Next Article in Journal
Mitochondrial Quality Control Processes at the Crossroads of Cell Death and Survival: Mechanisms and Signaling Pathways
Previous Article in Journal
Ceanothanes Derivatives as Peripheric Anionic Site and Catalytic Active Site Inhibitors of Acetylcholinesterase: Insights for Future Drug Design
Previous Article in Special Issue
Royal Jelly: Biological Action and Health Benefits
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Effect of Neuropsychiatric Drugs on the Oxidation-Reduction Balance in Therapy

by
Karina Sommerfeld-Klatta
1,*,
Wiktoria Jiers
1,
Szymon Rzepczyk
2,
Filip Nowicki
2,
Magdalena Łukasik-Głębocka
3,
Paweł Świderski
2,
Barbara Zielińska-Psuja
1,
Zbigniew Żaba
3 and
Czesław Żaba
2
1
Department of Toxicology, Poznań University of Medical Sciences, 3 Rokietnicka Street, 60-806 Poznań, Poland
2
Department of Forensic Medicine, Poznań University of Medical Sciences, 10 Rokietnicka Street, 60-806 Poznań, Poland
3
Department of Emergency Medicine, Poznań University of Medical Sciences, 7 Rokietnicka Street, 60-806 Poznań, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(13), 7304; https://doi.org/10.3390/ijms25137304
Submission received: 21 April 2024 / Revised: 30 June 2024 / Accepted: 1 July 2024 / Published: 3 July 2024
(This article belongs to the Special Issue Targeting Oxidative Stress for Disease)

Abstract

:
The effectiveness of available neuropsychiatric drugs in the era of an increasing number of patients is not sufficient, and the complexity of neuropsychiatric disease entities that are difficult to diagnose and therapeutically is increasing. Also, discoveries about the pathophysiology of neuropsychiatric diseases are promising, including those initiating a new round of innovations in the role of oxidative stress in the etiology of neuropsychiatric diseases. Oxidative stress is highly related to mental disorders, in the treatment of which the most frequently used are first- and second-generation antipsychotics, mood stabilizers, and antidepressants. Literature reports on the effect of neuropsychiatric drugs on oxidative stress are divergent. They are starting with those proving their protective effect and ending with those confirming disturbances in the oxidation–reduction balance. The presented publication reviews the state of knowledge on the role of oxidative stress in the most frequently used therapies for neuropsychiatric diseases using first- and second-generation antipsychotic drugs, i.e., haloperidol, clozapine, risperidone, olanzapine, quetiapine, or aripiprazole, mood stabilizers: lithium, carbamazepine, valproic acid, oxcarbazepine, and antidepressants: citalopram, sertraline, and venlafaxine, along with a brief pharmacological characteristic, preclinical and clinical studies effects.

1. Introduction

Mental disorders have constituted a burden on society for many years. The situation has deteriorated significantly as a result of the pandemic, as well as the risks associated with the increasing stress and direct neuropsychiatric effects of the SARS-CoV-2 virus. Unfortunately, the efficacy of available neuropsychiatric drugs is not sufficient in an era of increasing numbers of patients, and the involvement of both diagnostically and therapeutically complex diseases is rising. Psychiatric disorders are a range of various conditions associated with abnormal functioning of the nervous system, which are common in patients with nervous system disorders [1]. As a result of these abnormalities, in response to a stressful situation, the body activates maladaptive behavioral patterns that impede daily functioning [2]. The most common mental disorders include mood swings, neurotic disorders, dementia disorders, schizoaffective disorders, eating and personality disorders, sleep disorders, or addictions (Table 1) [3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23]. According to the World Health Report, mental disorders affect approximately 25% of people at some point during their lifetime. The co-occurrence of neurological diseases with mood disorders and anxiety syndromes is common in patients with neurodegenerative diseases (Parkinson’s, Alzheimer’s, and Huntington’s), stroke, epilepsy, and demyelinating diseases. The choice of treatment for neurological patients with co-occurring mood and anxiety disorders is often difficult, as it has to take into account the symptoms of the underlying disease and poorer drug tolerance [1]. According to the International Neuropsychiatry Association, discoveries in the pathophysiology of neuropsychiatric diseases are promising, including those initiating a new round of innovation within the contribution of oxidative stress to the etiology of neuropsychiatric diseases [2,24,25]. Oxidative stress is a significant risk factor for many psychiatric disorders, for the treatment of which first- and second-generation antipsychotics, mood stabilizers, and antidepressants are most commonly used (Table 2) [2].
Oxidative stress is a state of imbalance between antioxidants and free radicals in the body. Both oxidants and antioxidants, essential for vital processes, should remain in quantitative balance for the safety of the entire system. However, there are known factors under the influence of which one overpowers the other, resulting in changes, i.e., DNA damage, disruption of protein, lipid, or carbohydrate function and structure. In addition, oxidative stress may be involved in the etiology of many conditions, i.e., hypertension, obesity, atherosclerosis, or diabetes. Also, in terms of many neuropsychiatric diseases, despite advances in neurobiological research, the pathophysiology of many of these diseases has yet to be more extensively described. Advances in the understanding of the pathomechanism of many psychiatric disorders, as well as neuropsychiatric diseases, may help to discover effective therapies and indicators for their early diagnosis. Oxidative stress, which damages biomolecules and causes dysfunctions of, among others, cellular systems at the level of, e.g., mitochondria or dopamine receptors, is also referred to as a critical phenomenon in patients with mental disorders [2,26,27,28,29] (Figure 1).
Under conditions of homeostasis, and thus at safe concentrations, the presence of reactive oxygen species (ROS) is not harmful to the organism and acts as a regulator and mediator of physiological processes. ROS play an essential role in the ordinary course of inflammatory reactions, regulation of immune processes, or T-lymphocyte activation, with concomitant adhesion of leukocyte cells to the endothelium [27,28,29,30]. Currently, the diagnostic market makes available numerous analytical tools and biomarkers that can be used to assess oxidative stress. Examples of oxidative markers include myeloperoxidase (MPO), 4-hydroxynonenal (HNE), F2-isoprostanes (IsoP), 8-hydroxy-2-deoxyguanosine (8-OHdG), malondialdehyde (MDA), allantoin, and thiobarbituric acid reactive substances (TBARS). They are used to monitor tissue damage resulting from a loss of balance between antioxidants and free radicals [31,32,33]. Table 3 brings together the oxidative stress markers according to the adverse changes they are used to assess.
Knowledge of the biotransformation of drugs used to treat psychiatric disorders involving enzymatic systems may support the hypothesis of oxidative disorders generated by, among other things, reactive metabolites or radicals. However, the link between the effects of many drugs and oxidative stress is still the subject of few scientific publications aiming to confirm their antioxidant or pro-oxidant effects in groups of treated patients [35,36].
The presented publication reviews the state of knowledge on the share of oxidative stress in the most commonly used therapies for neuropsychiatric diseases using first- and second-generation antipsychotics, i.e., haloperidol (HLP), clozapine (CLO), risperidone (RIS), olanzapine (OLZ), quetiapine (QT) or aripiprazole (ARP), mood stabilizers: lithium (Li), carbamazepine (CBZ), valproic acid (VPA), oxcarbazepine (OXC), and antidepressants: citalopram (CIT), sertraline (SER), venlafaxine (VEN), along with an explanation of the obtained results and their comparison. The data provided and discussed come from the US National Library of Medicine (PubMed) bibliographic sources, selecting publications from the last ten years and older for analysis in order to discuss the results of more recent preclinical and clinical studies. The key search terms for the literature search were oxidative stress and neuropsychiatric drugs.

2. First and Second-Generations of Antipsychotic Drugs

2.1. Haloperidol (HLP)

2.1.1. Pharmacological HLP Data

Haloperidol (HLP), synthesized in 1958, belongs to the butyrophenone derivatives. Its discovery came during research into new derivatives of pethidine and methadone, both of which were claimed to have analgesic activity. However, it has been proven that this drug has significant efficacy in the treatment of psychiatric disorders, including schizophrenia, especially hallucinations and delusions [37,38,39]. The antipsychotic effect of the drug is due to the blockade of dopamine D2 receptors. Its high affinity for the receptor, together with a low cholinolytic effect, minimizes the occurrence of symptoms such as constipation, dry mouth, and visual disturbances [38,40,41,42,43].

2.1.2. Preclinical HLP Studies

Oxidative stress occurring with a first-generation antipsychotic drug such as haloperidol is not a fully understood issue. The use of the drug has been shown to increase oxidative stress, which has an impact on the pathophysiology of schizophrenia. On the one hand, oxidative imbalance may be part of the pathogenesis of the disease, but on the other hand, it can lead to patients experiencing extrapyramidal symptoms (EPS). Preclinical and in vitro (cell) studies conducted on HLP have revealed that the use of the drug is associated with a decrease in antioxidant enzyme activity and an increase in reactive oxygen species (ROS) levels [44,45,46,47,48,49,50,51]. Perera et al. demonstrated that biochemical changes in the rat brain after haloperidol administration are accompanied by histopathological changes such as necrosis and fibrosis. Although the exact pathophysiology of EPS is not known, the authors here point to an important role for ROS in inducing oxidative stress, representing one mechanism of haloperidol-induced long-term neurotoxicity [44]. When HLP is used, dopamine is increased, which deaminates and generates significant amounts of free radicals and hydrogen peroxide, among other things. Reactive metabolites leading to multicellular damage and lipid peroxidation contribute to the cytotoxic effects of the drug. Raudenska et al. showed that long-term haloperidol administration to rats leads to changes in sigma-1R receptor and inositol triphosphate receptor (IP3R) expression, modifying myocardial contractility. This may underlie cardiac arrhythmias and QT interval prolongation in patients using haloperidol [45]. Haloperidol can also lead to changes in brain tissue, as evidenced by the study by El-Awadan et al. After 14 days of administering the drug to rats at a dose of 1 mg/kg, the concentrations of GSH, malondialdehyde (MDA), and nitric oxide (NO) in the brain and liver were determined. Levels of the former were significantly reduced in both tissues. In contrast, there was an increase in NO and MDA levels in brain tissue [47]. The adverse effects of the drug on antioxidant enzymes in the brain were also confirmed by measuring the expression of the enzyme peroxiredoxin-6. After a 28-day administration of haloperidol (1 mg/kg) to rats, significantly increased levels of the test protein were observed in the grey matter of the brain, with a concomitant increase in lipid peroxidation. However, peroxiredoxin-6 concentrations were unchanged in the white matter of the brain and liver of rats [48].
Subsequent studies in human cell lines confirmed that, under prooxidant conditions, therapeutic concentrations of haloperidol increase TBARS levels. However, a significant correlation between the used drug and lipid peroxidation was not observed for cells not treated with hydrogen peroxide. In addition, the combination of lithium and haloperidol significantly reduced cell survival. Despite the lack of statistical significance of the results obtained, they indicate the need for particular caution when combining drugs used in the treatment of psychiatric disorders [50,51].

2.1.3. Clinial HLP Studies

Available clinical data on the relationship between HLP and oxidative stress points to the use of biomarkers such as superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH). The use of haloperidol results in a decrease in the activity of the above antioxidant enzymes, as confirmed by a study by Bošković et al. conducted on a group of patients treated with haloperidol for a minimum of six months. In addition, they observed significantly lower concentrations of the biomarkers determined in patients with extrapyramidal symptoms [46]. In contrast, Singh et al. further assessed markers of lipid peroxidation by determining serum thiobarbituric acid reactive substances (TBARS). TBARS levels were significantly elevated in patients treated with haloperidol (for at least six weeks) compared with patients taking the second-generation antipsychotic drug olanzapine. SOD has also been shown to be negatively correlated with serum TBARS [49].

2.2. Clozapine (CLO)

2.2.1. Pharmacological CLO Data

Clozapine (CLO), which belongs to the atypical antipsychotics, has structural similarities to tricyclic antidepressants. Its synthesis took place in the early 1960s. A clear advantage of clozapine appeared to be the absence of extrapyramidal side effects. Nonetheless, the drug was initially skeptically assessed and withdrawn from use, a factor contributed to by the significant risk of agranulocytosis during its use. However, continued research has confirmed significant effectiveness in the treatment of symptoms of psychosis, especially drug-resistant schizophrenia [52,53]. Regular blood tests of patients have also been shown to control the risk of agranulocytosis. Consequently, the drug was reintroduced to the pharmaceutical market in 1989 in Europe—and a year later in the USA. However, its use is limited, and it is not a first-line drug. The inclusion of CLO in pharmacotherapy occurs primarily when treatment with other antipsychotics is unsuccessful. In addition, its use is associated with adverse effects that are troublesome for patients, e.g., weight gain, constipation, metabolic disorders, and the development of diabetes [52,54,55]. The antipsychotic effect of clozapine is due to its diverse pharmacological profile. This is because it shows affinity for dopamine D1, D4, and D2 receptors. In the case of the D2 receptor, dissociation occurs very rapidly, thus reducing the risk of hyperprolactinemia and extrapyramidal symptoms. Clozapine also has an affinity for serotonin, adrenergic, histamine, and muscarinic receptors [52,54,55].

2.2.2. Preclinical CLO Studies

There are numerous attempts to link oxidative stress to adverse effects, including metabolic disorders occurring during clozapine use, in preclinical studies. This points to the involvement of free radicals leading to the oxidation of proteins, which results in their disruption or loss of function [56,57,58,59,60,61]. In the study by Walss-Bass et al., human neuroblastoma cells were used to identify changes in proteins under oxidative stress after administration of CLO (in three concentrations of 1, 10, or 20 µM). The level of reactive oxygen species increased with dose until reaching a maximum value at a drug concentration of 10 µM. Among others, malate dehydrogenase, calumenin, or the muscle isoenzyme of keratin kinase were found to be particularly susceptible to oxidation. Irreversible changes in enzymes involved in key metabolic processes can lead to the occurrence of multiple disorders in the body [57].
A study by Abdel-Wahab et al. in rats further revealed an association between oxidative stress and myocardial ischemia. Clozapine causes an increased release of catecholamines, which in turn increases the production of free radicals. As a consequence, cell damage occurs with the infiltration of neutrophils, which release cytokines, among others, TNF-α. Rats treated with CLO for 21 days showed a significant increase in MDA and 8-hydroxy-2-deoxyguanosine (8-OHdG), which was observed at doses of 15 and 25 mg/kg/day. This indicates lipid peroxidation and oxidative damage to DNA. The cardiotoxic effect was further confirmed by an increase in the activity of the biomarkers of cardiac damage—lactate dehydrogenase (LDH) and cardiac creatine kinase isoenzyme (CK-MB). At the same time, GSH levels decreased significantly [58]. Similar results were obtained during clozapine cardiotoxicity studies using striped danios. After a three-day exposure of embryos to the drug (concentration range 12.5–100 µM), cardiac morphological abnormalities and bradycardia were detected. In addition, levels of ROS, MDA, and pro-inflammatory cytokines increased. In contrast, CAT and SOD activity decreased. These results confirmed that CLO contributes to both oxidative stress and inflammation, thus leading to cardiotoxicity in preclinical studies [59].

2.2.3. Clinial CLO Studies

Clozapine-induced oxidative stress also affects blood cells in patients with agranulocytosis and in patients without agranulocytosis. A clinical study by Fehsel et al. suggests that the use of the drug is associated with an increase in ROS production in neutrophils, which may contribute to the induction of their apoptosis. In addition, increased expression of the proapoptotic gene p53 was observed, and this also occurred in patients treated with other antipsychotics, including olanzapine [60].
However, the results of a study comparing oxidative stress parameters in patients treated with perphenazine, risperidone, and clozapine led the authors to a completely different conclusion. Indeed, it has been shown that in patients treated with clozapine for at least three months, serum GSH and SOD levels increased, and MDA levels were significantly lower compared to patients treated with perphenazine and risperidone [61].

2.3. Risperidone (RIS)

2.3.1. Pharmacological RIS Data

During studies conducted in the 1960s and 1970s, it was observed that the administration of lysergic acid diethylamide (LSD) to rats produced symptoms similar to the negative symptoms of schizophrenia. This phenomenon has been linked to 5-HT2 receptor agonism and serotonin overactivation. A new target in the field of treatment of psychotic disorders has, therefore, become the development of compounds that block D2 and 5-HT2 receptors simultaneously. In the early 1980s, a benzoxazole derivative was obtained, which was classified as an atypical antipsychotic drug in the form of risperidone (RIS). The drug was approved by the US Food and Drug Administration in a relatively short time, as early as 1993, for the treatment of schizophrenia [62,63,64]. The drug also binds to α1adrenergic receptors and α2 and histamine H1 receptors. A particular advantage of the drug is its lack of affinity for muscarinic receptors; thus, there is little likelihood of anticholinergic symptoms developing. Although it is not without side effects and can lead to weight gain, hyperprolactinemia, hyperlipidemia, and diabetes, among others, it has a limited side effect profile compared to its predecessors [64,65,66,67,68].

2.3.2. Preclinical RIS Studies

With risperidone, the incidence of serious side effects such as extrapyramidal symptoms is limited, which is attributed to lower levels of oxidative stress [69,70,71,72]. Stojkovic et al. demonstrated that the drug affects parameters such as MDA, GSH, and SOD in the brain structures of rats. An increase in previously reduced GSH levels was observed in the brains of rats administered phencyclidine (PCP) initially for redox imbalance, followed by RIS for nine weeks. PCP led to oxidative damage in the brain and lipid peroxidation, but long-term administration of risperidone resulted in reduced levels of MDA and SOD [69].
However, some researchers indicate that risperidone has a cytotoxic effect by increasing the production of reactive oxygen species, thus inducing oxidative stress. In isolated hepatocytes of lab rats incubated with risperidone, a significant decrease in GSH levels was observed compared to the control group, as well as signs of cell damage, including, among others, a decrease in mitochondrial membrane potential and lipid peroxidation [72].

2.3.3. Clinial CLO Studies

Similar results were observed in a group of patients treated with RIS for 12 weeks at a dose of 6 mg/day. The concentration of SOD in the blood decreased significantly. However, the effect of the drug on NO levels remains inconclusive. The above study did not show a decrease in elevated NO levels in patients’ blood [70]. Different results were obtained in the blood of patients undergoing six weeks of pharmacotherapy with risperidone at individually selected doses in the range of 3–11 mg/day, as there was an increase in the levels of NO and its metabolites. At the same time, NO levels in the blood of patients with schizophrenia have also been shown to be significantly lower than in healthy individuals [71]. The aforementioned study by Hendouei et al. also assessed the effect of risperidone on oxidative stress parameters. Although clozapine showed a significant advantage in antioxidant activity, the total antioxidant capacity of plasma (FRAP, ferric reducing antioxidant power) reached the highest value compared to the patient group treated with risperidone [61].

2.4. Olanzapine (OLZ)

2.4.1. Pharmacological OLZ Data

In 1996, olanzapine (OLZ) was approved by the FDA for the treatment of schizophrenia, and in 2004, it was also approved for the treatment of bipolar disorder [73,74]. Due to its structural similarity to clozapine, olanzapine has a similar receptor profile. It shows high affinity for both dopamine receptors (e.g., D2, D4), serotonin (among others, 5-HT2A, 5H-T2C), adrenergic, histamine, as well as muscarinic. It has been shown that this drug also affects the glutamatergic system by interacting with NMDA receptors. However, the exact mechanism is not known [74,75]. The use of OLZ is associated with a limited risk of extrapyramidal symptoms and small increases in prolactin levels. Despite the above-mentioned advantages, the drug, especially during long-term therapy, significantly interferes with the body’s metabolic processes, leading to numerous disorders such as hyperlipidemia, diabetes, and weight gain. The latter is more common with olanzapine compared to other antipsychotics. Drowsiness, dry mouth, dizziness, and fatigue, as well as other symptoms, may also occur [73,74,75]. Olanzapine is classified as a second-choice drug due to its significant impact on metabolism. Nevertheless, due to its considerable efficacy, it continues to be one of the most commonly used second-generation antipsychotics, along with quetiapine, risperidone, and aripiprazole [41,68,76].

2.4.2. Preclinical OLZ Studies

There are numerous findings to compare oxidative stress with OLZ toxicity, including preclinical and clinical studies [77,78,79,80,81]. In numerous projects, olanzapine is compared with other antipsychotics, both first- and second-generation. In the animal study by Reinke et al., it was combined with haloperidol and clozapine. The obtained results indicate a significant advantage of atypical drugs over classical ones. Unlike haloperidol, they did not cause oxidative brain damage in the studied rats. A 28-day treatment with olanzapine administered intravenously at doses of 2.5, 5, and 10 mg/kg resulted in lower TBARS and carbonyl levels. Two-month treatment of rats with schizophrenia (doses of 10–20 mg/day) also resulted in improvements in oxidative stress parameters. Lipid peroxidation was assessed by analyzing MDA concentrations. Compared to pre-treatment values, biomarker levels decreased significantly [79].
Research from recent years also indicates an important role of the prefrontal cortex in the treatment of schizophrenia. Olanzapine-induced inflammation of this area and activation of the immune response are most likely linked to the pathogenesis of schizophrenia. Administration of the drug to rats for 1, 8, and 36 days (dose 3 mg/kg/day) resulted in activation of IKKβ/NF-κB inflammatory signaling pathways, increased expression of the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β, and immune system proteins including iNOS, TLR4, and CD14 in the prefrontal cortex [81].
OLZ also shows effects on hypothalamic cells, with dose- and time-dependent effects suggested. During in vitro studies on cell lines, a significant decrease in cell survival was observed at concentrations ≥100 μM after 24 h of incubation. Quantification of live hypothalamic cells confirmed the neurotoxicity of OLZ after both 24 and 48 h of incubation, which was for concentrations of 100 μM. No significant changes in cell survival were observed at a concentration of 25 μM. Based on the results, it can be concluded that olanzapine leads to dose-dependent neurotoxicity, where low concentrations of olanzapine (<100 µM) show beneficial antioxidant properties, but high doses (≥100 µM) result in cell damage [78].

2.4.3. Clinial OLZ Studies

Schizophrenic patients have been shown to have elevated levels of free radicals, with reduced levels of antioxidants. However, a significant number of studies show improvements in oxidative stress parameters in the blood of patients treated with OLZ. In the group of patients receiving the drug for 3 months at a dose of 5–20 mg/day, there was a decrease in the elevated MDA levels. The improvement was also noticeable for GSH, the concentration of which decreased significantly. In addition, the initially reduced levels of α-tocopheryl and ascorbic acid increased significantly. In this study, similar effects were obtained for risperidone, but better results in improving levels of oxidative stress biomarkers were obtained in the group of patients treated with olanzapine [77].
Total antioxidant status (TAS) was measured in schizophrenic patients. As a result of treatment, the blood levels of patients increased significantly. On the basis of the obtained data, researchers concluded that olanzapine showed a partial ability to regulate elevated levels of oxidative stress in patients with schizophrenia [80].

2.5. Quetiapine (QT)

2.5.1. Pharmacological QT Data

Quetiapine (QT), like clozapine and olanzapine, belongs to the dibenzoazepine derivatives. In 1997, it was approved by the FDA for the treatment of schizophrenia. In 2004, the registration indication was expanded to include the treatment of bipolar disorder and, a few years later, also the treatment of depression, with quetiapine being recognized as an adjunctive therapy drug [73,82,83]. It is characterized by strong antagonism to the 5-HT2A receptor. It binds to a small extent to the D2 receptor. Its sedative effect is most likely related to its very strong affinity for histamine H1 receptors. In turn, the occurrence of orthostatic hypotonia is explained by its affinity for α1-adrenergic receptors. Although the mechanisms of action of quetiapine are not fully understood, it is indicated that its metabolite—norquetiapine—influences antidepressant efficacy. This compound not only acts agonistically at 5-HT1A receptors but also inhibits norepinephrine transport factor (NET). Side effects include drowsiness, dry mouth, dyslipidemia, and weight gain. However, compared to olanzapine and clozapine, the intensity of metabolic disturbances occurring during quetiapine use is lower [41,68,73,83,84,85].

2.5.2. Preclinical QT Studies

Studies have shown that quetiapine presents different directions of action from the damaging effects of oxidative stress [86,87,88,89,90,91]. Han et al. induced free radical production by a seven-day intra-peritoneal administration of 20% ethanol (2 g/kg/day) to rats. An increase in total antioxidant capacity (TAC), SOD, and CAT activity was observed in brain areas of animals receiving the drug at a dose of 10 mg/kg/day. This effect was not observed in the liver, where there was an increase in MDA and ROS concentrations despite drug administration [86]. Similar results were also obtained with fourteen days of quetiapine therapy (20 mg/kg), which produced a significant increase in SOD activity and a decrease in TBARS levels in the rat brain areas studied (hippocampus, amygdala, and prefrontal cortex). This indicates a reduction in lipid peroxidation and protein damage, thereby reducing oxidative stress [87]. Due to the significant effectiveness of quetiapine in the treatment of cognitive deficits, studies were also conducted to assess its impact on behavioral changes resulting from anticancer treatment with doxorubicin. The results of the 30-day study revealed that oxidative stress and inflammation parameters were improved in the brain tissue of the rats. Quetiapine administration reduced high concentrations of MDA and GSH in the brains of rats [91].

2.5.3. Clinical QT Studies

In contrast to preclinical data, determination of myeloperoxidase (MPO) and C-reactive protein (CRP) activity in patients after QT overdose revealed oxidative dysfunction and the development of inflammation. Levels of both biomarkers increased significantly, which was further related to serum drug concentration and the patient’s clinical condition. Quetiapine concentrations above 1 μg/mL in blood correlated with both increased MPO and CRP levels [88]. However, in other studies conducted with quetiapine-treated patients, the antioxidant effect of the drug has been observed. After four weeks of therapy, a decrease in baseline-elevated plasma TBARS and urinary F2-isoprostane concentrations was noted. Initial levels of the lipid peroxidation marker 4-hydroxynonenal (4-HNE) in the schizophrenia group were not significantly different from the control group, and these levels did not change after quetiapine treatment. This study also allowed a comparison of the antioxidant properties of selected atypical antipsychotics. Quetiapine, together with clozapine and olanzapine, showed the most beneficial effect on oxidative stress parameters [89].
This effect was also confirmed during in vitro studies using blood drawn from healthy volunteers. Significant reductions in lipid peroxidation in the form of reduced TBARS levels occurred after both 1 and 24 h of plasma incubation with the drug [90].

2.6. Aripiprazole (ARP)

2.6.1. Pharmacological ARP Data

In 2002, the FDA approved the new drug aripiprazole (ARP) for the treatment of schizophrenia. As a quinoline derivative, it exhibits a unique mechanism of action, making it classed as a third-generation antipsychotic. Like quetiapine, aripiprazole has received approval for the treatment of bipolar affective disorder and depression as adjunctive therapy. This occurred in 2004 and 2007 [73]. It is called a dopamine system stabilizer. When the activity of the dopaminergic system is reduced, it acts as an agonist for D2 receptors by stimulating them. In contrast, when dopamine concentrations are too high, aripiprazole acts as an antagonist. This allows for antipsychotic effects while reducing the risk of EPS. This drug also has an affinity for 5-HT1A, 5-HT2A, 5-HT7, and D3 receptors and moderate activity at D4, H1, and α1-adrenergic receptors. A particular advantage of aripiprazole is its beneficial effect on prolactin. This is because it does not increase serum concentrations of the hormone. In addition, it can lead to the normalization of prolactin levels in patients whose levels were initially elevated. The most common side effects include drowsiness, akathisia, anxiety, nausea, vomiting, and headache [56,68,73,92,93,94,95,96].

2.6.2. Preclinical ARP Studies

Preclinical, in vitro, and clinical studies have shown that ARP can generate or reduce oxidative stress [97,98,99,100,101]. It was observed that in both isolated mouse liver mitochondria and in isolated blood cells from healthy patients, mitochondrial respiration was disrupted by aripiprazole administration. This drug led to the induction of reactive oxygen species production and lipid peroxidation, as indicated by increased levels of modified HNE proteins [97].
In contrast, some studies confirm the induction of an antioxidant response by ARP. In view of its potential antidepressant effect, an analysis was made of the drug’s effect on oxidative stress parameters that also play an important role in the pathogenesis of depression. Lipid peroxidation levels in the cerebral cortex of rats receiving aripiprazole (2.5 mg/kg) for four weeks did not differ from those of the control group. However, glutathione peroxidase activity, GSH, and vitamin C concentrations increased significantly [99].
Recent studies also indicate a potential antioxidant effect on neurons. Administration of aripiprazole to rats for 30 days at doses of 1 and 2 mg/kg resulted in reduced levels of the oxidative stress marker MDA and the inflammatory marker COX-2 for both drug concentrations. In addition, the antioxidant activities of GSH and CAT increased. According to the researchers, the results obtained demonstrate the neuroprotective mechanism of action of aripiprazole [100].
Equally beneficial effects of the drug on antioxidant protection and cell viability were observed in relation to rat hepatocytes. Under oxidative conditions, after eight weeks of treatment, there was an increase in the activity of the antioxidant enzymes, SOD and CAT, which applied to both 2.23 and 6 µM ARP concentrations. At the same time, the dose-dependent effect of the drug was shown, and the largest increase in enzymes occurred after the drug was administered at a higher concentration [101].

2.6.3. Clinical ARP Studies

In contrast, the results of a clinical study conducted to assess the effect of therapeutic doses of antipsychotics on oxidative stress parameters suggest that aripiprazole has a dose-dependent effect. Plasma samples from healthy volunteers were incubated with the drug for 24 h. For both drug concentrations of 163 and 242 ng/mL, no significant effect was observed on plasma TBARS levels. However, at lower concentrations, there was a slight induction of lipid peroxidation, indicating possible prooxidant properties [98].

3. Mood Stabilizers

3.1. Lithium (Li)

3.1.1. Pharmacological Li Data

The origins of the use of lithium (Li) date back to the 19th century, but the first publications demonstrating its effectiveness were published in the mid-20th century [102,103]. It is currently used mainly in the treatment of schizoaffective disorders, including bipolar mood disorders, as a mood stabilizer [104]. It is also suggested for use in the treatment of schizophrenia as a next-line treatment and in severe forms of depression due to its anti-suicidal properties [105,106]. Its use in neurodegenerative diseases has also been postulated [107,108,109]. Despite its long history of use, the mechanism of action of lithium is not yet fully understood. However, two main mechanisms of action have been postulated: by blocking inositol phosphatases involved in intracellular signal transduction and by inhibiting glycogen synthase kinase 3 [110,111]. In addition, it increases GABA-based inhibitory neurotransmission, which is of particular importance in the treatment of mania [112]. Side effects associated, particularly with long-term use, include kidney damage and endocrine disruption in the form of deterioration of thyroid functions [113,114].

3.1.2. Preclinical Li Studies

Preclinical, clinical, and in vitro studies have shown that lithium enhances or inhibits oxidative stress [115,116,117,118,119,120,121,122,123]. In preclinical studies conducted on rats induced into a state of mania by methamphetamine, it was shown that the oxidative effect of lithium is influenced by the dose of the substance inducing the manic state in the model and the site of action in the nervous tissue [118]. Results confirming the antioxidant properties of lithium were also shown in a study by Jornad et al., which was also conducted on rats introduced into a model of mania. SOD activity was statistically significantly reduced with lithium treatment compared to the group introduced into mania without lithium treatment in both the prefrontal and hippocampus. In the case of CAT, increased activity was also observed in both brain regions. In addition, a significant reduction in TBARS was also observed after lithium therapy in neural tissue. In contrast, in non-manic animals, the use of lithium did not affect oxidative stress parameters in any case [119]. Another study in rats showed a significant increase in SOD levels only in the group treated with higher concentrations of lithium, suggesting a dose-dependent increase in oxidative damage [121].
Additionally, in an in vitro model study, lithium was shown to inhibit oxidative stress-induced senescence in nerve cells [120]. In a study conducted on rat liver cells in vitro, lithium cytotoxicity associated with its participation in the formation of oxygen-free radicals was demonstrated. Moreover, cytotoxicity increased with increasing ion concentrations. In addition, the role of the CYP2E1 isoenzyme in oxidative stress associated with the intake of lithium was postulated [123].

3.1.3. Clinical Li Studies

A clinical study conducted on a group of bipolar disorder patients treated with lithium showed a statistically significant reduction in TBARS level, an exponent of increased lipid peroxidation, after starting therapy. Similar observations have been shown for superoxide dismutase (SOD). CAT and GPx levels did not change statistically significantly after lithium treatment but were significantly higher compared to the control group, which is explained by the pathophysiology of the disease. In addition, changes in oxidative stress parameters have been linked to the response to lithium therapy [115,116]. A study by Banerjee et al., also conducted on a group of bipolar disorder patients, showed significantly reduced lipid peroxidation exponents in lithium-treated patients compared to lithium-naive patients. Moreover, an increase in Na+-K+-ATPase activity associated with lithium treatment and affecting the reduction of oxidative stress was demonstrated [117]. In contrast, a study in healthy volunteers showed a statistically significant reduction in SOD after Li therapy, with no significant effect on CAT or TBARS levels [122].

3.2. Valproic Acid (VPA)

3.2.1. Pharmacological VPA Data

The action of valproic acid (VPA) is based on blocking voltage-gated ion channels and modulating GABA-nergic and monoamine-based transmission within neural tissue. Through the induction of glutamic acid decarboxylase, VPA increases levels of GABA, which is an inhibitory neurotransmitter. In addition, VPA inhibits the enzymes that break down GABA, which further contributes to its levels. This reduces impulse conduction, which translates into a reduction in the occurrence of convulsions. The effect on NMDA receptor-based conduction is also not insignificant. This is carried out by weakening the action of glutamate, which is a stimulating neurotransmitter. In addition, VPA reduces glucose utilization within the brain, which results in reduced ATP production and indirectly translates into a slower metabolism in the neural tissue. Common side effects associated with the use of VPA include weakness, lethargy, nausea, weight gain, and hair condition deterioration. In addition, hematological disorders and hepatotoxicity associated with VPA intake are observed. In addition, valproic acid has shown significant teratogenic properties [124,125,126,127,128,129,130,131,132,133,134,135,136].

3.2.2. Preclinical VPA Studies

The use of valproic acid affects the redox balance in the body [137,138,139,140,141,142,143,144,145]. Studies in rats have shown VPA-associated significant reductions in GTx, SOD, and CAT activity in neural tissue. In addition, oxidative stress exponents such as TBARS and carbonyls have been shown to be elevated [137]. However, this effect can be inhibited by selenium and L-carnitine supplementation, which has also been confirmed in studies conducted on rats [138,139]. In turn, work carried out on the same animal model showed neuroprotective properties of VPA against nerve cells after the occurrence of mechanical trauma in the mechanism of inhibiting oxidative stress [140]. VPA also has a similar effect in the case of nerve tissue damage as a result of stroke, which has been demonstrated in studies on the same animal model [141].
Studies conducted on human neuroblastoma cells in an in vitro model showed a reduction in hydrogen peroxide levels in cells subjected to stimulated excitotoxicity in the case of incubation with VPA. In addition, an increase in SOD and a slight increase in CAT were also observed in samples incubated with VPA. However, when excitotoxicity was induced, the relationships were not statistically significant [142]. Also, a study conducted on a mouse embryo model showed an increase in reactive oxygen species associated with VPA administration and a negative impact on the embryo. In addition, administration of exogenous CAT showed potential embryonic protective properties, which could not be demonstrated for SOD [144]. Elevated markers of oxidative stress associated with the use of VPA have also been demonstrated in work on glioma cell lines. VPA-related reactive oxygen species caused damage within mitochondria, affected cell–cell communication pathways, and regulated VPA-induced apoptosis [145].

3.2.3. Clinical VPA Studies

A clinical study conducted on a group of VPA-treated patients showed a significant reduction in antioxidant potential (FRAP) between the VPA-treated and the control group. Importantly, no such relationship was shown for reduced to oxidized glutathione (GSH/GSSG ratio) [143].

3.3. Carbamazepine (CBZ)

3.3.1. Pharmacological CBZ Data

The origins of the use of carbamazepine (CBZ) date back to the mid-20th century. Its mechanism is based on the modulation and blocking of potential-gated sodium channels. Its effects on serotonin- and catecholamine-based conductance have also been postulated. Among its other uses are the treatment of seizures, neuropathic pain, and manic episodes in the course of bipolar disorder. Side effects associated with its use include feelings of fatigue, headaches, blurred vision, and ataxia. Cases of Stevens–Johnson syndrome, severe hematological disorders, and toxic epidermal necrolysis associated with carbamazepine use have also been described [146,147,148,149,150,151,152].

3.3.2. Preclinical CBZ Studies

The studies of the effect of CBZ on the condition of oxidative stress are varied [153,154,155,156,157,158,159,160]. In vitro studies on erythrocytes showed no significant effect of the drug on oxidative stress in the cell [155]. In contrast, a study conducted on a plant cell model showed an increase in lipid peroxidation and levels of hydroperoxide and carbonyl proteins. In addition, a statistically non-significant increase in CAT levels was observed [160].
Studies of CBZ nephrotoxicity in rats suggest an association of renal cell damage with increased rates of oxidative stress [156]. In another study conducted on rats, no significant changes in malondialdehyde, glutathione, and SOD levels were observed in neural tissue during CBZ treatment. A significant decrease in GSH and SOD levels and an increase in MDA levels were only visible in the group with induced epilepsy [157]. A study in another animal model showed an increase in lipid peroxidation and hydroperoxide levels after exposure to CBZ. At the same time, a reduction in SOD, CAT, and GPx activity was confirmed [158]. Similar conclusions were reached in a similar study on the same animal model. A reduction in SOD, CAT, and GPx activity was shown with a concomitant increase in TBARS and carbonyl protein [159].

3.3.3. Clinical CBZ Studies

A study conducted on a pediatric population showed a significant reduction in antioxidant capacity and an increase in the oxidative stress index among epilepsy patients treated with carbamazepine compared to a healthy control group. In addition, an association was found between the use of CBZ and increased total oxidative status [160]. Similarly, a study by Varoglu et al. found a significant reduction in some antioxidative markers, with an increase in selected oxidative markers during carbamazepine monotherapy [154].

3.4. Oxcarbazepine (OXC)

3.4.1. Pharmacological OXC Data

The origins of the use of oxcarbazepine (OXC) in therapy date back to the 1990s. It is a structural derivative of carbamazepine, which is a pro-drug that is activated in the body as an active metabolite. It is postulated to act mainly by blocking and influencing voltage-gated sodium channels. The primary uses of the drug include the treatment of epilepsy and affective disorders. Compared to carbamazepine, OXC is characterized by greater safety and fewer side effects and interactions. Common side effects associated with OXC use include dizziness, headaches, and nausea [161,162,163,164,165,166].

3.4.2. Preclinical OXC Studies

The studies of the effect of OXC on the status of oxidative stress are not very numerous and clear [167,168,169,170,171]. Studies in mice have shown no significant effect of OXC use on levels of oxidative stress exponents such as MDA, GSH, CAT, and SOD determined in brain tissue. Elevated MDA levels and reduced glutathione levels were only observed with OXC use in individuals with pharmacologically induced epilepsy [168]. In turn, studies conducted in rats showed a significant protective effect of the use of OXC on nerve cells after hypoxia induced by cardiac arrest. It is postulated that these protective properties result from the attenuation of oxidative stress by increasing the presence of SOD [169]. The neuroprotective properties of OXC through its effect on redox balance are also confirmed by a rodent study by Park et al., which showed a reduction in the production of superoxide anions.
In addition, elevated levels of SOD, CAT, and GPx were observed in nerve cells [170]. An increase in the formation of ROS as a result of epileptic seizures has been demonstrated [171].

3.4.3. Clinical OXC Studies

A meta-analysis by Rezaei et al. found no significant statistical difference between OXC-treated and healthy individuals in blood-assessed homocysteine levels, which are directly related to redox balance regulation [167]. Similar observations have also been made regarding folic acid and cobalamin’s indirect involvement in the regulation of oxidative stress [168].

4. Antidepressants

4.1. Citalopram (CIT)

4.1.1. Pharmacological CIT Data

Citalopram (CIT) is a selective serotonin reuptake inhibitor (SSRI), authorized in 1998. In addition, it has a low affinity for dopaminergic D2 receptors, 5-HT1A and 5-HT2A receptors, α-adrenergic and β-adrenergic, muscarinic, and histamine H1 receptors [172]. It is used in the treatment of depressive disorders, anxiety disorders with or without anxiety attacks with agoraphobia, as well as obsessive-compulsive disorders. Common side effects include sleep disturbances, nausea, decreased sex drive, excessive sweating, and dry mouth [173].

4.1.2. Preclinical CIT Studies

The research results provide a lot of data on the impact of CIT on oxidative stress [174,175,176,177,178,179,180,181]. Hepatotoxicity of CIT, resulting from free radical effects, has been observed in rat studies. Treatment took place in vitro and in vivo. In the in vitro group, hepatocyte death was noted at 500 µM, as well as increased ROS formation, breakdown of mitochondrial potential, lysosomal membrane leakage, GSH depletion, and lipid peroxidation. The in vivo tests at a dose of citalopram (20 mg/kg), confirmed the above damage [174].
In addition, other studies conducted on male rats have shown that CIT causes testicular damage to the mechanisms of oxidative stress as well as hormonal changes. The drug was used at doses of 5, 10, and 20 mg/kg. Sperm showed abnormal morphology and DNA damage, while their concentration decreased. In addition, histopathological changes in the testicles were observed. Luteinizing hormone concentrations increased with each dose of citalopram, while testosterone concentrations increased with doses of 5 and 10 mg/kg. Reduced glutathione levels signaled increased oxidative stress in the male rats given citalopram at doses of 10 and 20 mg/kg [175]. Studies conducted on Daphnia magna have shown that citalopram causes oxidative damage. The microorganisms were exposed to citalopram in the water at a concentration of 1.03 mg/L. In Daphnia, an increase in the concentration of the antioxidant molecule glutathione S-transferase (GST) was observed, as well as an increase in the activity of SOD, CAT, and GPx. In addition, MDA, protein carbonyl, and 8-OHdG concentrations increased. CIT has been shown to cause oxidative damage [176].
Antitumor effects of citalopram on hepatocellular carcinoma cells have also been observed. The cytotoxic effect of the drug on HepG2 cells was manifested by decreased cell viability and increased ROS formation. An increase in mitochondrial Bax and a decrease in Bcl2, as well as cytochrome C release, were observed. It has been suggested that citalopram may exhibit apoptotic effects against a hepatocellular carcinoma cell line via induction of cell death via a cytochrome C release mechanism and ROS-dependent activation of NFκB [177].
In addition, studies on the effect of citalopram on the processing of amyloid-β precursor protein (AβPP) have suggested that the drug may increase AβPP processing and also reduce oxidative stress. The study was conducted on induced pluripotent stem cells (iPSCs). Citalopram was administered for 45 days at concentrations of 0.8, 5, and 10 µM. In cells with the PSEN1 gene mutation, O2 concentrations were significantly reduced after each dose of citalopram. In contrast, no change in the formation of terminal oxidative stress markers was observed after treatment [180].
In addition, in studies conducted in rats, it was observed that co-administration of citalopram combined with rosuvastatin intensifies oxidative stress. The concomitant medications were used for 14 days. The results showed an increase in peroxidase and glutathione reductase activity, with no effect on antioxidant concentrations [178]. On the other hand, in studies conducted in Wistar rats, a neuroprotective effect of citalopram was observed, induced by the alleviation of oxidative stress, inflammation, apoptosis, and an altered metabolic profile. Treatment with CIT at 4 mg/kg significantly altered 17 metabolites with attenuation of malondialdehyde, reduced glutathione, matrix metalloproteinases, and apoptosis markers [179].

4.1.3. Clinical CIT Studies

A limited number of studies examine oxidative stress measured in patient groups. There was a significant increase in serum SOD, serum MDA, and decrease in plasma ascorbic acid levels in patients treated with CIT for major depression as compared to control subjects [181].

4.2. Sertraline (SER)

4.2.1. Pharmacological SER Data

Sertraline (SER) was introduced in 1991. It is a naphthaleneamine derivative. The mechanism of action of the drug is to inhibit the presynaptic reuptake of serotonin from the synaptic cleft (SSRI). Norepinephrine and dopamine transport are also blocked to a small extent. Sertraline slightly binds to adrenergic, histamine, muscarinic, dopaminergic, serotonergic, benzodiazepine, and GABA receptors. It is used to treat depressive disorders, obsessive-compulsive disorder, post-traumatic stress disorder, and seizure disorders. Common side effects include insomnia, drowsiness, anorexia, tremors, dizziness and headaches, diarrhea, nausea, or sexual dysfunction. In addition, movement disorders, paresthesias, visual disturbances, or increased sweating have been noted. In addition, sertraline-induced hepatotoxicity has also been described [182,183,184].

4.2.2. Preclinical SER Studies

The effects of SER on lipoperoxidation and antioxidant enzyme activity in plasma and brain tissues were studied in Wistar albino rats. Animals were administered the drug at doses of 10, 40, and 80 mg/kg. Lipid peroxidation (MDA) levels in plasma and brain tissue increased in all rats treated with sertraline. SOD activity in brain tissue decreased, while plasma concentrations increased compared to the control group. In contrast, plasma and brain tissue CAT concentrations and plasma paraoxonase (PON) concentrations decreased compared to the control group. Administration of higher doses of sertraline has been found to increase oxidative stress [185]. Studies in Drosophila melanogaster have shown sertraline-induced DNA damage and cell toxicity. Antidepressant-treated larvae manifested delayed development and reduced survival. It was observed that, through the action of sertraline, mitotically active tissues had DNA breaks and underwent apoptosis with increased frequency. Interestingly, the toxicity of the drug was partially mitigated by the administration of ascorbic acid. It has been suggested that sertraline induces oxidative DNA damage [186]. Sertraline-induced hepatotoxicity is probably due to mitochondrial dysfunction. The study was conducted on male Sprague Dawley rats. It has been observed that at doses of 75 and 100 µM, sertraline inhibits mitochondrial function by uncoupling oxidative phosphorylation and inhibiting the activity of complexes I and V. This process is further mediated by the loss of ATP and the induction of microsomal TG transport protein (MPT) [184].
The effects of sertraline (also in combination with haloperidol) on oxidative stress in mice were also compared. SER was administered at doses of 10–20 mg/kg. In antidepressant monotherapy, brain concentrations of GSH, MDA, TAC, and nitrite remained unchanged, while catalase and PON1 activity decreased. It has been found that sertraline, through its effects, can expose the brain to further oxidative damage [187]. Importantly, some studies suggest using the oxidative properties of sertraline to treat certain diseases. Sertraline can be used in the treatment of prostate cancer. Antidepressant-induced apoptosis and autophagy by activating free radical production, hydrogen peroxide (H2O2) formation, lipid peroxidation, and decreasing GSH concentration. Furthermore, sertraline significantly reduced the expression of aldehyde dehydrogenase 1 (ALDH1) and the stem cell marker CD44 [188].
Other studies have investigated the oxidative properties of sertraline against the Leishmania infantum parasite. The drug-induced uncoupling of respiration, a significant decrease in intracellular ATP concentration, and oxidative stress in the promastigote L. infantum. In addition, prolonged metabolic dysfunction was demonstrated. Sertraline killed Leishmania through a multidirectional mechanism of action, eliminating the parasite’s primary metabolic pathways. The use of sertraline to treat visceral leishmaniasis in an off-label regimen has been suggested [189].

4.2.3. Clinical SER Studies

In contrast, a pilot study comparing the effects of sertraline on markers of oxidative stress (MDA) revealed a different effect on oxidative properties. People with depression, also diagnosed with heart failure, were divided into two groups: those receiving sertraline and those not receiving any antidepressant. After three months of treatment, patients treated with sertraline showed a significant reduction in MDA. It has been suggested that SER treatment reduces plasma markers of oxidative stress in depressed patients with additionally diagnosed heart failure [190].

4.3. Venlafaxine (VEN)

4.3.1. Pharmacological VEN Data

Venlafaxine (VEN) was launched in 1994 [191]. This substance, together with its metabolite—O-desmethylvenlafaxine—acts as a serotonin and norepinephrine reuptake inhibitor (SNRI). In addition, venlafaxine shows weak properties as a dopamine reuptake inhibitor. Its serotonergic effect contributes to sedative, drowsiness-increasing, and neutralizing effects, while its noradrenergic action can result in the opposite effects, i.e., agitation, deterioration of sleep quality, and sometimes even induce feelings of anxiety. The drug is approved for a variety of uses, such as the treatment of episodes of major depression, prevention of recurrent episodes of major depression, treatment of generalized anxiety disorder, treatment of social phobia, and treatment of paroxysmal anxiety [192]. Most patients tolerate VEN well, although symptoms such as dizziness and headache, nausea, dry mouth, and night sweats are most commonly seen in clinical practice. An increase in blood pressure is also observed, especially at the beginning of therapy or with high daily doses (300 mg/day) [193]. Other potential side effects mentioned in the literature include tinnitus, palpitations, sleep disturbances, vomiting, diarrhea, constipation, decreased appetite, and nervousness [194].

4.3.2. Preclinical VEN Studies

A study in male mice assessed venlafaxine’s ability to damage DNA in the brain and liver and its oxidative effects on DNA, lipids, and proteins. An intermediate dose of the drug (50 mg/kg) caused significant DNA damage at 2 and 6 h after exposure, while a high dose (250 mg/kg) caused even more pronounced oxidative damage over the same time interval. Damage was observed in both the brain and liver, with the liver being, however, more severe. Lipoperoxidation effects in the brain and liver were evident after 6 and 12 h at doses of 50 and 250 mg/kg. In contrast, liver nitrite content induced 2 h after exposure to venlafaxine (250 mg/kg) was elevated [195].
Similar observations were made on the effect of VEN on hepatocyte cytotoxicity—in this study, the drug was administered to male rats, and then liver cells were isolated from them. Venlafaxine has been observed to cause hepatotoxicity through the induction of oxidative stress and subsequent toxic effects, including GSH depletion, lipid peroxidation, breakdown of mitochondrial potential, and lysosomal membrane leakage in hepatocytes. In addition, functional mitochondrial damage was observed through inhibition of mitochondrial respiratory complexes II and IV. Lysosomes and mitochondria have been shown to be the earliest target structures for venlafaxine-induced hepatotoxicity [196].
In addition, chronic VEN administration affects the methylation of promoters of genes involved in oxidative and nitrosative stress. This includes mRNA expression of SOD1, SOD2, and NOS1 in peripheral blood mononuclear cells (PBMC) and in the brain, as well as gene expression of CAT, Gpx1, and Gpx4 only in the brain [197]. In a study in mice, co-administration of VEN (40 mg/kg) with morphine showed inhibition of naloxone-induced withdrawal symptoms. In addition, the drug reduces the expression of TNF-α, IL-1β, IL-6, NO, and the concentration of MDA in brain tissue. Repeated administration of venlafaxine inhibits the decline in brain-derived neurotrophic factor (BDNF), thiol, and GPx [198]. Studies evaluating the effects of venlafaxine on glucose homeostasis and oxidative stress showed that the drug’s effects were associated with a TBARS decrease and an increase in GSH levels in the brain. Mice with diabetes were studied; VEN was administered at doses of 8 and 16 mg/kg/day for 21 days. Glucose levels did not change significantly after administration [199]. No clinical VEN study results were found in the literature.

5. Discussion

Understanding the mechanisms and role of oxidative stress in the possible etiopathogenesis of psychiatric diseases makes it possible to develop new therapeutic approaches based on its modulation. Such research has been conducted in the case of bipolar disorder, where oxidative stress has been shown to affect molecule damage at the neurobiological level. This observation makes it possible to try to use drugs that affect the balance between antioxidative and oxidative processes to induce remission of the disease [1].
A compilation of the many publications on in vitro and animal experiments and the few findings from studies conducted in patient groups indicates considerable interest in the topic of the involvement of oxidative stress in the mechanism of many neurodegenerative diseases treated with older and newer generation drugs.
The findings indicate that significant differences can be observed in clinical practice regarding the prescribing of the classic drug haloperidol. Although second-generation drugs are preferred in most cases, haloperidol still stands out among the classic antipsychotics, as its prescription frequency has not changed significantly over the past decade [41,42,43]. Despite this, the contribution of oxidative stress during HLP use is still not fully understood. A substantial body of research allows the authors to conclude that haloperidol increases oxidative stress in brain tissue, showing a tendency to increase neuronal damage [47]. Hence, one of the main directions of recent research is the study of the interaction of antipsychotic drugs, including haloperidol, with other substances. This is because they can exhibit both adverse and beneficial joint effects on oxidative stress parameters [44,47,51].
Continued research among neuropsychiatric drugs has confirmed the significant efficacy of clozapine in the treatment of symptoms of psychosis, particularly drug-resistant schizophrenia [52,53]. Studies on the involvement of clozapine in oxidative stress further revealed the cardiotoxic effects of the drug, confirmed by elevated activities of biomarkers of cardiac damage, i.e., LDH and cardiac creatine kinase isoenzyme, CK-MB, with a concomitant decrease in GSH levels, such an important antioxidant in the body [58]. However, the results of a study comparing oxidative stress parameters in patients treated with perphenazine, risperidone, and clozapine led the authors to a completely different conclusion. Indeed, it was shown that in the group of patients treated with clozapine for at least three months, serum GSH and SOD levels increased, and MDA levels were significantly lower compared to patients treated with perphenazine and risperidone. It has, therefore, been suggested that clozapine, contributes to a significant alleviation of the negative symptoms of schizophrenia by exhibiting antioxidant effects [61].
The results suggest that atypical antipsychotics (clozapine, risperidone) have a more favorable effect on levels of oxidative stress biomarkers compared to classical drugs (perphenazine), which may be important in the context of both schizophrenia treatment and symptom relief [61]. The success of clozapine led to a search for its chemical analog, which would not be associated with such a high risk of developing agranulocytosis. This research allowed the synthesis of olanzapine, a new tricyclic dibenzoazepine derivative belonging to the second generation of antipsychotics. The drug quickly gained popularity after its approval and, along with risperidone, became one of the most widely used antipsychotics. Its prescribing frequency declined slightly in the 2000s, most likely driven by patient concerns about side effects [41,68,76].
Numerous studies have been conducted to clarify the relationship between OLZ treatment and changes in antioxidant and oxidant levels in the body [77]. Given the lack of oxidative damage induced by olanzapine, researchers point to a possible neuroprotective mechanism of action of the drug [79]. The results suggest that the use of olanzapine in the short term, unfortunately, leads to the development of inflammation in both the central and peripheral nervous system, which induces the occurrence of altered immunological responses in patients with schizophrenia. These changes may be related to the therapeutic effects of olanzapine, but their association with adverse side effects, e.g., weight gain, is also not excluded [81].
QT shows a complex effect on oxidative stress. As with the other second-generation drugs, research results indicate their antioxidant properties [86,87]. The authors pointed to the potential use of quetiapine to improve cognition and reduce oxidative damage to the brain, drawing additional attention to the drug’s neuroprotective properties. Indeed, a reduction in Bax and caspase-3 protein levels provides protection against neuronal apoptosis [91,96]. The favorable side-effect profile makes aripiprazole one of the drugs preferred by patients. Along with risperidone, quetiapine, and olanzapine, it is the most commonly prescribed atypical antipsychotic drug [56,68,96]. Given the unique mechanism of action of aripiprazole and its favorable side-effect profile, it is expected to have an equally favorable effect on oxidative stress parameters. However, the studies conducted do not provide a clear answer regarding the potential pro- or antioxidant effect of the drug [97]. Recent studies also indicate a potential antioxidant effect on neurons. According to the researchers, the results obtained demonstrate the neuroprotective mechanism of action of aripiprazole [100]. Based on the results, it was inferred that aripiprazole provides potential protection for liver cells under oxidative conditions found in people with schizophrenia [101].
The effect of lithium on oxidative status is still not fully understood. A number of factors have now been postulated to influence the effect of lithium on oxidative stress. Among them, factors related to the conduct of therapy should be distinguished, such as the doses used, the time of taking, or interactions with other drugs used. The type of tissue affected by lithium also has an impact. Furthermore, when assessing the effect of lithium on oxidative stress, the pre-existing redox balance disturbances, which form the pathophysiological basis of diseases, must also be taken into account [114,115]. A study in healthy volunteers showed a statistically significant reduction in SOD after lithium therapy, with no significant effect on CAT or TBARS levels [122]. In a study conducted on rat liver cells in vitro, Li cytotoxicity was demonstrated to be associated with participation in the formation of oxygen-free radicals. Moreover, cytotoxicity increased with increasing blood lithium concentrations. In addition, the role of the CYP2E1 isoenzyme in oxidative stress associated with the intake of lithium was postulated [123].
Valproic acid and carbamazepine were among the first drugs to be used in the treatment of epilepsy. Toxicity related to the oxidative stress effects of VPA is also seen in rat hepatocyte cells, which is related to the intensive and multi-step hepatic metabolism of the drug [139].
Capacity and increased oxidative stress index among epilepsy patients treated with carbamazepine compared to a healthy control group. In addition, a correlation was found between the use of carbamazepine and increased total oxidative status [153].
The effect of oxcarbazepine on oxidative stress remains a subject of research [166]. Also, in the case of epilepsy, the role of oxidative stress as one of the components in epileptogenesis has been demonstrated [171].
The effect of citalopram on the development of oxidative stress is important. Studies on Daphnia magna have shown that citalopram causes oxidative damage [176]. Antitumor effects of citalopram on hepatocellular carcinoma cells have also been observed. It has been suggested that citalopram may exhibit apoptotic effects against a hepatocellular carcinoma cell line via induction of cell death via a cytochrome C release mechanism and ROS-dependent activation of NFκB [177].
The topic of sertraline-induced oxidative stress is frequently addressed in research. Studies in Drosophila melanogaster have shown sertraline-induced DNA damage and cell toxicity. Sertraline-induced hepatotoxicity is probably due to mitochondrial dysfunction [184]. Significantly, some studies suggest using the oxidative properties of sertraline to treat certain diseases. Sertraline can be used in the treatment of prostate cancer [188]. In contrast, a pilot study comparing the effects of sertraline on markers of oxidative stress (MDA) revealed different effects on oxidative properties [190].
The relationship between venlafaxine and oxidative stress is a topic addressed in the literature. Studies evaluating the effects of venlafaxine on glucose homeostasis and oxidative stress showed that the drug’s effects were associated with a decrease in TBARS and an increase in brain GSH levels [198].

6. Conclusions

In conclusion, oxidative stress is an important risk factor for many psychiatric disorders in the treatment of which first- and second-generation antipsychotics are most commonly used, mood stabilizers, and antidepressants, and this publication reviews the state of the art of the contribution of oxidative stress to the most commonly used treatments of neuropsychiatric diseases, with first- and second-generation antipsychotics as important in treatment success as discoveries in the pathophysiology of neuropsychiatric diseases, including those initiating a new round of innovations in the contribution of oxidative stress to the etiology of neuropsychiatric diseases. For neurological patients, it is important to identify the cause of the oxidation–reduction disturbances, which can be led by pharmacological drug properties, mechanism of action, drug–drug interaction, dose, and nutritional status [2,6,8,77,99,138,139,167,168,181,200]. All changes should be taken into account in oxidative stress generation because it is necessary to understand the characteristics and scope of each neuropsychiatric drug used in therapy.

Author Contributions

Conceptualization, K.S.-K.; methodology, W.J.; software, S.R. and F.N.; formal analysis, K.S.-K., W.J., S.R. and F.N.; resources, M.Ł.-G. and P.Ś.; writing—original draft preparation, K.S.-K.; writing—review and editing, K.S.-K., W.J., S.R. and F.N.; visualization, K.S.-K.; supervision, B.Z.-P.; project administration, C.Ż.; funding acquisition, Z.Ż. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sachdev, P.S. Whither Neuropsychiatry? J. Neuropsychiatry Clin. Neurosci. 2005, 17, 140–144. [Google Scholar] [CrossRef] [PubMed]
  2. Salim, S. Oxidative Stress and Psychological Disorders. Curr. Neuropharmacol. 2014, 12, 140–147. [Google Scholar] [CrossRef] [PubMed]
  3. Morandi, A.; Davis, D.; Bellelli, G.; Arora, R.C.; Caplan, G.A.; Kamholz, B.; Kolanowski, A.; Fick, D.M.; Kreisel, S.; MacLullich, A.; et al. The Diagnosis of Delirium Superimposed on Dementia: An Emerging Challenge. J. Am. Med. Dir. Assoc. 2017, 18, 12–18. [Google Scholar] [CrossRef] [PubMed]
  4. Adamis, D.; Van Munster, B.C.; Macdonald, A.J.D. The Genetics of Deliria. Int. Rev. Psychiatry 2009, 21, 20–29. [Google Scholar] [CrossRef] [PubMed]
  5. Lees, B.; Meredith, L.R.; Kirkland, A.E.; Bryant, B.E.; Squeglia, L.M. Effect of Alcohol Use on the Adolescent Brain and Behavior. Pharmacol. Biochem. Behav. 2020, 192, 172906. [Google Scholar] [CrossRef]
  6. Jee, H.J.; Lee, S.G.; Bormate, K.J.; Jung, Y.-S. Effect of Caffeine Consumption on the Risk for Neurological and Psychiatric Disorders: Sex Differences in Human. Nutrients 2020, 12, 3080. [Google Scholar] [CrossRef] [PubMed]
  7. Cappelletti, S.; Piacentino, D.; Sani, G.; Aromatario, M. Caffeine: Cognitive and Physical Performance Enhancer or Psychoactive Drug? Curr. Neuropharmacol. 2015, 13, 71–88. [Google Scholar] [CrossRef]
  8. Urits, I.; Gress, K.; Charipova, K.; Li, N.; Berger, A.A.; Cornett, E.M.; Hasoon, J.; Kassem, H.; Kaye, A.D.; Viswanath, O. Cannabis Use and Its Association with Psychological Disorders. Psychopharmacol. Bull 2020, 50, 56–67. [Google Scholar] [PubMed]
  9. McCutcheon, R.A.; Reis Marques, T.; Howes, O.D. Schizophrenia-An Overview. JAMA Psychiatry 2020, 77, 201–210. [Google Scholar] [CrossRef]
  10. Rosell, D.R.; Futterman, S.E.; McMaster, A.; Siever, L.J. Schizotypal Personality Disorder: A Current Review. Curr. Psychiatry Rep. 2014, 16, 452. [Google Scholar] [CrossRef]
  11. Joseph, S.M.; Siddiqui, W. Delusional Disorder. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  12. Dailey, M.W.; Saadabadi, A. Mania. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  13. Phillips, M.L.; Kupfer, D.J. Bipolar Disorder Diagnosis: Challenges and Future Directions. Lancet 2013, 381, 1663–1671. [Google Scholar] [CrossRef]
  14. Klein, R.G. Anxiety Disorders. J. Child Psychol. Psychiatry 2009, 50, 153–162. [Google Scholar] [CrossRef]
  15. Goodman, W.K.; Grice, D.E.; Lapidus, K.A.B.; Coffey, B.J. Obsessive-Compulsive Disorder. Psychiatr. Clin. North Am. 2014, 37, 257–267. [Google Scholar] [CrossRef]
  16. Treasure, J.; Duarte, T.A.; Schmidt, U. Eating Disorders. Lancet 2020, 395, 899–911. [Google Scholar] [CrossRef]
  17. Szelenberger, W.; Soldatos, C. Sleep Disorders in Psychiatric Practice. World Psychiatry 2005, 4, 186–190. [Google Scholar] [PubMed]
  18. Cooper, R.J. Over-the-Counter Medicine Abuse—A Review of the Literature. J. Subst. Use 2013, 18, 82–107. [Google Scholar] [CrossRef]
  19. Schulte Holthausen, B.; Habel, U. Sex Differences in Personality Disorders. Curr. Psychiatry Rep. 2018, 20, 107. [Google Scholar] [CrossRef] [PubMed]
  20. Garg, G.; Elshimy, G.; Marwaha, R. Gender Dysphoria. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  21. Sharma, B.R. Disorders of Sexual Preference and Medicolegal Issues Thereof. Am. J. Forensic. Med. Pathol. 2003, 24, 277–282. [Google Scholar] [CrossRef]
  22. Mahone, E.M.; Denckla, M.B. Attention-Deficit/Hyperactivity Disorder: A Historical Neuropsychological Perspective. J. Int. Neuropsychol. Soc. 2017, 23, 916–929. [Google Scholar] [CrossRef] [PubMed]
  23. Martino, D.; Mink, J.W. Tic Disorders. Contin. (Minneap Minn) 2013, 19, 1287–1311. [Google Scholar] [CrossRef]
  24. Divac, N.; Prostran, M.; Jakovcevski, I.; Cerovac, N. Second-Generation Antipsychotics and Extrapyramidal Adverse Effects. Biomed. Res. Int. 2014, 2014, 656370. [Google Scholar] [CrossRef] [PubMed]
  25. Lehman, A.F.; Lieberman, J.A.; Dixon, L.B.; McGlashan, T.H.; Miller, A.L.; Perkins, D.O.; Kreyenbuhl, J.; American Psychiatric Association; Steering Committee on Practice Guidelines. Practice Guideline for the Treatment of Patients with Schizophrenia, Second Edition. Am. J. Psychiatry 2004, 161 (Suppl. 2), 1–56. [Google Scholar]
  26. Klier, C.M.; Mossaheb, N.; Saria, A.; Schloegelhofer, M.; Zernig, G. Pharmacokinetics and Elimination of Quetiapine, Venlafaxine, and Trazodone during Pregnancy and Postpartum. J. Clin. Psychopharmacol. 2007, 27, 720–722. [Google Scholar] [CrossRef]
  27. Yoshikawa, T.; Naito, Y. What Is Oxidative Stress? Jpn. Med. Assoc. J. 2002, 45, 271–276. [Google Scholar]
  28. Teleanu, D.M.; Niculescu, A.-G.; Lungu, I.I.; Radu, C.I.; Vladâcenco, O.; Roza, E.; Costăchescu, B.; Grumezescu, A.M.; Teleanu, R.I. An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 5938. [Google Scholar] [CrossRef]
  29. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
  30. Nishimura, Y.; Kanda, Y.; Sone, H.; Aoyama, H. Oxidative Stress as a Common Key Event in Developmental Neurotoxicity. Oxid. Med. Cell. Longev. 2021, 2021, 6685204. [Google Scholar] [CrossRef]
  31. Gonçalves, R.V.; Costa, A.M.A.; Grzeskowiak, L. Oxidative Stress and Tissue Repair: Mechanism, Biomarkers, and Therapeutics. Oxid. Med. Cell. Longev. 2021, 2021, 6204096. [Google Scholar] [CrossRef] [PubMed]
  32. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative Stress, Aging, and Diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef]
  33. Gyurászová, M.; Kovalčíková, A.; Janšáková, K.; Šebeková, K.; Celec, P.; Tóthová, Ľ. Markers of Oxidative Stress and Antioxidant Status in the Plasma, Urine and Saliva of Healthy Mice. Physiol. Res. 2018, 67, 921–934. [Google Scholar] [CrossRef]
  34. Marrocco, I.; Altieri, F.; Peluso, I. Measurement and Clinical Significance of Biomarkers of Oxidative Stress in Humans. Oxid. Med. Cell. Longev. 2017, 2017, 6501046. [Google Scholar] [CrossRef] [PubMed]
  35. Li, X.; Cameron, M.D. Potential Role of a Quetiapine Metabolite in Quetiapine-Induced Neutropenia and Agranulocytosis. Chem. Res. Toxicol. 2012, 25, 1004–1011. [Google Scholar] [CrossRef] [PubMed]
  36. Soeiro-DE-Souza, M.G.; Dias, V.V.; Missio, G.; Balanzá-Martinez, V.; Valiengo, L.; Carvalho, A.F.; Moreno, R.A. Role of Quetiapine beyond Its Clinical Efficacy in Bipolar Disorder: From Neuroprotection to the Treatment of Psychiatric Disorders (Review). Exp. Ther. Med. 2015, 9, 643–652. [Google Scholar] [CrossRef] [PubMed]
  37. Granger, B.; Albu, S. The Haloperidol Story. Ann. Clin. Psychiatry 2005, 17, 137–140. [Google Scholar] [CrossRef] [PubMed]
  38. Tyler, M.W.; Zaldivar-Diez, J.; Haggarty, S.J. Classics in Chemical Neuroscience: Haloperidol. ACS Chem. Neurosci. 2017, 8, 444–453. [Google Scholar] [CrossRef] [PubMed]
  39. López-Muñoz, F.; Alamo, C. The Consolidation of Neuroleptic Therapy: Janssen, the Discovery of Haloperidol and Its Introduction into Clinical Practice. Brain Res. Bull 2009, 79, 130–141. [Google Scholar] [CrossRef]
  40. Joy, C.B.; Adams, C.E.; Lawrie, S.M. Haloperidol versus Placebo for Schizophrenia. Cochrane Database Syst. Rev. 2006, 4, CD003082. [Google Scholar]
  41. Radojčić, M.R.; Pierce, M.; Hope, H.; Senior, M.; Taxiarchi, V.P.; Trefan, L.; Swift, E.; Abel, K.M. Trends in Antipsychotic Prescribing to Children and Adolescents in England: Cohort Study Using 2000-19 Primary Care Data. Lancet Psychiatry 2023, 10, 119–128. [Google Scholar] [CrossRef] [PubMed]
  42. Anczewska, M.; Balicki, M.; Droździkowska, A.; Gorczyca, P.; Janus, J.; Paciorek, S.; Plisko, R.; Zięba, M. Practice of Prescribing Antipsychotics in Schizophrenia during 2013-2018 Based on Data from the National Health Fund. Psychiatr. Pol. 2022, 56, 751–766. [Google Scholar] [CrossRef]
  43. Haloperidol—Drug Usage Statistics, ClinCalc DrugStats Database. Available online: https://clincalc.com/DrugStats/Drugs/Haloperidol (accessed on 26 January 2024).
  44. Perera, J.; Tan, J.H.; Jeevathayaparan, S.; Chakravarthi, S.; Haleagrahara, N. Neuroprotective Effects of Alpha Lipoic Acid on Haloperidol-Induced Oxidative Stress in the Rat Brain. Cell Biosci. 2011, 1, 12. [Google Scholar] [CrossRef]
  45. Raudenska, M.; Gumulec, J.; Babula, P.; Stracina, T.; Sztalmachova, M.; Polanska, H.; Adam, V.; Kizek, R.; Novakova, M.; Masarik, M. Haloperidol Cytotoxicity and Its Relation to Oxidative Stress. Mini Rev. Med. Chem. 2013, 13, 1993–1998. [Google Scholar] [CrossRef] [PubMed]
  46. Bošković, M.; Grabnar, I.; Terzič, T.; Kores Plesničar, B.; Vovk, T. Oxidative Stress in Schizophrenia Patients Treated with Long-Acting Haloperidol Decanoate. Psychiatry Res. 2013, 210, 761–768. [Google Scholar] [CrossRef] [PubMed]
  47. El-Awdan, S.A.; Abdel Jaleel, G.A.; Saleh, D.O. Alleviation of Haloperidol Induced Oxidative Stress in Rats: Effects of Sucrose vs Grape Seed Extract. Bull. Fac. Pharm. Cairo Univ. 2015, 53, 29–35. [Google Scholar] [CrossRef]
  48. Andreazza, A.C.; Barakauskas, V.E.; Fazeli, S.; Feresten, A.; Shao, L.; Wei, V.; Wu, C.H.; Barr, A.M.; Beasley, C.L. Effects of Haloperidol and Clozapine Administration on Oxidative Stress in Rat Brain, Liver and Serum. Neurosci. Lett. 2015, 591, 36–40. [Google Scholar] [CrossRef] [PubMed]
  49. Singh, O.P.; Chakraborty, I.; Dasgupta, A.; Datta, S. A Comparative Study of Oxidative Stress and Interrelationship of Important Antioxidants in Haloperidol and Olanzapine Treated Patients Suffering from Schizophrenia. Indian J. Psychiatry 2008, 50, 171–176. [Google Scholar] [CrossRef]
  50. Gawlik-Kotelnicka, O.; Mielicki, W.; Rabe-Jabłońska, J.; Strzelecki, D. Impact of Lithium Alone or in Combination with Haloperidol on Selected Oxidative Stress Parameters in Human Plasma in Vitro. Redox Rep. 2016, 21, 45–49. [Google Scholar] [CrossRef]
  51. Kajero, J.A.; Seedat, S.; Ohaeri, J.U.; Akindele, A.; Aina, O. The Effects of Cannabidiol on Behavioural and Oxidative Stress Parameters Induced by Prolonged Haloperidol Administration. Acta Neuropsychiatr. 2022, 4, 1–11. [Google Scholar] [CrossRef]
  52. Khokhar, J.Y.; Henricks, A.M.; Sullivan, E.D.K.; Green, A.I. Unique Effects of Clozapine: A Pharmacological Perspective. Adv. Pharmacol. 2018, 82, 137–162. [Google Scholar]
  53. Crilly, J. The History of Clozapine and Its Emergence in the US Market: A Review and Analysis. Hist. Psychiatry 2007, 18, 39–60. [Google Scholar] [CrossRef]
  54. Seeman, P. Clozapine, a Fast-off-D2 Antipsychotic. ACS Chem. Neurosci. 2014, 5, 24–29. [Google Scholar] [CrossRef]
  55. Masdrakis, V.G.; Baldwin, D.S. Prevention of Suicide by Clozapine in Mental Disorders: Systematic Review. Eur. Neuropsychopharmacol. 2023, 69, 4–23. [Google Scholar] [CrossRef]
  56. Griffiths, K.; Martínez-Gutiérrez, A.D.; Rentería, M.E.; Patel, R. Antipsychotic Prescribing Trends in Schizophrenia between 2011 and 2021: Real-World Data from a US Electronic Health Record Database. Neurosci. Appl. 2022, 1, 269. [Google Scholar] [CrossRef]
  57. Walss-Bass, C.; Weintraub, S.T.; Hatch, J.; Mintz, J.; Chaudhuri, A.R. Clozapine Causes Oxidation of Proteins Involved in Energy Metabolism: A Possible Mechanism for Antipsychotic-Induced Metabolic Alterations. Int. J. Neuropsychopharmacol. 2008, 11, 1097–1104. [Google Scholar] [CrossRef] [PubMed]
  58. Abdel-Wahab, B.; Abdalla, M.; El-Khawanky, M. Does Clozapine Induce Myocarditis, Myocardial Oxidative Stress and DNA Damage in Rats. Egypt. J. Forensic Sci. 2014, 4, 75–82. [Google Scholar] [CrossRef]
  59. Zhang, F.; Han, L.; Wang, J.; Shu, M.; Liu, K.; Zhang, Y.; Hsiao, C.; Tian, Q.; He, Q. Clozapine Induced Developmental and Cardiac Toxicity on Zebrafish Embryos by Elevating Oxidative Stress. Cardiovasc. Toxicol. 2021, 21, 399–409. [Google Scholar] [CrossRef] [PubMed]
  60. Fehsel, K.; Loeffler, S.; Krieger, K.; Henning, U.; Agelink, M.; Kolb-Bachofen, V.; Klimke, A. Clozapine Induces Oxidative Stress and Proapoptotic Gene Expression in Neutrophils of Schizophrenic Patients. J. Clin. Psychopharmacol. 2005, 25, 419–426. [Google Scholar] [CrossRef]
  61. Hendouei, N.; Farnia, S.; Mohseni, F.; Salehi, A.; Bagheri, M.; Shadfar, F.; Barzegar, F.; Hoseini, S.D.; Charati, J.Y.; Shaki, F. Alterations in Oxidative Stress Markers and Its Correlation with Clinical Findings in Schizophrenic Patients Consuming Perphenazine, Clozapine and Risperidone. Biomed. Pharmacother. 2018, 103, 965–972. [Google Scholar] [CrossRef] [PubMed]
  62. Awouters, F.H.L.; Lewi, P.J. Forty Years of Antipsychotic Drug Research--from Haloperidol to Paliperidone—with Dr. Paul Janssen. Arzneimittelforschung 2007, 57, 625–632. [Google Scholar] [CrossRef]
  63. Colpaert, F.C. Discovering Risperidone: The LSD Model of Psychopathology. Nat. Rev. Drug Discov. 2003, 2, 315–320. [Google Scholar] [CrossRef]
  64. Chopko, T.C.; Lindsley, C.W. Classics in Chemical Neuroscience: Risperidone. ACS Chem. Neurosci. 2018, 9, 1520–1529. [Google Scholar] [CrossRef]
  65. Pajonk, F.-G. Risperidone in Acute and Long-Term Therapy of Schizophrenia—A Clinical Profile. Prog Neuropsychopharmacol. Biol. Psychiatry 2004, 28, 15–23. [Google Scholar] [CrossRef]
  66. Taurines, R.; Fekete, S.; Preuss-Wiedenhoff, A.; Warnke, A.; Wewetzer, C.; Plener, P.; Burger, R.; Gerlach, M.; Romanos, M.; Egberts, K.M. Therapeutic Drug Monitoring in Children and Adolescents with Schizophrenia and Other Psychotic Disorders Using Risperidone. J. Neural. Transm. 2022, 129, 689–701. [Google Scholar] [CrossRef] [PubMed]
  67. Yunusa, I.; El Helou, M.L. The Use of Risperidone in Behavioral and Psychological Symptoms of Dementia: A Review of Pharmacology, Clinical Evidence, Regulatory Approvals, and Off-Label Use. Front. Pharmacol. 2020, 11, 596. [Google Scholar] [CrossRef]
  68. Atypical Antipsychotics—Drug Usage Statistics, ClinCalc DrugStats Database. Available online: https://clincalc.com/DrugStats/TC/AtypicalAntipsychotics (accessed on 26 January 2024).
  69. Stojkovic, M.; Radmanovic, B.; Jovanovic, M.; Janjic, V.; Muric, N.; Ristic, D.I. Risperidone Induced Hyperprolactinemia: From Basic to Clinical Studies. Front. Psychiatry 2022, 13, 874705. [Google Scholar] [CrossRef]
  70. Zhang, X.Y.; Zhou, D.F.; Shen, Y.C.; Zhang, P.Y.; Zhang, W.F.; Liang, J.; Chen, D.C.; Xiu, M.H.; Kosten, T.A.; Kosten, T.R. Effects of Risperidone and Haloperidol on Superoxide Dismutase and Nitric Oxide in Schizophrenia. Neuropharmacology 2012, 62, 1928–1934. [Google Scholar] [CrossRef]
  71. Lee, B.-H.; Kim, Y.-K. Reduced Plasma Nitric Oxide Metabolites before and after Antipsychotic Treatment in Patients with Schizophrenia Compared to Controls. Schizophr. Res. 2008, 104, 36–43. [Google Scholar] [CrossRef] [PubMed]
  72. Eftekhari, A.; Ahmadian, E.; Azarmi, Y.; Parvizpur, A.; Hamishehkar, H.; Eghbal, M.A. In Vitro/Vivo Studies towards Mechanisms of Risperidone-Induced Oxidative Stress and the Protective Role of Coenzyme Q10 and N-Acetylcysteine. Toxicol. Mech. Methods 2016, 26, 520–528. [Google Scholar] [CrossRef] [PubMed]
  73. Pryor, K.; Storer, K. Chapter 11—Drugs for Neuropsychiatric Disorders. In Pharmacology and Physiology for Anesthesia: Foundations and Clinical Application; Elsevier: Amsterdam, The Netherlands, 2013; pp. 180–207. [Google Scholar]
  74. Citrome, L.; McEvoy, J.P.; Todtenkopf, M.S.; McDonnell, D.; Weiden, P.J. A Commentary on the Efficacy of Olanzapine for the Treatment of Schizophrenia: The Past, Present, and Future. Neuropsychiatr. Dis. Treat. 2019, 15, 2559–2569. [Google Scholar] [CrossRef] [PubMed]
  75. McCormack, P.L.; Wiseman, L.R. Olanzapine: A Review of Its Use in the Management of Bipolar I Disorder. Drugs 2004, 64, 2709–2726. [Google Scholar] [CrossRef]
  76. Meftah, A.M.; Deckler, E.; Citrome, L.; Kantrowitz, J.T. New Discoveries for an Old Drug: A Review of Recent Olanzapine Research. Postgrad. Med. 2020, 132, 80–90. [Google Scholar] [CrossRef]
  77. Zerin Khan, F.; Sultana, S.P.; Akhter, N.; Mosaddek, A.S.M. Effect of Olanzapine and Risperidone on Oxidative Stress in Schizophrenia Patients. Int. Biol. Biomed. J. 2018, 4, 89–97. [Google Scholar]
  78. Boz, Z.; Hu, M.; Yu, Y.; Huang, X.-F. N-Acetylcysteine Prevents Olanzapine-Induced Oxidative Stress in mHypoA-59 Hypothalamic Neurons. Sci. Rep. 2020, 10, 19185. [Google Scholar] [CrossRef] [PubMed]
  79. Reinke, A.; Martins, M.R.; Lima, M.S.; Moreira, J.C.; Dal-Pizzol, F.; Quevedo, J. Haloperidol and Clozapine, but Not Olanzapine, Induces Oxidative Stress in Rat Brain. Neurosci. Lett. 2004, 372, 157–160. [Google Scholar] [CrossRef] [PubMed]
  80. Al-Chalabi, B.M.; Thanoon, I.A.J.; Ahmed, F.A. Potential Effect of Olanzapine on Total Antioxidant Status and Lipid Peroxidation in Schizophrenic Patients. Neuropsychobiology 2009, 59, 8–11. [Google Scholar] [CrossRef] [PubMed]
  81. Li, W.-T.; Huang, X.-F.; Deng, C.; Zhang, B.-H.; Qian, K.; He, M.; Sun, T.-L. Olanzapine Induces Inflammation and Immune Response via Activating ER Stress in the Rat Prefrontal Cortex. Curr. Med. Sci. 2021, 41, 788–802. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, P.; Si, T. Use of Antipsychotics in the Treatment of Depressive Disorders. Shanghai Arch. Psychiatry 2013, 25, 134–140. [Google Scholar]
  83. Ignácio, Z.M.; Calixto, A.V.; da Silva, R.H.; Quevedo, J.; Réus, G.Z. The Use of Quetiapine in the Treatment of Major Depressive Disorder: Evidence from Clinical and Experimental Studies. Neurosci. Biobehav. Rev. 2018, 86, 36–50. [Google Scholar] [CrossRef]
  84. El-Khalili, N. Update on Extended Release Quetiapine Fumarate in Schizophrenia and Bipolar Disorders. Neuropsychiatr. Dis. Treat. 2012, 8, 523–536. [Google Scholar] [CrossRef]
  85. Brett, J. Concerns about Quetiapine. Aust. Prescr. 2015, 38, 95–97. [Google Scholar] [CrossRef]
  86. Han, J.-H.; Tian, H.-Z.; Lian, Y.-Y.; Yu, Y.; Lu, C.-B.; Li, X.-M.; Zhang, R.-L.; Xu, H. Quetiapine Mitigates the Ethanol-Induced Oxidative Stress in Brain Tissue, but Not in the Liver, of the Rat. Neuropsychiatr. Dis. Treat. 2015, 11, 1473–1482. [Google Scholar]
  87. Ignácio, Z.M.; Réus, G.Z.; Abelaira, H.M.; de Moura, A.B.; de Souza, T.G.; Matos, D.; Goldim, M.P.; Mathias, K.; Garbossa, L.; Petronilho, F.; et al. Acute and Chronic Treatment with Quetiapine Induces Antidepressant-like Behavior and Exerts Antioxidant Effects in the Rat Brain. Metab. Brain Dis. 2017, 32, 1195–1208. [Google Scholar] [CrossRef] [PubMed]
  88. Sommerfeld-Klatta, K.; Łukasik-Głębocka, M.; Krawczak, E.; Stodolska, A.; Zielińska-Psuja, B. Participation of quetiapine in oxidative stress and inflammation status in the treatment of drug overdose. Acta Pol. Pharm. 2023, 80, 327–333. [Google Scholar] [CrossRef] [PubMed]
  89. Dietrich-Muszalska, A.; Kolodziejczyk-Czepas, J.; Nowak, P. Comparative Study of the Effects of Atypical Antipsychotic Drugs on Plasma and Urine Biomarkers of Oxidative Stress in Schizophrenic Patients. Neuropsychiatr. Dis. Treat. 2021, 17, 555–565. [Google Scholar] [CrossRef] [PubMed]
  90. Dietrich-Muszalska, A.; Kontek, B.; Rabe-Jabłońska, J. Quetiapine, Olanzapine and Haloperidol Affect Human Plasma Lipid Peroxidation in Vitro. Neuropsychobiology 2011, 63, 197–201. [Google Scholar] [CrossRef] [PubMed]
  91. Mani, V.; Alshammeri, B.S. Quetiapine Moderates Doxorubicin-Induced Cognitive Deficits: Influence of Oxidative Stress, Neuroinflammation, and Cellular Apoptosis. Int. J. Mol. Sci. 2023, 24, 11525. [Google Scholar] [CrossRef] [PubMed]
  92. Stark, A.D.; Jordan, S.; Allers, K.A.; Bertekap, R.L.; Chen, R.; Mistry Kannan, T.; Molski, T.F.; Yocca, F.D.; Sharp, T.; Kikuchi, T.; et al. Interaction of the Novel Antipsychotic Aripiprazole with 5-HT1A and 5-HT 2A Receptors: Functional Receptor-Binding and in Vivo Electrophysiological Studies. Psychopharmacology 2007, 190, 373–382. [Google Scholar] [CrossRef] [PubMed]
  93. Preda, A.; Shapiro, B.B. A Safety Evaluation of Aripiprazole in the Treatment of Schizophrenia. Expert Opin. Drug Saf. 2020, 19, 1529–1538. [Google Scholar] [CrossRef] [PubMed]
  94. Prommer, E. Aripiprazole. Am. J. Hosp. Palliat. Care 2017, 34, 180–185. [Google Scholar] [CrossRef] [PubMed]
  95. Cuomo, A.; Beccarini Crescenzi, B.; Goracci, A.; Bolognesi, S.; Giordano, N.; Rossi, R.; Facchi, E.; Neal, S.M.; Fagiolini, A. Drug Safety Evaluation of Aripiprazole in Bipolar Disorder. Expert Opin. Drug Saf. 2019, 18, 455–463. [Google Scholar] [CrossRef]
  96. Khanna, P.; Suo, T.; Komossa, K.; Ma, H.; Rummel-Kluge, C.; El-Sayeh, H.G.; Leucht, S.; Xia, J. Aripiprazole versus Other Atypical Antipsychotics for Schizophrenia. Cochrane Database Syst. Rev. 2014, 2014, CD006569. [Google Scholar] [CrossRef]
  97. Doblado, L.; Patel, G.; Yildiz, R.; Sellinger, L.; Koller, D.; Abad-Santos, F.; Peral, B.; Ben, A.; Monsalve, M. The SGAs Olanzapine and Aripiprazole Inhibit Mitochondrial Respiration and Induce Oxidative Stress. Free. Radic. Biol. Med. 2021, 165, 58–59. [Google Scholar] [CrossRef]
  98. Dietrich-Muszalska, A.; Kolińska-Łukaszuk, J. Comparative Effects of Aripiprazole and Selected Antipsychotic Drugs on Lipid Peroxidation in Plasma. Psychiatry Clin. Neurosci. 2018, 72, 329–336. [Google Scholar] [CrossRef] [PubMed]
  99. Eren, I.; Naziroğlu, M.; Demirdaş, A. Protective Effects of Lamotrigine, Aripiprazole and Escitalopram on Depression-Induced Oxidative Stress in Rat Brain. Neurochem. Res. 2007, 32, 1188–1195. [Google Scholar] [CrossRef] [PubMed]
  100. Mani, V.; Alshammeri, B.S. Aripiprazole Attenuates Cognitive Impairments Induced by Lipopolysaccharide in Rats through the Regulation of Neuronal Inflammation, Oxidative Stress, and Apoptosis. Medicina 2023, 60, 46. [Google Scholar] [CrossRef] [PubMed]
  101. Kramar, B.; Pirc Marolt, T.; Monsalve, M.; Šuput, D.; Milisav, I. Antipsychotic Drug Aripiprazole Protects Liver Cells from Oxidative Stress. Int. J. Mol. Sci. 2022, 23, 8292. [Google Scholar] [CrossRef] [PubMed]
  102. Cade, J.F.J. Lithium Salts in the Treatment of Psychotic Excitement. Med. J. Aust. 1949, 2, 349–352. [Google Scholar] [CrossRef] [PubMed]
  103. Shorter, E. The History of Lithium Therapy. Bipolar. Disord. 2009, 11 (Suppl. 2), 4–9. [Google Scholar] [CrossRef] [PubMed]
  104. Licht, R.W. Lithium: Still a Major Option in the Management of Bipolar Disorder. CNS Neurosci. Ther. 2012, 18, 219–226. [Google Scholar] [CrossRef]
  105. Leucht, S.; Helfer, B.; Dold, M.; Kissling, W.; McGrath, J.J. Lithium for Schizophrenia. Cochrane Database Syst. Rev. 2015, 2015, CD003834. [Google Scholar] [CrossRef]
  106. Bschor, T. Lithium in the Treatment of Major Depressive Disorder. Drugs 2014, 74, 855–862. [Google Scholar] [CrossRef]
  107. Lazzara, C.A.; Kim, Y.-H. Potential Application of Lithium in Parkinson’s and Other Neurodegenerative Diseases. Front. Neurosci. 2015, 9, 403. [Google Scholar] [CrossRef]
  108. Matsunaga, S.; Kishi, T.; Annas, P.; Basun, H.; Hampel, H.; Iwata, N. Lithium as a Treatment for Alzheimer’s Disease: A Systematic Review and Meta-Analysis. J. Alzheimers Dis. 2015, 48, 403–410. [Google Scholar] [CrossRef] [PubMed]
  109. Forlenza, O.V.; de Paula, V.J.; Machado-Vieira, R.; Diniz, B.S.; Gattaz, W.F. Does Lithium Prevent Alzheimer’s Disease? Drugs Aging 2012, 29, 335–342. [Google Scholar] [CrossRef]
  110. Codogno, P.; Meijer, A.J. Chapter 306—Signaling in Autophagy Related Pathways. In Handbook of Cell Signaling, 2nd ed.; Bradshaw, R.A., Dennis, E.A., Eds.; Academic Press: San Diego, CA, USA, 2010; pp. 2583–2588. [Google Scholar]
  111. Brown, K.M.; Tracy, D.K. Lithium: The Pharmacodynamic Actions of the Amazing Ion. Ther. Adv. Psychopharmacol. 2013, 3, 163–176. [Google Scholar] [CrossRef]
  112. Malhi, G.S.; Tanious, M.; Das, P.; Coulston, C.M.; Berk, M. Potential Mechanisms of Action of Lithium in Bipolar Disorder. Current Understanding. CNS Drugs 2013, 27, 135–153. [Google Scholar] [CrossRef]
  113. Wen, J.; Sawmiller, D.; Wheeldon, B.; Tan, J. A Review for Lithium: Pharmacokinetics, Drug Design, and Toxicity. CNS Neurol. Disord. Drug Targets 2019, 18, 769–778. [Google Scholar] [CrossRef] [PubMed]
  114. McKnight, R.F.; Adida, M.; Budge, K.; Stockton, S.; Goodwin, G.M.; Geddes, J.R. Lithium Toxicity Profile: A Systematic Review and Meta-Analysis. Lancet 2012, 379, 721–728. [Google Scholar] [CrossRef]
  115. Rabe-Jabłońska, J.; Dietrich-Muszalska, A. Effects of Lithium on Oxidative Stress. In Studies on Psychiatric Disorders; Dietrich-Muszalska, A., Chauhan, V., Grignon, S., Eds.; Springer: New York, NY, USA, 2015; pp. 567–573. [Google Scholar]
  116. De Sousa, R.T.; Zarate, C.A.; Zanetti, M.V.; Costa, A.C.; Talib, L.L.; Gattaz, W.F.; Machado-Vieira, R. Oxidative Stress in Early Stage Bipolar Disorder and the Association with Response to Lithium. J. Psychiatr. Res. 2014, 50, 36–41. [Google Scholar] [CrossRef] [PubMed]
  117. Banerjee, U.; Dasgupta, A.; Rout, J.K.; Singh, O.P. Effects of Lithium Therapy on Na+-K+-ATPase Activity and Lipid Peroxidation in Bipolar Disorder. Prog. Neuropsychopharmacol. Biol. 2012, 37, 56–61. [Google Scholar] [CrossRef]
  118. da-Rosa, D.D.; Valvassori, S.S.; Steckert, A.V.; Ornell, F.; Ferreira, C.L.; Lopes-Borges, J.; Varela, R.B.; Dal-Pizzol, F.; Andersen, M.L.; Quevedo, J. Effects of Lithium and Valproate on Oxidative Stress and Behavioral Changes Induced by Administration of M-AMPH. Psychiatry Res. 2012, 198, 521–526. [Google Scholar] [CrossRef]
  119. Jornada, L.K.; Valvassori, S.S.; Steckert, A.V.; Moretti, M.; Mina, F.; Ferreira, C.L.; Arent, C.O.; Dal-Pizzol, F.; Quevedo, J. Lithium and Valproate Modulate Antioxidant Enzymes and Prevent Ouabain-Induced Oxidative Damage in an Animal Model of Mania. J. Psychiatr. Res. 2011, 45, 162–168. [Google Scholar] [CrossRef] [PubMed]
  120. Tufekci, K.U.; Alural, B.; Tarakcioglu, E.; San, T.; Genc, S. Lithium Inhibits Oxidative Stress-Induced Neuronal Senescence through miR-34a. Mol. Biol. Rep. 2021, 48, 4171–4180. [Google Scholar] [CrossRef] [PubMed]
  121. Toplan, S.; Dariyerli, N.; Ozdemir, S.; Ozcelik, D.; Zengin, E.U.; Akyolcu, M.C. Lithium-Induced Hypothyroidism: Oxidative Stress and Osmotic Fragility Status in Rats. Biol. Trace Elem. Res. 2013, 152, 373–378. [Google Scholar] [CrossRef] [PubMed]
  122. Khairova, R.; Pawar, R.; Salvadore, G.; Juruena, M.F.; de Sousa, R.T.; Soeiro-de-Souza, M.G.; Salvador, M.; Zarate, C.A.; Gattaz, W.F.; Machado-Vieira, R. Effects of Lithium on Oxidative Stress Parameters in Healthy Subjects. Mol. Med. Rep. 2012, 5, 680–682. [Google Scholar] [PubMed]
  123. Eskandari, M.R.; Fard, J.K.; Hosseini, M.-J.; Pourahmad, J. Glutathione Mediated Reductive Activation and Mitochondrial Dysfunction Play Key Roles in Lithium Induced Oxidative Stress and Cytotoxicity in Liver. Biometals 2012, 25, 863–873. [Google Scholar] [CrossRef]
  124. Franceschetti, S.; Hamon, B.; Heinemann, U. The Action of Valproate on Spontaneous Epileptiform Activity in the Absence of Synaptic Transmission and on Evoked Changes in [Ca2+]o and [K+]o in the Hippocampal Slice. Brain Res. 1986, 386, 1–11. [Google Scholar] [CrossRef]
  125. Johannessen, C.U.; Johannessen, S.I. Valproate: Past, Present, and Future. CNS Drug Rev. 2003, 9, 199–216. [Google Scholar] [CrossRef] [PubMed]
  126. Lloyd, K.A. A Scientific Review: Mechanisms of Valproate-Mediated Teratogenesis. Biosci. Horiz. Int. J. Stud. Res. 2013, 6, hzt003. [Google Scholar] [CrossRef]
  127. Tomson, T.; Battino, D.; Bonizzoni, E.; Craig, J.; Lindhout, D.; Perucca, E.; Sabers, A.; Thomas, S.V.; Vajda, F.; EURAP Study Group. Dose-Dependent Teratogenicity of Valproate in Mono- and Polytherapy: An Observational Study. Neurology 2015, 85, 866–872. [Google Scholar] [CrossRef]
  128. Bromfield, E.B.; Dworetzky, B.A.; Wyszynski, D.F.; Smith, C.R.; Baldwin, E.J.; Holmes, L.B. Valproate Teratogenicity and Epilepsy Syndrome. Epilepsia 2008, 49, 2122–2124. [Google Scholar] [CrossRef]
  129. Gerstner, T.; Bell, N.; König, S. Oral Valproic Acid for Epilepsy--Long-Term Experience in Therapy and Side Effects. Expert Opin. Pharmacother. 2008, 9, 285–292. [Google Scholar] [CrossRef] [PubMed]
  130. Nanau, R.M.; Neuman, M.G. Adverse Drug Reactions Induced by Valproic Acid. Clin. Biochem. 2013, 46, 1323–1338. [Google Scholar] [CrossRef] [PubMed]
  131. Marson, A.G.; Sills, G.J. Valproate. In The Treatment of Epilepsy; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2015; pp. 652–666. [Google Scholar]
  132. Andrade, C. Valproate in Pregnancy: Recent Research and Regulatory Responses. J. Clin. Psychiatry 2018, 79, 18f12351. [Google Scholar] [CrossRef] [PubMed]
  133. Chateauvieux, S.; Morceau, F.; Dicato, M.; Diederich, M. Molecular and Therapeutic Potential and Toxicity of Valproic Acid. J. Biomed. Biotechnol. 2010, 2010, 479364. [Google Scholar] [CrossRef] [PubMed]
  134. Gobbi, G.; Janiri, L. Sodium- and Magnesium-Valproate in Vivo Modulate Glutamatergic and GABAergic Synapses in the Medial Prefrontal Cortex. Psychopharmacology 2006, 185, 255–262. [Google Scholar] [CrossRef]
  135. Sher, Y.; Miller Cramer, A.C.; Ament, A.; Lolak, S.; Maldonado, J.R. Valproic Acid for Treatment of Hyperactive or Mixed Delirium: Rationale and Literature Review. Psychosomatics 2015, 56, 615–625. [Google Scholar] [CrossRef] [PubMed]
  136. VanDongen, A.M.; VanErp, M.G.; Voskuyl, R.A. Valproate Reduces Excitability by Blockage of Sodium and Potassium Conductance. Epilepsia 1986, 27, 177–182. [Google Scholar] [CrossRef] [PubMed]
  137. Chaudhary, S.; Parvez, S. An in Vitro Approach to Assess the Neurotoxicity of Valproic Acid-Induced Oxidative Stress in Cerebellum and Cerebral Cortex of Young Rats. Neuroscience 2012, 225, 258–268. [Google Scholar] [CrossRef]
  138. Salimi, A.; Alyan, N.; Akbari, N.; Jamali, Z.; Pourahmad, J. Selenium and L-Carnitine Protects from Valproic Acid-Induced Oxidative Stress and Mitochondrial Damages in Rat Cortical Neurons. Drug Chem. Toxicol. 2022, 45, 1150–1157. [Google Scholar] [CrossRef]
  139. Kiang, T.K.L.; Teng, X.W.; Karagiozov, S.; Surendradoss, J.; Chang, T.K.H.; Abbott, F.S. Role of Oxidative Metabolism in the Effect of Valproic Acid on Markers of Cell Viability, Necrosis, and Oxidative Stress in Sandwich-Cultured Rat Hepatocytes. Toxicol. Sci. 2010, 118, 501–509. [Google Scholar] [CrossRef]
  140. Lee, J.Y.; Maeng, S.; Kang, S.R.; Choi, H.Y.; Oh, T.H.; Ju, B.G.; Yune, T.Y. Valproic Acid Protects Motor Neuron Death by Inhibiting Oxidative Stress and Endoplasmic Reticulum Stress-Mediated Cytochrome C Release after Spinal Cord Injury. J. Neurotrauma 2014, 31, 582–594. [Google Scholar] [CrossRef]
  141. Suda, S.; Katsura, K.; Kanamaru, T.; Saito, M.; Katayama, Y. Valproic Acid Attenuates Ischemia-Reperfusion Injury in the Rat Brain through Inhibition of Oxidative Stress and Inflammation. Eur. J. Pharmacol. 2013, 707, 26–31. [Google Scholar] [CrossRef] [PubMed]
  142. Terzioğlu Bebitoğlu, B.; Oğuz, E.; Gökçe, A. Effect of Valproic Acid on Oxidative Stress Parameters of Glutamate-Induced Excitotoxicity in SH-SY5Y Cells. Exp. Ther. Med. 2020, 20, 1321–1328. [Google Scholar] [CrossRef] [PubMed]
  143. Sommerfeld-Klatta, K.; Łukasik-Głębocka, M.; Kobyłka, S.; Zielińska-Psuja, B. The impact of valproic acid and 2-propyl-4-pentenoic acid on antioxidant status measured by GSH/GSSG ratio and FRAP levels in acute and chronic exposure. Acta Pol. Pharm. Drug Res. 2023, 80, 335–340. [Google Scholar] [CrossRef] [PubMed]
  144. Tung, E.W.Y.; Winn, L.M. Valproic Acid Increases Formation of Reactive Oxygen Species and Induces Apoptosis in Postimplantation Embryos: A Role for Oxidative Stress in Valproic Acid-Induced Neural Tube Defects. Mol. Pharmacol. 2011, 80, 979–987. [Google Scholar] [CrossRef]
  145. Fu, J.; Shao, C.-J.; Chen, F.-R.; Ng, H.-K.; Chen, Z.-P. Autophagy Induced by Valproic Acid Is Associated with Oxidative Stress in Glioma Cell Lines. Neuro. Oncol. 2010, 12, 328–340. [Google Scholar] [CrossRef] [PubMed]
  146. Alrashood, S.T. Carbamazepine. Profiles Drug Subst. Excip. Relat. Methodol. 2016, 41, 133–321. [Google Scholar] [PubMed]
  147. Pellock, J.M. Carbamazepine Side Effects in Children and Adults. Epilepsia 1987, 28 (Suppl. 3), S64–S70. [Google Scholar] [CrossRef] [PubMed]
  148. Thorn, C.F.; Leckband, S.G.; Kelsoe, J.; Leeder, J.S.; Müller, D.J.; Klein, T.E.; Altman, R.B. PharmGKB Summary: Carbamazepine Pathway. Pharm. Genom. 2011, 21, 906–910. [Google Scholar] [CrossRef]
  149. Fricke-Galindo, I.; LLerena, A.; Jung-Cook, H.; López-López, M. Carbamazepine Adverse Drug Reactions. Expert Rev. Clin. Pharmacol. 2018, 11, 705–718. [Google Scholar] [CrossRef]
  150. Wiffen, P.J.; Derry, S.; Moore, R.A.; Kalso, E.A. Carbamazepine for Chronic Neuropathic Pain and Fibromyalgia in Adults. Cochrane Database Syst. Rev. 2014, 2014, CD005451. [Google Scholar] [CrossRef] [PubMed]
  151. Dailey, J.W.; Reith, M.E.; Yan, Q.S.; Li, M.Y.; Jobe, P.C. Carbamazepine Increases Extracellular Serotonin Concentration: Lack of Antagonism by Tetrodotoxin or Zero Ca2+. Eur. J. Pharmacol. 1997, 328, 153–162. [Google Scholar] [CrossRef] [PubMed]
  152. Lemos, L.; Fontes, R.; Flores, S.; Oliveira, P.; Almeida, A. Effectiveness of the Association between Carbamazepine and Peripheral Analgesic Block with Ropivacaine for the Treatment of Trigeminal Neuralgia. J. Pain. Res. 2010, 3, 201–212. [Google Scholar] [PubMed]
  153. Tutanc, M.; Aras, M.; Dokuyucu, R.; Altas, M.; Zeren, C.; Arica, V.; Ozturk, O.H.; Motor, S.; Yilmaz, C. Oxidative Status in Epileptic Children Using Carbamazepine. Iran J. Pediatr. 2015, 25, e3885. [Google Scholar] [CrossRef] [PubMed]
  154. Varoglu, A.O.; Yildirim, A.; Aygul, R.; Gundogdu, O.L.; Sahin, Y.N. Effects of Valproate, Carbamazepine, and Levetiracetam on the Antioxidant and Oxidant Systems in Epileptic Patients and Their Clinical Importance. Clin. Neuropharmacol. 2010, 33, 155–157. [Google Scholar] [CrossRef] [PubMed]
  155. Ficarra, S.; Misiti, F.; Russo, A.; Carelli-Alinovi, C.; Bellocco, E.; Barreca, D.; Laganà, G.; Leuzzi, U.; Toscano, G.; Giardina, B.; et al. Antiepileptic Carbamazepine Drug Treatment Induces Alteration of Membrane in Red Blood Cells: Possible Positive Effects on Metabolism and Oxidative Stress. Biochimie 2013, 95, 833–841. [Google Scholar] [CrossRef] [PubMed]
  156. Erdem Guzel, E.; Kaya Tektemur, N.; Tektemur, A.; Etem Önalan, E. Carbamazepine-Induced Renal Toxicity May Be Associated with Oxidative Stress and Apoptosis in Male Rat. Drug Chem. Toxicol. 2023, 46, 136–143. [Google Scholar] [CrossRef] [PubMed]
  157. Arora, T.; Mehta, A.K.; Sharma, K.K.; Mediratta, P.K.; Banerjee, B.D.; Garg, G.R.; Sharma, A.K. Effect of Carbamazepine and Lamotrigine on Cognitive Function and Oxidative Stress in Brain during Chemical Epileptogenesis in Rats. Basic Clin. Pharmacol. Toxicol. 2010, 106, 372–377. [Google Scholar] [CrossRef]
  158. Gasca-Pérez, E.; Galar-Martínez, M.; García-Medina, S.; Pérez-Coyotl, I.A.; Ruiz-Lara, K.; Cano-Viveros, S.; Pérez-Pastén Borja, R.; Gómez-Oliván, L.M. Short-Term Exposure to Carbamazepine Causes Oxidative Stress on Common Carp (Cyprinus Carpio). Environ. Toxicol. Pharmacol. 2019, 66, 96–103. [Google Scholar] [CrossRef]
  159. Li, Z.-H.; Li, P.; Rodina, M.; Randak, T. Effect of Human Pharmaceutical Carbamazepine on the Quality Parameters and Oxidative Stress in Common Carp (Cyprinus carpio L.) Spermatozoa. Chemosphere 2010, 80, 530–534. [Google Scholar] [CrossRef]
  160. García-Medina, S.; Galar-Martínez, M.; Gómez-Oliván, L.M.; Torres-Bezaury, R.M.D.C.; Islas-Flores, H.; Gasca-Pérez, E. The Relationship between Cyto-Genotoxic Damage and Oxidative Stress Produced by Emerging Pollutants on a Bioindicator Organism (Allium Cepa): The Carbamazepine Case. Chemosphere 2020, 253, 126675. [Google Scholar] [CrossRef] [PubMed]
  161. Shorvon, S. Oxcarbazepine: A Review. Seizure 2000, 9, 75–79. [Google Scholar] [CrossRef]
  162. Shorvon, S.D. Drug Treatment of Epilepsy in the Century of the ILAE: The Second 50 Years, 1959–2009. Epilepsia 2009, 50 (Suppl. S3), 93–130. [Google Scholar] [CrossRef] [PubMed]
  163. Tecoma, E.S. Oxcarbazepine. Epilepsia 1999, 40 (Suppl. 5), S37–S46. [Google Scholar] [CrossRef] [PubMed]
  164. Beydoun, A.; Kutluay, E. Oxcarbazepine. Expert Opin. Pharmacother. 2002, 3, 59–71. [Google Scholar] [CrossRef] [PubMed]
  165. Faught, E.; Kim, H. Oxcarbazepine. In The Treatment of Epilepsy; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2015; pp. 533–545. [Google Scholar]
  166. Cárdenas-Rodríguez, N.; Coballase-Urrutia, E.; Rivera-Espinosa, L.; Romero-Toledo, A.; Sampieri, A.; Ortega-Cuellar, D.; Montesinos-Correa, H.; Floriano-Sánchez, E.; Carmona-Aparicio, L. Modulation of Antioxidant Enzymatic Activities by Certain Antiepileptic Drugs (Valproic Acid, Oxcarbazepine, and Topiramate): Evidence in Humans and Experimental Models. Oxid Med. Cell Longev. 2013, 2013, 598493. [Google Scholar] [CrossRef] [PubMed]
  167. Rezaei, S.; Shab-Bidar, S.; Abdulahi Abdurahman, A.; Djafarian, K. Oxcarbazepine Administration and the Serum Levels of Homocysteine, Vitamin B12 and Folate in Epileptic Patients: A Systematic Review and Meta-Analysis. Seizure 2017, 45, 87–94. [Google Scholar] [CrossRef] [PubMed]
  168. Agarwal, N.B.; Agarwal, N.K.; Mediratta, P.K.; Sharma, K.K. Effect of Lamotrigine, Oxcarbazepine and Topiramate on Cognitive Functions and Oxidative Stress in PTZ-Kindled Mice. Seizure 2011, 20, 257–262. [Google Scholar] [CrossRef]
  169. Kim, Y.H.; Lee, T.-K.; Lee, J.-C.; Kim, D.W.; Hong, S.; Cho, J.H.; Shin, M.C.; Choi, S.Y.; Won, M.-H.; Kang, I.J. Therapeutic Administration of Oxcarbazepine Saves Cerebellar Purkinje Cells from Ischemia and Reperfusion Injury Induced by Cardiac Arrest through Attenuation of Oxidative Stress. Antioxidants 2022, 11, 2450. [Google Scholar] [CrossRef]
  170. Park, C.W.; Ahn, J.H.; Lee, T.-K.; Park, Y.E.; Kim, B.; Lee, J.-C.; Kim, D.W.; Shin, M.C.; Park, Y.; Cho, J.H.; et al. Post-Treatment with Oxcarbazepine Confers Potent Neuroprotection against Transient Global Cerebral Ischemic Injury by Activating Nrf2 Defense Pathway. Biomed. Pharmacother. 2020, 124, 109850. [Google Scholar] [CrossRef]
  171. Kośmider, K.; Kamieniak, M.; Czuczwar, S.J.; Miziak, B. Second Generation of Antiepileptic Drugs and Oxidative Stress. Int. J. Mol. Sci. 2023, 24, 3873. [Google Scholar] [CrossRef]
  172. Baumann, P. Pharmacology and Pharmacokinetics of Citalopram and Other SSRIs. Int. Clin. Psychopharmacol. 1996, 11 (Suppl. S1), 5–11. [Google Scholar] [CrossRef]
  173. Citalopram Monograph for Professionals. Drugs.com. Available online: https://www.drugs.com/monograph/citalopram.html (accessed on 7 February 2024).
  174. Ahmadian, E.; Eftekhari, A.; Fard, J.K.; Babaei, H.; Nayebi, A.M.; Mohammadnejad, D.; Eghbal, M.A. In Vitro and in Vivo Evaluation of the Mechanisms of Citalopram-Induced Hepatotoxicity. Arch. Pharm. Res. 2017, 40, 1296–1313. [Google Scholar] [CrossRef] [PubMed]
  175. Ilgin, S.; Kilic, G.; Baysal, M.; Kilic, V.; Korkut, B.; Ucarcan, S.; Atli, O. Citalopram Induces Reproductive Toxicity in Male Rats. Birth Defects Res. 2017, 109, 475–485. [Google Scholar] [CrossRef] [PubMed]
  176. Duan, S.; Fu, Y.; Dong, S.; Ma, Y.; Meng, H.; Guo, R.; Chen, J.; Liu, Y.; Li, Y. Psychoactive Drugs Citalopram and Mirtazapine Caused Oxidative Stress and Damage of Feeding Behavior in Daphnia Magna. Ecotoxicol. Environ. Saf. 2022, 230, 113147. [Google Scholar] [CrossRef] [PubMed]
  177. Ahmadian, E.; Eftekhari, A.; Babaei, H.; Nayebi, A.M.; Eghbal, M.A. Anti-Cancer Effects of Citalopram on Hepatocellular Carcinoma Cells Occur via Cytochrome C Release and the Activation of NF-kB. Anticancer. Agents Med. Chem. 2017, 17, 1570–1577. [Google Scholar] [CrossRef] [PubMed]
  178. Herbet, M.; Izdebska, M.; Piątkowska-Chmiel, I.; Poleszak, E.; Jagiełło-Wójtowicz, E. Estimation of Oxidative Stress Parameters in Rats after Simultaneous Administration of Rosuvastatin with Antidepressants. Pharmacol. Rep. 2016, 68, 172–176. [Google Scholar] [CrossRef] [PubMed]
  179. Gupta, S.; Upadhayay, D.; Sharma, U.; Jagannathan, N.R.; Gupta, Y.K. Citalopram Attenuated Neurobehavioral, Biochemical, and Metabolic Alterations in Transient Middle Cerebral Artery Occlusion Model of Stroke in Male Wistar Rats. J. Neurosci. Res. 2018, 96, 1277–1293. [Google Scholar] [CrossRef] [PubMed]
  180. Elsworthy, R.J.; Crowe, J.A.; King, M.C.; Dunleavy, C.; Fisher, E.; Ludlam, A.; Parri, H.R.; Hill, E.J.; Aldred, S. The Effect of Citalopram Treatment on Amyloid-β Precursor Protein Processing and Oxidative Stress in Human hNSC-Derived Neurons. Transl. Psychiatry 2022, 12, 285. [Google Scholar] [CrossRef]
  181. Khanzode, S.D.; Dakhale, G.N.; Khanzode, S.S.; Saoji, A.; Palasodkar, R. Oxidative damage and major depression: The potential antioxidant action of selective serotonin re-uptake inhibitors. Redox Rep. 2003, 8, 365–370. [Google Scholar] [CrossRef]
  182. DeVane, C.L.; Liston, H.L.; Markowitz, J.S. Clinical Pharmacokinetics of Sertraline. Clin. Pharm. 2002, 41, 1247–1266. [Google Scholar] [CrossRef] [PubMed]
  183. Cipriani, A.; La Ferla, T.; Furukawa, T.A.; Signoretti, A.; Nakagawa, A.; Churchill, R.; McGuire, H.; Barbui, C. Sertraline versus Other Antidepressive Agents for Depression. Cochrane Database Syst. Rev. 2010, 4, CD006117. [Google Scholar] [CrossRef] [PubMed]
  184. Li, Y.; Couch, L.; Higuchi, M.; Fang, J.-L.; Guo, L. Mitochondrial Dysfunction Induced by Sertraline, an Antidepressant Agent. Toxicol. Sci. 2012, 127, 582–591. [Google Scholar] [CrossRef] [PubMed]
  185. Battal, D.; Yalin, S.; Eker, E.D.; Aktas, A.; Sahin, N.O.; Cebo, M.; Berköz, M. Possible Role of Selective Serotonin Reuptake Inhibitor Sertraline on Oxidative Stress Responses. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 477–484. [Google Scholar] [PubMed]
  186. Jajoo, A.; Donlon, C.; Shnayder, S.; Levin, M.; McVey, M. Sertraline Induces DNA Damage and Cellular Toxicity in Drosophila That Can Be Ameliorated by Antioxidants. Sci. Rep. 2020, 10, 4512. [Google Scholar] [CrossRef]
  187. Abdel-Salam, O.M.E.; Youness, E.R.; Khadrawy, Y.A.; Sleem, A.A. Brain and Liver Oxidative Stress after Sertraline and Haloperidol Treatment in Mice. J. Basic Clin. Physiol. Pharmacol. 2013, 24, 115–123. [Google Scholar] [CrossRef] [PubMed]
  188. Chinnapaka, S.; Bakthavachalam, V.; Munirathinam, G. Repurposing Antidepressant Sertraline as a Pharmacological Drug to Target Prostate Cancer Stem Cells: Dual Activation of Apoptosis and Autophagy Signaling by Deregulating Redox Balance. Am. J. Cancer Res. 2020, 10, 2043–2065. [Google Scholar] [PubMed]
  189. Lima, M.L.; Abengózar, M.A.; Nácher-Vázquez, M.; Martínez-Alcázar, M.P.; Barbas, C.; Tempone, A.G.; López-Gonzálvez, Á.; Rivas, L. Molecular Basis of the Leishmanicidal Activity of the Antidepressant Sertraline as a Drug Repurposing Candidate. Antimicrob. Agents Chemother. 2018, 62, e01928-18. [Google Scholar] [CrossRef]
  190. Michalakeas, C.A.; Parissis, J.T.; Douzenis, A.; Nikolaou, M.; Varounis, C.; Andreadou, I.; Antonellos, N.; Markantonis-Kiroudis, S.; Paraskevaidis, I.; Ikonomidis, I.; et al. Effects of Sertraline on Circulating Markers of Oxidative Stress in Depressed Patients with Chronic Heart Failure: A Pilot Study. J. Card Fail. 2011, 17, 748–754. [Google Scholar] [CrossRef]
  191. Cipriani, A.; Furukawa, T.A.; Salanti, G.; Geddes, J.R.; Higgins, J.P.; Churchill, R.; Watanabe, N.; Nakagawa, A.; Omori, I.M.; McGuire, H.; et al. Comparative Efficacy and Acceptability of 12 New-Generation Antidepressants: A Multiple-Treatments Meta-Analysis. Lancet 2009, 373, 746–758. [Google Scholar] [CrossRef]
  192. Mierzejewski, P. Wenlafaksyna XR, zastosowanie kliniczne większych dawek (>150 mg). Psychiatria 2020, 17, 76–79. [Google Scholar] [CrossRef]
  193. Kıvrak, Y.; Güvenç, T.S.; Akbulut, N.; Yağcı, I.; Cığşar, G.; Gündüz, S.; Balcı, B. Accelerated Hypertension after Venlafaxine Usage. Case Rep. Psychiatry 2014, 2014, 659715. [Google Scholar] [CrossRef] [PubMed]
  194. Ballenger, J.C. Clinical Evaluation of Venlafaxine. J. Clin. Psychopharmacol. 1996, 16 (3 Suppl. 2), 29S–35S; discussion 35S–36S. [Google Scholar] [CrossRef] [PubMed]
  195. Madrigal-Bujaidar, E.; Paniagua-Pérez, R.; Rendón-Barrón, M.J.; Morales-González, J.A.; Madrigal-Santillán, E.O.; Álvarez-González, I. Investigation of the DNA Damage and Oxidative Effect Induced by Venlafaxine in Mouse Brain and Liver Cells. Toxics 2022, 10, 737. [Google Scholar] [CrossRef] [PubMed]
  196. Ahmadian, E.; Babaei, H.; Mohajjel Nayebi, A.; Eftekhari, A.; Eghbal, M.A. Venlafaxine-Induced Cytotoxicity Towards Isolated Rat Hepatocytes Involves Oxidative Stress and Mitochondrial/Lysosomal Dysfunction. Adv. Pharm. Bull 2016, 6, 521–530. [Google Scholar] [CrossRef] [PubMed]
  197. Wigner, P.; Synowiec, E.; Czarny, P.; Bijak, M.; Jóźwiak, P.; Szemraj, J.; Gruca, P.; Papp, M.; Śliwiński, T. Effects of Venlafaxine on the Expression Level and Methylation Status of Genes Involved in Oxidative Stress in Rats Exposed to a Chronic Mild Stress. J. Cell Mol. Med. 2020, 24, 5675–5694. [Google Scholar] [CrossRef] [PubMed]
  198. Mansouri, M.T.; Naghizadeh, B.; Ghorbanzadeh, B.; Amirgholami, N.; Houshmand, G.; Alboghobeish, S. Venlafaxine Inhibits Naloxone-Precipitated Morphine Withdrawal Symptoms: Role of Inflammatory Cytokines and Nitric Oxide. Metab. Brain Dis. 2020, 35, 305–313. [Google Scholar] [CrossRef] [PubMed]
  199. Khanam, R.; Najfi, H.; Akhtar, M.; Vohora, D. Evaluation of Venlafaxine on Glucose Homeostasis and Oxidative Stress in Diabetic Mice. Hum. Exp. Toxicol. 2012, 31, 1244–1250. [Google Scholar] [CrossRef]
  200. Lee, H. The Importance of Nutrition in Neurological Disorders and Nutrition Assessment Methods. Brain Neurorehabil. 2022, 15, e1. [Google Scholar] [CrossRef]
Figure 1. First (FGA) and second-generation (SGA) antipsychotics and extrapyramidal adverse effects—receptor scheme and mechanism [24,25,26].
Figure 1. First (FGA) and second-generation (SGA) antipsychotics and extrapyramidal adverse effects—receptor scheme and mechanism [24,25,26].
Ijms 25 07304 g001
Table 1. Mental and behavioural disorders in selected categories with clinical details [3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23].
Table 1. Mental and behavioural disorders in selected categories with clinical details [3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23].
Mental and Behavioural DisordersSelected CategoriesClinical DetailsPublications
Organic, including symptomatic, mental disordersDementia in Alzheimer’s disease Progressive, neurodegenerative disease
Loss of cognitive function
[3,4]
Vascular dementia Result of infraction of the brain due to vascular disease
Delirium, not induced by alcohol and other psychoactive substances Non-specific organic cerebral syndrome
Including: disturbances of consciousness, attention, perception and emotional disturbances
Unspecified organic or symptomatic mental disorder Brain disorder, injury, or toxicity
Mental and behavioural disorders
Mental and behavioural disorders due to psychoactive substance useMental and behavioural disorders due to use of alcoholWide variety of disorders resulting from abuse or misuse of alcohol/psychoactive substances[5,6,7,8]
Mental and behavioural disorders due to use of sedatives or hypnotics
Mental and behavioural disorders due to use of other stimulants, including caffeine
Mental and behavioural disorders due to multiple drug use and use of other psychoactive substances
Schizophrenia, schizotypal, and delusional disordersSchizophreniaMultifactorial, combination of genetic and environmental factors
Abnormalities in the perception or expression of reality
[9,10,11]
Schizotypal disorder Eccentric thoughts, inappropriate affect and behaviour, extreme social anxiety, and limited interpersonal interaction
Mood (affective) disordersManic episode Extreme changes in behaviour, and mood that drastically affect their functioning [12,13]
Bipolar affective disorder Severe mood swings (mania and depression)
Neurotic,
stress-related and somatoform disorders
Phobic anxiety disorders Strong, irrational fear, anxiety [14,15]
Obsessive-compulsive disorderIntrusive ideas, thoughts and images
Behavioural syndromes associated with physiological disturbances and physical factorsEating disordersPhysiological and psychological disturbances in appetite or food intake[16,17,18]
Nonorganic sleep disorders Disturbance of normal sleep patterns
Abuse of non-dependence-producing substancesThe use of non-addictive drugs, particularly those purchased without a prescription (antacids, herbal, folk remedies, steroids, hormones and vitamins)
Disorders of adult personality and behaviourSpecific personality disorders Disorders characterized by disturbances in the personality[19,20,21]
Gender identity disordersIncongruence between experienced or expressed gender and the one assigned at birth
Disorders of sexual preferenceIntense sexually arousing fantasies, urges, or behaviours
Behavioural and emotional disorders with onset usually occurring in childhood and adolescenceHyperkinetic disordersDifficulty sustaining cognitive engagement in activities[22,23]
Tic disordersRecurrent tics (involuntary, rapid, nonrhythmic motor movement or vocal)
Table 2. Antipsychotic, mood stabilizers and antidepressants drugs (SSRI, selective serotonin reuptake inhibitor) and (SNRI, serotonin and norepinephrine reuptake inhibitors)—generations and applications [1,3,9,24,25].
Table 2. Antipsychotic, mood stabilizers and antidepressants drugs (SSRI, selective serotonin reuptake inhibitor) and (SNRI, serotonin and norepinephrine reuptake inhibitors)—generations and applications [1,3,9,24,25].
Antipsychotic DrugsMood StabilizersAntidepressants
SchizophreniaBipolar DisorderDepression
I II III First-Line DrugsSecond-Line DrugsI II First-Line
Drugs
Add-on TherapySSRISNRIFirst-Line Drugs
HaloperidolClozapine Quetiapine
Risperidone
Olanzapine
AripiprazoleRisperidone
Olanzapine
Quetiapine
Aripiprazole
ClozapineLithium
Valproate
Carbamazepine
OxcarbazepineLithium
Valproate
Carbamazepine
OxcarbazepineSertraline CitalopramVenlafaxineSertraline Citalopram Venlafaxine
Table 3. Biological markers of oxidative stress [31,33,34].
Table 3. Biological markers of oxidative stress [31,33,34].
Destructive ChangesMarkers of Oxidative Stress
Lipid peroxidationMalondialdehyde
F2-isoprostanes
Oxidized low density lipoproteins
Oxidized LDL (low-density lipoprotein) antibodies
4-hydroxynonenal
Acrolein
Oxidation of nucleic acids8-hydroxy-2-deoxyguanosine
Reactive aldehydes
Reduced sugars, e.g., ribose
Carbohydrate oxidation3-nitrotyrosine
Protein oxidationSubstances that react with thiobarbituric acid
Glycation end products
Oxidized thiol groups
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sommerfeld-Klatta, K.; Jiers, W.; Rzepczyk, S.; Nowicki, F.; Łukasik-Głębocka, M.; Świderski, P.; Zielińska-Psuja, B.; Żaba, Z.; Żaba, C. The Effect of Neuropsychiatric Drugs on the Oxidation-Reduction Balance in Therapy. Int. J. Mol. Sci. 2024, 25, 7304. https://doi.org/10.3390/ijms25137304

AMA Style

Sommerfeld-Klatta K, Jiers W, Rzepczyk S, Nowicki F, Łukasik-Głębocka M, Świderski P, Zielińska-Psuja B, Żaba Z, Żaba C. The Effect of Neuropsychiatric Drugs on the Oxidation-Reduction Balance in Therapy. International Journal of Molecular Sciences. 2024; 25(13):7304. https://doi.org/10.3390/ijms25137304

Chicago/Turabian Style

Sommerfeld-Klatta, Karina, Wiktoria Jiers, Szymon Rzepczyk, Filip Nowicki, Magdalena Łukasik-Głębocka, Paweł Świderski, Barbara Zielińska-Psuja, Zbigniew Żaba, and Czesław Żaba. 2024. "The Effect of Neuropsychiatric Drugs on the Oxidation-Reduction Balance in Therapy" International Journal of Molecular Sciences 25, no. 13: 7304. https://doi.org/10.3390/ijms25137304

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