer JW. Hyperglycemia and antipsychotic medications. J Clin Psychiatry. 2001;62(Suppl 27):15–26; discussion 40–41.
  • Colli A, Cocciolo M, Francobandiera F, Rogantin F, Cattalini N. Diabetic ketoacidosis associated with clozapine treatment. Diabetes Care. 1999;22:176–177. doi:10.2337/diacare.22.1.176a
  • Newcomer JW, Haupt DW, Fucetola R, et al. Abnormalities in glucose regulation during antipsychotic treatment of schizophrenia. Arch Gen Psychiatry. 2002;59:337–345.11926934
  • Wetterling T, Schneider B, Weber B. Dyslipidemia and schizophrenia. Psychiatr Prax. 2007;34:223–229. doi:10.1055/s-2006-94005617160750
  • Galling B, Roldán A, Nielsen RE, et al. Type 2 diabetes mellitus in youth exposed to antipsychotics: a systematic review and meta-analysis. JAMA Psychiatry. 2016;73:247–259. doi:10.1001/jamapsychiatry.2015.292326792761
  • Atmaca M, Kuloglu M, Tezcan E, Ustundag B. Serum leptin and triglyceride levels in patients on treatment with atypical antipsychotics. J Clin Psychiatry. 2003;64:598–604.12755665
  • Meyer JM, Koro CE. The effects of antipsychotic therapy on serum lipids: a comprehensive review. Schizophr Res. 2004;70:1–17. doi:10.1016/j.schres.2004.01.01415246458
  • Wirshing DA, Boyd JA, Meng LR, Ballon JS, Marder SR, Wirshing WC. The effects of novel antipsychotics on glucose and lipid levels. J Clin Psychiatry. 2002;63:856–865.12416594
  • Meyer JM. A retrospective comparison of weight, lipid, and glucose changes between risperidone- and olanzapine-treated inpatients: metabolic outcomes after 1 year. J Clin Psychiatry. 2002;63:425–433.12019668
  • Olfson M, Marcus SC, Corey-Lisle P, Tuomari AV, Hines P, L’Italien GJ. Hyperlipidemia following treatment with antipsychotic medications. Am J Psychiatry. 2006;163:1821–1825. doi:10.1176/ajp.2006.163.10.182117012695
  • Weiden PJ, Daniel DG, Simpson G, Romano SJ. Improvement in indices of health status in outpatients with schizophrenia switched to ziprasidone. J Clin Psychopharmacol. 2003;23:595–600. doi:10.1097/01.jcp.0000095347.32154.0814624190
  • Doucette DE, Grenier JP, Robertson PS. Olanzapine-induced acute pancreatitis. Ann Pharmacother. 2000;34:1128–1131. doi:10.1345/aph.1939011054978
  • Gupta A, Dadheech G, Yadav D, Sharma P, Gautam S. Metabolic issues in schizophrenic patients receiving antipsychotic treatment. Indian J Clin Biochem. 2014;29:196–201. doi:10.1007/s12291-013-0415-z24757302
  • Kerr TA, Jonnalagadda S, Prakash C, Azar R. Pancreatitis following olanzapine therapy: a report of three cases. Case Rep Gastroenterol. 2007;1:15–20. doi:10.1159/00010422221487466
  • Rizos E, Tournikioti K, Alevyzakis E, et al. Acute necrotizing pancreatitis following olanzapine treatment and 759C/T polymorphism of HTR2C gene: a case report. In Vivo. 2015;29:529–531.26359410
  • Rossor AM, Leech N, Neely RD. Olanzapine-induced chylomicronemia presenting as acute pancreatitis. J Clin Psychopharmacol. 2007;27:395–396. doi:10.1097/01.jcp.0000264988.55603.bb17632226
  • Targher G, Bertolini L, Padovani R, et al. Prevalence of non-alcoholic fatty liver disease and its association with cardiovascular disease in patients with type 1 diabetes. J Hepatol. 2010;53:713–718. doi:10.1016/j.jhep.2010.04.03020619918
  • Carney CP, Jones L, Woolson RF. Medical comorbidity in women and men with schizophrenia: a population-based controlled study. J Gen Intern Med. 2006;21:1133–1137. doi:10.1111/j.1525-1497.2006.00563.x17026726
  • Fuller BE, Rodriguez VL, Linke A, Sikirica M, Dirani R, Hauser P. Prevalence of liver disease in veterans with bipolar disorder or schizophrenia. Gen Hosp Psychiatry. 2011;33:232–237. doi:10.1016/j.genhosppsych.2011.03.00621601719
  • Sokal J, Messias E, Dickerson FB, et al. Comorbidity of medical illnesses among adults with serious mental illness who are receiving community psychiatric services. J Nerv Ment Dis. 2004;192:421–427.15167405
  • Hsu JH, Chien IC, Lin CH, Chou YJ, Chou P. Increased risk of chronic liver disease in patients with schizophrenia: a population-based cohort study. Psychosomatics. 2014;55:163–171. doi:10.1016/j.psym.2013.06.00123932529
  • Morlán-Coarasa MJ, Arias-Loste MT, de la Foz VO, et al. Incidence of non-alcoholic fatty liver disease and metabolic dysfunction in first episode schizophrenia and related psychotic disorders: a 3-year prospective randomized interventional study. Psychopharmacology (Berl). 2016;233:3947–3952. doi:10.1007/s00213-016-4422-727620899
  • Liu H, Lu HY. Nonalcoholic fatty liver disease and cardiovascular disease. World J Gastroenterol. 2014;20:8407–8415. doi:10.3748/wjg.v20.i26.840725024598
  • Moore N, Hall G, Sturkenboom M, Mann R, Lagnaoui R, Begaud B. Biases affecting the proportional reporting ratio (PPR) in spontaneous reports pharmacovigilance databases: the example of sertindole. Pharmacoepidemiol Drug Saf. 2003;12:271–281. doi:10.1002/pds.84812812006
  • Hale AS. A review of the safety and tolerability of sertindole. Int Clin Psychopharmacol. 1998;13(Suppl 3):S65–S70.9690973
  • Haddad PM, Anderson IM. Antipsychotic-related QTc prolongation, torsade de pointes and sudden death. Drugs. 2002;62:1649–1671. doi:10.2165/00003495-200262110-0000612109926
  • Azorin JM, Strub N, Loft H. A double-blind, controlled study of sertindole versus risperidone in the treatment of moderate-to-severe schizophrenia. Int Clin Psychopharmacol. 2006;21:49–56.16317317
  • Daniel DG, Wozniak P, Mack RJ, McCarthy BG. Long-term efficacy and safety comparison of sertindole and haloperidol in the treatment of schizophrenia. The Sertindole Study Group. Psychopharmacol Bull. 1998;34:61–69.9564200
  • Zimbroff DL, Kane JM, Tamminga CA, et al. Controlled, dose-response study of sertindole and haloperidol in the treatment of schizophrenia. Sertindole study group. Am J Psychiatry. 1997;154:782–791. doi:10.1176/ajp.154.6.7829167505
  • Gallhofer B, Jaanson P, Mittoux A, Tanghøj P, Lis S, Krieger S. Course of recovery of cognitive impairment in patients with schizophrenia: a randomised double-blind study comparing sertindole and haloperidol. Pharmacopsychiatry. 2007;40:275–286. doi:10.1055/s-2007-99029118030652
  • Idris N, Neill J, Grayson B, et al. Sertindole improves sub-chronic PCP-induced reversal learning and episodic memory deficits in rodents: involvement of 5-HT(6) and 5-HT (2A) receptor mechanisms. Psychopharmacology (Berl). 2010;208:23–36. doi:10.1007/s00213-009-1702-519851757
  • Rodefer JS, Nguyen TN, Karlsson JJ, Arnt J. Reversal of subchronic PCP-induced deficits in attentional set shifting in rats by sertindole and a 5-HT6 receptor antagonist: comparison among antipsychotics. Neuropsychopharmacology. 2008;33:2657–2666. doi:10.1038/sj.npp.130165418094666
  • Karamatskos E, Lambert M, Mulert C, Naber D. Drug safety and efficacy evaluation of sertindole for schizophrenia. Expert Opin Drug Saf. 2012;11:1047–1062. doi:10.1517/14740338.2012.72698422992213
  • De Hert M, Mittoux A, He Y, Peuskens J. Metabolic parameters in the short- and long-term treatment of schizophrenia with sertindole or risperidone. Eur Arch Psychiatry Clin Neurosci. 2011;261:231–239. doi:10.1007/s00406-010-0142-x20820795
  • Boyda HN, Tse L, Procyshyn RM, Honer WG, Barr AM. Preclinical models of antipsychotic drug-induced metabolic side effects. Trends Pharmacol Sci. 2010;31:484–497. doi:10.1016/j.tips.2010.07.00220674990
  • Baptista T, de Baptista EA, Ying KNM, et al. Comparative effects of the antipsychotics sulpiride or risperidone in rats. I: bodyweight, food intake, body composition, hormones and glucose tolerance. Brain Res. 2002;957:144–151. doi:10.1016/s0006-8993(02)03616-812443990
  • Goudie AJ, Smith JA, Halford JC. Characterization of olanzapine-induced weight gain in rats. J Psychopharmacol. 2002;16:291–296. doi:10.1177/02698811020160040212503827
  • Cooper GD, Pickavance LC, Wilding JP, Harrold JA, Halford JC, Goudie AJ. Effects of olanzapine in male rats: enhanced adiposity in the absence of hyperphagia, weight gain or metabolic abnormalities. J Psychopharmacol. 2007;21:405–413. doi:10.1177/026988110606963717050655
  • Pouzet B, Mow T, Kreilgaard M, Velschow S. Chronic treatment with antipsychotics in rats as a model for antipsychotic-induced weight gain in human. Pharmacol Biochem Behav. 2003;75:133–140. doi:10.1016/S0091-3057(03)00042-X12759121
  • Cooper GD, Harrold JA, Halford JC, Goudie AJ. Chronic clozapine treatment in female rats does not induce weight gain or metabolic abnormalities but enhances adiposity: implications for animal models of antipsychotic-induced weight gain. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:428–436. doi:10.1016/j.pnpbp.2007.09.01217933447
  • Tulipano G, Rizzetti C, Bianchi I, Fanzani A, Spano P, Cocchi D. Clozapine-induced alteration of glucose homeostasis in the rat: the contribution of hypothalamic-pituitary-adrenal axis activation. Neuroendocrinology. 2007;85:61–70. doi:10.1159/00010098117374945
  • Victoriano M, Hermier D, Even PC, et al. Early perturbation in feeding behaviour and energy homeostasy in olanzapine-treated rats. Psychopharmacology (Berl). 2009;206:167–176. doi:10.1007/s00213-009-1593-519572122
  • Smith GC, Chaussade C, Vickers M, Jensen J, Shepherd PR. Atypical antipsychotic drugs induce derangements in glucose homeostasis by acutely increasing glucagon secretion and hepatic glucose output in the rat. Diabetologia. 2008;51:2309–2317. doi:10.1007/s00125-008-1152-318843478
  • Smith GC, Vickers MH, Cognard E, Shepherd PR. Clozapine and quetiapine acutely reduce glucagon-like peptide-1 production and increase glucagon release in obese rats: implications for glucose metabolism and food choice behaviour. Schizophr Res. 2009;115:30–40. doi:10.1016/j.schres.2009.07.01119679451
  • Houseknecht KL, Robertson AS, Zavadoski W, Gibbs EM, Johnson DE, Rollema H. Acute effects of atypical antipsychotics on whole-body insulin resistance in rats: implications for adverse metabolic effects. Neuropsychopharmacology. 2007;32:289–297. doi:10.1038/sj.npp.130120917035934
  • Chintoh AF, Mann SW, Lam L, et al. Insulin resistance and decreased glucose-stimulated insulin secretion after acute olanzapine administration. J Clin Psychopharmacol. 2008;28:494–499. doi:10.1097/JCP.0b013e318184b4c518794643
  • Chintoh AF, Mann SW, Lam L, et al. Insulin resistance and secretion in vivo: effects of different antipsychotics in an animal model. Schizophr Res. 2009;108:127–133. doi:10.1016/j.schres.2008.12.01219157785
  • Vestri HS, Maianu L, Moellering DR, Garvey WT. Atypical antipsychotic drugs directly impair insulin action in adipocytes: effects on glucose transport, lipogenesis, and antilipolysis. Neuropsychopharmacology. 2007;32:765–772. doi:10.1038/sj.npp.130114216823387
  • Yang LH, Chen TM, Yu ST, Chen YH. Olanzapine induces SREBP-1-related adipogenesis in 3T3-L1 cells. Pharmacol Res. 2007;56:202–208. doi:10.1016/j.phrs.2007.05.00717651982
  • Minet-Ringuet J, Even PC, Valet P, et al. Alterations of lipid metabolism and gene expression in rat adipocytes during chronic olanzapine treatment. Mol Psychiatry. 2007;12:562–571. doi:10.1038/sj.mp.400194817211438
  • Fernø J, Vik-Mo AO, Jassim G, et al. Acute clozapine exposure in vivo induces lipid accumulation and marked sequential changes in the expression of SREBP, PPAR, and LXR target genes in rat liver. Psychopharmacology (Berl). 2009;203:73–84. doi:10.1007/s00213-008-1370-x18989661
  • Raeder MB, Fernø J, Vik-Mo AO, Steen VM. SREBP activation by antipsychotic- and antidepressant-drugs in cultured human liver cells: relevance for metabolic side-effects. Mol Cell Biochem. 2006;289:167–173. doi:10.1007/s11010-006-9160-416718372
  • Monteiro S, Dias P, Madeira S, et al. Metabolic syndrome in dyslipidemia consultations. Rev Port Cardiol. 2006;25:821–831.17100172
  • Yang Z, Yin JY, Gong ZC, et al. Evidence for an effect of clozapine on the regulation of fat-cell derived factors. Clin Chim Acta. 2009;408:98–104. doi:10.1016/j.cca.2009.07.02119682446
  • Hemmrich K, Gummersbach C, Pallua N, Luckhaus C, Fehsel K. Clozapine enhances differentiation of adipocyte progenitor cells. Mol Psychiatry. 2006;11:980–981. doi:10.1038/sj.mp.400189217063181
  • Hemmrich K, Kappel BA, Paul NE, et al. Antipsychotic drugs increase adipose stem cell differentiation–implications for treatment with antipsychotic drugs. J Clin Psychopharmacol. 2011;31:663–665. doi:10.1097/JCP.0b013e31822bf0a521881453
  • Sertié AL, Suzuki AM, Sertié RA, et al. Effects of antipsychotics with different weight gain liabilities on human in vitro models of adipose tissue differentiation and metabolism. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35:1884–1890. doi:10.1016/j.pnpbp.2011.07.01721840366
  • Bulua AC, Simon A, Maddipati R, et al. Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS). J Exp Med. 2011;208:519–533. doi:10.1084/jem.2010204921282379
  • Pillai A, Parikh V, Terry AV, Mahadik SP. Long-term antipsychotic treatments and crossover studies in rats: differential effects of typical and atypical agents on the expression of antioxidant enzymes and membrane lipid peroxidation in rat brain. J Psychiatr Res. 2007;41:372–386. doi:10.1016/j.jpsychires.2006.01.01116564057
  • Polydoro M, Schröder N, Lima MN, et al. Haloperidol- and clozapine-induced oxidative stress in the rat brain. Pharmacol Biochem Behav. 2004;78:751–756. doi:10.1016/j.pbb.2004.05.01815301931
  • Schaffer LF, de Freitas CM, Chiapinotto CAP, et al. Harpagophytum procumbens ethyl acetate fraction reduces fluphenazine-induced vacuous chewing movements and oxidative stress in rat brain. Neurochem Res. 2016;41:1170–1184. doi:10.1007/s11064-015-1811-y26732278
  • Lauressergues E, Martin F, Helleboid A, et al. Overweight induced by chronic risperidone exposure is correlated with overexpression of the SREBP-1c and FAS genes in mouse liver. Naunyn Schmiedebergs Arch Pharmacol. 2011;383:423–436. doi:10.1007/s00210-010-0597-321336545
  • Lauressergues E, Staels B, Valeille K, et al. Antipsychotic drug action on SREBPs-related lipogenesis and cholesterogenesis in primary rat hepatocytes. Naunyn Schmiedebergs Arch Pharmacol. 2010;381:427–439. doi:10.1007/s00210-010-0499-420333360
  • Canfrán-Duque A, Casado ME, Pastor O, et al. Atypical antipsychotics alter cholesterol and fatty acid metabolism in vitro. J Lipid Res. 2013;54:310–324. doi:10.1194/jlr.M02694823175778
  • Soliman HM, Wagih HM, Algaidi SA, Hafiz AH. Histological evaluation of the role of atypical antipsychotic drugs in inducing non-alcoholic fatty liver disease in adult male albino rats (light and electron microscopic study). Folia Biol (Praha). 2013;59:173–180.24280139
  • Bilgic S, Tastemir KD, Azirak S, Guvenc AN, Kocaman N, Ozer MK. The protective effect of thymoquinone over olanzapine-induced side effects in liver, and metabolic side effects. Bratisl Lek Listy. 2017;118:618–625. doi:10.4149/BLL_2017_11929198130
  • Li Y, Su R, Xu S, Huang Q, Xu H. Artesunate prevents rats from the clozapine-induced hepatic steatosis and elevation in plasma triglycerides. Neuropsychiatr Dis Treat. 2017;13:2477–2487. doi:10.2147/NDT.S14506929026311
  • Coccurello R, Moles A. Potential mechanisms of atypical antipsychotic-induced metabolic derangement: clues for understanding obesity and novel drug design. Pharmacol Ther. 2010;127:210–251. doi:10.1016/j.pharmthera.2010.04.00820493213
  • Balt SL, Galloway GP, Baggott MJ, Schwartz Z, Mendelson J. Mechanisms and genetics of antipsychotic-associated weight gain. Clin Pharmacol Ther. 2011;90:179–183. doi:10.1038/clpt.2011.9721633343
  • Correll CU, Malhotra AK. Pharmacogenetics of antipsychotic-induced weight gain. Psychopharmacology (Berl). 2004;174:477–489. doi:10.1007/s00213-004-1949-915243737
  • Field BC, Chaudhri OB, Bloom SR. Obesity treatment: novel peripheral targets. Br J Clin Pharmacol. 2009;68:830–843. doi:10.1111/j.1365-2125.2009.03522.x20002077
  • Gil-Campos M, Cañete RR, Gil A. Adiponectin, the missing link in insulin resistance and obesity. Clin Nutr. 2004;23:963–974. doi:10.1016/j.clnu.2004.04.01015380884
  • Li S, Shin HJ, Ding EL, van Dam RM. Adiponectin levels and risk of type 2 diabetes: a systematic review and meta-analysis. JAMA. 2009;302:179–188. doi:10.1001/jama.2009.97619584347
  • Sapra M, Lawson D, Iranmanesh A, Varma A. Adiposity-independent hypoadiponectinemia as a potential marker of insulin resistance and inflammation in schizophrenia patients treated with second generation antipsychotics. Schizophr Res. 2016;174:132–136. doi:10.1016/j.schres.2016.04.05127211515
  • Bartoli F, Crocamo C, Clerici M, Carrà G. Second-generation antipsychotics and adiponectin evels in schizophrenia: a comparative meta-analysis. Eur Neuropsychopharmacol. 2015;25:1767–1774. doi:10.1016/j.euroneuro.2015.06.01126164075
  • Tagami K, Kashiwase Y, Yokoyama A, et al. The atypical antipsychotic, olanzapine, potentiates ghrelin-induced receptor signaling: an in vitro study with cells expressing cloned human growth hormone secretagogue receptor. Neuropeptides. 2016;58:93–101. doi:10.1016/j.npep.2015.12.01026775231
  • Hegedűs C, Kovács D, Kiss R, et al. Effect of long-term olanzapine treatment on meal-induced insulin sensitization and on gastrointestinal peptides in female Sprague-Dawley rats. J Psychopharmacol. 2015;29:1271–1279. doi:10.1177/026988111560295226349558
  • Virkkunen M, Wahlbeck K, Rissanen A, Naukkarinen H, Franssila-Kallunki A. Decrease of energy expenditure causes weight increase in olanzapine treatment - a case study. Pharmacopsychiatry. 2002;35:124–126. doi:10.1055/s-2002-3152112107860
  • Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796–1808. doi:10.1172/JCI1924614679176
  • Van Gaal LF. Long-term health considerations in schizophrenia: metabolic effects and the role of abdominal adiposity. Eur Neuropsychopharmacol. 2006;16(Suppl 3):S142–S148. doi:10.1016/j.euroneuro.2006.06.00516863690
  • Victoriano M, de Beaurepaire R, Naour N, et al. Olanzapine-induced accumulation of adipose tissue is associated with an inflammatory state. Brain Res. 2010;1350:167–175. doi:10.1016/j.brainres.2010.05.06020570665
  • Devlin MJ, Yanovski SZ, Wilson GT. Obesity: what mental health professionals need to know. Am J Psychiatry. 2000;157:854–866. doi:10.1176/appi.ajp.157.6.85410831462
  • Monji A, Kato T, Kanba S. Cytokines and schizophrenia: microglia hypothesis of schizophrenia. Psychiatry Clin Neurosci. 2009;63:257–265.19579286
  • O’Connell KE, Thakore J, Dev KK. Pro-inflammatory cytokine levels are raised in female schizophrenia patients treated with clozapine. Schizophr Res. 2014;156:1–8. doi:10.1016/j.schres.2014.03.02024742875
  • Sobiś J, Rykaczewska-Czerwińska M, Świętochowska E, Gorczyca P. Therapeutic effect of aripiprazole in chronic schizophrenia is accompanied by anti-inflammatory activity. Pharmacol Rep. 2015;67:353–359. doi:10.1016/j.pharep.2014.09.00725712663
  • Hatziagelaki E, Tsiavou A, Gerasimou C, et al. Effects of olanzapine on cytokine profile and brain-derived neurotrophic factor in drug-naive subjects with first-episode psychosis. Exp Ther Med. 2019;17:3071–3076. doi:10.3892/etm.2019.728530906479
  • Drzyzga L, Obuchowicz E, Marcinowska A, Herman ZS. Cytokines in schizophrenia and the effects of antipsychotic drugs. Brain Behav Immun. 2006;20:532–545. doi:10.1016/j.bbi.2006.02.00216580814
  • Baker GF, Rogers HJ. Effects of psychotropic drugs on the erythrocyte permeability to glucose and ethylidene glucose. Biochem Pharmacol. 1972;21:1871–1878. doi:10.1016/0006-2952(72)90183-94646186
  • Ardizzone TD, Bradley RJ, Freeman AM, Dwyer DS. Inhibition of glucose transport in PC12 cells by the atypical antipsychotic drugs risperidone and clozapine, and structural analogs of clozapine. Brain Res. 2001;923:82–90. doi:10.1016/s0006-8993(01)03026-811743975
  • Dwyer DS, Liu Y, Bradley RJ. Dopamine receptor antagonists modulate glucose uptake in rat pheochromocytoma (PC12) cells. Neurosci Lett. 1999;274:151–154. doi:10.1016/s0304-3940(99)00712-010548412
  • Dwyer DS, Pinkofsky HB, Liu Y, Bradley RJ. Antipsychotic drugs affect glucose uptake and the expression of glucose transporters in PC12 cells. Prog Neuropsychopharmacol Biol Psychiatry. 1999;23:69–80.10368857
  • Dwyer DS, Donohoe D. Induction of hyperglycemia in mice with atypical antipsychotic drugs that inhibit glucose uptake. Pharmacol Biochem Behav. 2003;75:255–260.12873613
  • Jassim G, Skrede S, Vázquez MJ, et al. Acute effects of orexigenic antipsychotic drugs on lipid and carbohydrate metabolism in rat. Psychopharmacology (Berl). 2012;219:783–794. doi:10.1007/s00213-011-2397-y21748251
  • Hou JC, Pessin JE. Ins (endocytosis) and outs (exocytosis) of GLUT4 trafficking. Curr Opin Cell Biol. 2007;19:466–473. doi:10.1016/j.ceb.2007.04.01817644329
  • Beaulieu JM, Sotnikova TD, Marion S, Lefkowitz RJ, Gainetdinov RR, Caron MG. An Akt/beta-arrestin 2/PP2A signaling complex mediates dopaminergic neurotransmission and behavior. Cell. 2005;122:261–273. doi:10.1016/j.cell.2005.05.01216051150
  • Girault JA, Greengard P. The neurobiology of dopamine signaling. Arch Neurol. 2004;61:641–644. doi:10.1001/archneur.61.5.64115148138
  • Martínez JA. Mitochondrial oxidative stress and inflammation: an slalom to obesity and insulin resistance. J Physiol Biochem. 2006;62:303–306.17615956
  • Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219–246. doi:10.1146/annurev-physiol-021909-13584620148674
  • Baig MR, Navaira E, Escamilla MA, Raventos H, Walss-Bass C. Clozapine treatment causes oxidation of proteins involved in energy metabolism in lymphoblastoid cells: a possible mechanism for antipsychotic-induced metabolic alterations. J Psychiatr Pract. 2010;16:325–333.20859109
  • Casademont J, Garrabou G, Miró O, et al. Neuroleptic treatment effect on mitochondrial electron transport chain: peripheral blood mononuclear cells analysis in psychotic patients. J Clin Psychopharmacol. 2007;27:284–288. doi:10.1097/JCP.0b013e318054753e17502776
  • Ji B, La Y, Gao L, et al. A comparative proteomics analysis of rat mitochondria from the cerebral cortex and hippocampus in response to antipsychotic medications. J Proteome Res. 2009;8:3633–3641. doi:10.1021/pr800876z19441803
  • Kotzka J, Müller-Wieland D. Sterol regulatory element-binding protein (SREBP)-1: gene regulatory target for insulin resistance. Expert Opin Ther Targets. 2004;8:141–149. doi:10.1517/14728222.8.2.14115102555
  • Shimano H. Sterol regulatory element-binding proteins (SREBPs): transcriptional regulators of lipid synthetic genes. Prog Lipid Res. 2001;40:439–452.11591434
  • Fernø J, Raeder MB, Vik-Mo AO, et al. Antipsychotic drugs activate SREBP-regulated expression of lipid biosynthetic genes in cultured human glioma cells: a novel mechanism of action. Pharmacogenomics J. 2005;5:298–304. doi:10.1038/sj.tpj.650032316027736
  • Cai HL, Tan QY, Jiang P, et al. A potential mechanism underlying atypical antipsychotics-induced lipid disturbances. Transl Psychiatry. 2015;5:e661. doi:10.1038/tp.2015.16126485545
  • McKenney JM, Sica D. Prescription omega-3 fatty acids for the treatment of hypertriglyceridemia. Am J Health Syst Pharm. 2007;64:595–605. doi:10.2146/ajhp06016417353568
  • Hassanali Z, Ametaj BN, Field CJ, Proctor SD, Vine DF. Dietary supplementation of n-3 PUFA reduces weight gain and improves postprandial lipaemia and the associated inflammatory response in the obese JCR:LA-cp rat. Diabetes Obes Metab. 2010;12:139–147. doi:10.1111/j.1463-1326.2009.01130.x19917068
  • Pachikian BD, Neyrinck AM, Cani PD, et al. Hepatic steatosis in n-3 fatty acid depleted mice: focus on metabolic alterations related to tissue fatty acid composition. BMC Physiol. 2008;8:21. doi:10.1186/1472-6793-8-2119046413
  • Pachikian BD, Essaghir A, Demoulin JB, et al. Hepatic n-3 polyunsaturated fatty acid depletion promotes steatosis and insulin resistance in mice: genomic analysis of cellular targets. PLoS One. 2011;6:e23365. doi:10.1371/journal.pone.002336521853118
  • Qi K, Fan C, Jiang J, et al. Omega-3 fatty acid containing diets decrease plasma triglyceride concentrations in mice by reducing endogenous triglyceride synthesis and enhancing the blood clearance of triglyceride-rich particles. Clin Nutr. 2008;27:424–430. doi:10.1016/j.clnu.2008.02.00118362042
  • Araya J, Rodrigo R, Videla LA, et al. Increase in long-chain polyunsaturated fatty acid n - 6/n - 3 ratio in relation to hepatic steatosis in patients with non-alcoholic fatty liver disease. Clin Sci (Lond). 2004;106:635–643. doi:10.1042/CS2003032614720121
  • Burrows T, Collins CE, Garg ML. Omega-3 index, obesity and insulin resistance in children. Int J Pediatr Obes. 2011;6:e532–e539. doi:10.3109/17477166.2010.54948921226540
  • Evans DR, Parikh VV, Khan MM, Coussons C, Buckley PF, Mahadik SP. Red blood cell membrane essential fatty acid metabolism in early psychotic patients following antipsychotic drug treatment. Prostaglandins Leukot Essent Fatty Acids. 2003;69:393–399.14623492
  • Ranjekar PK, Hinge A, Hegde MV, et al. Decreased antioxidant enzymes and membrane essential polyunsaturated fatty acids in schizophrenic and bipolar mood disorder patients. Psychiatry Res. 2003;121:109–122.14656446
  • Reddy RD, Keshavan MS, Yao JK. Reduced red blood cell membrane essential polyunsaturated fatty acids in first episode schizophrenia at neuroleptic-naive baseline. Schizophr Bull. 2004;30:901–911. doi:10.1093/oxfordjournals.schbul.a00714015957200
  • Caniato RN, Alvarenga ME, Garcia-Alcaraz MA. Effect of omega-3 fatty acids on the lipid profile of patients taking clozapine. Aust N Z J Psychiatry. 2006;40:691–697. doi:10.1080/j.1440-1614.2006.01869.x16866765
  • Peet M, Horrobin DF. A dose-ranging exploratory study of the effects of ethyl- eicosapentaenoate in patients with persistent schizophrenic symptoms. J Psychiatr Res. 2002;36:7–18.11755456
  • McNamara RK, Magrisso IJ, Hofacer R, et al. Omega-3 fatty acid deficiency augments risperidone-induced hepatic steatosis in rats: positive association with stearoyl-CoA desaturase. Pharmacol Res. 2012;66:283–291. doi:10.1016/j.phrs.2012.06.01022750665