Elsevier

Psychoneuroendocrinology

Volume 104, June 2019, Pages 286-299
Psychoneuroendocrinology

Olanzapine-induced endoplasmic reticulum stress and inflammation in the hypothalamus were inhibited by an ER stress inhibitor 4-phenylbutyrate

https://doi.org/10.1016/j.psyneuen.2019.03.017Get rights and content

Highlights

  • Olanzapine treatment activated hypothalamic ER stress PERK-eIF2α and inflammatory IKKβ-NFκB signaling.

  • Co-treatment with an ER stress inhibitor, 4-PBA, decreased olanzapine-induced weight gain.

  • 4-PBA inhibited olanzapine-induced activated PERK-eIF2α and IKKβ-NFκB signaling in the hypothalamus.

  • Hypothalamic ER stress was related to olanzapine-induced weight gain.

Abstract

Antipsychotics are the most important treatment for schizophrenia. However, antipsychotics, particularly olanzapine and clozapine, are associated with severe weight gain/obesity side-effects. Although numerous studies have been carried out to identify the exact mechanisms of antipsychotic-induced weight gain, it is still important to consider other pathways. Endoplasmic reticulum (ER) stress signaling and its associated inflammation pathway is one of the most important pathways involved in regulation of energy balance. In the present study, we examined the role of hypothalamic protein kinase R like endoplasmic reticulum kinase- eukaryotic initiation factor 2α (PERK-eIF2α) signaling and the inflammatory IkappaB kinase β- nuclear factor kappa B (IKKβ-NFκB) signaling pathway in olanzapine-induced weight gain in female rats. In this study, we found that olanzapine significantly activated PERK-eIF2α and IKKβ-NFκB signaling in SH-SY5Y cells in a dose-dependent manner. Olanzapine treatment for 8 days in rats was associated with activated PERK-eIF2α signaling and IKKβ-NFκB signaling in the hypothalamus, accompanied by increased food intake and weight gain. Co-treatment with an ER stress inhibitor, 4-phenylbutyrate (4-PBA), decreased olanzapine-induced food intake and weight gain in a dose- and time-dependent manner. Moreover, 4-PBA dose-dependently inhibited olanzapine-induced activated PERK-eIF2α and IKKβ-NFκB signaling in the hypothalamus. These results suggested that hypothalamic ER stress may play an important role in antipsychotic-induced weight gain.

Introduction

Antipsychotics (APs) are widely used in the treatment of schizophrenia. However, APs particularly olanzapine and clozapine, are associated with weight gain and other metabolic side-effects. Previous studies have suggested that multiple factors are involved in AP-induced weight gain, including the histamine H1 receptor and its associated 5′ AMP-activated protein kinase (AMPK) signaling (He et al., 2014), H3 receptor (Lian et al., 2014; Poyurovsky et al., 2013, 2005), serotonin receptor 2C (Lord et al., 2017), ghrelin-mediated growth hormone secretagog receptor (GHS-R) signaling (Tagami et al., 2016; Zhang et al., 2014b), and neuropeptides (Huang et al., 2006; Zhang et al., 2014b). It has long been established that the cannabinoid receptor 1 (CB1 receptor) is involved in the metabolic mechanisms of food intake (Lazzari et al., 2011; Manca et al., 2013; Mastinu et al., 2012, 2013). Recently, a study in female rats reported that olanzapine-induced food intake and weight gain was markedly inhibited by the CB1 receptor inverse agonists rimonabant or NESS06SM (Lazzari et al., 2017), suggesting the importance of the CB1 receptor in AP-induced weight gain. Moreover, interesting research demonstrated that the microbial fermentation of prebiotics, B-GOS® attenuated olanzapine-induced weight gain in female rats (Kao et al., 2018). However, in the clinic, there is still a lack of medication for preventing and treating AP-induced weight gain. To find out the interaction of APs with other signaling pathways that mediate energy balance is very important for treating AP-induced weight gain and for development of novel non-obesogenic APs.

The endoplasmic reticulum (ER) is a continuous membrane system in the cells of eukaryotic organisms. ER stress refers to a pathophysiological process in which ER function is disordered under conditions of hypoxia, oxidative stress, undernutrition or imbalance of calcium ion homeostasis (Cnop et al., 2012). ER stress is mediated by three proteins: protein kinase R-like ER kinase (PERK), inositol requiring enzyme 1(IRE1) and activating transcription factor 6 (ATF6) (Cnop et al., 2012). Under non-stress conditions, the three proteins are inactive in combination with glucose-regulated protein 78 (GRP78). GRP78 is also referred to as the immunoglobulin heavy chain-binding protein (BiP) (Lee, 2005). When ER stress occurs, PERK, IRE1, and ATF6 dissociate from GRP78, and PERK is then activated by phosphorylation. pPERK thereby phosphorylates the downstream eukaryotic initiation factor 2α (eIF2α) to inhibit the translation and synthesis of transcription factor 4 (ATF4). ATF4 up-regulates GRP78/BiP expression (PERK-eIF2α signaling), and this effect contributes to the restoration of endoplasmic reticulum homeostasis (Cnop et al., 2012). Therefore, up-regulation of GRP78/BiP expression is a widely used marker of ER stress (Cnop et al., 2012). It is noteworthy that activation of hypothalamic ER stress PERK- eIF2α signaling leads to obesity (Cnop et al., 2012; Ozcan et al., 2009; Ramirez and Claret, 2015; Zhang et al., 2008). In high-fat diet (HFD) induced obese rodents (both males and females), the expression of pPERK, peIF2α and GRP78/BiP in the hypothalamus was significantly increased (Cakir et al., 2013; Melo et al., 2014). In male mice model, activation of hypothalamic ER stress leads to hyperphagia and inhibition of intra-capsular brown adipose tissue (IBAT) decomposition and heat production, resulting in severe obesity (Ozcan et al., 2009; Qiang et al., 2017). Conversely, inhibition of the hypothalamic ER stress by 4-phenylbutyrate (4-PBA) and tauroursodeoxycholic acid (TUDCA) reduces food intake and increases heat production in IBAT in male obese mice, thereby inhibiting obesity (Melo et al., 2014; Ozcan et al., 2009; Ramirez and Claret, 2015)

In the pathological process of obesity, hypothalamic ER stress is related to inflammation (Zhang et al., 2008). It has been demonstrated that activation of hypothalamic PERK- eIF2α signaling activates nuclear factor kappa B (NFκB), one of the most important transcriptional regulators in inflammatory response (Zhang et al., 2008). HFD feeding significantly activated hypothalamic ER stress and IKKβ-NFκB signaling, whereas intraventricular injection of an ER stress inhibitor TUDCA markedly inhibited hypothalamic IKKβ-NFκB signaling, accompanied by decreased food intake and weight gain in mice (gender not shown) (Zhang et al., 2008). Activation of hypothalamic IKKβ-NFκB signaling by constitutively active IKKβ cloned lentiviral vector induced increased food intake and weight gain in mice. On the contrary, inhibition of hypothalamic IKKβ-NFκB signaling by dominant-negative IKKβ cloned lentiviral vector significantly protected mice from obesity (Zhang et al., 2008). These results suggested that overnutrition activates hypothalamic IKKβ-NFκB signaling at least partly via elevating hypothalamic ER stress. Moreover, it has been found in male mice that NFκB activation increased the transcriptional activity of pro-opiomelanocortin (POMC) in the hypothalamus, suggesting involvement of NFκB in feeding control (Jang et al., 2010). Furthermore, the involvement of NFκB in a male murine model of neurodevelopment disorder has been reported recently (Bonini et al., 2016).

Previous studies have illustrated that the most obesogenic APs, including olanzapine, clozapine and risperidone, significantly activated ER stress PERK- eIF2α signaling in β-pancreatic cells (Ozasa et al., 2013) and the male mouse liver (Lauressergues et al., 2012), therefore leading to glucose and lipid metabolism dysfunction. A recent study in a female rat model reported that acute clozapine treatment (treated once and rats were sacrificed 1 h after the treatment) increased the protein expression of eIF2α and IRE1 in the liver (Weston-Green et al., 2018). This evidence suggested that AP treatment did activate ER stress PERK-eIF2α signaling. However, the effect of olanzapine on hypothalamic ER stress PERK-eIF2α signaling and IKKβ-NFκB signaling is unclear. The present study aimed to examine the role of hypothalamic ER stress PERK- elF2α signaling and its related IKKβ-NFκB signaling in olanzapine-induced weight gain.

Section snippets

Cell culture and olanzapine treatment

The human neuroblastoma SH-SY5Y cell line was purchased from American Type Culture Collection (Rockville, MD, USA). The cells were cultured in Dulbecco`s Modified Eagle Medium (DMEM) (Thermo Fisher Scientific, Wuhan, Hubei, China, #31,966,021) supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin (Thermo Fisher Scientific, #10,270,098 and #15070063, respectively) at 37 °C in a humidified 5% CO2 incubator. The cells were regularly subcultured to maintain them in the logarithmic

Olanzapine dose-dependently activated PERK- eIF2α and IKKβ-NFκB signaling in human neuroblastoma SH-SY5Y cells

The protein expression of key molecules of PERK- eIF2α and IKKβ-NFκB signaling was examined by Western blot. As shown in Fig. 1A-B, compared with the control group (treated with DMSO), olanzapine 100 μM treated for 24 h markedly increased the protein expression of pPERK (increased by 107.3 ± 26.9%, p <  0.05), peIF2α (by 70.7 ± 9.3%, p < 0.01), ATF4 (by 57.7 ±14.8%, p < 0.01) and GRP78/BiP (by 44.6 ± 2.0%, p <  0.01). 50 μM olanzapine did not significantly increase the protein expression of

Discussion

The present study firstly demonstrated that olanzapine induced aberrant activation of ER stress PERK-eIF2α signaling in human neuroblastoma SH-SY5Y cells, suggesting that olanzapine might directly act on the neurons to induce ER stress. The animal study provides the first evidence that sub-chronic olanzapine treatment activated the hypothalamic PERK-eIF2α signaling. The protein expression of pPERK positively correlated with food intake, weight gain and feeding efficiency. Previous studies

Conclusions

In conclusion, our study revealed that olanzapine treatment markedly activated the hypothalamic ER stress PERK-eIF2α signaling and inflammatory IKKβ-NFκB signaling, and these effects were related to olanzapine-induced weight gain. Co-treatment with an ER stress inhibitor, 4-PBA, inhibited olanzapine-induced weight gain via suppressing PERK-eIF2α signaling and the related inflammatory IKKβ-NFκB signaling in the hypothalamus. This study may shed light on future possibilities for development of

List of author contributions

Prof T Sun and X-F Huang designed this study. Drs M He, J Li, G Gao, Ms T Zhou, W Li, J Hu and J Chen performed and managed this project study. M He and J Li undertook the statistical analysis. M He wrote the draft of the manuscript. Drs Sun, Huang and He contributed in the discussion of interpretation of the results. All authors have contributed to and have approved the final manuscript.

Conflicts of interest

None.

Acknowledgements

The work was supported by the National Natural Science Foundation of China (81803515, 21805218 and 21703163), the Wuhan University of Technology fund for First-class University and First-class Discipline Construction Projects (472-20162008), the Natural Science Foundation of Hubei Province (2018CFB342 and 2018CFB348) and the Fundamental Research Funds for the Central Universities (173120003). We thank Ms Hongqin Wang and Prof. Chao Deng (University of Wollongong) for their kind help with the

References (103)

  • F. Gressier et al.

    Pharmacogenetics of clozapine response and induced weight gain: a comprehensive review and meta-analysis

    Eur. Neuropsychopharmacol.

    (2016)
  • H. Hakko et al.

    Are females at special risk of obesity if they become psychotic? The longitudinal Northern Finland 1966 birth cohort study

    Schizophr. Res.

    (2006)
  • M. He et al.

    Hypothalamic histamine H1 receptor-AMPK signaling time-dependently mediates olanzapine-induced hyperphagia and weight gain in female rats

    Psychoneuroendocrinology

    (2014)
  • M. He et al.

    Time-dependent effects of olanzapine treatment on the expression of histidine decarboxylase, H1 and H3 receptor in the rat brain: The roles in olanzapine-induced obesity

    Psychoneuroendocrinology

    (2017)
  • X.F. Huang et al.

    Neuropeptide Y mRNA expression levels following chronic olanzapine, clozapine and haloperidol administration in rats

    Neuropeptides

    (2006)
  • M. Ikegami et al.

    Olanzapine induces glucose intolerance through the activation of AMPK in the mouse hypothalamus

    Eur. J. Pharmacol.

    (2013)
  • P.-G. Jang et al.

    NF-κB activation in hypothalamic pro-opiomelanocortin neurons Is essential in illness- and leptin-induced anorexia

    J. Biol. Chem.

    (2010)
  • N.R. Kim et al.

    Protective effects of olanzapine and haloperidol on serum withdrawal-induced apoptosis in SH-SY5Y cells

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2008)
  • E. Lauressergues et al.

    Does endoplasmic reticulum stress participate in APD-induced hepatic metabolic dysregulation?

    Neuropharmacology

    (2012)
  • P. Lazzari et al.

    Weight loss induced by rimonabant is associated with an altered leptin expression and hypothalamic leptin signaling in diet-induced obese mice

    Behav. Brain Res.

    (2011)
  • P. Lazzari et al.

    Metabolic side effects induced by olanzapine treatment are neutralized by CB1 receptor antagonist compounds co-administration in female rats

    Eur. Neuropsychopharmacol.

    (2017)
  • A.S. Lee

    The ER chaperone and signaling regulator GRP78/BiP as a monitor of endoplasmic reticulum stress

    Methods

    (2005)
  • J.G. Lee et al.

    Effects of olanzapine on brain-derived neurotrophic factor gene promoter activity in SH-SY5Y neuroblastoma cells

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2010)
  • J. Lian et al.

    Betahistine ameliorates olanzapine-induced weight gain through modulation of histaminergic, NPY and AMPK pathways

    Psychoneuroendocrinology

    (2014)
  • X. Liu et al.

    Betahistine co-treatment ameliorates dyslipidemia induced by chronic olanzapine treatment in rats through modulation of hepatic AMPKalpha-SREBP-1 and PPARalpha-dependent pathways

    Pharmacol. Res.

    (2015)
  • I. Manca et al.

    Novel pyrazole derivatives as neutral CB(1) antagonists with significant activity towards food intake

    Eur. J. Med. Chem.

    (2013)
  • G. Marwarha et al.

    Endoplasmic reticulum stress-induced CHOP activation mediates the down-regulation of leptin in human neuroblastoma SH-SY5Y cells treated with the oxysterol 27-hydroxycholesterol

    Cell. Signal.

    (2012)
  • A. Mastinu et al.

    NESS038C6, a novel selective CB1 antagonist agent with anti-obesity activity and improved molecular profile

    Behav. Brain Res.

    (2012)
  • A. Mastinu et al.

    NESS06SM reduces body weight with an improved profile relative to SR141716A

    Pharmacol. Res.

    (2013)
  • A.C. Maurin et al.

    Hypothalamic eIF2alpha signaling regulates food intake

    Cell. Rep.

    (2014)
  • A.M. Melo et al.

    Hypothalamic endoplasmic reticulum stress and insulin resistance in offspring of mice dams fed high-fat diet during pregnancy and lactation

    Metabolism

    (2014)
  • M. Monda et al.

    Quetiapine lowers sympathetic and hyperthermic reactions due to cerebral injection of orexin A

    Neuropeptides

    (2006)
  • L. Ozcan et al.

    Endoplasmic reticulum stress plays a central role in development of leptin resistance

    Cell. Metab.

    (2009)
  • S. Ramirez et al.

    Hypothalamic ER stress: a bridge between leptin resistance and obesity

    FEBS Lett.

    (2015)
  • B.M. Razavi et al.

    Protective effects of green tea on olanzapine-induced-metabolic syndrome in rats

    Biomed. Pharmacother.

    (2017)
  • E. Rojczyk et al.

    Effect of short and long-term treatment with antipsychotics on orexigenic/anorexigenic neuropeptides expression in the rat hypothalamus

    Neuropeptides

    (2015)
  • T. Romanatto et al.

    TNF-alpha acts in the hypothalamus inhibiting food intake and increasing the respiratory quotient--effects on leptin and insulin signaling pathways

    Peptides

    (2007)
  • K. Tagami 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)
  • M.J. Waterson et al.

    Neuronal regulation of energy homeostasis: beyond the hypothalamus and feeding

    Cell. Metab.

    (2015)
  • K. Weston-Green et al.

    Olanzapine treatment and metabolic dysfunction: a dose response study in female sprague Dawley rats

    Behavioural. Brain Res.

    (2011)
  • K. Weston-Green et al.

    Olanzapine treatment and metabolic dysfunction: a dose response study in female sprague Dawley rats

    Behav. Brain Res.

    (2011)
  • K.W. Williams et al.

    Xbp1s in pomc neurons connects ER stress with energy balance and glucose homeostasis

    Cell. Metab.

    (2014)
  • Q. Wu et al.

    Loss of GABAergic signaling by AgRP neurons to the parabrachial nucleus leads to starvation

    Cell

    (2009)
  • X. Zhang et al.

    Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity

    Cell

    (2008)
  • V.L. Albaugh et al.

    Hormonal and metabolic effects of olanzapine and clozapine related to body weight in rodents

    Obesity (Silver Spring)

    (2006)
  • V.L. Albaugh et al.

    Olanzapine promotes fat accumulation in male rats by decreasing physical activity, repartitioning energy and increasing adipose tissue lipogenesis while impairing lipolysis

    Mol. Psychiatry

    (2011)
  • M. Aravagiri et al.

    Pharmacokinetics and tissue distribution of olanzapine in rats

    Biopharm. Drug Dispos.

    (1999)
  • L. Benarroch et al.

    Atypical antipsychotics and effects on feeding: from mice to men

    Psychopharmacology (Berl)

    (2016)
  • N. Bergemann et al.

    Olanzapine plasma concentration, average daily dose, and interaction with co-medication in schizophrenic patients

    Pharmacopsychiatry

    (2004)
  • S.A. Bonini et al.

    Cortical structure alterations and social behavior impairment in p50-deficient mice

    Cereb. Cortex

    (2016)
  • Cited by (24)

    • Network pharmacology-based exploration of therapeutic mechanism of Liu-Yu-Tang in atypical antipsychotic drug-induced metabolic syndrome

      2021, Computers in Biology and Medicine
      Citation Excerpt :

      The specific mechanisms by which AADs induce metabolic syndrome are not yet fully understood. There are several influencing factors such as elevated inflammatory cytokines in circulation, altered hypothalamic neuropeptides and pituitary hormones, altered striatal and mesolimbic dopamine reward pathways, endoplasmic reticulum stress, intestinal flora imbalance, and gene polymorphisms [29–31]. To explore the underlying mechanisms of LYT's curative effects on AADs-induced MetS, we focus on two aspects: 1) Identify key drug targets that have major regulatory roles in treatment, and 2) identify key biological pathways through which key molecules conduct cascade signals and exert therapeutic effects.

    • Longdan Xiegan Tang attenuates liver injury and hepatic insulin resistance by regulating the angiotensin-converting enzyme 2/Ang (1–7)/Mas axis-mediated anti-inflammatory pathway in rats

      2021, Journal of Ethnopharmacology
      Citation Excerpt :

      The atypical antipsychotic drug olanzapine has been reported to induce liver injury manifested by jaundice with increased blood bilirubin, and alanine aminotransferase (ALT) and aspartate aminostransferase (AST) hyperactivities, fatty liver and hepatitis (including hepatocellular injury, cholestatic, or mixed type) (Mouradian-Stamatiadis et al., 2002; Ozcanli et al., 2006; Liu et al., 2015a, b; Gómez-Lumbreras et al., 2018; Jiang et al., 2019; Ren et al., 2019b). Besides, olanzapine frequently causes other inflammation-associated metabolic abnormalities, such as insulin resistance and type 2 diabetes both in humans and in rodents (Ballon et al., 2014; Chen et al., 2018; He et al., 2019; Li et al., 2019; Singh et al., 2019; Ren et al., 2019a). In this context, the present study investigated the involvement of the ACE2/Ang (1–7)/Mas axis in LXT-elicited improvement of liver injury using the olanzapine-induced rat model.

    • The protein kinase D1-mediated inflammatory pathway is involved in olanzapine-induced impairment of skeletal muscle insulin signaling in rats

      2021, Life Sciences
      Citation Excerpt :

      Olanzapine also suppressed chronic social isolation-induced hepatic NFκB activation in male rats [60]. However, olanzapine treatment activated IKKβ/NFκB in the hypothalamus of female rats [61]. It has been reported that long-term treatment of schizophrenia patients with olanzapine induced systemic insulin resistance, accompanied by increases in plasma concentrations, and adipose, hypothalamic and aortic expression of pro-inflammatory cytokines including IL1ß, IL6, TNFα and NFκB in humans, rodents or 3T3-L1 adipocytes [62–64].

    View all citing articles on Scopus
    View full text