Elsevier

Psychoneuroendocrinology

Volume 104, June 2019, Pages 42-48
Psychoneuroendocrinology

Antipsychotics differentially regulate insulin, energy sensing, and inflammation pathways in hypothalamic rat neurons

https://doi.org/10.1016/j.psyneuen.2019.01.029Get rights and content

Highlights

  • Antipsychotic (AP)s directly impact insulin and inflammatory pathways in hypothalamic neurons.

  • The MAPK pathway was consistently upregulated by all APs.

  • Only clozapine and aripiprazole inhibited pAKT and increased pAMPK.

  • Olanzapine and aripiprazole upregulated inflammatory pathways and BDNF.

  • APs have differential effects in the hypothalamus which do not align with clinical metabolic risk.

Abstract

Introduction

Second generation antipsychotic (AP)s remain the gold-standard treatment for schizophrenia and are widely used on- and off-label for other psychiatric illnesses. However, these agents cause serious metabolic side-effects. The hypothalamus is the primary brain region responsible for whole body energy regulation, and disruptions in energy sensing (e.g. insulin signaling) and inflammation in this brain region have been implicated in the development of insulin resistance and obesity. To elucidate mechanisms by which APs may be causing metabolic dysregulation, we explored whether these agents can directly impact energy sensing and inflammation in hypothalamic neurons.

Methods

The rat hypothalamic neuronal cell line, rHypoE-19, was treated with olanzapine (0.25–100 uM), clozapine (2.5–100 uM) or aripiprazole (5–20 uM). Western blots measured the energy sensing protein AMPK, components of the insulin signaling pathway (AKT, GSK3β), and components of the MAPK pathway (ERK1/2, JNK, p38). Quantitative real-time PCR was performed to determine changes in the mRNA expression of interleukin (IL)-6, IL-10 and brain derived neurotrophic factor (BDNF).

Results

Olanzapine (100 uM) and clozapine (100, 20 uM) significantly increased pERK1/2 and pJNK protein expression, while aripiprazole (20 uM) only increased pJNK. Clozapine (100 uM) and aripiprazole (5 and 20 uM) significantly increased AMPK phosphorylation (an orexigenic energy sensor), and inhibited insulin-induced phosphorylation of AKT. Olanzapine (100 uM) treatment caused a significant increase in IL-6 while aripiprazole (20 uM) significantly decreased IL-10. Olanzapine (100 uM) and aripiprazole (20 uM) increased BDNF expression.

Conclusions

We demonstrate that antipsychotics can directly regulate insulin, energy sensing, and inflammatory pathways in hypothalamic neurons. Increased MAPK activation by all antipsychotics, alongside olanzapine-associated increases in IL-6, and aripiprazole-associated decreases in IL-10, suggests induction of pro-inflammatory pathways. Clozapine and aripiprazole inhibition of insulin-stimulated pAKT and increases in AMPK phosphorylation (an orexigenic energy sensor) suggests impaired insulin action and energy sensing. Conversely, olanzapine and aripiprazole increased BDNF, which would be expected to be metabolically beneficial. Overall, our findings suggest differential effects of antipsychotics on hypothalamic neuroinflammation and energy sensing.

Introduction

Antipsychotics (APs) are the gold-standard treatment for schizophrenia and are increasingly being prescribed both on- and off-label for other psychiatric conditions. However, these medications cause serious metabolic side effects. In turn, patients with schizophrenia have a two-fold increase in standardized mortality ratio from cardiovascular disease (Hennekens et al., 2005). Although the liability for metabolic side effects differs between APs (Newcomer, 2005), it has recently become clear that no single AP is devoid of this risk, particularly in younger individuals who are AP-naïve (Correll et al., 2009). Additionally, it is found that regardless of class or agent, APs increase risk of diabetes 2–3 fold, above and beyond the risk that is conferred by the illness of schizophrenia itself (Rajkumar et al., 2017).

The mechanisms of AP-induced metabolic dysregulation are unclear, but are understood to occur at least in part through the central nervous system (Kowalchuk et al., 2018). In addition, the question has arisen whether APs could be causing metabolic perturbations by directly impact brain insulin signalling (Kowalchuk et al., 2017). Insulin receptors are expressed at high levels in many brain areas, where signalling pathways (mediated through AKT, GSK3β, and the MAPKs) play a significant role in neuronal growth, memory, and energy regulation. Brain studies examining AP administration in vitro and in rodents in vivo have variably suggested upregulation of pathways downstream of the insulin receptor (i.e. AKT phosphorylation/ activation; GSK phosphorylation /deactivation), which is difficult to reconcile with the adverse metabolic effects of APs observed clinically. However, many in vitro studies have not been conducted under conditions of insulin stimulation, and thus fail to directly test effects of APs on insulin response. Insulin stimulation has been used when investigating AP effects in peripheral tissues, with variable results (Del Campo et al., 2018; Engl et al., 2005). in vivo rodent data on central insulin signalling is similarly difficult to interpret as most APs cause acute elevations in blood glucose and insulin (Boyda et al., 2010; Smith et al., 2014). Hyperglycemia and hyperinsulinemia has been shown to increase central AKT and GSK-3 phosphorylation, even in the face of insulin deficiency or disrupted insulin action (Clodfelder-Miller et al., 2005; Smith et al., 2014). Thus, it is difficult to determine if changes in activation of these proteins are due to the AP or secondary to the elevations in blood glucose. There is also the possibility that APs upregulate central insulin signalling but impair alternate energy sensing signals such as AMP-activated protein kinase (AMPK), which has been associated with AP-induced glucose dysregulation (Ikegami et al., 2013; Martins et al., 2010).

Alternatively, inflammation is also an area of interest as neuroinflammation is considered a causal factor of metabolic disease (Thaler and Schwartz, 2010). However, clinical studies evaluating AP effects on inflammation are highly inconclusive due to the many confounds which can influence inflammatory state and which are associated with the illness of schizophrenia (i.e. smoking, diet, obesity) (Tourjman et al., 2013). The heterogeneity among studies examining effects of APs on insulin and inflammatory pathways may also be attributable to a differential response of brain regions to AP exposure. Only a limited number of studies have examined APs effects on insulin signalling or energy sensing specifically in the hypothalamus (Kowalchuk et al., 2018), which is the primary brain region involved in whole body energy regulation. In addition, the hypothalamus is composed of highly heterogenous neuronal populations, and the limited in vivo papers available have examined the entire hypothalamus (Obuchowicz et al., 2006; Zhang et al., 2014). Thus, we do not know how APs may act on specific cell types. Finally, as these were whole-body experiments, we do not know if the effects seen are due to the direct impact of APs on the hypothalamus, or occur in response to feedback from other tissues signalling to this brain region.

In the present study, we set out to elucidate the direct effects of different APs (clozapine, olanzapine, aripiprazole) on insulin signalling, energy sensing and inflammation in the hypothalamus. In order to circumvent the confounding variables present in clinical studies, and to avoid changes in adiposity or acute hyperglycemia observed in rodents administered APs, we chose to employ an invitro hypothalamic cell model. We hypothesized that APs would differentially impair insulin signalling, upregulate AMPK, and induce inflammation, according to their clinical metabolic liability (clozapine = olanzapine > aripiprazole).

Section snippets

Cell culture and reagents

Rat hypothalamic neurons were immortalized using SV-40 T-antigen as previously described (Belsham et al., 2004; Gingerich et al., 2009). The immortalized rat hypothalamic embryonic neuronal cell line, rHypoE-19, was selected as it has been previously characterized to express the insulin receptor, essential components of the insulin signalling pathway, and cytokine receptors (Dhillon et al., 2012; Mayer and Belsham, 2009; Nazarians-Armavil et al., 2013). The cell line also expresses

AKT & GSK3β

Phosphorylation of AKT is a key step in the canonical insulin signalling pathway; GSK3β is downstream of AKT and phosphorylation inhibits GSK3β action (Clodfelder-Miller et al., 2005). To determine if APs impair insulin response, the rHypoE-19 cells were treated with one of the three APs, with and without insulin, for 15 min. As a positive control, insulin stimulation alone significantly increased pAKT protein expression compared to vehicle. Clozapine (100 μM) treatment significantly decreased

Discussion

This study provides the first evidence that antipsychotics (AP)s can directly impact insulin signalling, AMPK, MAPKs, and inflammatory mediators in hypothalamic neurons. Given that the hypothalamus is a key regulator of whole body energy homeostasis, direct action of APs on the hypothalamus could account in part for metabolic dysregulation, such as the insulin resistance and obesity, observed in patients taking APs. AP-induced hypothalamic dysregulation is also a concern as it has been proposed

Conclusions

This study is a first step towards disentangling the proposed association between AP-induced metabolic effects, therapeutic response, and schizophrenia (which may be associated with underlying disturbances in insulin signalling or inflammatory pathways). We have found that the APs olanzapine, clozapine, and aripiprazole can directly regulate insulin signalling, AMPK, MAPKs, and inflammatory mediators in rat hypothalamic neurons. These direct effects of APs on the hypothalamus could contribute

Funding

This work was supported by a University of Toronto, Department of Psychiatry Excellence Funds Grant, and a Banting and Best Diabetes Centre (BBDC) Reuben and Helene Dennis Diabetes Scholar Award awarded to MH. CK was supported by the BBDC Novo Nordisk Fellowship, a University of Toronto, Cleghorn Fellowship award, and a CAMH Discovery fund Award.

Conflicts of interest

MH received consultant fees from Alkermes. All other authors declare that they have no conflicts of interest.

Acknowledgements

The authors thank the members of the Belsham lab for their technical support and advice throughout the project. The authors also thank Dr. Gary Remington and Dr. Paul Fletcher for their advice throughout the project, as well as editing the final manuscript.

References (62)

  • M. Ikegami et al.

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

    Eur. J. Pharmacol.

    (2013)
  • M. Kluge et al.

    Effects of clozapine and olanzapine on cytokine systems are closely linked to weight gain and drug-induced fever

    Psychoneuroendocrinology

    (2009)
  • S. Kyosseva et al.

    Differential and region-specific activation of mitogen-activated protein kinases following chronic administration of phencyclidine in rat brain

    Neuropsychopharmacology

    (2001)
  • J.G. Lee et al.

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

    Prog. Neuro-Psychopharmacol. Biol. Psychiatry

    (2010)
  • J. Lian et al.

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

    Psychoneuroendocrinology

    (2014)
  • X.-H. Lu et al.

    Olanzapine produces trophic effects in vitro and stimulates phosphorylation of Akt/PKB, ERK1/2, and the mitogen-activated protein kinase p38

    Brain Res.

    (2004)
  • C.M. Mayer et al.

    Insulin directly regulates NPY and AgRP gene expression via the MAPK MEK/ERK signal transduction pathway in mHypoE-46 hypothalamic neurons

    Mol. Cell. Endocrinol.

    (2009)
  • S.A. McFadden et al.

    Glucose responsiveness in a novel adult-derived GnRH cell line, mHypoA-GnRH/GFP: involvement of AMP-activated protein kinase

    Mol. Cell. Endocrinol.

    (2013)
  • M. Nakata et al.

    AAV-mediated IL-10 gene transfer counteracts inflammation in the hypothalamic arcuate nucleus and obesity induced by high-fat diet

    Neuropeptides

    (2017)
  • C.D. Pandya et al.

    BDNF-TrkB signaling and neuroprotection in schizophrenia

    Asian J. Psychiatry

    (2013)
  • V. Parikh et al.

    Olanzapine counteracts reduction of brain-derived neurotrophic factor and TrkB receptors in rat hippocampus produced by haloperidol

    Neurosci. Lett.

    (2004)
  • V. Tourjman et al.

    Antipsychotics’ effects on blood levels of cytokines in schizophrenia: a meta-analysis

    Schizophr. Res.

    (2013)
  • E. Tsaousidou et al.

    Distinct roles for JNK and IKK activation in agouti-related peptide neurons in the development of obesity and insulin resistance

    Cell Rep.

    (2014)
  • M.H. Xiu et al.

    Decreased interleukin-10 serum levels in first-episode drug-naïve schizophrenia: relationship to psychopathology

    Schizophr. Res.

    (2014)
  • P. Yuan et al.

    Altered levels of extracellular signal-regulated kinase signaling proteins in postmortem frontal cortex of individuals with mood disorders and schizophrenia

    J. Affect. Disord.

    (2010)
  • G. Zhang et al.

    Sensitive liquid chromatography/tandem mass spectrometry method for the simultaneous determination of olanzapine, risperidone, 9-hydroxyrisperidone, clozapine, haloperidol and ziprasidone in rat brain tissue

    J. Chromatogr. B Analyt. Technol. Biomed. Life Sci.

    (2007)
  • R.D. Almeida et al.

    Neuroprotection by BDNF against glutamate-induced apoptotic cell death is mediated by ERK and PI3-kinase pathways

    Cell Death Differ.

    (2005)
  • M. Aravagiri et al.

    Pharmacokinetics and tissue distribution of olanzapine in rats

    Biopharm. Drug Dispos.

    (1999)
  • O. Bai et al.

    Expression of brain-derived neurotrophic factor mRNA in rat hippocampus after treatment with antipsychotic drugs

    J. Neurosci. Res.

    (2003)
  • M. Bak et al.

    Almost all antipsychotics result in weight gain: a meta-analysis

    PLoS One

    (2014)
  • B.F. Belgardt et al.

    Hypothalamic and pituitary c-jun N-terminal kinase 1 signaling coordinately regulates glucose metabolism

    PNAS

    (2010)
  • Cited by (34)

    • Serum hepcidin / ferroportin levels in bipolar disorder and schizophrenia

      2021, Journal of Trace Elements in Medicine and Biology
    • Association between the improvement in depressive symptoms and serum BDNF levels in drug-naive first episode patients with schizophrenia: A longitudinal follow-up

      2021, Psychoneuroendocrinology
      Citation Excerpt :

      In the present study, we found that OLA can increase BDNF levels after a 12-week treatment with OLA, which is consistent with several previous studies (Jena et al., 2019). For example, a study in hypothalamic rat neurons found that OLA administration upregulated the expression of pERK1/2 and pJNK, and BDNF levels (Kowalchuk et al., 2019). Also, increased BDNF upregulated the expression of pERK1/2 and pJNK and induced the neuronal survival signaling pathway via ERK1/2 activation.

    View all citing articles on Scopus
    1

    Shared senior authorship.

    View full text