Elsevier

Brain Research Bulletin

Volume 170, May 2021, Pages 246-253
Brain Research Bulletin

Haloperidol elicits oxidative damage in the brain of rats submitted to the ketamine-induced model of schizophrenia

https://doi.org/10.1016/j.brainresbull.2021.01.021Get rights and content

Highlights

  • Several side effects were assigned to haloperidol, an antipsychotic drug.

  • Molecular alterations underlying the side effects of this drug are poorly understood.

  • Haloperidol induced remarkable oxidative damage to lipids and proteins in rat brain.

  • Redox imbalance is one of the potential mechanisms involved in the oxidative damage.

Abstract

The present study aims to evaluate the effects of haloperidol, an important first-generation antipsychotic, on the antioxidant system parameters in the brain of animals subjected to a model of schizophrenia induced by ketamine. Adult rats intraperitoneally received saline (1 mL/kg) or ketamine (25 mg/kg body weight) for 15 days, and saline or haloperidol (0.1 mg/kg body weight) via gavage once a day, between the 9th and 14th days. In the frontal cortex, hippocampus, and striatum, assessments of lipid (4-hydroxy-2-nonenal and 8-isoprostane levels) and protein (protein carbonyl content) oxidative damage were conducted. It was also measured the glutathione peroxidase and glutathione reductase activities in the same cerebral structures. Increases in the 4-hydroxy-2-nonenal and 8-isoprostane levels were detected in rats receiving haloperidol and ketamine. An increase in the carbonyl content was also observed in animals receiving ketamine, haloperidol, or a combination thereof. In animals receiving the antipsychotic, there was a decrease in the activity of the enzymes. Therefore, both ketamine and haloperidol induced oxidative damage. A possible energy dysfunction or a haloperidol effect targeting the glutathione enzymes, and then disrupting the redox homeostasis in neurons, could not be ruled out, although further studies are required to confirm or refute a direct interaction.

Introduction

Schizophrenia (SZ) is a lifelong mental disorder characterized by positive and negative symptoms, as well as cognitive impairment (Krzystanek and Pałasz, 2019). The SZ prevalence is 0.3–1% worldwide (Cattane et al., 2018; World Health Organization (WHO), 2018). SZ pathophysiology remains poorly understood, while the alterations observed in several brain structures and the lack of responsiveness of negative and cognitive symptoms to antipsychotics are still a challenge (Dean, 2017; Rodrigues-Amorim et al., 2017). Nevertheless, it was hypothesized that an excess or depletion of dopamine, serotonin, glutamate, and γ-aminobutyric acid (GABA) collaborates for the pathophysiology of the disorder (Patel et al., 2014). There is evidence of immune system dysfunction in schizophrenic patients, which could lead to neuroinflammation (Miller et al., 2011). Moreover, studies have indicated that an impairment in the antioxidant defenses is also detectable in patients (Pavlović et al., 2002; Gysin et al., 2007; Dadheech et al., 2008).

In the present study, an SZ model was induced using ketamine (Ket), a well-known anesthetic drug (Canever et al., 2010). Ket administration is regarded as a suitable SZ animal model since it mimics the pathogen mechanisms associated with the symptoms of the disorder. Indeed, Ket administration enables the study of positive, negative, and cognitive alterations observed in SZ, which contributes to providing face validity to this model (Chatterjee et al., 2011; Frohlich and Van Horn, 2014). Neurochemical effects induced by Ket include oxidative stress and blockade of N-methyl-D-aspartic acid (NMDA) receptors, preventing the influx of calcium in neurons, which induces behavioral changes similar to that observed in the patients, collaborating to provide construct validity to the model (Chatterjee et al., 2011). Additional evidence about how to induce the Ket model and how it works regarding the behavioral analysis is showed in recent studies (Zugno et al., 2016; Damazio et al., 2017; Canever et al., 2018; Supp et al., 2020). Data from these papers enable the reproduction of the model to assess potential neurochemical alterations similar to that observed in schizophrenic patients using haloperidol or other drugs.

The therapy of SZ and other psychotic disorders can be carried out by first-generation antipsychotics. These drugs primarily act by inhibiting dopaminergic receptors (Dean, 2017). Among the drugs of this class, haloperidol (Hal) is recommended for acute episodes of positive symptoms and as maintenance therapy for SZ (Haddad and Correll, 2018; Chokhawala and Stevens, 2019). Although the antipsychotics have persisted as the standard therapy for SZ and are effective in treating the positive symptoms, these drugs may induce several side effects, including tardive dyskinesia, muscle rigidity, and tremors (Buchanan et al., 2007; Chatterjee et al., 2011; Dean, 2017). At least in part, oxidative stress seems to be involved in such effects.

In this regard, Hal (2 mg kg−1 day−1) was found to impair the activity of enzymes such as catalase and superoxide dismutase as well as elicit an increase in the levels of the lipid peroxidation by-product hydroxyalkenal in rat brain (Parikh et al., 2003). Besides, Hal (1.5 mg kg−1 day−1) induced significant increases in the content of thiobarbituric acid-reactive species (TBARS) in the striatum as well as in the protein carbonyl content in hippocampus of rats (Reinke et al., 2004). Hal was also linked to increased serum TBARS in SZ patients, in comparison with olanzapine (Singh et al., 2008). In the Raudenska and coworkers' review (2013) is outlined a more detailed picture of the mechanisms involved in the cytotoxicity of Hal-induced oxidative stress.

Other antipsychotics with a possible role in oxidative stress include clozapine, chlorpromazine, risperidone, and ziprasidone (Martins et al., 2008; Elmorsy et al., 2017; Dietrich-Muszalska and Kolińska-Łukaszuk, 2018). The first-generation drugs are more prone to induce lipid peroxidation than the most recent ones (Kropp et al., 2005). Despite the repertory of alterations mediated by reactive oxygen species described to date, the precise mechanisms underlying the toxicity of antipsychotics are still incomplete, even at the lowest doses of the drugs. In this scenario, the present study aimed to evaluate the effects of a low dose of Hal on oxidative stress parameters in the brain of rats submitted to an animal model of SZ induced by Ket.

Section snippets

Animals

Twenty adult male Wistar rats (Rattus norvegicus; 250–350 g body weight; heterogenic strain) acquired from the Central Animal House at the University of Southern Santa Catarina were kept as 5 per cage and received ad libitum chow and drinking water. The cages were maintained in a room with a 12 h-light/dark cycle (lights on at 7:00 a.m.) at 22 ± 1 °C. All procedures were conducted following the National Institutes of Health (US) “Guide for the Care and Use of Laboratory Animals” (National

Results

The present study was designed to investigate the effects of the Hal administration in an animal model of SZ induced by Ket. First, it was evaluated the oxidative damage to lipids in the cerebral structures, in terms of 4-HNE and 8-ISO contents. The administration of Ket elicited an increase in the 4-HNE levels in the frontal cortex, hippocampus, and striatum, as compared to the control group. Interestingly, Hal did not alter the increase in the 4-HNE levels induced by Ket in any brain

Discussion

In the present study, a significant increase in the lipid peroxidation parameters was found in the brain structures in rats receiving Hal itself or combined with Ket. The evaluation of protein carbonylation in the samples was also carried out and revealed that all groups receiving Hal, Ket, or a combination thereof presented an increase in this parameter in the three cerebral structures analyzed. Finally, the evaluation of the activity of GPx and GR demonstrated a decrease in these parameters

Funding sources

Translational Psychiatry Program (USA) is funded by a grant from the National Institute of Health/National Institute of Mental Health (1R21MH117636-01A1, to JQ). Center of Excellence on Mood Disorders (USA) is funded by the Pat Rutherford Jr. Chair in Psychiatry, John S. Dunn Foundation and Anne and Don Fizer Foundation Endowment for Depression Research. Translational Psychiatry Laboratory (Brazil) is funded by grants from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq),

CRediT authorship contribution statement

Samira S. Valvassori: Conceptualization, Methodology, Supervision, Project administration. José H. Cararo: Writing - original draft, Visualization. Samira Menegas: Writing - original draft, Visualization. Taise Possamai-Della: Investigation, Validation. Jorge M. Aguiar-Geraldo: Investigation, Validation. Simone Lespinasse Araujo: Investigation. Gustavo Antunes Mastella: Investigation. João Quevedo: Writing - review & editing, Funding acquisition. Alexandra I. Zugno: Writing - review & editing,

Declaration of Competing Interest

JQ has the following declarations of interest: Clinical research support: Janssen Pharmaceutical (Clinical Trial), Allergan (Clinical Trial); Advisory boards, speaker bureaus, expert witness, or consultant: Daiichi Sankyo (Speaker Bureau); Patent, equity, or royalty: Instituto de Neurociências Dr. João Quevedo (Stockholder); Other: Artmed Editora (Copyright), Artmed Panamericana (Copyright). All the other authors have no conflicts of interest.

Acknowledgments

Translational Psychiatry Program (USA) is supported by the Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth). Translational Psychiatry Laboratory (Brazil) is one of the centers of the National Institute for Molecular Medicine (INCT-MM) and one of the members of the Center of Excellence in Applied Neurosciences of Santa Catarina (NENASC).

References (50)

  • M. Kronbauer et al.

    Influence of magnesium supplementation on movement side effects related to typical antipsychotic treatment in rats

    Behav. Brain Res.

    (2017)
  • A.Y. Onaolapo et al.

    Melatonin attenuates behavioural deficits and reduces brain oxidative stress in a rodent model of schizophrenia

    Biomed. Pharmacother.

    (2017)
  • V. Parikh et al.

    Differential effects of antipsychotics on expression of antioxidant enzymes and membrane lipid peroxidation in rat brain

    J. Psychiatr. Res.

    (2003)
  • A. Pillai et al.

    Long-term antipsychotic treatments and crossover studies in rats: differential effects of typical and atypical agents on the expression of antioxidant enzymes and membrane lipid peroxidation in rat brain

    J. Psychiatr. Res.

    (2007)
  • A. Reinke et al.

    Haloperidol and clozapine, but not olanzapine, induces oxidative stress in rat brain

    Neurosci. Lett.

    (2004)
  • D. Rodrigues-Amorim et al.

    Schizophrenia: a review of potential biomarkers

    J. Psychiatr. Res.

    (2017)
  • A.B. Sonego et al.

    Cannabidiol prevents haloperidol-induced vacuos chewing movements and inflammatory changes in mice via PPARγ receptors

    Brain Behav. Immun.

    (2018)
  • M. Vairetti et al.

    Nicergoline reverts haloperidol-induced loss of detoxifying-enzyme activity

    Eur. J. Pharmacol.

    (2004)
  • M.C. Yang et al.

    Generalized estimating equation model and long-term exposure effect of antipsychotics on SH-SY5Y cells against oxidative stressors

    Eur. J. Pharmacol.

    (2014)
  • A.I. Zugno et al.

    Effect of folic acid on oxidative stress and behavioral changes in the animal model of schizophrenia induced by ketamine

    J. Psychiatr. Res.

    (2016)
  • O.M. Abdel-Salam et al.

    Brain oxidative stress and neurodegeneration in the ketamine model of schizophrenia during antipsychotic treatment: effects of N-acetylcysteine treatment

    ROS

    (2018)
  • L. Canever et al.

    A rodent model of schizophrenia reveals increase in creatine kinase activity with associated behavior changes

    Oxid. Med. Cell. Longev.

    (2010)
  • N. Castagnoli et al.

    Enzyme-catalyzed bioactivation of cyclic tertiary amines to form potential neurotoxins

    Pol. J. Pharmacol.

    (1999)
  • N. Cattane et al.

    Prenatal exposure to environmental insults and enhanced risk of developing Schizophrenia and Autism Spectrum disorder: focus on biological pathways and epigenetic mechanisms

    Neurosci. Biobehav. Rev.

    (2018)
  • K. Chokhawala et al.

    Antipsychotic Medications

    (2019)
  • Cited by (0)

    View full text