Revision Antipsicoticos Alteraciones Metabolicas OJO Imagenes 2019

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

European Journal of Pharmacology 844 (2019) 231–240

Contents lists available at ScienceDirect

European Journal of Pharmacology


journal homepage: www.elsevier.com/locate/ejphar

Review

Antipsychotics-induced metabolic alterations: Recounting the mechanistic T


insights, therapeutic targets and pharmacological alternatives

Raghunath Singha, Yashika Bansala, Bikash Medhib, Anurag Kuhada,
a
Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh
160014, India
b
Department of Pharmacology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India

A R T I C LE I N FO A B S T R A C T

Keywords: Atypical antipsychotics (AAPs) are the drug of choice in the management of mental illnesses by virtue of their
Antipsychotics advantage over typical antipsychotics i.e. least tendency of producing extrapyramidal motor symptoms (EPS) or
Weight gain pseudoparkinsonism. Despite the clinical efficacy, AAPs produces troublesome adverse effects, particularly
Hyperphagia hyperphagia, hyperglycemia, dyslipidemia weight gain, diabetes mellitus, insulin resistance and QT prolonga-
Diabetes mellitus
tion which further develops metabolic and cardiac complications with subsequent reduction in life expectancy,
Neuropeptides
poor patient compliance, and sudden death. AAPs-induced weight gain and metabolic alterations are increasing
Uncoupling proteins
Gut-microbiota at an alarming rate and became an utmost matter of concern for psychopharmacotherapy. Diverse underlying
mechanisms have been explored such as the interaction of AAPs with neurotransmitter receptors, alteration in
food reward anticipation behavior, altered expressions of hypothalamic orexigenic and anorexigenic neuro-
peptides, histamine H1 receptor-mediated hypothalamic AMP-activated protein kinase (AMPK) activation, in-
creased blood leptin, ghrelin, pro-inflammatory cytokines. Antipsychotics induced imbalance in energy home-
ostasis, reduction in energy expenditure which is linked to altered expression of uncoupling proteins (UCP-1) in
brown adipose tissue and reduced hypothalamic orexin expressions are emerging insights. In addition, alteration
in gut-microbiota and subsequent inflammation, dyslipidemia, obesity, and diabetes after AAPs treatment are
also associated with weight gain and metabolic alterations. Oral hypoglycemics and lipid-lowering drugs are
mainly prescribed in the clinical management of weight gain associated with AAPs while many other phar-
macological and nonpharmacological interventions also have been explored in different clinical and preclinical
studies. In this review, we critically discuss the current scenario, mechanistic insights, biomarkers, and ther-
apeutic alternatives for metabolic alterations associated with antipsychotics.

1. Introduction antipsychotics (AAPs) or second-generation antipsychotics (SGA) were


originally defined as the drugs effective against positive symptoms of
The exponential surge in antipsychotic prescription is attributed to a schizophrenia without inducing EPS. AAPs have least or no tendency of
drastic increase in the prevalence of neuropsychiatric disorders producing EPS and also didn’t provoke catalepsy (Hippius, 1989). Hy-
worldwide (Gonçalves et al., 2015). Chlorpromazine was introduced as perprolactinemia which is result of D2 receptor blockade in anterior
the first antipsychotic drug in 1950s which has effectively cured the pituitary, was absent with AAPs except risperidone and its active me-
positive symptoms of schizophrenia. With the span of time, more potent tabolite paliperidone (Bostwick et al., 2009; Kusumi et al., 2015). Ad-
antipsychotics were added in this category and termed as ‘typical’ or ditionally, AAPs are effective in treatment of negative symptoms as well
first-generation antipsychotics (Farde et al., 1992). Typical anti- as cognitive impairment associated with schizophrenia (Leucht et al.,
psychotics pronouncedly produced extrapyramidal side-effects (EPSs, 2009; Woodward et al., 2005). By virtue of their clinical efficacy, AAPs
which includes tardive dyskinesia, akathisia, and pseudoparkinsonism) are drug of choice in the management of mental illnesses like schizo-
and hyperprolactinemia. These adverse events are consequences of ni- phrenia, bipolar disorder, psychotic depression, schizoaffective dis-
grostriatal dopaminergic (D2) receptor blockade (Klawans et al., 1980). orders and autism spectrum disorder, etc. (Lian et al., 2016a). Despite
In order to overcome those severe adverse effects, development of their strong clinical efficacy in controlling symptoms of the psychiatric
‘atypical’ antipsychotics took place in 1990s. However, atypical disorders, AAPs having a propensity of producing severe metabolic


Correspondence to: University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh 160014, India.
E-mail address: anurag_pu@yahoo.com (A. Kuhad).

https://doi.org/10.1016/j.ejphar.2018.12.003
Received 13 October 2018; Received in revised form 2 December 2018; Accepted 5 December 2018
Available online 07 December 2018
0014-2999/ © 2018 Published by Elsevier B.V.
R. Singh et al. European Journal of Pharmacology 844 (2019) 231–240

alterations like increased BMI, central obesity, hyperphagia, hypergly- (Panariello et al., 2011). Unlike other AAPs, ziprasidone and ar-
cemia, dyslipidemia, insulin resistance, and diabetes mellitus, which ipiprazole have less tendency to produce weight gain and metabolic
are collectively termed as metabolic syndrome (MetS) (Cuerda et al., syndrome due to their partial agonistic property on 5-HT2C receptors.
2014; Reynolds and Kirk, 2010). Among all, olanzapine and clozapine Olanzapine is an inverse agonist of 5-HT2C receptors and exhibits the
has highest likelihood of inducing weight gain which may be due to highest tendency to induce weight gain (Leucht et al., 2009) therefore,
their higher affinity to 5-HT2C and H1 receptors (Reynolds and Kirk, it is widely used to induce metabolic syndrome in the experimental
2010). animal.
Antipsychotic-induced weight gain (AIWG) and metabolic altera- The adrenergic system plays a very important role in metabolism
tions lead to cardiovascular complications, socio-occupational in- and energy balance. Adrenergic system stimulates glycogenolysis, glu-
abilities, decreased life expectancy and poor patient compliances. coneogenesis, and lipolysis which ultimately leads to hyperglycemia
Weight gain and MetS are observed as a severe side effect observed (Peterson et al., 1988). α-2 adrenoceptor present at pre-synaptic nerve
in > 40% of patients receiving AAPs (Lett et al., 2011; Mittal et al., endings are antagonized by several atypical antipsychotics (risperidone,
2017). Patients with the first episode of psychosis (FEP) are more sus- clozapine, aripiprazole) may plausible role in weight gain. α-2R also
ceptible to getting obese as compared to those who had previous ex- present on pancreatic β-cell inhibits insulin release once activated
posure to AAPs (Almandil et al., 2013). Recently, a study suggested that (Starrenburg and Bogers, 2009). Postural hypotension is another major
monitoring of weight change (1 kg or more of baseline weight) with side effect of risperidone and quetiapine which is because of α-1 re-
olanzapine treatment at week 2 can be useful in predicting weight gain ceptor antagonism (Haddad and Sharma, 2007). A higher incidence
at week 6 (Lin et al., 2018). The underlying mechanism of AIWG is not (> 8%) of QTc prolongation in patients receiving AAPs is a major in-
understood clearly as it is multifactorial. Several mechanistic insights sight for cardiovascular complications like arrhythmia and coronary
underlying AIWG have been extensively investigated like interaction of artery diseases (Alvarez and Pahissa, 2010). Additionally, AAPs an-
AAPs with neurotransmitters and their receptors, altered reward cir- tagonizes β-3 adrenoceptor (ADRB3), present mainly on adipocyte,
cuitry and food reward anticipation behaviors, alterations in expression pancreatic beta-cells and disrupt energy homeostasis. Physiologically,
of hypothalamic orexigenic/anorexigenic neuropeptides, gut and adi- ADRB3 activated by norepinephrine leads to lipolysis thus useful in
pose tissues driven peptides and hormones, pituitary hormones, weight control, maintaining energy homeostasis (Basile et al., 2001;
proinflammatory cytokines, energy balance, thermoregulation, and al- Starrenburg and Bogers, 2009). β-3 activation also reduces insulin-
teration in gut microbiota. In different sections of this review, we have mediated leptin release (Gettys et al., 1996) so their antagonism can be
discussed the updated mechanistic insights, biomarkers and pharma- correlated with hyperleptinemia associated with AAPs. Alongside the
cotherapeutic alternatives for antipsychotic-induced metabolic altera- sympathetic nervous system, interaction of AAPs with parasympathetic
tions. nervous system has been correlated with metabolic side effects.
Blockade of muscarinic cholinergic (mACh) receptors by antipsychotics
2. AAPs: neurotransmitter-receptor interactions lead to weight gain through anticholinergic effect (as shown in Fig. 2).
Clozapine has a variable effect on subtypes of muscarinic receptors. It
AAPs have heterogeneous receptor interaction which makes it dif- acts as a partial agonist (M1, M2, and M4) and antagonist (M3, M5) to
ficult to understand the mechanism behind metabolic alterations. muscarinic receptors whereas N-desmethylclozapine (an active meta-
Mesolimbic and mesocortical dopaminergic hyperactivity is a major bolite of clozapine) acts as a full agonist (M1), partial agonist (M5), and
pathophysiological aspect of schizophrenia. Based on pharmacody- also as an antagonist (M3) (Davies et al., 2005). Olanzapine and clo-
namics profiling of ‘typical’ antipsychotics, dopamine (D2) receptor is a zapine in low concentrations impair cholinergic-mediated insulin se-
major target of antipsychotic action and extrapyramidal motor symp- cretion by M3 receptors blockade in isolated rat islet cells (Coccurello
toms (EPS) (Kapur and Remington, 2001). Projections of dopaminergic and Moles, 2010). Atropine attenuated the acute insulin release after
neurons from ventral tegmental area (VTA) to the striatum are crucial olanzapine treatment which confirms the involvement of muscarinic
players in brain reward modulation and appetite regulation. CNS sti- receptor augmentation in olanzapine-induced hyperinsulinemia as a
mulants are found to decrease appetite leading to weight loss by de- result of glucose intolerance (Rickels et al., 2017). Olanzapine and
creasing dopamine (DA) turnover in striatal neurons. Conversely, pre- clozapine increase feed intake and weight gain which is associated with
clinical studies show that antipsychotics (amisulpride) with high D2 H1 antagonism mediated by activation of hypothalamic AMPK whereas,
receptor antagonism in the striatum lead to increased appetite, hyper- clozapine did not increase pAMPK in H1 knockout mice (Kim et al.,
phagia and weight gain (Nielsen et al., 2016). DA antagonists impair 2007). An H1 receptor agonist, 2-(3-trifluoromethylphenyl) has been
the reward circuitry leading to altered feeding, hyperphagia, and shown to inhibit hyperphagia-AMPK activation induced by olanzapine
weight gain. Functional magnetic resonance imaging (fMRI) studies in in a dose-dependent manner (He et al., 2014). There are several clinical
the brain of obese individuals showed reduced reward response in and preclinical studies on betahistine (a H1 agonist, and H3 antagonist)
dorsal striatum which corroborated in the schizophrenic patients re- showing its protective effects in AAPs-induced weight gain and meta-
ceiving acute or no antipsychotic treatment (Nielsen et al., 2012). Other bolic alterations. Betahistine co-treatment has been reported to de-
DAergic projections involved are dorsal striatal, cortical, limbic and crease feeding and increase thermogenesis in BAT by altering H1 re-
hypothalamic (Volkow et al., 2011). Hypothalamic DAergic neurons ceptor mediated neuropeptide-Y (NPY) and AMPKα pathways in the
express D1 and D2, which may be linked to metabolic conditions. Hy- hypothalamus and protects against olanzapine-induced dyslipidemia
pothalamic D1 are highly expressed in the ventromedial nucleus hy- through modulation of AMPKα-SREBP-1 (Sterol regulatory element
pothalamus (VMN) of obese rats whereas D2 found in lesser amount in binding protein-1)-PPARα-dependent pathways (Lian et al., 2016b,
VMN of lean rats (Fetissov et al., 2002). Second to the dopamine, 5HT 2014a, 2014b).
receptors (mainly 5-HT2A and 5-HT2C) are major sites for the anti- Furthermore, cannabinoid neurotransmission is also associated with
psychotic effect of AAPs and also for ‘atypicality’ (Kusumi et al., 2015; appetite regulation which is mainly due to the abundance of the re-
Meltzer, 2004). 5-HT2C antagonism is particularly related to hyper- ceptors in the brainstem. Cannabinoid receptor (CB1) downregulates
phagia and weight gain (as shown in Fig. 1). 5-HT2C antagonism by anorexigenic neuropeptides like cocaine and amphetamine-regulated
olanzapine has been reported to induce hyperphagia, glucose intoler- transcript (CART) in the Arc, corticotropin-releasing hormone (CRH) in
ance and altered energy homeostasis which were reversed by selective PVN, while upregulates orexigenic neuropeptide Y (NPY) and melanin-
5-HT2C agonist (lorcaserin) (Lord et al., 2017). Polymorphism in the 5- concentrating hormone (MCH) in the lateral hypothalamus (LH) (Cota
HT2C receptor promoter gene (-759C/T) has been associated with et al., 2003; Osei-Hyiaman et al., 2005). A gene polymorphism study
weight gain and elevated leptin levels after antipsychotics treatment showed that weight gain in individuals with chronic schizophrenia

232
R. Singh et al. European Journal of Pharmacology 844 (2019) 231–240

Fig. 1. Schematic representation of interaction of AAPs with major neurotransmitter-receptors and association with metabolic alterations. 5-HT-serotonin,
GABA-γ-aminobutyric acid, Glut- glutamate, CB- cannabinoids, ECs-endocannabinoids, NMDA- n-methyl D-aspartic acid, VGLUT- vesicular glutamate transporter,
CB1R- cannabinoid receptor 1, α-1& α-2- α-adrenergic receptor 1 & 2, M1, M2 & M3,- muscarinic receptor 1, 2 & 3, H1, H3-histamine 1 and 3 receptor, CVD-
cardiovascular diseases, MetS- Metabolic syndrome.

receiving AAPs has been associated with a polymorphism in CB1R gene found to increase hepatic glucose production by activation of hy-
(Tiwari et al., 2010). Chronic treatment in male rats with high-dose pothalamic AMP-activated protein kinase (Ikegami et al., 2013). In-
risperidone increases cannabinoid binding density of CB1 receptor creased BMI following AAPs treatment is positively correlated with
agonist [3H]CP-55940 in Arc while olanzapine-treated female rats increased levels of insulin, leptin, and HOMA-IR (Baeza et al., 2017).
showed decreased binding in DVC (Weston-Green et al., 2012, 2008). Hypoadiponectinemia associated with insulin resistance and increased
CB1R antagonist (AVE1625) has shown to be useful in treating cogni- leptin levels have been reported in schizophrenia patients receiving
tive impairments associated with schizophrenia additionally reduced AAPs treatment (Sapra et al., 2016). Additionally, chronic olanzapine
olanzapine-induced weight gain in mice (Black et al., 2011). Hence, treatment has been found to increase serum glucagon levels in human
targeting CB receptors could be another therapeutic approach in the (Teff et al., 2013) as well as in rats (Smith et al., 2011). A single in-
management of weight gain associated with antipsychotics. jection of olanzapine in rats failed to increase glucagon levels (Nagata
et al., 2016) while clozapine did (Jassim et al., 2012). Recently, glu-
cagon receptor knockout mice (Gcgr(-/-)) were found to be protected
3. AAPs: altered lipid and carbohydrate homeostasis against hyperglycemia induced by acute olanzapine treatment which
was associated with reduced liver glucose production (Castellani et al.,
Schizophrenia patients are at increased risk of developing dyslipi- 2017). GLP-1 (Glucagon-like peptide-1) agonist (exenatide) given in
demia due to genetic inclination (Gough and O’Donovan, 2005) along adjunctive with AAPs reduced weight gain and delayed development of
with dietary habits and AAPs treatment. Clinical studies have reported diabetes (Ishøy et al., 2014). Glucose homeostasis alterations need to be
dyslipidemia following AAPs treatment in patients with schizophrenia monitored with ongoing AAPs treatment, early monitoring of glucose
(Mackin et al., 2007). Olanzapine has been reported to have the highest levels can prevent or reduce chances of developing T2DM, insulin re-
likelihood of producing dyslipidemia (Takeuchi et al., 2015). AAPs sistance, cardiovascular complications and development of MetS. A
treatment leads to insulin resistance followed by increased synthesis of possible correlation between altered glucose homeostasis and AAPs has
fatty acids and triglycerides from liver and causes hypertriglyceridemia. been depicted in Fig. 2.
AAPs treatment increases the synthesis of free fatty acids, phospholipid,
triacylglycerides in rodents and human hepatocytes (Canfrán-Duque
et al., 2013). Further, schizophrenic patients are susceptible to diabetes 4. AAPs: folate and one carbon metabolism
mellitus and AAPs play a synergistic role in this context (Lindenmayer
et al., 2001). AAPs increases fasting glucose in patients which leads to Folate, a cofactor for enzymes involved in DNA and RNA bio-
glucose intolerance (Fernandez-Egea et al., 2009) and ultimately im- synthesis and donor of methyl groups to the methylation cycle for the
pairs glucose homeostasis. Altered glucose homeostasis developed after synthesis of homocysteine (Hcy) using methionine. N5-methyltetrahy-
certain hours-days of AAPs treatment which is independent of weight drofolate (MTHF) donates its methyl group to vitamin B12-dependent
gain in both rodents and human (Albaugh et al., 2011). Olanzapine enzyme (methionine synthase) which recycles Hcy back to methionine

233
R. Singh et al. European Journal of Pharmacology 844 (2019) 231–240

Fig. 2. Schematic representation of antipsychotic drugs-induced metabolic alteration. Effects of AAPs on major organ/systems, alteration in biomarkers
(Peptides, hormones, adipokines, cytokines) and corresponding metabolic alterations leading to weight gain and MetS associated with AAPs treatment. GH-growth
hormone, TSH-thyroid stimulating hormone, MSH-melanocyte-stimulating hormone, T3-triiodothyronine, T4-thyroxine, UCP-uncoupling proteins, BAT-brown
adipose tissue, WAT-white adipose tissue, NPY-neuropeptide-Y, AgRP-agouty related peptide, POMC- pro-opiomelanocortin, CART-cocaine-and amphetamine-
regulated transcript, MCH-melanin-concentrating hormone, GLP-1-glucagon like peptide, MetS-metabolic syndrome.

(Scott et al., 1994). Hyperhomocysteinemia has been associated with 5. AAPs: adipokines and cytokines
risk of schizophrenia (del Rosario García-Miss et al., 2010). Many re-
ports suggest disrupted one-carbon metabolism (OCM) and hyperho- Adiponectin is an adipocyte-derived hormone that is secreted by
mocysteinemia together with depleted levels of folate and vitamin B12 mature adipocytes. It controls the physiological pathways of carbohy-
levels among FEP patients (Ayesa-Arriola et al., 2012). Mechanisms drate and lipid metabolism and has anti-inflammatory, insulin sensi-
depicting associations between OCM alterations and schizophrenia may tizer and anti-atherogenic properties to mediate various vascular pro-
be due to the interaction of Hcy with NMDA receptors. OCM alterations cesses (Trujillo and Scherer, 2005). Adiponectin inhibits local pro-
may lead to anomalous DNA methylation of genes responsible for inflammatory cytokines production regulated via activation of toll-like
neurodevelopment and neurotransmission has been associated with receptors (TLRs) (Neumeier et al., 2006) and inhibition of nuclear
schizophrenia pathophysiology (Misiak et al., 2013). Association of factor-κB (NF-κB) (Lira et al., 2012; Robinson et al., 2011). Reduced
MetS and AAPs with MTHFR gene polymorphisms have been frequently adiponectin levels have been reported in obese patients and also in
studied (Roffeei et al., 2014). MTHFR C677T polymorphism found to be high-fat diet fed rats which subsequently associated with the develop-
associated with AIWG in patients with schizophrenia (Srisawat et al., ment of insulin resistance (Yamauchi et al., 2001). Reduced adipokines
2014). Hcy is known in advancing of schizophrenia following several and metabolic anomalies are found to be associated with AAPs treat-
mechanisms especially for negative symptoms and cognitive impair- ment (Gonçalves et al., 2015). Meta-analyses clearly conclude the
ment with schizophrenia (CIAS). A study reported that elevated serum correlation of hypoadiponectinemia and AAPs, particularly olanzapine
Hcy and triglyceride and lower folate and HDL levels in FES patients and clozapine, but not with quetiapine and risperidone (Bartoli et al.,
than in healthy controls. Higher Hcy and lower B12 in patients with 2015a, 2015b). Further, leptin is another adipokine, synthesized in
metabolic syndrome were found to be associated with negative symp- white adipose tissue (WAT), possesses cytokine-like properties and
toms and CIAS (Baeza et al., 2017; Misiak et al., 2016). Folate sup- anorexigenic in nature. Elevated leptin levels have been observed in
plementation has been stated to reduce AAP-associated metabolic risks schizophrenic patients treated with AAPs and its associated with BMI
and also to reduce the risk of endothelial dysfunction (Ellingrod et al., might be a result of a negative feedback mechanism to compensate
2015). Moreover, hyperhomocysteinemia and disrupted folate meta- increased fat mass (Potvin et al., 2015). Hyperleptinemia associated
bolism could be a biomarker for severity of negative symptoms and with adipogenecity is often complemented by hypoadiponectinemia.
cognitive impairment associated with schizophrenia and further de- These alterations are believed to be a predisposing factor for metabolic
velopment of metabolic alterations. anomalies and an elevated inflammatory state. Risperidone treatment
in children with autism has been found to increase blood levels of
glucose, insulin, prolactin, leptin, and HOMA-IR significantly

234
R. Singh et al. European Journal of Pharmacology 844 (2019) 231–240

(Srisawasdi et al., 2017). Recently, a study suggested that leptin/adi- feeding area (lateral and dorsomedial hypothalamus and perifornical
ponectin ratio could be a preferential indicator of AIWG and MetS area) have abundance of orexins (hypocretins) and their receptors
(Chen et al., 2018). (orexin 1 and 2, OX1R and OX2R). The orexins are regulators of
Altered levels of pro-inflammatory cytokines are associated with feeding, arousal, reward and implicated extensively for alcohol and
schizophrenia (Martínez-Gras et al., 2012). Earlier, Miller et al. have opioid-seeking (Baimel et al., 2015). Orexin fibers co-expressed with
carried out a meta-analysis reporting the correlation of altered cyto- dopaminergic neurons medial pre-frontal cortex (mPFC) and the medial
kines levels and antipsychotic treatment. They have reported that first shell of the NAc, which suggests its link with dopamine and reward.
episode of schizophrenia (FEP) and acutely relapsed inpatients showed AAPs found to activate hypothalamic orexin neurons, which activate
an increase in interleukin (IL-1 and IL-6), and transforming growth dopamine neurons in VTA leading to increase dopamine levels in PFC
factor-β (TGF-β) levels which were normalized with antipsychotic and striatum, which was found to be associated with weight gain (Fadel
treatment. Conversely, IL-12, interferon-γ (IFN-γ), tumor necrosis et al., 2002). Activation of dopaminergic neurons by activated orexins
factor-α (TNF-α) and IL-2 receptor appeared as trait markers because neurons in PFC by AAPs has been implicated for antipsychotic action
their levels were not normalized with antipsychotic treatment whereas (Chien et al., 2015). Rats treated with olanzapine showed activated
unaffected IL-6 level and decreased the level of IL-1β were found in orexins positive neurons in the PFC and perifornical region of LH which
schizophrenia patients (Miller et al., 2011). Further, in another meta- is associated with weight gain (Stefanidis et al., 2009). Quetiapine,
analysis, Capuzzi et al. have shown a decrease in IL-2 and IL-6 levels haloperidol, and clozapine lowers sympathetic hyperthermic reactions
after four weeks of APD treatment. IL-2, IL-6, and IL-1β were suggested in rats treated with a cerebral injection of orexin A. Risperidone in-
as state markers whilst TNF-α, IFN-γ and IL-17 were considered as trait creases the hyperthermic sympathetic reaction in rats, this explains the
markers (Capuzzi et al., 2017). Haloperidol and risperidone increased effect of AAPs on thermoregulation and weight gain (Monda et al.,
interleukin (IL-10, which is an anti-inflammatory cytokine) levels while 2006). Hence, modulators of these neuropeptides, hormones and their
no significant change was seen in levels of tumor necrosis factor-α receptors could be of great interest in management of obesity and
(TNF-α) and IL-1β (Obuchowicz et al., 2017). A seven-month follow-up metabolic alterations associated with AAPs treatment. Association of
study showed increased levels of C-peptide, leptin and decreased levels these neuropeptides- hormones with AIWG has been illustrated in
of adiponectin which were found to be associated with increased BMI Figs. 1 and 2).
and APD treatment (Balõtšev et al., 2017). AAPs induces activation of
NF-κB and it's target genes which transcripts for TNF-α, IL-1β, IL-8 and 7. AAPs: altered gut-microbiota
MCP-1 in human and rodent's adipocytes (Sárvári et al., 2014). In a
nutshell, it could be said that APD treatment alters the levels if adipo- Altered gut-brain axis has been implicated in neuropsychiatric dis-
kines and cytokines which further involve in development of metabolic orders like schizophrenia and depression and also with weight gain
alteration. The possible interaction between adipokines-cytokines and associated with AAPs. Microbial colonization has been found to reverse
AAPs have been illustrated in Fig. 2. altered behavior and synaptic plasticity in mice. Gut microbiome has a
direct correlation with adaptive immune response and inflammation
6. AAPs: appetite-regulating neuropeptides and hormones which are closely associated with the pathogenesis of schizophrenia.
Gut-dysbiosis leads to obesity and metabolic alterations which are
Central neuroendocrine system is the controller of appetite, energy common with schizophrenia (Nemani et al., 2015). The negative impact
intake, and expenditure. Arc controls feeding by two interconnected of AAPs on gut microbiota has been correlated with weight gain (Kanji
groups of orexigenic (NPY/AgRP) and anorexigenic (POMC/CART) et al., 2018). Olanzapine-induced weight gain in germ-free mouse
neuronal circuits. These neuronal circuits control secretion, metabo- suggests a plausible role of gut microbiota in weight gain which is
lism, and action of hormones which regulate metabolism in the body possible because of its antimicrobial potential to residential enteric
(Palou et al., 2009; Trayhurn and Bing, 2006). The orexigenic, NPY/ bacteria (Morgan et al., 2014). Chronic olanzapine treatment has shown
AgRP neurons group corresponds to the feeling of hunger along with significant weight gain in female rats and also showed elevated plasma
some co-expressed orexins receptors (OX1R and OX2R). OXR are co- proinflammatory cytokines (IL-8, and IL-1β) and gut-dysbiosis was
expressed in GABAergic neurons which makes them inhibitory factors found to be positively association (Davey et al., 2012). Children re-
for a network of hypothalamic neurons (Abizaid and Horvath, 2008). ceiving risperidone for 12 months showed an increase in BMI and a
On another hand, the anorexigenic group of neurons, consist of the reduced ratio of Bacteroidetes: Firmicutes as compared with AAPs naïve
CART, POMC, α-MSH, which are satitation factors and inhibits food patients (Bahr et al., 2015). Olanzapine-induced increased body weight,
intake (Kohno and Yada, 2012). Histamine receptor (H1 and H2) are fat deposition, and increased plasma free fatty acids have been reversed
expressed in PVN and VMH and produce the anorexic effect. Olanza- by co-administration of antibiotic cocktail (neomycin, metronidazole,
pine and clozapine are a blocker of histaminergic receptors and pro- polymyxin B) in rats, which affirms the role of gut microbiota in AIWG
duces hyperphagia. Histaminergic transmission can be enhanced by (Davey et al., 2013). A recent study reported that adjunction of the
blocking of presynaptic H3 receptor and H3 antagonists could be tar- prebiotic formulation (BGOS) with olanzapine has reduced the risk of
geted for anorexigenic their potential (Deng et al., 2010; Rojczyk et al., metabolic alteration and improve cognitive function in rats (Kao et al.,
2015). Further, ghrelin is an orexigenic hormone mainly secreted in the 2018). Along with other novel insights, altered gut microbiota with
stomach (Nakazato et al., 2001) binds to the ghrelin receptor (GHS- AAPs treatment could be of ample interest. Maintaining the gut-mi-
R1a) which is highly expressed in the Arc and regulates food intake and crobiota with probiotics and probiotics could be of great advantage in
energy expenditure (Andrews, 2011). In the hypothalamic networks order to enhance the safety of psychophramacotherapy.
(Arc), ghrelin found to upregulate orexigenic neuropeptide (NPY and
AgRP) whereas downregulate anorexigenic peptides (POMC and CART) 8. AAPs: energy homeostasis
expressions, which can directly contribute to the orexigenic effect.
Olanzapine and clozapine increases ghrelin release as well it's receptors Energy (calorie) intake, expenditure, and storage are major com-
expression which clearly indicate hyperphagia and weight gain ponents of energy balance. Body weight changes only when there is an
(Albaugh et al., 2006; Murashita et al., 2007; Van Der Zwaal et al., imbalance in energy consumption and expenditure i.e. higher intake
2012; Zhang et al., 2014). Hence, AAPs-induced upregulates expression and lesser expenditure. Obesity could be considered as consequences of
of ghrelin and its receptors upregulate hypothalamic orexigenic neu- high intake and less expenditure of energy (Hill et al., 2012). There are
ropeptides and down-regulates anorexigenic neuropeptides. several factors that regulate food intake and energy expenditure are
In addition to these neuropeptides and ghrelin, hypothalamic discussed well in neuropeptide section. Melanocortin, leptin, orexin,

235
R. Singh et al. European Journal of Pharmacology 844 (2019) 231–240

NPY/AgRP, ghrelin, endocannabinoids, sympathetic nervous system, Additional to all other mechanistic insights discussed in this review,
histamine, dopamine, acetylcholine, pituitary hormones, and many alteration in reward anticipation could be an alternative mechanism in
others, regulates food intake and energy balance in either way. Central weight gain associated with AAPs.
control of food intake also involved in reward circuitry and cognition
which gives a neuronal basis for food intake regulation. Palatable food, 10. Management of AIWG
spices, triggers reward signals and stimulates the mesolimbic system
(Liu et al., 2014). Rats treated with risperidone showed a reduction in Many non-pharmacological approaches have been implicated in
body temperature without weight gain (Ota et al., 2005). In humans, management of AIWG. Behavioral interventions have found to be ef-
risperidone has conflicting results on body temperature, in some cases fective in psychiatric patients on APDs and development of metabolic
extremely high fever and muscle rigidity was observed. Mice treated syndrome. Caregivers need to take care of patients, record their daily
with risperidone shows weight gain associated with reduced locomo- food intake, exercise and medication intake. Controlled dietary intake
tion and hyperphagia along with higher levels of uncoupling protein (dietary intervention), regular psychoeducational sessions and regular
(UCP-1) in the BAT (Li et al., 2013). UCP1 and UCP2 regulates adaptive exercise could prevent and/or reduce the incidences of weight gain,
thermogenesis and have been investigated for their possible roles in diabetes, and CVDs (Amiaz et al., 2016). A recent study reported that
body temperature regulation and energy balance and associated with lifestyle modification along with metformin treatment can significantly
AIWG (Cope et al., 2009; Stefanidis et al., 2009). Mice treated with reduce the risk of weight gain and metabolic risk associated with AAPs
risperidone shows weight gain as a consequence of energy imbalance, (Zheng et al., 2018).
while hyperthermia is due to elevated UCP1 in BAT, UCP3 in gastro- Pharmacological interventions used clinically in management of
cnemius, depleted orexin expression in the hypothalamus (Cope et al., antipsychotics-induced metabolic alterations are mainly oral hypogly-
2009). Additionally, reduced BDNF levels in hypothalamus and serum cemic and antihyperlipidemic drugs. Metformin, a safest oral hy-
are associated with AIWG. Polymorphism in the BDNF Val66Met gene poglycemic drug, which has been extensively studied and is used
has been positively correlated with AIWG (Fonseka et al., 2015). BDNF clinically for this purpose. It increases peripheral utilization of glucose,
after sympathetic stimulation increases mRNA expression of UCP1 inhibits hepatic glucose production and prevents the development of
(Tsuchida et al., 2001). Hence, the role of AAPs in thermoregulation, insulin resistance. It can reduce prevent and reduces weight gain in
energy expenditure, altered BDNF levels can be assimilated hyper- psychiatric patients receiving AAPs (Arman et al., 2008; Klein et al.,
phagia and weight gain. 2006; Lee and Jeong, 2011). However, a study showed no improvement
in olanzapine-induced weight gain but some lipid parameters were re-
9. AAPs: reward system modulation versed (Baptista et al., 2006). Early metformin prescribing with AAPs
has been found preventive against AIWG (Goltz and Rice, 2017;
The DAergic neurons projecting from VTA to striatum are crucial Hendrick et al., 2017). Antidiabetic and antihyperlipidemics are the
players in the brain reward system which controls appetite. Drugs in- drug of choice in the management of AIWG (Varghese et al., 2016).
creasing DAergic turnover in the striatum, reduces body weight by Statins prescribed in patients with severe mental illness has been found
decreasing appetite whereas animal studies show amisulpride with high to control lipid profile but failed to protect against cardiovascular
D2 receptor affinity in the striatum lead to increased appetite, hyper- complications (Blackburn et al., 2017). H1 agonist (betahistine) is the
phagia and weight gain (Nielsen et al., 2016). Obesity and weight most explored drug against AIWG in clinical trials, and many studies
dysregulations are most likely comorbidities with psychopathology and are reported its clinical efficacy in the management of the problem
are persistent with the severity of the psychiatric illnesses and showed (Poyurovsky et al., 2013, 2005). Along with the above, following
considerable overlapping substance abuse and reward system. Altered pharmacological interventions have also been tested clinically for
reward anticipation in psychiatric illnesses has been implicated in in- AIWG (Mizuno et al., 2014), amantadine (Deberdt et al., 2005; Graham
creased food intake, weight gain, and susceptibility towards substance et al., 2005), aripiprazole (Fan et al., 2013), atomoxetine (Ball et al.,
abuse and addiction. Striatal DAergic neurotransmission is a major site 2011), D-fenfluramine (Goodall et al., 1988), famotidine (Poyurovsky
for regulation of eating behavior, motivation, reinforcement, and re- et al., 2004), fluoxetine (Bustillo et al., 2003), sibutramine (Biedermann
ward anticipation (Grimm et al., 2017). The reward deficiency hy- et al., 2014), metformin-sibutramine combination (Baptista et al.,
pothesis suggested reduced DA activity impairs reward circuitry and 2008), modafinil (Sudhakar et al., 2008), nizatidine (Assunção et al.,
lead to abnormal craving/eating behavior, and obesity (Blum et al., 2006), orlistat (Joffe et al., 2008), phenylpropanolamine (White et al.,
2014). Volkow et al. unveiled the role of dopamine in food reward, 2002), reboxetine (Poyurovsky et al., 2007), betahistine-reboxetine
reinforcement, and obesity and also reported associations of striatal D2 combination (Poyurovsky et al., 2013), rosiglitazone (Baptista et al.,
receptors with prefrontal metabolism and overeating in obese patients. 2009), topiramate (Afshar et al., 2009), zonisamide (Ghanizadeh et al.,
Reduction in striatal D2 receptors could contribute to overeating via 2013), exenatide (Ishøy et al., 2014), naltrexone (Tek et al., 2014) and
their modulation of striatal prefrontal pathways (Volkow et al., 2011). α-lipoic acid (Kim et al., 2016). Most recently, melatonin found to re-
PET studies in human proved the role of DA in obesity. A reduced duce AIWG in adolescent patients with bipolar disorder (Mostafavi
striatal D2 density was found in obese patients (Wang et al., 2001). This et al., 2017), bergamot-derived polyphenolic fraction in low dose im-
indicates, overlapping in the neurobiology of obesity and addiction proved metabolic parameters after use of AAPs (Bruno et al., 2017). The
disorders. Additionally, chronic consumption of high-fat (HFD) and focus of current pharmacotherapy of neuropsychiatric disorders should
high-sugar diet found to down-regulate DA receptors which shows be on maximizing the efficacy and minimizing the development of
phenotype like of chronic drugs abuse related increased dopaminergic adverse effects especially metabolic alterations.
signaling (Val-Laillet et al., 2015).
Almost all antipsychotics produce weight gain which is because of 11. Conclusion
their D2 receptors blockade and inhibition of phasic dopaminergic ef-
fect in the striatum and other areas (Bak et al., 2014). AAPs leads to the The dramatic surge in neuropsychiatric illness has significantly in-
reduced motivation for physical activity and increased motivation for creased antipsychotic prescription. With higher clinical efficacy as
feeding rich in calories. A longitudinal study highlighted the correlation compared to typical antipsychotics and less tendency to induce EPS
between AAPs treatment, weight gain, and striatal activity. Patient re- makes them first-line drugs in the management of schizophrenia, bi-
ceiving amisulpride for more than six weeks showed an association of polar, psychotic depression, autism, and other neuropsychiatric dis-
weight gain with increased putamen activity and reward anticipatory orders. Besides the advantages, AAPs induce metabolic abnormalities
response in monetary incentive delay task (Nielsen et al., 2016). which are of utmost concern for current psychopharamcotherapy.

236
R. Singh et al. European Journal of Pharmacology 844 (2019) 231–240

Mechanistic insight includes alterations in hypothalamic neuropep- other addictions. Br. J. Pharmacol. 172, 334–348.
tides, striatal and mesolimbic dopamine reward pathways, gut hor- Bak, M., Fransen, A., Janssen, J., van Os, J., Drukker, M., 2014. Almost all antipsychotics
result in weight gain: a meta-analysis. PLoS One 9, e94112.
mones, peptides, pituitary hormones, elevated proinflammatory cyto- Ball, M., Warren, K., Feldman, S., McMahon, R., Kelly, D., Buchanan, R., 2011. Placebo-
kine, altered energy homeostasis, and gut-dysbiosis are involve in controlled trial of atomoxetine for weight reduction in people with schizophrenia
AAPs-induced MetS. The focus should be given to the development of treated with clozapine or olanzapine. Clin. Schizophr. Relat. Psychoses 5, 17–25.
Balõtšev, R., Haring, L., Koido, K., Leping, V., Kriisa, K., Zilmer, M., Vasar, V., Piir, A.,
antipsychotics with partial agonistic effect on the 5-HT2C receptor or Lang, A., Vasar, E., 2017. Antipsychotic treatment is associated with inflammatory
unlike existing AAPs, the newer one should have minimal effect on and metabolic biomarkers alterations among first-episode psychosis patients: a 7-
other neurotransmitter systems. Pre/probiotics need to be incorporated month follow-up study. Early Interv. Psychiatry. https://doi.org/10.1111/eip.12457.
Baptista, T., Martínez, J., Lacruz, A., Rangel, N., Beaulieu, S., Serrano, A., Arapé, Y.,
into the diet of the patient while taking AAPs, which can prevent the Martinez, M., de Mendoza, S., Teneud, L., 2006. Metformin for prevention of weight
gut dysbiosis and could prevent the progression of MetS. Monitoring of gain and insulin resistance with olanzapine: a double-blind placebo-controlled trial.
biomarkers discussed here need to be given more emphasis while an- Can. J. Psychiatry 51, 192–196.
Baptista, T., Rangel, N., El Fakih, Y., Uzcategui, E., Galeazzi, T., Beaulieu, S., De Baptista,
tipsychotic therapy is ongoing. Early diagnosis could be preventive and
E.A., 2009. Rosiglitazone in the assistance of metabolic control during olanzapine
can delay or restrict the development of MetS in a patient receiving administration in schizophrenia: a pilot double-blind, placebo-controlled, 12-week
AAPs. trial. Pharmacopsychiatry 42, 14–19.
Baptista, T., Uzcátegui, E., Rangel, N., El Fakih, Y., Galeazzi, T., Beaulieu, S., de Baptista,
E.A., 2008. Metformin plus sibutramine for olanzapine-associated weight gain and
Acknowledgments metabolic dysfunction in schizophrenia: a 12-week double-blind, placebo-controlled
pilot study. Psychiatry Res. 159, 250–253.
Financial support from Indian Council of medical research (ICMR)/ Bartoli, F., Crocamo, C., Clerici, M., Carrà, G., 2015a. Second-generation antipsychotics
and adiponectin levels in schizophrenia: a comparative meta-analysis. Eur.
Department of health research (DHR), New Delhi, India is gratefully Neuropsychopharmacol. 25, 1767–1774. https://doi.org/10.1016/j.euroneuro.2015.
acknowledged for research grant awarded to Dr Anurag Kuhad under 06.011.
the grant number GIA/59/2014-DHR. We gratefully acknowledge re- Bartoli, F., Lax, A., Crocamo, C., Clerici, M., Carrà, G., 2015b. Plasma adiponectin levels
in schizophrenia and role of second-generation antipsychotics: a meta-analysis.
search grant sanctioned by SERB, DST, and UGC New Delhi. Council of Psychoneuroendocrinology. https://doi.org/10.1016/j.psyneuen.2015.03.012.
Scientific and Industrial Research (CSIR), New Delhi, India is gratefully Basile, V.S., Masellis, M., McIntyre, R.S., Meltzer, H.Y., Lieberman, J.A., Kennedy, J.L.,
acknowledged for awarding senior research fellowship to Mr 2001. Genetic dissection of atypical antipsychotic-induced weight gain: novel pre-
liminary data on the pharmacogenetic puzzle. J. Clin. Psychiatry 62, 45–66.
Raghunath Singh. Biedermann, F., Fleischhacker, W.W., Kemmler, G., Ebenbichler, C.F., Lechleitner, M.,
Hofer, A., 2014. Sibutramine in the treatment of antipsychotic-induced weight gain: a
Conflict of interest pilot study in patients with schizophrenia. Int. Clin. Psychopharmacol. 29, 181–184.
Black, M.D., Stevens, R.J., Rogacki, N., Featherstone, R.E., Senyah, Y., Giardino, O.,
Borowsky, B., Stemmelin, J., Cohen, C., Pichat, P., Arad, M., Barak, S., De Levie, A.,
Authors have none to declare. Weiner, I., Griebel, G., Varty, G.B., 2011. AVE1625, a cannabinoid CB1 receptor
antagonist, as a co-treatment with antipsychotics for schizophrenia: improvement in
References cognitive function and reduction of antipsychotic-side effects in rodents.
Psychopharmacology 215, 149–163. https://doi.org/10.1007/s00213-010-2124-0.
Blackburn, R., Osborn, D., Walters, K., Falcaro, M., Nazareth, I., Petersen, I., 2017. Statin
Abizaid, A., Horvath, T.L., 2008. Brain circuits regulating energy homeostasis. Regul. prescribing for people with severe mental illnesses: a staggered cohort study of ‘real-
Pept. 149, 3–10. world’impacts. BMJ Open 7, e013154.
Afshar, H., Roohafza, H., Mousavi, G., Golchin, S., Toghianifar, N., Sadeghi, M., Talaei, Blum, K., Thanos, P.K., Gold, M.S., 2014. Dopamine and glucose, obesity, and reward
M., 2009. Topiramate add-on treatment in schizophrenia: a randomised, double- deficiency syndrome. Front. Psychol. 5.
blind, placebo-controlled clinical trial. J. Psychopharmacol. 23, 157–162. Bostwick, J.R., Guthrie, S.K., Ellingrod, V.L., 2009. Antipsychotic‐induced hyperpro-
Albaugh, V.L., Henry, C.R., Bello, N.T., Hajnal, A., Lynch, S.L., Halle, B., Lynch, C.J., lactinemia. Pharmacother. J. Hum. Pharmacol. Drug Ther. 29, 64–73.
2006. Hormonal and metabolic effects of olanzapine and clozapine related to body Bruno, A., Pandolfo, G., Crucitti, M., Cacciola, M., Santoro, V., Spina, E., Zoccali, R.A.,
weight in rodents. Obesity 14, 36–51. Muscatello, M.R.A., 2017. Low-dose of bergamot-derived polyphenolic fraction (BPF)
Albaugh, V.L., Singareddy, R., Mauger, D., Lynch, C.J., 2011. A double blind, placebo- did not improve metabolic parameters in second generation antipsychotics-treated
controlled, randomized crossover study of the acute metabolic effects of olanzapine patients: results from a 60-days open-label study. Front. Pharmacol. 8.
in healthy volunteers. PLoS One 6, e22662. Bustillo, J.R., Lauriello, J., Parker, K., Hammond, R., Rowland, L., Bogenschutz, M.,
Almandil, N.B., Liu, Y., Murray, M.L., Besag, F.M.C., Aitchison, K.J., Wong, I.C.K., 2013. Samuel, K., 2003. Treatment of weight gain with fluoxetine in olanzapine-treated
Weight gain and other metabolic adverse effects associated with atypical anti- schizophrenic outpatients. Neuropsychopharmacology 28, 527.
psychotic treatment of children and adolescents: a systematic review and meta- Canfrán-Duque, A., Casado, M.E., Pastor, O., Sánchez-Wandelmer, J., de la Peña, G.,
analysis. Pediatr. Drugs 15, 139–150. Lerma, M., Mariscal, P., Bracher, F., Lasunción, M.A., Busto, R., 2013. Atypical an-
Alvarez, P.A., Pahissa, J., 2010. QT alterations in psychopharmacology: proven candi- tipsychotics alter cholesterol and fatty acid metabolism in vitro. J. Lipid Res. 54,
dates and suspects. Curr. Drug Saf. 5, 97–104. 310–324. https://doi.org/10.1194/jlr.M026948.
Amiaz, R., Rubinstein, K., Czerniak, E., Karni, Y., Weiser, M., 2016. A diet and fitness Capuzzi, E., Bartoli, F., Crocamo, C., Clerici, M., Carrà, G., 2017. Acute variations of
program similarly affects weight reduction in schizophrenia patients treated with cytokine levels after antipsychotic treatment in drug-naïve subjects with a first-epi-
typical or atypical medications. Pharmacopsychiatry 26, 112–116. sode psychosis: a meta-analysis. Neurosci. Biobehav. Rev. https://doi.org/10.1016/j.
Andrews, Z.B., 2011. The extra-hypothalamic actions of ghrelin on neuronal function. neubiorev.2017.03.003.
Trends Neurosci. 34, 31–40. Castellani, L.N., Peppler, W.T., Sutton, C.D., Whitfield, J., Charron, M.J., Wright, D.C.,
Arman, S., Sadramely, M.R., Nadi, M., Koleini, N., 2008. A randomized, double-blind, 2017. Glucagon receptor knockout mice are protected against acute olanzapine-in-
placebo-controlled trial of metformin treatment for weight gain associated with in- duced hyperglycemia. Psychoneuroendocrinology 82, 38–45. https://doi.org/10.
itiation of risperidone in children and adolescents. Saudi Med. J. 29, 1130–1134. 1016/j.psyneuen.2017.05.005.
Assunção, S.S.M., Ruschel, S.I., Rosa, L., de, C.R., Campos, J.A.O., Alves, M.J.O., Bracco, Chen, V.C.-H., Chen, C.-H., Chiu, Y.-H., Lin, T.-Y., Li, F.-C., Lu, M.-L., 2018. Leptin/
O.L., Lima, M.S. de, 2006. Weight gain management in patients with schizophrenia Adiponectin ratio as a potential biomarker for metabolic syndrome in patients with
during treatment with olanzapine in association with nizatidine. Rev. Bras. Psiquiatr. schizophrenia. Psychoneuroendocrinology 92, 34–40.
28, 270–276. Chien, Y.-L., Liu, C.-M., Shan, J.-C., Lee, H.-J., Hsieh, M.H., Hwu, H.-G., Chiou, L.-C.,
Ayesa-Arriola, R., Pérez-Iglesias, R., Rodríguez-Sánchez, J.M., Mata, I., Gómez-Ruiz, E., 2015. Elevated plasma orexin A levels in a subgroup of patients with schizophrenia
García-Unzueta, M., Martínez-García, O., Tabares-Seisdedos, R., Vázquez-Barquero, associated with fewer negative and disorganized symptoms.
J.L., Crespo-Facorro, B., 2012. Homocysteine and cognition in first-episode psychosis Psychoneuroendocrinology 53, 1–9. https://doi.org/10.1016/j.psyneuen.2014.12.
patients. Eur. Arch. Psychiatry Clin. Neurosci. 262, 557–564. 012.
Baeza, I., Vigo, L., de la Serna, E., Calvo-Escalona, R., Merchán-Naranjo, J., Rodríguez- Coccurello, R., Moles, A., 2010. Potential mechanisms of atypical antipsychotic-induced
Latorre, P., Arango, C., Castro-Fornieles, J., 2017. The effects of antipsychotics on metabolic derangement: clues for understanding obesity and novel drug design.
weight gain, weight-related hormones and homocysteine in children and adolescents: Pharmacol. Ther. 127, 210–251. https://doi.org/10.1016/j.pharmthera.2010.04.
a 1-year follow-up study. Eur. Child Adolesc. Psychiatry 26, 35–46. 008.
Bahr, S.M., Weidemann, B.J., Castro, A.N., Walsh, J.W., deLeon, O., Burnett, C.M.L., Cope, M.B., Li, X., Jumbo-Lucioni, P., DiCostanzo, C.A., Jamison, W.G., Kesterson, R.A.,
Pearson, N.A., Murry, D.J., Grobe, J.L., Kirby, J.R., 2015. Risperidone-induced Allison, D.B., Nagy, T.R., 2009. Risperidone alters food intake, core body tempera-
weight gain is mediated through shifts in the gut microbiome and suppression of ture, and locomotor activity in mice. Physiol. Behav. 96, 457–463.
energy expenditure. EBioMedicine 2, 1725–1734. https://doi.org/10.1016/j.ebiom. Cota, D., Marsicano, G., Tschöp, M., Grübler, Y., Flachskamm, C., Schubert, M., Auer, D.,
2015.10.018. Yassouridis, A., Thöne-Reineke, C., Ortmann, S., 2003. The endogenous cannabinoid
Baimel, C., Bartlett, S.E., Chiou, L., Lawrence, A.J., Muschamp, J.W., Patkar, O., Tung, L., system affects energy balance via central orexigenic drive and peripheral lipogenesis.
Borgland, S.L., 2015. Orexin/hypocretin role in reward: implications for opioid and J. Clin. Investig. 112, 423–431.

237
R. Singh et al. European Journal of Pharmacology 844 (2019) 231–240

Cuerda, C., Velasco, C., Merchán-Naranjo, J., García-Peris, P., Arango, C., 2014. The ef- J., 2013. Olanzapine increases hepatic glucose production through the activation of
fects of second-generation antipsychotics on food intake, resting energy expenditure hypothalamic adenosine 5′‐monophosphate‐activated protein kinase. Diabetes Obes.
and physical activity. Eur. J. Clin. Nutr. https://doi.org/10.1038/ejcn.2013.253. Metab. 15, 1128–1135.
Davey, K.J., Cotter, P.D., O’sullivan, O., Crispie, F., Dinan, T.G., Cryan, J.F., O’mahony, Ishøy, P.L., Knop, F.K., Broberg, B.V., Baandrup, L., Fagerlund, B., Jørgensen, N.R.,
S.M., 2013. Antipsychotics and the gut microbiome: olanzapine-induced metabolic Andersen, U.B., Rostrup, E., Glenthøj, B.Y., Ebdrup, B.H., 2014. Treatment of anti-
dysfunction is attenuated by antibiotic administration in the rat. Transl. Psychiatry 3, psychotic-associated obesity with a GLP-1 receptor agonist—protocol for an in-
e309. vestigator-initiated prospective, randomised, placebo-controlled, double-blinded in-
Davey, K.J., O’Mahony, S.M., Schellekens, H., O’Sullivan, O., Bienenstock, J., Cotter, P.D., tervention study: the TAO study protocol. BMJ Open 4. https://doi.org/10.1136/
Dinan, T.G., Cryan, J.F., 2012. Gender-dependent consequences of chronic olanza- bmjopen-2013-004158.
pine in the rat: effects on body weight, inflammatory, metabolic and microbiota Jassim, G., Skrede, S., Vázquez, M.J., Wergedal, H., Vik-Mo, A.O., Lunder, N., Diéguez, C.,
parameters. Psychopharmacology 221, 155–169. Vidal-Puig, A., Berge, R.K., López, M., 2012. Acute effects of orexigenic antipsychotic
Davies, M.A., Compton-Toth, B.A., Hufeisen, S.J., Meltzer, H.Y., Roth, B.L., 2005. The drugs on lipid and carbohydrate metabolism in rat. Psychopharmacology 219,
highly efficacious actions of N-desmethylclozapine at muscarinic receptors are un- 783–794.
ique and not a common property of either typical or atypical antipsychotic drugs: is Joffe, G., Takala, P., Tchoukhine, E., Hakko, H., Raidma, M., Putkonen, H., Eronen, M.,
M1 agonism a pre-requisite for mimicking clozapine's actions? Psychopharmacology Räsänen, P., 2008. Orlistat in clozapine-or olanzapine-treated patients with over-
178, 451–460. https://doi.org/10.1007/s00213-004-2017-1. weight or obesity: a 16-week randomized, double-blind, placebo-controlled trial. J.
Deberdt, W., Winokur, A., Cavazzoni, P.A., Trzaskoma, Q.N., Carlson, C.D., Bymaster, Clin. Psychiatry 69, 706–711.
F.P., Wiener, K., Floris, M., Breier, A., 2005. Amantadine for weight gain associated Kanji, S., Fonseka, T.M., Marshe, V.S., Sriretnakumar, V., Hahn, M.K., Müller, D.J., 2018.
with olanzapine treatment. Eur. Neuropsychopharmacol. 15, 13–21. The microbiome-gut-brain axis: implications for schizophrenia and antipsychotic
del Rosario García-Miss, M., Pérez-Mutul, J., López-Canul, B., Solís-Rodríguez, F., Puga- induced weight gain. Eur. Arch. Psychiatry Clin. Neurosci. 268, 3–15.
Machado, L., Oxté-Cabrera, A., Gurubel-Maldonado, J., Arankowsky-Sandoval, G., Kao, A.C.-C., Spitzer, S., Anthony, D.C., Lennox, B., Burnet, P.W.J., 2018. Prebiotic at-
2010. Folate, homocysteine, interleukin-6, and tumor necrosis factor alfa levels, but tenuation of olanzapine-induced weight gain in rats: analysis of central and periph-
not the methylenetetrahydrofolate reductase C677T polymorphism, are risk factors eral biomarkers and gut microbiota. Transl. Psychiatry 8, 66.
for schizophrenia. J. Psychiatr. Res. 44, 441–446. Kapur, S., Remington, G., 2001. Dopamine D 2 receptors and their role in atypical anti-
Deng, C., Weston-Green, K., Huang, X.-F., 2010. The role of histaminergic H1 and H3 psychotic action: still necessary and may even be sufficient. Biol. Psychiatry 50,
receptors in food intake: a mechanism for atypical antipsychotic-induced weight 873–883.
gain? Prog. Neuro-Psychopharmacol. Biol. Psychiatry 34, 1–4. Kim, N.W., Song, Y.-M., Kim, E., Cho, H.-S., Cheon, K.-A., Kim, S.J., Park, J.Y., 2016.
Ellingrod, V.L., Grove, T.B., Burghardt, K.J., Taylor, S.F., Dalack, G., 2015. The effect of Adjunctive α-lipoic acid reduces weight gain compared with placebo at 12 weeks in
folate supplementation and genotype on cardiovascular and epigenetic measures in schizophrenic patients treated with atypical antipsychotics: a double-blind rando-
schizophrenia subjects. NPJ Schizophr. 1, 15046. mized placebo-controlled study. Int. Clin. Psychopharmacol. 31, 265–274.
Fadel, J., Bubser, M., Deutch, A.Y., 2002. Differential activation of orexin neurons by Kim, S.F., Huang, A.S., Snowman, A.M., Teuscher, C., Snyder, S.H., 2007. Antipsychotic
antipsychotic drugs associated with weight gain. J. Neurosci. 22, 6742–6746. drug-induced weight gain mediated by histamine H1 receptor-linked activation of
Fan, X., Borba, C.P.C., Copeland, P., Hayden, D., Freudenreich, O., Goff, D.C., Henderson, hypothalamic AMP-kinase. Proc. Natl. Acad. Sci. 104, 3456–3459.
D.C., 2013. Metabolic effects of adjunctive aripiprazole in clozapine‐treated patients Klawans, H.L., Goetz, C.G., Perlik, S., 1980. Tardive dyskinesia: review and update. Am. J.
with schizophrenia. Acta Psychiatr. Scand. 127, 217–226. Psychiatry 137, 900–908.
Farde, L., Nordström, A.-L., Wiesel, F.-A., Pauli, S., Halldin, C., Sedvall, G., 1992. Positron Klein, D.J., Cottingham, E.M., Sorter, M., Barton, B.A., Morrison, J.A., 2006. A rando-
emission tomographic analysis of central D1 and D2 dopamine receptor occupancy in mized, double-blind, placebo-controlled trial of metformin treatment of weight gain
patients treated with classical neuroleptics and clozapine: relation to extrapyramidal associated with initiation of atypical antipsychotic therapy in children and adoles-
side effects. Arch. Gen. Psychiatry 49, 538–544. cents. Am. J. Psychiatry 163, 2072–2079.
Fernandez-Egea, E., Bernardo, M., Donner, T., Conget, I., Parellada, E., Justicia, A., Kohno, D., Yada, T., 2012. Arcuate NPY neurons sense and integrate peripheral metabolic
Esmatjes, E., Garcia-Rizo, C., Kirkpatrick, B., 2009. Metabolic profile of anti- signals to control feeding. Neuropeptides 46, 315–319.
psychotic-naive individuals with non-affective psychosis. Br. J. Psychiatry 194, Kusumi, I., Boku, S., Takahashi, Y., 2015. Psychopharmacology of atypical antipsychotic
434–438. drugs: from the receptor binding profile to neuroprotection and neurogenesis.
Fetissov, S.O., Meguid, M.M., Sato, T., Zhang, L.-H., 2002. Expression of dopaminergic Psychiatry Clin. Neurosci. 69, 243–258.
receptors in the hypothalamus of lean and obese Zucker rats and food intake. Am. J. Lee, Y.J., Jeong, J.H., 2011. A systematic review of metformin to limit weight‐gain with
Physiol. Integr. Comp. Physiol. 283, R905–R910. atypical antipsychotics. J. Clin. Pharm. Ther. 36, 537–545.
Fonseka, T.M., Tiwari, A.K., Gonçalves, V.F., Lieberman, J.A., Meltzer, H.Y., Goldstein, Lett, T. A.P., Wallace, T.J.M., Chowdhury, N.I., Tiwari, a.K., Kennedy, J.L., Müller, D.J.,
B.I., Kennedy, J.L., Kennedy, S.H., Müller, D.J., 2015. The role of genetic variation 2011. Pharmacogenetics of antipsychotic-induced weight gain: review and clinical
across IL-1β, IL-2, IL-6, and BDNF in antipsychotic-induced weight gain. World J. implications. Mol. Psychiatry 10, 1–25. https://doi.org/10.1038/mp.2011.109.
Biol. Psychiatry 16, 45–56. Leucht, S., Corves, C., Arbter, D., Engel, R.R., Li, C., Davis, J.M., 2009. Second-generation
Gettys, T.W., Harkness, P.J., Watson, P.M., 1996. The beta 3-adrenergic receptor inhibits versus first-generation antipsychotic drugs for schizophrenia: a meta-analysis. Lancet
insulin-stimulated leptin secretion from isolated rat adipocytes. Endocrinology 137, 373, 31–41.
4054–4057. Li, X., Johnson, M.S., Smith, D.L., Li, Y., Kesterson, R.A., Allison, D.B., Nagy, T.R., 2013.
Ghanizadeh, A., Nikseresht, M.S., Sahraian, A., 2013. The effect of zonisamide on anti- Effects of risperidone on energy balance in female C57BL/6J mice. Obesity 21,
psychotic-associated weight gain in patients with schizophrenia: a randomized, 1850–1857.
double-blind, placebo-controlled clinical trial. Schizophr. Res. 147, 110–115. Lian, J., Huang, X.-F., Pai, N., Deng, C., 2016a. Ameliorating antipsychotic-induced
Goltz, J.S., Rice, T.R., 2017. Commentary: a randomized, double-blind, placebo-con- weight gain by betahistine: mechanisms and clinical implications. Pharmacol. Res.
trolled trial of metformin treatment of weight gain associated with initiation of 106, 51–63.
atypical antipsychotic therapy in children and adolescents. Front. Psychiatry 8. Lian, J., Huang, X.-F., Pai, N., Deng, C., 2016b. Ameliorating antipsychotic-induced
Gonçalves, P., Araújo, J.R., Martel, F., 2015. Antipsychotics-induced metabolic altera- weight gain by betahistine: mechanisms and clinical implications. Pharmacol. Res.
tions: focus on adipose tissue and molecular mechanisms. Eur. 106, 51–63. https://doi.org/10.1016/j.phrs.2016.02.011.
Neuropsychopharmacol. 25, 1–16. https://doi.org/10.1016/j.euroneuro.2014.11. Lian, J., Huang, X.F., Pai, N., Deng, C., 2014a. Betahistine ameliorates olanzapine-in-
008. duced weight gain through modulation of histaminergic, NPY and AMPK pathways.
Goodall, E., Oxtoby, C., Richards, R., Watkinson, G., Brown, D., Silverstone, T., 1988. A Psychoneuroendocrinology 48, 77–86. https://doi.org/10.1016/j.psyneuen.2014.06.
clinical trial of the efficacy and acceptability of D-fenfluramine in the treatment of 010.
neuroleptic-induced obesity. Br. J. Psychiatry 153, 208–213. Lian, J., Huang, X.F., Pai, N., Deng, C., 2014b. Preventing olanzapine-induced weight gain
Gough, S.C.L., O’Donovan, M.C., 2005. Clustering of metabolic comorbidity in schizo- using betahistine: a study in a rat model with chronic olanzapine treatment. PLoS One
phrenia: a genetic contribution? J. Psychopharmacol. https://doi.org/10.1177/ 9, e104160. https://doi.org/10.1371/journal.pone.0104160.
0269881105058380. Lin, C.-H., Lin, S.-C., Huang, Y.-H., Wang, F.-C., Huang, C.-J., 2018. Early prediction of
Graham, K.A., Perkins, D.O., Edwards, L.J., Barrier Jr, R.C., Lieberman, J.A., Harp, J.B., olanzapine-induced weight gain for schizophrenia patients. Psychiatry Res. 263,
2005. Effect of olanzapine on body composition and energy expenditure in adults 207–211.
with first-episode psychosis. Am. J. Psychiatry 162, 118–123. Lindenmayer, J.-P., Nathan, A.-M., Smith, R.C., 2001. Hyperglycemia associated with the
Grimm, O., Kaiser, S., Plichta, M.M., Tobler, P.N., 2017. Altered reward anticipation: use of atypical antipsychotics. J. Clin. Psychiatry.
potential explanation for weight gain in schizophrenia? Neurosci. Biobehav. Rev. Lira, F.S., Rosa, J.C., Pimentel, G.D., Seelaender, M., Damaso, A.R., Oyama, L.M., do
Haddad, P.M., Sharma, S.G., 2007. Adverse effects of atypical antipsychotics: differential Nascimento, C.O., 2012. Both adiponectin and interleukin-10 inhibit LPS-induced
risk and clinical implications. CNS Drugs 21, 911–936. activation of the NF-κB pathway in 3T3-L1 adipocytes. Cytokine 57, 98–106.
He, M., Zhang, Q., Deng, C., Wang, H., Lian, J., Huang, X.-F., 2014. Hypothalamic his- Liu, C., Lee, S., Elmquist, J.K., 2014. Circuits controlling energy balance and mood: in-
tamine H1 receptor-AMPK signaling time-dependently mediates olanzapine-induced herently intertwined or just complicated intersections? Cell Metab. 19, 902–909.
hyperphagia and weight gain in female rats. Psychoneuroendocrinology 42, 153–164. https://doi.org/10.1016/j.cmet.2014.02.008.
Hendrick, V., Dasher, R., Gitlin, M., Parsi, M., 2017. Minimizing weight gain for patients Lord, C.C., Wyler, S.C., Wan, R., Castorena, C.M., Ahmed, N., Mathew, D., Lee, S., Liu, C.,
taking antipsychotic medications: the potential role for early use of metformin. Ann. Elmquist, J.K., 2017. The atypical antipsychotic olanzapine causes weight gain by
Clin. Psychiatry 29, 120–124. targeting serotonin receptor 2C. J. Clin. Investig. 127, 3402–3406. https://doi.org/
Hill, J.O., Wyatt, H.R., Peters, J.C., 2012. Energy balance and obesity. Circulation 126, 10.1172/JCI93362.
126–132. Mackin, P., Bishop, D., Watkinson, H., Gallagher, P., Ferrier, I.N., 2007. Metabolic disease
Hippius, H., 1989. The history of clozapine. Psychopharmacology 99, S3–S5. and cardiovascular risk in people treated with antipsychotics in the community. Br. J.
Ikegami, M., Ikeda, H., Ohashi, T., Ohsawa, M., Ishikawa, Y., Kai, M., Kamei, A., Kamei, Psychiatry 191, 23–29. https://doi.org/10.1192/bjp.bp.106.031716.

238
R. Singh et al. European Journal of Pharmacology 844 (2019) 231–240

Martínez-Gras, I., García-Sánchez, F., Guaza, C., Rodríguez-Jiménez, R., Andrés-Esteban, Poyurovsky, M., Fuchs, C., Pashinian, A., Levi, A., Weizman, R., Weizman, A., 2013.
E., Palomo, T., Rubio, G., Borrell, J., 2012. Altered immune function in unaffected Reducing antipsychotic-induced weight gain in schizophrenia: a double-blind pla-
first-degree biological relatives of schizophrenia patients. Psychiatry Res. 200, cebo-controlled study of reboxetine–betahistine combination. Psychopharmacology
1022–1025. 226, 615–622.
Meltzer, H.Y., 2004. What's atypical about atypical antipsychotic drugs? Curr. Opin. Poyurovsky, M., Pashinian, A., Levi, A., Weizman, R., Weizman, A., 2005. The effect of
Pharmacol. 4, 53–57. https://doi.org/10.1016/j.coph.2003.09.010. betahistine, a histamine H1 receptor agonist/H3 antagonist, on olanzapine-induced
Miller, B.J., Buckley, P., Seabolt, W., Mellor, A., Kirkpatrick, B., 2011. Meta-analysis of weight gain in first-episode schizophrenia patients. Int. Clin. Psychopharmacol. 20,
cytokine alterations in schizophrenia: clinical status and antipsychotic effects. Biol. 101–103.
Psychiatry 70, 663–671. https://doi.org/10.1016/J.BIOPSYCH.2011.04.013. Poyurovsky, M., Tal, V., Maayan, R., Gil-Ad, I., Fuchs, C., Weizman, A., 2004. The effect
Misiak, B., Frydecka, D., Piotrowski, P., Kiejna, A., 2013. The multidimensional nature of of famotidine addition on olanzapine-induced weight gain in first-episode schizo-
metabolic syndrome in schizophrenia: lessons from studies of one-carbon metabolism phrenia patients: a double-blind placebo-controlled pilot study. Eur.
and DNA methylation. Epigenomics 5, 317–329. Neuropsychopharmacol. 14, 332–336.
Misiak, B., Łaczmański, Ł., Słoka, N.K., Szmida, E., Piotrowski, P., Loska, O., Ślęzak, R., Reynolds, G.P., Kirk, S.L., 2010. Metabolic side effects of antipsychotic drug treat-
Kiejna, A., Frydecka, D., 2016. Metabolic dysregulation in first-episode schizophrenia ment–pharmacological mechanisms. Pharmacol. Ther. 125, 169–179.
patients with respect to genetic variation in one-carbon metabolism. Psychiatry Res. Rickels, M.R., Perez, E.M., Peleckis, A.J., Alshehabi, E., Nguyen, H.-L., Stefanovski, D.,
238, 60–67. https://doi.org/10.1016/j.psychres.2016.01.077. Rickels, K., Teff, K.L., 2017. Contribution of parasympathetic muscarinic augmen-
Mittal, K., Gonçalves, V.F., Harripaul, R., Cuperfain, A.B., Rollins, B., Tiwari, A.K., Zai, tation of insulin secretion to olanzapine-induced hyperinsulinemia. Am. J. Physiol.
C.C., Maciukiewicz, M., Müller, D.J., Vawter, M.P., Kennedy, J.L., 2017. A compre- Metab.
hensive analysis of mitochondrial genes variants and their association with anti- Robinson, K., Prins, J., Venkatesh, B., 2011. Clinical review: adiponectin biology and its
psychotic-induced weight gain. Schizophr. Res. https://doi.org/10.1016/j.schres. role in inflammation and critical illness. Crit. Care 15, 221.
2017.06.046. Roffeei, S.N., Mohamed, Z., Reynolds, G.P., Said, M.A., Hatim, A., Mohamed, E.H.M.,
Mizuno, Y., Suzuki, T., Nakagawa, A., Yoshida, K., Mimura, M., Fleischhacker, W.W., Aida, S.A., Zainal, N.Z., 2014. Association of FTO, LEPR and MTHFR gene poly-
Uchida, H., 2014. Pharmacological strategies to counteract antipsychotic-induced morphisms with metabolic syndrome in schizophrenia patients receiving anti-
weight gain and metabolic adverse effects in schizophrenia: a systematic review and psychotics. Pharmacogenomics 15, 477–485.
meta-analysis. Schizophr. Bull. 40, 1385–1403. https://doi.org/10.1093/schbul/ Rojczyk, E., Palasz, A., Wiaderkiewicz, R., 2015. Effect of short and long-term treatment
sbu030. with antipsychotics on orexigenic/anorexigenic neuropeptides expression in the rat
Monda, M., Viggiano, A., Viggiano, A., Viggiano, E., Messina, G., Tafuri, D., De Luca, V., hypothalamus. Neuropeptides 51, 31–42. https://doi.org/10.1016/j.npep.2015.04.
2006. Quetiapine lowers sympathetic and hyperthermic reactions due to cerebral 001.
injection of orexin A. Neuropeptides 40, 357–363. https://doi.org/10.1016/j.npep. Sapra, M., Lawson, D., Iranmanesh, A., Varma, A., 2016. Adiposity-independent hypoa-
2006.07.003. diponectinemia as a potential marker of insulin resistance and inflammation in
Morgan, A.P., Crowley, J.J., Nonneman, R.J., Quackenbush, C.R., Miller, C.N., Ryan, A.K., schizophrenia patients treated with second generation antipsychotics. Schizophr. Res.
Bogue, M.A., Paredes, S.H., Yourstone, S., Carroll, I.M., 2014. The antipsychotic 174, 132–136. https://doi.org/10.1016/j.schres.2016.04.051.
olanzapine interacts with the gut microbiome to cause weight gain in mouse. PLoS Sárvári, A.K., Veréb, Z., Uray, I.P., Fésüs, L., Balajthy, Z., 2014. Atypical antipsychotics
One 9, e115225. induce both proinflammatory and adipogenic gene expression in human adipocytes
Mostafavi, S.-A., Solhi, M., Mohammadi, M.-R., Akhondzadeh, S., 2017. Melatonin for in vitro. Biochem. Biophys. Res. Commun. 450, 1383–1389.
reducing weight gain following administration of atypical antipsychotic olanzapine Scott, J.M., Weir, D.G., Molloy, A., McPartlin, J., Daly, L., Kirke, P., 1994. Folic acid
for adolescents with bipolar disorder: a randomized, double-blind, placebo-controlled metabolism and mechanisms of neural tube defects. Ciba Found. Symp. 181,
trial. J. Child Adolesc. Psychopharmacol. 180–191.
Murashita, M., Kusumi, I., Hosoda, H., Kangawa, K., Koyama, T., 2007. Acute adminis- Smith, G.C., Vickers, M.H., Shepherd, P.R., 2011. Olanzapine effects on body composi-
tration of clozapine concurrently increases blood glucose and circulating plasma tion, food preference, glucose metabolism and insulin sensitivity in the rat. Arch.
ghrelin levels in rats. Psychoneuroendocrinology 32, 777–784. Physiol. Biochem. 117, 241–249.
Nagata, M., Nakajima, M., Ishiwata, Y., Takahashi, Y., Takahashi, H., Negishi, K., Srisawasdi, P., Vanwong, N., Hongkaew, Y., Puangpetch, A., Vanavanan, S., Intachak, B.,
Yasuhara, M., 2016. Mechanism underlying induction of hyperglycemia in rats by Ngamsamut, N., Limsila, P., Sukasem, C., Kroll, M.H., 2017. Impact of risperidone on
single administration of olanzapine. Biol. Pharm. Bull. 39, 754–761. leptin and insulin in children and adolescents with autistic spectrum disorders. Clin.
Nakazato, M., Murakami, N., Date, Y., Kojima, M., Matsuo, H., Kangawa, K., Matsukura, Biochem. 50, 678–685. https://doi.org/10.1016/j.clinbiochem.2017.02.003.
S., 2001. A role for ghrelin in the central regulation of feeding. Nature 409, 194–198. Srisawat, U., Reynolds, G.P., Zhang, Z.J., Zhang, X.R., Arranz, B., San, L., Dalton, C.F.,
https://doi.org/10.1038/35051587. 2014. Methylenetetrahydrofolate reductase (MTHFR) 677C/T polymorphism is as-
Nemani, K., Hosseini Ghomi, R., McCormick, B., Fan, X., 2015. Schizophrenia and the sociated with antipsychotic-induced weight gain in first-episode schizophrenia. Int. J.
gut–brain axis. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 56, 155–160. https:// Neuropsychopharmacol. 17, 485–490.
doi.org/10.1016/j.pnpbp.2014.08.018. Starrenburg, F.C.J., Bogers, J.P.A.M., 2009. How can antipsychotics cause diabetes mel-
Neumeier, M., Weigert, J., Schäffler, A., Wehrwein, G., Müller-Ladner, U., Schölmerich, litus? Insights based on receptor-binding profiles, humoral factors and transporter
J., Wrede, C., Buechler, C., 2006. Different effects of adiponectin isoforms in human proteins. Eur. Psychiatry 24, 164–170. https://doi.org/10.1016/j.eurpsy.2009.01.
monocytic cells. J. Leukoc. Biol. 79, 803–808. 001.
Nielsen, M., Rostrup, E., Wulff, S., et al., 2012. Improvement of brain reward abnorm- Stefanidis, A., Verty, A.N.A., Allen, A.M., Owens, N.C., Cowley, M.A., Oldfield, B.J., 2009.
alities by antipsychotic monotherapy in schizophrenia. Arch. Gen. Psychiatry 69, The role of thermogenesis in antipsychotic drug‐induced weight gain. Obesity 17,
1195–1204. https://doi.org/10.1001/archgenpsychiatry.2012.847. 16–24.
Nielsen, M.Ø., Rostrup, E., Wulff, S., Glenthøj, B., Ebdrup, B.H., 2016. Striatal reward Sudhakar, T.P., Rao, G.P., Prasuna, P.L., Sagar, K.J.V., 2008. Study of effects of modafinil
activity and antipsychotic-associated weight change in patients with schizophrenia add-on therapy on excessive day time drowsiness and weight gain in patients on
undergoing initial treatment. JAMA Psychiatry 73, 121–128. atypical antipsychotics. Indian J. Psychol. Med. 30, 24.
Obuchowicz, E., Bielecka-Wajdman, A.M., Paul-Samojedny, M., Nowacka, M., 2017. Takeuchi, Y., Kajiyama, K., Ishiguro, C., Uyama, Y., 2015. Atypical antipsychotics and the
Different influence of antipsychotics on the balance between pro- and anti-in- risk of hyperlipidemia: a sequence symmetry analysis. Drug Saf. 38, 641–650.
flammatory cytokines depends on glia activation: an in vitro study. Cytokine 94, Teff, K.L., Rickels, M.R., Grudziak, J., Fuller, C., Nguyen, H.-L., Rickels, K., 2013.
37–44. https://doi.org/10.1016/j.cyto.2017.04.004. Antipsychotic-induced insulin resistance and postprandial hormonal dysregulation
Osei-Hyiaman, D., DePetrillo, M., Pacher, P., Liu, J., Radaeva, S., Bátkai, S., Harvey- independent of weight gain or psychiatric disease. Diabetes 62, 3232–3240.
White, J., Mackie, K., Offertáler, L., Wang, L., 2005. Endocannabinoid activation at Tek, C., Ratliff, J., Reutenauer, E., Ganguli, R., O’Malley, S.S., 2014. A randomized,
hepatic CB 1 receptors stimulates fatty acid synthesis and contributes to diet-induced double-blind, placebo-controlled pilot study of naltrexone to counteract anti-
obesity. J. Clin. Investig. 115, 1298–1305. psychotic-associated weight gain: proof of concept. J. Clin. Psychopharmacol. 34,
Ota, M., Mori, K., Nakashima, A., Kaneko, Y.S., Takahashi, H., Ota, A., 2005. Resistance to 608.
excessive bodyweight gain in risperidone‐injected rats. Clin. Exp. Pharmacol. Physiol. Tiwari, A.K., Zai, C.C., Likhodi, O., Lisker, A., Singh, D., Souza, R.P., Batra, P., Zaidi,
32, 279–287. S.H.E., Chen, S., Liu, F., 2010. A common polymorphism in the cannabinoid receptor
Palou, M., Sánchez, J., Rodríguez, A.M., Priego, T., Picó, C., Palou, A., 2009. Induction of 1 (CNR1) gene is associated with antipsychotic-induced weight gain in schizophrenia.
NPY/AgRP orexigenic peptide expression in rat hypothalamus is an early event in Neuropsychopharmacology 35, 1315.
fasting: relationship with circulating leptin, insulin and glucose. Cell. Physiol. Trayhurn, P., Bing, C., 2006. Appetite and energy balance signals from adipocytes. Philos.
Biochem. 23, 115–124. Trans. R. Soc. Lond. B Biol. Sci. 361, 1237–1249.
Panariello, F., De Luca, V., de Bartolomeis, A., 2011. Weight gain, schizophrenia and Trujillo, M.E., Scherer, P.E., 2005. Adiponectin–journey from an adipocyte secretory
antipsychotics: new findings from animal model and pharmacogenomic studies. protein to biomarker of the metabolic syndrome. J. Intern. Med. 257, 167–175.
Schizophr. Res. Treat. 2011, 16. https://doi.org/10.1155/2011/459284. Tsuchida, A., Nonomura, T., Ono-Kishino, M., Nakagawa, T., Taiji, M., Noguchi, H., 2001.
Peterson, H.R., Rothschild, M., Weinberg, C.R., Fell, R.D., McLeish, K.R., Pfeifer, M.A., Acute effects of brain-derived neurotrophic factor on energy expenditure in obese
1988. Body fat and the activity of the autonomic nervous system. N. Engl. J. Med. diabetic mice. Int. J. Obes. 25, 1286.
318, 1077–1083. Val-Laillet, D., Aarts, E., Weber, B., Ferrari, M., Quaresima, V., Stoeckel, L.E., Alonso-
Potvin, S., Zhornitsky, S., Stip, E., 2015. Antipsychotic-induced changes in blood levels of Alonso, M., Audette, M., Malbert, C.-H., Stice, E., 2015. Neuroimaging and neuro-
leptin in schizophrenia: a meta-analysis. Can. J. Psychiatry 60, S26. modulation approaches to study eating behavior and prevent and treat eating dis-
Poyurovsky, M., Fuchs, C., Pashinian, A., Levi, A., Faragian, S., Maayan, R., Gil-Ad, I., orders and obesity. NeuroImage Clin. 8, 1–31.
2007. Attenuating effect of reboxetine on appetite and weight gain in olanzapine- Van Der Zwaal, E.M., Merkestein, M., Lam, Y.K., Brans, M.A.D., Luijendijk, M.C.M., Bok,
treated schizophrenia patients: a double-blind placebo-controlled study. L.I.H., Verheij, E.R., La Fleur, S.E., Adan, R.A.H., 2012. The acute effects of olanza-
Psychopharmacology 192, 441–448. pine on ghrelin secretion, CCK sensitivity, meal size, locomotor activity and body

239
R. Singh et al. European Journal of Pharmacology 844 (2019) 231–240

temperature. Int. J. Obes. 36, 254. promote weight loss in patients with schizophrenia who have gained weight during
Varghese, D., Kirkwood, C.K., Carroll, N.V., 2016. Prevalence of antidiabetic and ntili- clozapine treatment. J. Clin. Psychiatry 63, 345–348.
pidemic medications in children and adolescents treated with atypical antipsychotics Woodward, N.D., Purdon, S.E., Meltzer, H.Y., Zald, D.H., 2005. A meta-analysis of neu-
in a virginia medicaid population. Ann. Pharmacother. 50, 463–470. ropsychological change to clozapine, olanzapine, quetiapine, and risperidone in
Volkow, N.D., Wang, G.-J., Baler, R.D., 2011. Reward, dopamine and the control of food schizophrenia. Int. J. Neuropsychopharmacol. 8, 457–472.
intake: implications for obesity. Trends Cogn. Sci. 15, 37–46. Yamauchi, T., Kamon, J., Waki, H., Terauchi, Y., Kubota, N., Hara, K., Mori, Y., Ide, T.,
Wang, G.-J., Volkow, N.D., Logan, J., Pappas, N.R., Wong, C.T., Zhu, W., Netusll, N., Murakami, K., Tsuboyama-Kasaoka, N., 2001. The fat-derived hormone adiponectin
Fowler, J.S., 2001. Brain dopamine and obesity. Lancet 357, 354–357. reverses insulin resistance associated with both lipoatrophy and obesity. Nat. Med. 7,
Weston-Green, K., Huang, X.-F., Deng, C., 2012. Alterations to melanocortinergic, 941.
GABAergic and cannabinoid neurotransmission associated with olanzapine-induced Zhang, Q., He, M., Deng, C., Wang, H., Lian, J., Huang, X.-F., 2014. Hypothalamic ghrelin
weight gain. PLoS One 7, e33548. signalling mediates olanzapine-induced hyperphagia and weight gain in female rats.
Weston-Green, K., Huang, X.F., Han, M., Deng, C., 2008. The effects of antipsychotics on Int. J. Neuropsychopharmacol. 17, 807–818.
the density of cannabinoid receptors in the dorsal vagal complex of rats: implications Zheng, W., Zhang, Q.-E., Cai, D.-B., Yang, X.-H., Ungvari, G.S., Ng, C.H., Wu, R.-R., Xiang,
for olanzapine-induced weight gain. Int. J. Neuropsychopharmacol. 11, 827–835. Y.-T., 2018. Combination of metformin and lifestyle intervention for antipsychotic-
https://doi.org/10.1017/s1461145708008560. related weight gain: a meta-analysis of randomized controlled trials.
White, J.C., Steele, V.M., Jaskiw, G.E., 2002. Phenylpropanolamine appears not to Pharmacopsychiatry.

240

You might also like