Next Article in Journal
An Objective Measure of Patellar Tendon Thickness Based on Ultrasonography and MRI in University Athletes
Next Article in Special Issue
New Opportunities and Challenges of Early Psychosis
Previous Article in Journal
Differences in Kinematic and Kinetic Patterns According to the Bone Tumor Location after Endoprosthetic Knee Replacement Following Bone Tumor Resection: A Comparative Gait Analysis between Distal Femur and Proximal Tibia
Previous Article in Special Issue
Fronto-Parietal Gray Matter Volume Loss Is Associated with Decreased Working Memory Performance in Adolescents with a First Episode of Psychosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Biological Mechanism(s) Underpinning the Association between Antipsychotic Drugs and Weight Gain

1
Institute for Innovation in Physical and Mental Health and Clinical Translation, IMPACT, School of Medicine, Deakin University, Geelong 3220, Australia
2
Genomics Centre, School of Life and Environmental Sciences, Deakin University, Geelong 3220, Australia
3
Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia
4
Department of Psychiatry, Royal Melbourne Hospital, University of Melbourne, Parkville 3052, Australia
5
Centre of Youth Mental Health, University of Melbourne, Parkville 3052, Australia
6
Orygen Youth Health Research Centre, Parkville 3052, Australia
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(18), 4095; https://doi.org/10.3390/jcm10184095
Submission received: 28 July 2021 / Revised: 4 September 2021 / Accepted: 8 September 2021 / Published: 10 September 2021
(This article belongs to the Special Issue New Opportunities and Challenges of Early Psychosis)

Abstract

:
Weight gain and consequent metabolic alterations are common side-effects of many antipsychotic drugs. Interestingly, several studies have suggested that improvement in symptoms and adverse metabolic effects are correlated. We used next generation sequencing data from NT-2 (human neuronal) cells treated with aripiprazole, amisulpride, risperidone, quetiapine, clozapine, or vehicle control, and compared with the Pillinger P-score (ranked from 0 to 1, indicating greater increase in weight gain and related metabolic parameters) to identify the genes most associated with the drugs’ propensity to cause weight gain. The top 500 genes ranked for their correlation with the drugs’ propensity to cause weight gain were subjected to pathway analysis using DAVID (NIH). We further investigated transcription factors (TFs) that are more likely to regulate the genes involved in these processes using the prediction tool of key TFs from TRRUST. The results suggest an enrichment for genes involved in lipid biosynthesis and metabolism, which are of interest for mechanisms underpinning weight-gain. The list of genes involved in the lipid pathways that correlated with weight gain was enriched for genes transcriptionally regulated by SREBF1 and SREBF2. Furthermore, quetiapine significantly increased the expression of SREBF1 and SREBF2 in NT-2 cells. Our results suggest that the effects of these antipsychotic drugs on lipid metabolism may be mediated, at least in part, via regulation of SREBF1/SREBF2 expression, with evidence of a direct effect of quetiapine on the expression of SREBF1/2. The effects of antipsychotic drugs on lipid metabolism may influence white matter structure (therapeutic effect) and the risk of weight gain, lipid disturbances, and, consequently, metabolic syndrome (adverse effects). Understanding the different molecular effects of these drugs could inform a personalized medicine approach in treating patients with schizophrenia.

1. Introduction

Antipsychotic-Induced Weight Gain (AIWG) is a debilitating and common adverse effect of antipsychotic treatment, and negatively impacts on life expectancy, quality of life, treatment adherence, and the likelihood of developing the metabolic syndrome and type-2 diabetes [1]. Weight gain also adversely affects clinical outcomes such as readmission [1].
The molecular mechanisms behind AIWG remain mostly unknown. Several studies have investigated receptor binding profiles to explain the metabolic abnormalities related to weight-gain. Involvement of the dopamine-based reward stimulation might underlie the changes in food consumption [2]; preclinical models also indicate the involvement of histamine H1 receptors, through activation of the hypothalamic AMP-kinase signaling to increase food intake [3]. The participation of the serotonin 5-HT2 receptors in control of feeding behavior is well established [4], and more recently genetic polymorphisms in the serotonin receptor 2C were associated with an increase in AIWG [5]. With respect to receptor binding profiles olanzapine and clozapine, which block histamine H1, serotonin 5-HT2A/C and dopamine D2/3 receptors, cause the greatest weight gain [6]; while compounds with little or no histamine and serotonin affinity—lurasidone and aripiprazole for example—have a lower risk of weight gain [7]. The effects of neurohormones such as leptin, adiponectin, glucagon like protein 1 (GLP-1), and insulin suggests the involvement of the gut–brain axis might underly both the therapeutic and weight-gain sides [8,9].
Besides receptor binding profile and antipsychotic side-effects, the weight gain in patients treated with antipsychotics has been linked to individual and environmental characteristics. A meta-analysis identified 13 single-nucleotide polymorphisms that were significantly associated with AIWG [10], and the pharmacogenomics associated with drug-induced weight gain was reviewed by Sneha Singh and colleagues [11] and Soria-Chacartegui [12]. Children and adolescents seem to be especially vulnerable to AIWG [13,14]. Increased risk for AIWG was also linked to first exposure to antipsychotic medication, longer use of antipsychotics, baseline weight, gender, and other individual characteristics [6,15,16].
In an effort to understand the effects of different antipsychotics on the metabolic alterations that occur in patients treated with these drugs, Pillinger and colleagues performed a systematic review and network meta-analysis comparing 18 antipsychotic medications [17]. One of the parameters investigated was change in body weight. In 83 studies included in the analysis, with 18,750 patients using antipsychotic medication for a median of 6 weeks, and 4210 patients in the placebo group, the authors were able to rank the antipsychotics for the degree of weight-gain using a P-score with a scale of 0 to 1, where the higher P-score indicated greater increase in weight-gain.
Therefore, using the P-score generated by the Pillinger [17] network meta-analysis and differential gene expression in neuronal like cells treated with antipsychotics we aimed to identify pathways or gene sets linked to biological mechanism(s) underpinning the propensity of antipsychotic drugs to cause weight gain.

2. Materials and Methods

2.1. Cell Culture

NT2 human teratocarcinoma cells (CVCL_0034, ATCC, Manassas, VA, USA) were cultured as previously described [18]. Briefly, the cells were maintained in standard cell culture media and then differentiated into neuronal-like cells using 1 × 10−5 M retinoic acid (Sigma-Aldrich, Sydney, Australia) for 28 days with media refreshed every 2–3 days. For experiments, cells were seeded at 2 × 105 cells/well (24-well plates) and treated with mitotic inhibitors (1 µM cytosine and 10 µM uridine; Sigma-Aldrich) every 2–3 days for a total of seven days. The cells were then treated with aripiprazole (0.1 µM), amisulpride (10 µM), clozapine (10 µM), quetiapine (50 µM), or risperidone (0.10 µM) for 24 h. Treatment doses and intervals were determined in previous dose–response studies in our lab, so that, when used in combination, no single drug dominated the effect on gene expression or affected cell viability; and were carried out throughout the following projects [18,19]. All drugs were purchased from Sigma-Aldrich (Sydney, Australia). Vehicle control cells were treated with 0.2% dimethyl sulfoxide (DMSO).

2.2. Gene Expression

Following the 24-h drug treatment, cells were harvested using Trizol, and total RNA was extracted using RNeasy® mini kits (Qiagen, Melbourne, Australia) and quantified by spectrophotometry (NanoDrop 1000 Thermo Fisher Scientific, Waltham, MA, USA). The quality of the extracted RNA was evaluated using an Agilent 2100 Bioanalyzer (Agilent Technologies, Melbourne, Australia). RNAseq libraries were prepared from 1ug of total RNA using a TruSeq RNA samples Preparation kit (Illumina, Victoria, Australia). The libraries were sequenced using a HiSeq 2500 flow cell (50 bp single end reads; Illumina) according to the manufacturer’s instructions.

2.3. Genome-Wide Gene Expression Analysis

The raw data were obtained in fastq format and processed using the Deakin Genomics Centre RNAseq alignment and expression quantification pipeline (https://github.com/m-richardson/RNASeq_pipe, accessed on 1 July 2017). In summary, this involves: Raw read quality filtering and adapter trimming (ILLUMINACLIP:2:30:10:4, SLIDINGWINDOW:5:20, AVGQUAL:20 MINLEN:36) with Trimmomatic v35 [20], and alignment to the reference genome using STAR v2.5 in 2-pass mode (Human genome version GRCh38) [21]. The expression was quantified at the gene level, and individual sample counts were collated into a m × n matrix for differential abundance testing. Normalization (TMM) and removal of low expressed gene were performed using edgeR [22] in R [23] following the edgeR manual (<1 cpm in n samples, where n is the number of samples in the smallest group for comparison). Differential gene expression analysis was assessed using edgeR in R, and the Benjamini–Hochberg [24] corrected p-values were calculated to account for multiple testing. Genes with corrected p-values of <0.05 were considered to be differentially expressed (Supplementary Table S1).

2.4. Pathway Analysis

We used parametric correlation to identify the genes most associated with the drug’s propensity to cause weight-gain. Within the list of differentially expressed genes (logFC) and the P-scores calculated by Pillinger (Table 1) we identified the top 500 genes that were positively correlated with the propensity to cause weight gain and subjected these genes to pathway analysis using the Database for Annotation, Visualization and Integrated Discovery (DAVID; National Institutes of Health) [25].

2.5. Transcriptional Regulatory Relationships Unravelled by Sentence-Based Text-Mining (TRRUST)

TRRUST is a manually curated database for transcriptional regulatory networks that can predict key transcription factors (TF) regulating gene expression [26]. The differentially expressed genes in the pathways of interest were submitted for analysis using TRRUST.

3. Results

3.1. DAVID Pathways

The top enriched pathways resulting from the top 500 genes correlated with the propensity of the antipsychotics to cause weight gain are listed in Table 2. Although the Benjamini adjusted p-value was not significant (<0.05), there appears to be enrichment for genes involved in lipid and cholesterol metabolism, which are of interest for mechanisms underpinning weight-gain.

3.2. TRRUST

Next, we submitted the differentially expressed genes in the pathways of interest, i.e., lipid biosynthesis and lipid metabolism (Table 3) to TRRUST.
The list of lipid metabolism genes that were correlated with weight gain was enriched for genes that are transcriptionally regulated by SREBF1 (FDR = 1.76 × 10−5) (Table 4). This suggests that the antipsychotic drugs’ effects on lipid metabolism may be mediated, at least in part, via regulation of SREBF1/SREBF2 expression.

3.3. Antipsychotic Drug Effects on SREBFs

We also investigated whether the antipsychotic drugs used to treat the NT-2 cells had effects on the gene expression of SREBFs (Table 5). Quetiapine was the only drug to significantly increase the expression of SREBF1 and SREBF2 (FDR = 1.03 × 10−11 and FDR = 10.9 × 10−22).

4. Discussion

We demonstrated that five commonly prescribed antipsychotic drugs change the expression of genes involved in the lipid biosynthesis and metabolic pathways, and that these genes might be regulated by SREBF1 and SREBF2. SREB proteins are transcription factors that play a key role in cholesterol biosynthesis influencing both uptake and fatty acid biosynthesis as well as upregulating the synthesis of sterol biosynthesis enzymes [27].
Our results are in line with previous experiments demonstrating the upregulation of genes regulated by SREB transcription factors after antipsychotic drug treatment in glial cells [28] and central nervous system related cell lines [29]. In glial cells, HMGCR, FASN, SREBP-1, and SREBP-2 expression was increased after treatment with haloperidol and clozapine [28]. Subsequent studies showed effects of chlorpromazine, haloperidol, olanzapine, risperidone, and ziprasidone on SREBPs and downstream genes, in GaMg and CCF-STTG1 glial cells, HCN2 cortical neurons and SH-SY5Y neuroblastoma cell lines [29]. In the context of the CNS these findings can be interpreted in line with the abnormalities in myelination and white matter identified in patients with schizophrenia [30], suggesting that antipsychotics might target symptoms at least in part through lipogenic activation and consequently increase myelination [31,32].
These results were also confirmed in adipocytes. Studies showed that olanzapine induced adipogenesis through the overexpression of genes regulated by SREBF1 [33]; and clozapine, olanzapine, and risperidone increased lipogenesis through Insig/SCAP/SREBP signalling [34]. This suggests that the therapeutic effect of antipsychotics on the CNS are associated with systemic effects that are at least in part responsible for the side-effects related to weight-gain and metabolic syndrome [35].
The association of polymorphisms in the SREBF1 and SREBF2 genes with schizophrenia was first reported in 2010 and has been replicated in three independent samples [36]. Further corroborations were reported in a Chinese cohort treated with clozapine; with two SREBF2 SNPs being associated with increased risk for the drug-induced metabolic syndrome [37]. The same group later reported an association of SREF1 and SCAP (SREBP cleavage-activation protein) SNPs with elevated risk of drug-induced metabolic syndrome in people with schizophrenia [38]. Altogether these studies point towards interindividual variances for AIWG that may be associated with genetic variation in SREBF genes.
Our results add to the existing knowledge by showing that aripiprazole, amisulpride, and quetiapine also affect the expression of genes involved in lipid and cholesterol pathways, with quetiapine having a direct effect on the expression of SREBF1 and SREBF2.
Baptista and colleagues investigated the efficacy of weight loss interventions [39], with the meta-analysis showing that amantadine, metformin, reboxetine, sibutramine and topiramate were partially effective in reducing AIWG. This was corroborated recently in a study showing that treatment augmentation with metformin and GLP-1RA cause reduction in weight with minimal reports of severe side effects [40]. Later analysis suggested that metformin had a greater effect in weight loss [41]. Due to the differences between antipsychotics in terms of the propensity to cause weight gain, a recent study investigated whether switching to an antipsychotic with lesser metabolic effects, such as aripiprazole, amisulpride, and ziprasidone [15] would be beneficial for weight loss. The results indicated that weight gain was mild in patients that did change treatments, although antipsychotic switch did not necessarily result in weight loss.
We acknowledge some limitations of our study. Pillinger et al. [17] used a comprehensive list of drugs in their study, however due to resource limitations, we prioritised the five drugs used in this study because they are commonly prescribed and thought to be mechanistically diverse. We tested a single dose of each drug and measured acute effects; therefore, these finds do not capture effects of chronic administration or drug–drug interactions. In addition, our experiments used neuronal-like cells without inducing of any disease model, underlying pathophysiological processes specific for each disease might influence the drug effects.
Here we showed that the propensity of antipsychotic drugs to cause weight gain was associated with differential effects on lipid metabolic genes at a transcriptional level independent of receptor binding profile, and that SREBF transcription factors may play a role in these effects. Taken together, the data on efficacy [7] adverse events and risk factors [17], and knowledge of the underlying pathways can inform a personalised medicine approach for treating patients with schizophrenia.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/jcm10184095/s1, Table S1: Differentially expressed genes per treatment.

Author Contributions

Conceptualization; O.M.D., M.B. and K.W.; methodology; B.P., C.C.B., B.S. and K.W.; formal analysis; T.T.T.T. and K.W.; investigation; B.P., C.C.B., B.S., S.K. and T.C.; data curation; M.F.R.; writing—original draft preparation; B.P.; writing—review and editing; B.P., C.C.B., B.S., S.K., T.C., M.F.R., T.T.T.T., Z.S.J.L., L.G., J.H.K., O.M.D., M.B. and K.W.; visualization; T.T.T.T. and Z.S.J.L.; funding acquisition; K.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NHMRC Project Grant (GNT1078928) and NHMRC Centre of Research Excellence (CREDIT) (GNT1153607).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data available from author upon request.

Acknowledgments

M.B. is supported by a NHMRC Senior Principal Research Fellowship (1156072). O.M.D. is an R.D. Wright Biomedical NHMRC Career Development Fellow (APP1145634) and has received grant support from the Brain and Behavior Foundation, Simons Autism Foundation, Stanley Medical Research Institute, Deakin University, Lilly, NHMRC, and ASBDD/Servier. She has also received in kind support from BioMedica Nutracuticals, NutritionCare and Bioceuticals.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Correll, C.U.; Solmi, M.; Veronese, N.; Bortolato, B.; Rosson, S.; Santonastaso, P.; Thapa-Chhetri, N.; Fornaro, M.; Gallicchio, D.; Collantoni, E.; et al. Prevalence, Incidence and Mortality from Cardiovascular Disease in Patients with Pooled and Specific Severe Mental Illness: A Large-scale Meta-analysis of 3,211,768 Patients and 113,383,368 Controls. World Psychiatry 2017, 16, 163–180. [Google Scholar] [CrossRef] [Green Version]
  2. Grimm, O.; Kaiser, S.; Plichta, M.M.; Tobler, P.N. Altered Reward Anticipation: Potential Explanation for Weight Gain in Schizophrenia? Neurosci. Biobehav. Rev. 2017, 75, 91–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kim, S.F.; Huang, A.S.; Snowman, A.M.; Teuscher, C.; Snyder, S.H. Antipsychotic Drug-Induced Weight Gain Mediated by Histamine H1 Receptor-Linked Activation of Hypothalamic AMP-Kinase. Proc. Natl. Acad. Sci. USA 2007, 104, 3456–3459. [Google Scholar] [CrossRef] [Green Version]
  4. Vickers, S.P.; Dourish, C.T.; Kennett, G.A. Evidence That Hypophagia Induced by D-Fenfluramine and d-Norfenfluramine in the Rat Is Mediated by 5-HT2C Receptors. Neuropharmacology 2001, 41, 200–209. [Google Scholar] [CrossRef]
  5. Wallace, T.J.M.; Zai, C.C.; Brandl, E.J.; Müller, D.J. Role of 5-HT 2C Receptor Gene Variants in Antipsychotic-Induced Weight Gain. Pharm. Pers. Med. 2011, 4, 83–93. [Google Scholar]
  6. Leucht, S.; Cipriani, A.; Spineli, L.; Mavridis, D.; Örey, D.; Richter, F.; Samara, M.; Barbui, C.; Engel, R.R.; Geddes, J.R.; et al. Comparative Efficacy and Tolerability of 15 Antipsychotic Drugs in Schizophrenia: A Multiple-Treatments Meta-Analysis. Lancet 2013, 382, 951–962. [Google Scholar] [CrossRef]
  7. Rummel-Kluge, C.; Komossa, K.; Schwarz, S.; Hunger, H.; Schmid, F.; Lobos, C.A.; Kissling, W.; Davis, J.M.; Leucht, S. Head-to-Head Comparisons of Metabolic Side Effects of Second Generation Antipsychotics in the Treatment of Schizophrenia: A Systematic Review and Meta-Analysis. Schizophr. Res. 2010, 123, 225–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Bioque, M.; González-Rodríguez, A.; Garcia-Rizo, C.; Cobo, J.; Monreal, J.A.; Usall, J.; Soria, V.; Labad, J. Targeting the Microbiome-Gut-Brain Axis for Improving Cognition in Schizophrenia and Major Mood Disorders: A Narrative Review. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 105, 110130. [Google Scholar] [CrossRef]
  9. Leptin and Psychiatric Illnesses: Does Leptin Play a Role in Antipsychotic-Induced Weight Gain? Available online: https://pubmed.ncbi.nlm.nih.gov/32033608/ (accessed on 23 July 2021).
  10. Zhang, J.-P.; Lencz, T.; Zhang, R.X.; Nitta, M.; Maayan, L.; John, M.; Robinson, D.G.; Fleischhacker, W.W.; Kahn, R.S.; Ophoff, R.A.; et al. Pharmacogenetic Associations of Antipsychotic Drug-Related Weight Gain: A Systematic Review and Meta-Analysis. Schizophr. Bull 2016, 42, 1418–1437. [Google Scholar] [CrossRef] [Green Version]
  11. Singh, S.; Ricardo-Silgado, M.L.; Bielinski, S.J.; Acosta, A. Pharmacogenomics of Medication-Induced Weight Gain and Antiobesity Medications. Obesity 2021, 29, 265–273. [Google Scholar] [CrossRef]
  12. Soria-Chacartegui, P.; Villapalos-García, G.; Zubiaur, P.; Abad-Santos, F.; Koller, D. Genetic Polymorphisms Associated With the Pharmacokinetics, Pharmacodynamics and Adverse Effects of Olanzapine, Aripiprazole and Risperidone. Front. Pharm. 2021, 12, 711940. [Google Scholar] [CrossRef]
  13. De Hert, M.; Dobbelaere, M.; Sheridan, E.M.; Cohen, D.; Correll, C.U. Metabolic and Endocrine Adverse Effects of Second-Generation Antipsychotics in Children and Adolescents: A Systematic Review of Randomized, Placebo Controlled Trials and Guidelines for Clinical Practice. Eur. Psychiatry 2011, 26, 144–158. [Google Scholar] [CrossRef] [PubMed]
  14. Bussell, K.; Reeves, G.; Hager, E.; Zhu, S.; Correll, C.U.; Riddle, M.A.; Sikich, L. Dietary Consumption Among Youth with Antipsychotic-Induced Weight Gain and Changes Following Healthy Lifestyle Education. J. Child. Adolesc. Psychopharmacol. 2021, 31, 364–375. [Google Scholar] [CrossRef]
  15. Bak, M.; Drukker, M.; Cortenraad, S.; Vandenberk, E.; Guloksuz, S. Antipsychotics Result in More Weight Gain in Antipsychotic Naive Patients than in Patients after Antipsychotic Switch and Weight Gain Is Irrespective of Psychiatric Diagnosis: A Meta-Analysis. PLoS ONE 2021, 16, e0244944. [Google Scholar] [CrossRef]
  16. Chen, Y.Q.; Li, X.R.; Zhang, L.; Zhu, W.B.; Wu, Y.Q.; Guan, X.N.; Xiu, M.H.; Zhang, X.Y. Therapeutic Response Is Associated With Antipsychotic-Induced Weight Gain in Drug-Naive First-Episode Patients With Schizophrenia: An 8-Week Prospective Study. J. Clin. Psychiatry 2021, 82, 20m13469. [Google Scholar] [CrossRef]
  17. Pillinger, T.; McCutcheon, R.A.; Vano, L.; Mizuno, Y.; Arumuham, A.; Hindley, G.; Beck, K.; Natesan, S.; Efthimiou, O.; Cipriani, A.; et al. Comparative Effects of 18 Antipsychotics on Metabolic Function in Patients with Schizophrenia, Predictors of Metabolic Dysregulation, and Association with Psychopathology: A Systematic Review and Network Meta-Analysis. Lancet Psychiatry 2020, 7, 64–77. [Google Scholar] [CrossRef]
  18. Panizzutti, B.; Bortolasci, C.C.; Spolding, B.; Kidnapillai, S.; Connor, T.; Richardson, M.F.; Truong, T.T.T.; Liu, Z.S.J.; Morris, G.; Gray, L.; et al. Transcriptional Modulation of the Hippo Signaling Pathway by Drugs Used to Treat Bipolar Disorder and Schizophrenia. Int. J. Mol. Sci. 2021, 22, 7164. [Google Scholar] [CrossRef]
  19. Kidnapillai, S.; Bortolasci, C.C.; Udawela, M.; Panizzutti, B.; Spolding, B.; Connor, T.; Sanigorski, A.; Dean, O.M.; Crowley, T.; Jamain, S.; et al. The Use of a Gene Expression Signature and Connectivity Map to Repurpose Drugs for Bipolar Disorder. World J. Biol. Psychiatry 2020, 21, 775–783. [Google Scholar] [CrossRef]
  20. Bolger, A.M.; Lohse, M.; Usadel, B. Trimmomatic: A Flexible Trimmer for Illumina Sequence Data. Bioinformatics 2014, 30, 2114–2120. [Google Scholar] [CrossRef] [Green Version]
  21. STAR: Ultrafast Universal RNA-Seq Aligner/Bioinformatics/Oxford Academic. Available online: https://academic.oup.com/bioinformatics/article/29/1/15/272537 (accessed on 2 June 2021).
  22. Robinson, M.D.; McCarthy, D.J.; Smyth, G.K. EdgeR: A Bioconductor Package for Differential Expression Analysis of Digital Gene Expression Data. Bioinformatics 2010, 26, 139–140. [Google Scholar] [CrossRef] [Green Version]
  23. R: A Language and Environment for Statistical Computing. Available online: https://www.gbif.org/tool/81287/r-a-language-and-environment-for-statistical-computing (accessed on 2 June 2021).
  24. Benjamini, Y.; Hochberg, Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J. R. Stat. Soc. Ser. B 1995, 57, 289–300. [Google Scholar] [CrossRef]
  25. Huang, D.W.; Sherman, B.T.; Lempicki, R.A. Systematic and Integrative Analysis of Large Gene Lists Using DAVID Bioinformatics Resources. Nat. Protoc. 2009, 4, 44–57. [Google Scholar] [CrossRef]
  26. Han, H.; Cho, J.-W.; Lee, S.; Yun, A.; Kim, H.; Bae, D.; Yang, S.; Kim, C.Y.; Lee, M.; Kim, E.; et al. TRRUST v2: An Expanded Reference Database of Human and Mouse Transcriptional Regulatory Interactions. Nucleic Acids Res. 2018, 46, D380–D386. [Google Scholar] [CrossRef] [PubMed]
  27. Steen, V.M.; Skrede, S.; Polushina, T.; López, M.; Andreassen, O.A.; Fernø, J.; Hellard, S.L. Genetic Evidence for a Role of the SREBP Transcription System and Lipid Biosynthesis in Schizophrenia and Antipsychotic Treatment. Eur. Neuropsychopharmacol. 2017, 27, 589–598. [Google Scholar] [CrossRef]
  28. Fernø, J.; Raeder, M.B.; Vik-Mo, A.O.; Skrede, S.; Glambek, M.; Tronstad, K.-J.; Breilid, H.; Løvlie, R.; Berge, R.K.; Stansberg, C.; et al. Antipsychotic Drugs Activate SREBP-Regulated Expression of Lipid Biosynthetic Genes in Cultured Human Glioma Cells: A Novel Mechanism of Action? Pharm. J 2005, 5, 298–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Fernø, J.; Skrede, S.; Vik-Mo, A.O.; Håvik, B.; Steen, V.M. Drug-Induced Activation of SREBP-Controlled Lipogenic Gene Expression in CNS-Related Cell Lines: Marked Differences between Various Antipsychotic Drugs. BMC Neurosci 2006, 7, 69. [Google Scholar] [CrossRef] [Green Version]
  30. Smirnova, L.P.; Yarnykh, V.L.; Parshukova, D.A.; Kornetova, E.G.; Semke, A.V.; Usova, A.V.; Pishchelko, A.O.; Khodanovich, M.Y.; Ivanova, S.A. Global Hypomyelination of the Brain White and Gray Matter in Schizophrenia: Quantitative Imaging Using Macromolecular Proton Fraction. Transl. Psychiatry 2021, 11, 365. [Google Scholar] [CrossRef] [PubMed]
  31. Garver, D.L.; Holcomb, J.A.; Christensen, J.D. Compromised Myelin Integrity during Psychosis with Repair during Remission in Drug-Responding Schizophrenia. Int. J. Neuropsychopharmacol. 2008, 11, 49–61. [Google Scholar] [CrossRef] [Green Version]
  32. Bartzokis, G.; Lu, P.H.; Nuechterlein, K.H.; Gitlin, M.; Doi, C.; Edwards, N.; Lieu, C.; Altshuler, L.L.; Mintz, J. Differential Effects of Typical and Atypical Antipsychotics on Brain Myelination in Schizophrenia. Schizophr. Res. 2007, 93, 13–22. [Google Scholar] [CrossRef] [Green Version]
  33. Yang, L.-H.; Chen, T.-M.; Yu, S.-T.; Chen, Y.-H. Olanzapine Induces SREBP-1-Related Adipogenesis in 3T3-L1 Cells. Pharm. Res 2007, 56, 202–208. [Google Scholar] [CrossRef]
  34. Chen, C.-C.; Hsu, L.-W.; Huang, K.-T.; Goto, S.; Chen, C.-L.; Nakano, T. Overexpression of Insig-2 Inhibits Atypical Antipsychotic-Induced Adipogenic Differentiation and Lipid Biosynthesis in Adipose-Derived Stem Cells. Sci. Rep. 2017, 7, 10901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Ferno, J. Lipogenic Effects of Psychotropic Drugs: Focus on the SREBP System. Front. Biosci. 2011, 16, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Le Hellard, S.; Theisen, F.M.; Haberhausen, M.; Raeder, M.B.; Fernø, J.; Gebhardt, S.; Hinney, A.; Remschmidt, H.; Krieg, J.C.; Mehler-Wex, C.; et al. Association between the Insulin-Induced Gene 2 (INSIG2) and Weight Gain in a German Sample of Antipsychotic-Treated Schizophrenic Patients: Perturbation of SREBP-Controlled Lipogenesis in Drug-Related Metabolic Adverse Effects? Mol. Psychiatry 2009, 14, 308–317. [Google Scholar] [CrossRef]
  37. Yang, L.; Chen, J.; Liu, D.; Yu, S.; Cong, E.; Li, Y.; Wu, H.; Yue, Y.; Zuo, S.; Wang, Y.; et al. Association between SREBF2 Gene Polymorphisms and Metabolic Syndrome in Clozapine-Treated Patients with Schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry 2015, 56, 136–141. [Google Scholar] [CrossRef] [PubMed]
  38. Yang, L.; Chen, J.; Li, Y.; Wang, Y.; Liang, S.; Shi, Y.; Shi, S.; Xu, Y. Association between SCAP and SREBF1 Gene Polymorphisms and Metabolic Syndrome in Schizophrenia Patients Treated with Atypical Antipsychotics. World J. Biol. Psychiatry 2016, 17, 467–474. [Google Scholar] [CrossRef]
  39. Baptista, T.; ElFakih, Y.; Uzcátegui, E.; Sandia, I.; Tálamo, E.; de Baptista, E.A.; Beaulieu, S. Pharmacological Management of Atypical Antipsychotic-Induced Weight Gain. CNS Drugs 2008, 22, 477–495. [Google Scholar] [CrossRef] [PubMed]
  40. Marteene, W.; Winckel, K.; Hollingworth, S.; Kisely, S.; Gallagher, E.; Hahn, M.; Ebdrup, B.H.; Firth, J.; Siskind, D. Strategies to Counter Antipsychotic-Associated Weight Gain in Patients with Schizophrenia. Expert Opin. Drug Saf. 2019, 18, 1149–1160. [Google Scholar] [CrossRef] [PubMed]
  41. Maayan, L.; Vakhrusheva, J.; Correll, C.U. Effectiveness of Medications Used to Attenuate Antipsychotic-Related Weight Gain and Metabolic Abnormalities: A Systematic Review and Meta-Analysis. Neuropsychopharmacology 2010, 35, 1520–1530. [Google Scholar] [CrossRef] [Green Version]
Table 1. Antipsychotic Propensity to Cause Weight-Gain P-Scores.
Table 1. Antipsychotic Propensity to Cause Weight-Gain P-Scores.
DrugP-Score
Aripiprazole0.26
Amisulpride0.41
Risperidone0.58
Quetiapine0.65
Clozapine0.90
P-score: value resulted from network metanalysis performed by Pillinger and colleagues [17]. P-score ranges from 0 to 1, with higher scores indicating a greater degree of weight gain and metabolic disturbance.
Table 2. Top Pathways Enriched in Genes Positively Correlated with the Drugs Propensity to Cause Weight Gain.
Table 2. Top Pathways Enriched in Genes Positively Correlated with the Drugs Propensity to Cause Weight Gain.
CategoryTermCountp-ValueFDR
UP_KEYWORDSLipid biosynthesis130.000200.054
UP_KEYWORDSLipid metabolism230.000310.054
UP_KEYWORDSCholesterol biosynthesis50.00070.084
UP_KEYWORDSSteroid biosynthesis60.00100.088
GOTERM_CC_DIRECTEndoplasmic reticulum340.00120.38
UP_KEYWORDSCholesterol metabolism70.00160.10
UP_KEYWORDSAlternative splicing2680.00170.10
GOTERM_BP_DIRECTCholesterol biosynthesis process60.00190.41
KEGG_PATHWAYLysosome100.00190.41
UP_KEYWORDSSterol biosynthesis50.00210.11
Count = number of genes in the pathway; DR = false discovery rate.
Table 3. Correlations between Differentially Expressed Genes in the Lipid Biosynthesis and Lipid Metabolism Pathways with P-Score.
Table 3. Correlations between Differentially Expressed Genes in the Lipid Biosynthesis and Lipid Metabolism Pathways with P-Score.
Gener2p-Value
LIPID BIOSYNTHESIS
PCYT20.930.0032
ACACA0.690.061
HMGCR0.660.074
MCAT0.660.074
FDPS0.630.087
DHCR240.620.093
LIPID METABOLISM
SREBF10.960.00091
PCYT20.930.0032
PCSK90.760.038
LPIN10.730.045
ACACA0.690.061
PLIN10.670.068
MCAT0.660.074
HMGCR0.660.074
FDPS0.630.087
DHCR240.620.093
r2 = parametric correlation between different expressed genes and P-score.
Table 4. TRRUST Transcription Factors Regulating Lipid Biosynthesis and Metabolism Genes.
Table 4. TRRUST Transcription Factors Regulating Lipid Biosynthesis and Metabolism Genes.
Key TFDescriptionp-ValueFDRList of Overlapped Genes
LIPID BIOSYNTHESIS
SREBF2sterol regulatory element binding transcription factor 25.84 × 10−55.84 × 10−5HMGCR, FDPS
LIPID METABOLISM
SREBF2sterol regulatory element binding transcription factor 22.31 × 10−61.38 × 10−5HMGCR, PCSK9, FDPS
SREBF1sterol regulatory element binding transcription factor 15.88 × 10−61.76 × 10−5LPIN1, PCSK9, FDPS
NFYCnuclear transcription factor Y, gamma0.000130.000261FDPS, LPIN1
RELAv-rel reticuloendotheliosis viral oncogene homolog A (avian)0.05980.0726PCYT2, PLIN1
NFKB1nuclear factor of kappa light polypeptide gene enhancer in B-cells 10.06050.0726PCYT2, PLIN1
SP1Sp1 transcription factor0.1290.129SREBF1, DHCR24
TF = transcriptional factor; FDR = false discovery rate.
Table 5. Antipsychotic Effect of SREB Gene Expression.
Table 5. Antipsychotic Effect of SREB Gene Expression.
SREBF1SREBF2
logFClogCPMp-ValueFDRlogFClogCPMp-ValueFDR
Quetiapine0.535.353.11 × 10−131.03 × 10−110.268.811.01 × 10−241.09 × 10−22
Amisulpride−0.154.930.170.50−0.108.500.04990.30
Aripiprazole−0.234.930.100.34−0.168.490.0250.18
Clozapine0.235.230.0260.200.058.630.410.72
Risperidone0.057.970.220.83−0.068.530.170.80
Log FC = logarithmic fold change; logCPM = logarithmic count per million; FDR = false discovery rate.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Panizzutti, B.; Bortolasci, C.C.; Spolding, B.; Kidnapillai, S.; Connor, T.; Richardson, M.F.; Truong, T.T.T.; Liu, Z.S.J.; Gray, L.; Kim, J.H.; et al. Biological Mechanism(s) Underpinning the Association between Antipsychotic Drugs and Weight Gain. J. Clin. Med. 2021, 10, 4095. https://doi.org/10.3390/jcm10184095

AMA Style

Panizzutti B, Bortolasci CC, Spolding B, Kidnapillai S, Connor T, Richardson MF, Truong TTT, Liu ZSJ, Gray L, Kim JH, et al. Biological Mechanism(s) Underpinning the Association between Antipsychotic Drugs and Weight Gain. Journal of Clinical Medicine. 2021; 10(18):4095. https://doi.org/10.3390/jcm10184095

Chicago/Turabian Style

Panizzutti, Bruna, Chiara C. Bortolasci, Briana Spolding, Srisaiyini Kidnapillai, Timothy Connor, Mark F. Richardson, Trang T. T. Truong, Zoe S. J. Liu, Laura Gray, Jee Hyun Kim, and et al. 2021. "Biological Mechanism(s) Underpinning the Association between Antipsychotic Drugs and Weight Gain" Journal of Clinical Medicine 10, no. 18: 4095. https://doi.org/10.3390/jcm10184095

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop