Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 72
1.
Expert Rev Neurother ; 24(5): 465-476, 2024 May.
Article En | MEDLINE | ID: mdl-38536761

INTRODUCTION: Major Depressive Disorder (MDD) is a mental health issue that significantly affects patients' quality of life and functioning. Despite available treatments, many patients continue to suffer due to incomplete symptom resolution and side effects. AREAS COVERED: This manuscript examines Vortioxetine's role in Major Depressive Disorder (MDD) treatment, highlighting its potential to reshape therapeutic strategies due to its unique Multimodal action and proven broad-spectrum efficacy in multiple depressive domains. A detailed examination of Vortioxetine's pharmacological aspects, including indications, dosage, pharmacodynamics, and pharmacokinetics, is provided, emphasizing its safety and effectiveness. The discussion extends to Vortioxetine's role in acute-phase treatment and maintenance of MDD and its profound impact on specialized depression domains. EXPERT OPINION: Vortioxetine is distinguished for its novel multimodal serotonin modulation mechanism, showcasing significant promise as an innovative treatment for MDD. Its efficacy, which is dose-dependent, along with a commendable tolerability profile, positions it as a potential leading option for initial treatment strategies. The discourse on dosage titration, particularly the strategy of initiating treatment at lower doses followed by gradual escalation, underscores the approach toward minimizing initial adverse effects while optimizing therapeutic outcomes, aligning with the principles of personalized medicine in psychiatric care.


Depressive Disorder, Major , Vortioxetine , Antidepressive Agents/administration & dosage , Antidepressive Agents/therapeutic use , Anxiety/complications , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Depressive Disorder, Major/complications , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/psychology , Emotions/drug effects , Escitalopram/administration & dosage , Escitalopram/therapeutic use , Post-Acute COVID-19 Syndrome/complications , Precision Medicine , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/therapeutic use , Serotonin/metabolism , Vortioxetine/administration & dosage , Vortioxetine/adverse effects , Vortioxetine/pharmacokinetics , Vortioxetine/pharmacology , Vortioxetine/therapeutic use , Humans , Neurotransmitter Agents/metabolism , Animals
2.
Immunopharmacol Immunotoxicol ; 46(2): 264-275, 2024 Apr.
Article En | MEDLINE | ID: mdl-38284357

BACKGROUND: Increasing evidence suggests that early life stress (ELS) and neuroinflammation are associated with the pathophysiology of depression. The purpose of this study was to determine the effects of Vortioxetine (VOR), a novel antidepressant, on ELS-induced behavioral changes and neuroinflammation. METHOD: Wistar Albino 4-week-old male rats were divided into four groups: control; chronic unpredictable stress (CUMS), VOR, CUMS + VOR. Neurobehavioral assessment was performed on the first, 21st, and 42nd days. RT-PCR was used to detect the expression of P2X7, NLRP3, IL1ß, IL18 in the prefrontal cortex. To assess the microglial activities of the prefrontal cortex, immunohistochemically stained CD68, and leukocyte common antigen (LCA) preparations were scanned with Manual WSI software, Basler camera, and scored. RESULT AND DISCUSSION: Exposure to CUMS was associated with depression and anxiety-like behaviors, and administration of VOR led to improvement in these behaviors. NLRP3, IL-1ß, and IL-18 were shown to be upregulated in the prefrontal cortex of CUMS rats, while their high expression was inhibited by VOR treatment. CD68 and LCA expressions were significantly higher in the CUMS group compared to the other groups. CONCLUSION: According to these results, it may be considered that NLRP3 inflammasome-associated neuroinflammatory response and microglial activation may play a role in the etiopathogenesis of ELS.


Depression , NLR Family, Pyrin Domain-Containing 3 Protein , Rats , Male , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Depression/metabolism , Vortioxetine/metabolism , Vortioxetine/pharmacology , Microglia/metabolism , Neuroinflammatory Diseases , Rats, Wistar , Inflammasomes/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Models, Theoretical
3.
CNS Neurol Disord Drug Targets ; 23(3): 395-401, 2024.
Article En | MEDLINE | ID: mdl-36944623

INTRODUCTION: Vortioxetine is a multimodal antidepressant drug that has been reported to have a positive impact on cognition, social function, and fatigue. Nevertheless, it has not been widely studied. Our objective was to explore the effects of vortioxetine on these and other parameters in patients with multiple sclerosis (MS) and depression. PATIENTS AND METHODOLOGY: This observational case series study included patients with MS and depression who received treatment with vortioxetine for at least 6 months. The patient history of depression and depressive symptoms was assessed. A neuropsychiatric evaluation was carried out using different scales, both before and after treatment. RESULTS: Of the 25 patients who enrolled in the study, 17 completed the treatment. Significant improvements were observed in health status (EQ-5D; p = 0.002), mood (Beck's Depression Inventory, BDI-II; p = 0.006), anxiety (State-Trait Anxiety Inventory, STAI-State; p = 0.021, and STAI-Trait; p = 0.011), and in the general health test (Short Form Health Survey, SF-36) for the vitality (p = 0.028) and mental health (p = 0.025) domains of the patients who completed the treatment. However, no statistically significant differences were observed in the cognitive tests related to attention, information processing speed, or fatigue. CONCLUSION: In this population, vortioxetine treatment was effective in reducing the symptoms of depression and improving anxiety, vitality, and mental health. In contrast, it did not produce any improvement in cognition or fatigue but an increase in sample size would be necessary to confirm these results.


Multiple Sclerosis , Humans , Vortioxetine/therapeutic use , Vortioxetine/pharmacology , Multiple Sclerosis/complications , Multiple Sclerosis/drug therapy , Depression/drug therapy , Depression/psychology , Cognition , Fatigue/drug therapy , Fatigue/etiology
4.
Neuroendocrinology ; 114(3): 279-290, 2024.
Article En | MEDLINE | ID: mdl-38104552

INTRODUCTION: Androgen deprivation therapy (ADT) is a mainstay treatment for prostate cancer, but many patients experience cognitive impairment in domains mediated by the medial prefrontal cortex (mPFC) and hippocampus. Prostate cancer typically occurs in older patients (>65 years). As age is often accompanied by cognitive decline, it may impact the efficacy of any treatment aimed at restoring cognitive impairment induced by ADT. Vortioxetine, a multimodal antidepressant that improves cognition in depression, has been shown to be efficacious in elderly patients. Therefore, vortioxetine may improve cognition in older patients who experience cognitive decline after ADT. METHODS: Young (3 months) and middle-aged (13 months) rats were used to investigate the influence of age on treating ADT-induced cognitive decline. As our previous studies used surgical castration, we tested if vortioxetine would reverse cognitive deficits associated with more translationally relevant chemical castration using degarelix. Vortioxetine was given in the diet for 21 days. Animals underwent behavioral testing to assess visuospatial memory mediated by the hippocampus and cognitive flexibility mediated by the mPFC. We also investigated changes in afferent-evoked responses in these regions in middle-aged rats. RESULTS: Degarelix induced impairments in both visuospatial memory and cognitive flexibility that were reversed by vortioxetine. Vortioxetine also rescued afferent-evoked responses in the mPFC and hippocampus. However, modest age-related reductions in baseline visuospatial memory limited our ability to detect further decreases induced by degarelix in middle-aged rats due to a floor effect. CONCLUSION: These results suggest that vortioxetine may be a treatment option for older prostate cancer patients who experience cognitive decline after ADT.


Androgen Antagonists , Oligopeptides , Prostatic Neoplasms , Male , Aged , Middle Aged , Humans , Rats , Animals , Vortioxetine/pharmacology , Androgen Antagonists/pharmacology , Androgens , Cognition
5.
Georgian Med News ; (342): 122-124, 2023 Sep.
Article En | MEDLINE | ID: mdl-37991966

The aim of this study was to evaluate current approaches to the pharmacotherapy of posttraumatic stress disorder. An information search was carried out in the databases PubMed, Ovid, EMBASE by keywords: "posttraumatic stress disorder", "treatment", and "medications". Search depth 2012-2022 years. From the general data (4877 articles) there were selected 14 articles with the highest degree of relevance. A content analysis of selected articles was carried out with the formation of recommendations for the use of pharmacotherapy in posttraumatic stress disorder. Currently, there are no unified approaches to the pharmacotherapy of posttraumatic stress disorder. Antidepressants (SSRI SNRIs) are primarily considered as first-line drugs, but only sertraline, paroxetine, and fluoxetine are approved by the FDA. But these drugs have a fairly wide range of side effects, including suicidal thoughts. The use of benzodiazepines should be limited as they increase the risk of developing posttraumatic stress disorder. Vortioxetine becomes a very promising option. The most significant benefits of vortioxetine are the significant positive effects of vortioxetine on attention, memory, and executive function. There is some evidence for the use of alpha-1 adrenoceptor antagonists and alpha-2 adrenoceptor agonists in therapy. In insomnia the use of prazosin and trazodone is recommended. Pharmacotherapy of posttraumatic stress disorder requires administration of medications with multimodal action. Currently, there are no unified approaches to the pharmacotherapy of posttraumatic stress disorder. Further randomized clinical trials are necessary for developing effective treatment of posttraumatic stress disorder.


Stress Disorders, Post-Traumatic , Humans , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Receptors, Adrenergic/therapeutic use , Stress Disorders, Post-Traumatic/drug therapy , Vortioxetine/pharmacology , Vortioxetine/therapeutic use
6.
Transl Psychiatry ; 13(1): 307, 2023 10 03.
Article En | MEDLINE | ID: mdl-37788996

Advances in prostate cancer treatment have significantly improved survival, but quality of life for survivors remains an under-studied area of research. Androgen deprivation therapy (ADT) is a foundational treatment for advanced prostate cancer and is used as an adjuvant for prolonged periods in many high-risk, localized tumors. More than half of patients treated with ADT experience debilitating cognitive impairments in domains such as spatial learning and working memory. In this study, we investigated the effects of androgen deprivation on hippocampal-mediated cognition in rats. Vortioxetine, a multimodal antidepressant, has been shown to improve cognition in depressed patients. Thus, we also tested the potential efficacy of vortioxetine in restoring impaired cognition after ADT. We further investigated mechanisms that might contribute to these effects, measuring changes in the circuitry and gene expression within the dorsal hippocampus. ADT via surgical castration induced impairments in visuospatial cognition on the novel object location test and attenuated afferent-evoked local field potentials recorded in the CA1 region of the dorsal hippocampus. Chronic dietary administration of vortioxetine effectively reversed these deficits. Castration significantly altered gene expression in the hippocampus, whereas vortioxetine had little effect. Pathway analysis revealed that androgen depletion altered pathways related to synaptic plasticity. These results suggest that the hippocampus may be vulnerable to ADT, contributing to cognitive impairment in prostate cancer patients. Further, vortioxetine may be a candidate to improve cognition in patients who experience cognitive decline after androgen deprivation therapy for prostate cancer and may do so by restoring molecular and circuit-level plasticity-related mechanisms compromised by ADT.


Cognitive Dysfunction , Prostatic Neoplasms , Humans , Male , Rats , Animals , Vortioxetine/metabolism , Vortioxetine/pharmacology , Androgen Antagonists/adverse effects , Androgen Antagonists/metabolism , Prostatic Neoplasms/drug therapy , Androgens/metabolism , Androgens/pharmacology , Quality of Life , Cognitive Dysfunction/metabolism , Hippocampus/metabolism
7.
J Clin Psychiatry ; 84(6)2023 10 04.
Article En | MEDLINE | ID: mdl-37796652

Objective: Major depressive disorder (MDD) remains difficult to treat, with many patients resistant to existing treatments or experiencing relapse. Cognitive dysfunction is associated with more severe clinical outcomes. Vortioxetine has shown efficacy in remediating depression-associated cognitive impairment. Anti-inflammatory augmentation of antidepressants is a new strategy in treating depression and has not previously been assessed for effects on cognition in depression.Methods: Exploratory analyses were performed on secondary outcome cognitive data from the PREDDICT parallel-group, randomized, double-blind, placebo-controlled trial at the University of Adelaide (Australia). Participants (N = 119) with MDD (validated with Mini-International Neuropsychiatric Interview for DSM-IV) were treated with vortioxetine and celecoxib or vortioxetine and placebo for 6 weeks between December 2017 and April 2020. Measures included objective cognition composite scores (Choice Reaction Time, N-Back, Digit Symbol Substitution Test, Trail Making Task Part B), subjective cognition scores (Perceived Deficits Questionnaire), and global cognition composite scores (combined objective and subjective scores) derived from the THINC integrated tool (THINC-it). High-sensitivity C-reactive protein (hsCRP) measured at baseline and week 6 was tested for a predictive relationship with cognitive outcomes.Results: Cognition composite scores demonstrated improvement by week 6 in both treatment groups. However, there was no significant interaction between change over time and treatment group. HsCRP did not have a significant relationship with any tested cognition measures.Conclusions: Both treatment groups showed a reduction in depression-associated cognitive impairment. No superior clinical effect was reported for the add-on celecoxib group. HsCRP was modulated by neither vortioxetine nor add-on celecoxib.Trial Registration: ANZCTR identifier: ACTRN12617000527369.


Depressive Disorder, Major , Humans , Vortioxetine/pharmacology , Vortioxetine/therapeutic use , Depressive Disorder, Major/psychology , Celecoxib/adverse effects , C-Reactive Protein , Treatment Outcome , Cognition , Double-Blind Method
8.
Psychopharmacology (Berl) ; 240(12): 2499-2513, 2023 Dec.
Article En | MEDLINE | ID: mdl-37555927

BACKGROUND: Depression, one of the most significant mental disorders, is still poorly understood in terms of its pathogenetic mechanisms despite its well-recognized association with stress. OBJECTIVES: The current study's goal was to ascertain how the novel antidepressant drug vortioxetine (VOR) affected the BDNF (brain-derived neurotrophic factor), S100, amyloid ß (Aß), CREB (cAMP response element-binding protein), and NR2B, as well as its impact on depression-like behaviors, and tissue damage in an experimental rodent model of depression caused by chronic unpredictable stress. METHODS: We employed twenty-eight Wistar albino male rats, and we randomly divided them into four groups, each consisting of 7 rats: control, CUMS (chronic unpredictable mild stress), CUMS+vortioxetine (CUMS+VOR), and CUMS+fluoxetine (CUMS+FLU). Sucrose preference and forced swimming tests (SPT and FST, respectively), PCR, ELISA, and histopathological and immunohistochemical evaluation were made on brains. RESULTS: The behaviors of reduced immobility in the FST and increased sucrose preference were observed in the CUMS group and they improved in the groups treated with VOR and FLU. Compared with the control group, the group exposed to CUMS showed increased Aß and decreased BDNF, CREB, and S-100 expressions, as well as neuronal degeneration (p<0.001). VOR and FLU treatment ameliorate the findings. CONCLUSIONS: This study demonstrated significant ameliorative effects of VOR in an experimental model of chronic unpredictable depression to reduce brain tissue damage and depression-like behaviors in rats. Effects of CUMS on the brain and possible effects of VOR.


Amyloid beta-Peptides , Depression , Humans , Rats , Animals , Vortioxetine/pharmacology , Depression/drug therapy , Depression/metabolism , Amyloid beta-Peptides/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Rats, Wistar , Sucrose/pharmacology , Glutamates/metabolism , Stress, Psychological/complications , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Disease Models, Animal , Hippocampus , S100 Calcium Binding Protein beta Subunit/metabolism , S100 Calcium Binding Protein beta Subunit/pharmacology
9.
Int J Neuropsychopharmacol ; 26(10): 655-668, 2023 10 19.
Article En | MEDLINE | ID: mdl-37025079

BACKGROUND: Although thought of as a multimodal-acting antidepressant targeting the serotonin system, more molecules are being shown to participate in the antidepressant mechanism of vortioxetine. A previous report has shown that vortioxetine administration enhanced the expression of rapamycin complex 1 (mTORC1) in neurons. It has been well demonstrated that mTORC1 participates in not only the pathogenesis of depression but also the pharmacological mechanisms of many antidepressants. Therefore, we speculate that the antidepressant mechanism of vortioxetine may require mTORC1. METHODS: Two mouse models of depression (chronic social defeat stress and chronic unpredictable mild stress) and western blotting were first used together to examine whether vortioxetine administration produced reversal effects against the chronic stress-induced downregulation in the whole mTORC1 signaling cascade in both the hippocampus and medial prefrontal cortex (mPFC). Then, LY294002, U0126, and rapamycin were used together to explore whether the antidepressant effects of vortioxetine in mouse models of depression were attenuated by pharmacological blockade of the mTORC1 system. Furthermore, lentiviral-mTORC1-short hairpin RNA-enhanced green fluorescence protein (LV-mTORC1-shRNA-EGFP) was adopted to examine if genetic blockade of mTORC1 also abolished the antidepressant actions of vortioxetine in mice. RESULTS: Vortioxetine administration produced significant reversal effects against the chronic stress-induced downregulation in the whole mTORC1 signaling cascade in both the hippocampus and mPFC. Both pharmacological and genetic blockade of the mTORC1 system notably attenuated the antidepressant effects of vortioxetine in mice. CONCLUSIONS: Activation of the mTORC1 system in the hippocampus and mPFC is required for the antidepressant actions of vortioxetine in mice.


Antidepressive Agents , Hippocampus , Mice , Animals , Vortioxetine/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Antidepressive Agents/pharmacology , Antidepressive Agents/metabolism , Prefrontal Cortex/metabolism , Sirolimus/pharmacology
10.
Behav Brain Res ; 444: 114365, 2023 04 27.
Article En | MEDLINE | ID: mdl-36858318

Schizophrenia is a devastating psychiatric disorder with complex symptoms and neurobiology. Serotonergic dysregulation is known to contribute to the pathogenesis of schizophrenia although dopaminergic and glutamatergic systems are thought to have central roles in neurobiology. No significant success can be achieved in the treatment of negative and cognitive symptoms while positive symptoms can be significantly reduced with current pharmacotherapy. Vortioxetine is a new multimodal antidepressant with 5-HT1A agonism, 5-HT1B partial agonism, 5-HT3, 5-HT7, and 5-HT1D antagonism, and serotonin reuptake inhibition. A limited number of studies suggest its therapeutic effect on the negative and cognitive symptoms of schizophrenia. Therefore, we investigated the potential beneficial effects of vortioxetine on behavioral and molecular deficits in the MK-801 model of schizophrenia in rats. Female Wistar albino rats (10-12 weeks) were grouped as saline, MK-801 (0.2 mg/kg), MK-801 + vortioxetine (2.5 mg/kg), MK-801 + vortioxetine (5 mg/kg), MK-801 + vortioxetine (10 mg/kg), MK-801 + risperidone (0.3 mg/kg), MK-801 + haloperidol (1 mg/kg) (n = 8 in each group). MK-801 has been daily administered (i.p.) for 14 days. Vortioxetine and antipsychotic treatments were injected for 21 days after a washout period of MK-801 and locomotor activity (LA), social interaction (SI), novel object recognition (NOR), Y-maze and prepulse inhibition (PPI) tests were performed at the 16-20th days of treatments, respectively. ELISA test was conducted to evaluate molecular analyses. MK-801 decreased PPI (%), social behaviors, and discrimination index in NOR and alternation (%) in the Y-maze test. In NOR and Y-maze tests, especially vortioxetine 5 and 10 mg/kg increased discrimination index and alternation (%) compared to MK-801. In addition, vortioxetine administration increased social behaviors. Moreover, MK-801 decreased GAD67 and parvalbumin levels while vortioxetine increased these protein levels compared to MK-801. Herein, we first suggested a potential therapeutic effect of vortioxetine, a new multimodal antidepressant, on negative and cognitive symptoms and neurobiological deficits including GAD67 and parvalbumin low expression in the MK-801 model in rats. It would be beneficial to confirm our results in different rodent models and to shed light on the possible mechanisms underlying these effects.


Dizocilpine Maleate , Schizophrenia , Animals , Rats , Female , Vortioxetine/pharmacology , Schizophrenia/drug therapy , Parvalbumins , Piperazines/pharmacology , Rats, Wistar , Antidepressive Agents/pharmacology , Serotonin 5-HT1 Receptor Agonists , Cognition
11.
Cells ; 12(6)2023 03 08.
Article En | MEDLINE | ID: mdl-36980178

Parkinson's Disease (PD) is a neurodegenerative disorder characterized by motor symptoms that result from loss of nigrostriatal dopamine (DA) cells. While L-DOPA provides symptom alleviation, its chronic use often results in the development of L-DOPA-induced dyskinesia (LID). Evidence suggests that neuroplasticity within the serotonin (5-HT) system contributes to LID onset, persistence, and severity. This has been supported by research showing 5-HT compounds targeting 5-HT1A/1B receptors and/or the 5-HT transporter (SERT) can reduce LID. Recently, vortioxetine, a multimodal 5-HT compound developed for depression, demonstrated acute anti-dyskinetic effects. However, the durability and underlying pharmacology of vortioxetine's anti-dyskinetic actions have yet to be delineated. To address these gaps, we used hemiparkinsonian rats in Experiment 1, examining the effects of sub-chronic vortioxetine on established LID and motor performance. In Experiment 2, we applied the 5-HT1A antagonist WAY-100635 or 5-HT1B antagonist SB-224289 in conjunction with L-DOPA and vortioxetine to determine the contributions of each receptor to vortioxetine's effects. The results revealed that vortioxetine consistently and dose-dependently attenuated LID while independently, 5-HT1A and 5-HT1B receptors each partially reversed vortioxetine's effects. Such findings further support the promise of pharmacological strategies, such as vortioxetine, and indicate that broad 5-HT actions may provide durable responses without significant side effects.


Dyskinesia, Drug-Induced , Levodopa , Rats , Animals , Levodopa/adverse effects , Vortioxetine/pharmacology , Vortioxetine/therapeutic use , Serotonin , Rats, Sprague-Dawley , Dyskinesia, Drug-Induced/drug therapy
12.
Am J Geriatr Psychiatry ; 31(6): 385-397, 2023 06.
Article En | MEDLINE | ID: mdl-36739247

OBJECTIVE: Age-related cognitive decline is common and potentially modifiable with cognitive training. Combining cognitive training with pro-cognitive medication offers an opportunity to modify brain networks to mitigate age-related cognitive decline. We tested the hypothesis that the efficacy of cognitive training could be amplified by combining it with vortioxetine, a pro-cognitive and pro-neuroplastic multimodal antidepressant. METHODS: We evaluated the effects of 6 months of computerized cognitive training plus vortioxetine (versus placebo) on resting state functional connectivity in older adults (age 65+) with age-related cognitive decline. We first evaluated the association of functional connectivity with age and cognitive performance (N = 66). Then we compared the effects of vortioxetine plus cognitive training versus placebo plus cognitive training on connectivity changes over the training period (n = 20). RESULTS: At baseline, greater age was significantly associated with lower within-network strength and network segregation, and poorer cognitive function. Cognitive training plus vortioxetine over 6 months positively impacted the relationship between age to mean network segregation. These effects were not observed in the placebo group. In contrast, vortioxetine did not modify the relationship of age to change in mean within-network strength. Exploratory analyses identified the cingulo-opercular network as the network most affected by cognitive training plus vortioxetine. CONCLUSION: This preliminary study provides evidence that combining cognitive training with pro-cognitive medication may modulate the effects of aging on functional brain networks. Results indicate that for older adults experiencing age-related cognitive decline, vortioxetine has a potentially beneficial effect on the correspondence between aging and functional brain network segregation. These results await replication in a larger sample.


Cognition , Cognitive Training , Aged , Humans , Brain , Magnetic Resonance Imaging , Vortioxetine/pharmacology , Vortioxetine/therapeutic use
13.
Eur Neuropsychopharmacol ; 70: 21-28, 2023 05.
Article En | MEDLINE | ID: mdl-36808043

Major Depressive Episodes (MDE) following COVID-19 are frequent, can have a characteristic clinical picture, and are associated with immune-inflammatory changes. Vortioxetine is known to improve physical and cognitive performance in patients with depression and shows anti-inflammatory and anti-oxidative activities. This study aimed to retrospectively evaluate the effects of vortioxetine after 1 and 3 months of treatment in 80 patients (44.4% males, 54±17.2 years) with post-COVID-19 MDE. The primary outcome was improvement in physical and cognitive symptoms measured by specific items of Hamilton Depression Rating Scale (HDRS) and Hamilton Anxiety Rating Scale (HARS), Short Form-36 Health Survey Questionnaire (SF-36), Digit Symbol Substitution Test (DSST), Perceived Deficits Questionnaire for Depression (PDQ-D5). Changes in mood, anxiety, anhedonia, sleep, and quality of life were also investigated, as well as the underlying inflammatory status. Results show that, alongside reduction of depressive symptoms (HDRS, p<0.001), vortioxetine (mean dose: 10.1±4.1 mg/day) significantly improved physical features (all measurements p<0.001) and cognitive functioning (DDST, p=0.02; PDQ-D5, p<0.001) throughout treatment. We also observed significant reductions in inflammatory indexes. Therefore, vortioxetine might be a favorable therapeutic choice in post-COVID-19 patients with MDE because of its beneficial effects on physical complaints and cognition, features that appear to be specifically affected in relation to SARS-CoV-2 infection, and its good safety/tolerability profile. High prevalence and clinical and socioeconomic implications of COVID-19 consequences are a major public health concern and developing tailored, safe interventions is crucial to promote full functional recovery.


COVID-19 , Depressive Disorder, Major , Male , Humans , Female , Vortioxetine/therapeutic use , Vortioxetine/pharmacology , Depressive Disorder, Major/complications , Depressive Disorder, Major/drug therapy , Antidepressive Agents/therapeutic use , Retrospective Studies , Quality of Life , Treatment Outcome , COVID-19/complications , SARS-CoV-2 , Cognition , Double-Blind Method
14.
Pharmacol Rep ; 75(2): 266-275, 2023 Apr.
Article En | MEDLINE | ID: mdl-36637685

BACKGROUND: Many psychoactive compounds have been developed to have more beneficial clinical efficacy than conventional drugs by adding agonistic action at 5-HT1A receptors. The aim of the present study was to evaluate several psychotropic drugs that had been reported to behave as an agonist at 5-HT1A receptor (aripiprazole, brexpiprazole, asenapine, lurasidone, and vortioxetine) in both rat and postmortem human brain membranes. METHODS: The [35S]GTPγS binding assay for Gi/o proteins coupled with 5-HT1A receptors was performed in rat brain membranes and postmortem human brain membranes. RESULTS: The specific binding was stimulated by brexpiprazole in rat hippocampus, human hippocampus, and human prefrontal cortex. Aripiprazole also behaved as an agonist in the same brain regions. Interestingly, its potency was much higher in rat hippocampal membranes than in human brain membranes, indicating the possibility of species differences. Although vortioxetine was an efficacious stimulator at high concentrations, its potency was undeterminable because of a lack of saturability. In addition to 5-HT1A receptor agonism, involvement of other components, e.g., 5-HT1B receptor agonism, was speculated by the biphasic inhibitory effects of the selective 5-HT1A receptor neutral antagonist. Negligible stimulatory effects were obtained as to lurasidone and asenapine. CONCLUSIONS: Our previous studies have raised the concept of a psychoactive drug group with a common pharmacological mechanism of action, i.e., 5-HT1A receptor agonism, consisting of perospirone, aripiprazole, ziprasidone, clozapine, quetiapine, nemonapride, and trazodone. The present study demonstrates the data indicating that brexpiprazole and probably vortioxetine are included in this drug group. Lurasidone and asenapine are excluded from this group.


Receptor, Serotonin, 5-HT1A , Serotonin , Rats , Humans , Animals , Aripiprazole/pharmacology , Serotonin/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Vortioxetine/pharmacology , Receptor, Serotonin, 5-HT1A/metabolism , Lurasidone Hydrochloride/pharmacology , Serotonin Receptor Agonists/pharmacology , Brain/metabolism , Psychotropic Drugs/pharmacology
15.
Int J Mol Sci ; 23(22)2022 Nov 10.
Article En | MEDLINE | ID: mdl-36430306

This determination of the mitochondrial effect of pharmacologically different antidepressants (agomelatine, ketamine and vortioxetine) was evaluated and quantified in vitro in pig brain-isolated mitochondria. We measured the activity of mitochondrial complexes, citrate synthase, malate dehydrogenase and monoamine oxidase, and the mitochondrial respiratory rate. Total hydrogen peroxide production and ATP production were assayed. The most potent inhibitor of all mitochondrial complexes and complex I-linked respiration was vortioxetine. Agomelatine and ketamine inhibited only complex IV activity. None of the drugs affected complex II-linked respiration, citrate synthase or malate dehydrogenase activity. Hydrogen peroxide production was mildly increased by agomelatine, which might contribute to increased oxidative damage and adverse effects at high drug concentrations. Vortioxetine significantly reduced hydrogen peroxide concentrations, which might suggest antioxidant mechanism activation. All tested antidepressants were partial MAO-A inhibitors, which might contribute to their antidepressant effect. We observed vortioxetine-induced MAO-B inhibition, which might be linked to decreased hydrogen peroxide formation and contribute to its procognitive and neuroprotective effects. Mitochondrial dysfunction could be linked to the adverse effects of vortioxetine, as vortioxetine is the most potent inhibitor of mitochondrial complexes and complex I-linked respiration. Clarifying the molecular interaction between drugs and mitochondria is important to fully understand their mechanism of action and the connection between their mechanisms and their therapeutic and/or adverse effects.


Ketamine , Animals , Swine , Vortioxetine/pharmacology , Ketamine/pharmacology , Malate Dehydrogenase , Citrate (si)-Synthase , Hydrogen Peroxide , Antidepressive Agents/pharmacology , Electron Transport Complex I , Monoamine Oxidase
16.
Rev Esp Anestesiol Reanim (Engl Ed) ; 69(10): 640-648, 2022 Dec.
Article En | MEDLINE | ID: mdl-36241510

Neuropathic pain is an important and disabling clinical problem, its management constitutes a challenge for healthcare professionals. Vortioxetine is a new antidepressant drug with multimodal action, which gives it a unique profile. Tricyclic antidepressants, in particular amitriptyline, and serotonin and norepinephrine reuptake inhibitors venlafaxine and duloxetine are first-line drugs in the treatment of neuropathic pain. The interaction between the pain and depression binomial is very frequent, being the most frequent psychological complication in patients with chronic pain. This comprehensive and descriptive review summarizes the most relevant pharmacological data on vortioxetine, as well as the specific literature on vortioxetine in neuropathic pain and chronic pain.


Chronic Pain , Neuralgia , Humans , Vortioxetine/therapeutic use , Vortioxetine/pharmacology , Chronic Pain/drug therapy , Antidepressive Agents/therapeutic use , Duloxetine Hydrochloride/therapeutic use , Neuralgia/drug therapy
17.
Physiol Behav ; 257: 113989, 2022 Dec 01.
Article En | MEDLINE | ID: mdl-36206848

Vortioxetine is a serotoninergic multi-target antidepressant, approved to treat major depressive disorder, and carries out its behavioral, pharmacological, and physiological effects through the blocking of serotonin 5-HT1D, 5-HT3, and 5-HT7 receptors and by activating 5-HT1A receptors. Some studies report that the simultaneous activation of the 5-HT1A serotonin receptors or blockade of 5-HT3 serotonin receptors decreased cocaine-induced locomotor activity and cocaine self-administration. Recent studies showed that vortioxetine decreased alcohol consumption. This studio aimed to evaluate the effects of vortioxetine dosing on cocaine-induced behavioral (cocaine-induced locomotor activity and cocaine-induced locomotor sensitization) and neurochemical (dopamine levels) effects. Male Wistar rats received cocaine during the induction and expression of locomotor sensitization. Vortioxetine was administered 30 min before cocaine. After each treatment, the locomotor activity was recorded for 30 min. Dopamine levels were determined in the ventral striatum, the prefrontal cortex (PFC), and the ventral tegmental area (VTA) by high-performance liquid chromatographic (HPLC) in animals treated with vortioxetine and cocaine. In this study, we found that vortioxetine decreased cocaine-induced locomotor activity, as well as the induction and expression of locomotor sensitization. As well as the amount of cocaine-induced dopamine decreased. Vortioxetine can be a useful therapeutic agent to reduce cocaine abuse.


Cocaine , Depressive Disorder, Major , Animals , Male , Rats , Cocaine/pharmacology , Dopamine/metabolism , Rats, Wistar , Receptors, Serotonin , Serotonin/metabolism , Vortioxetine/pharmacology , Locomotion
18.
Medicina (Kaunas) ; 58(10)2022 Sep 28.
Article En | MEDLINE | ID: mdl-36295524

Background and objectives: Vortioxetine (VRT) is a relatively new selective serotonin reuptake inhibitor (SSRI) antidepressant and serotonin receptor modulator, approved for the treatment of major depression and generalized anxiety disorder. Depression has been linked with psychomotor disengagement, oxidative stress burden and decreased blood levels of brain-derived neurotrophic factor (BDNF). In our study we performed the experimental investigation of VRT, magnesium and of their association on the rats' endurance capacity, motor behavior and blood biological disturbances in rats subjected to forced exercise in treadmill test. Materials and Methods: The substances were administered orally for 14 consecutive days, as follows: group 1 (control): distilled water 0.3 mL/100 g body; group 2 (Mg): magnesium chloride 200 mg/kg body; group 3 (VRT): VRT 20 mg/kg body; group 4 (VRT+Mg): VRT 20 mg/kg body + magnesium chloride 200 mg/kg body. Magnesium was used as positive control substance with known effects in treadmill test. The consequences of VRT treatment on glucose, cortisol, BDNF and oxidative stress biomarkers (superoxide-dismutase, malondialdehyde, glutathione-peroxidase, lactate dehydrogenase) were also assessed. Results and conclusions: The use of VRT resulted in an improvement in motor capacity and an increase of the rats' endurance to physical effort. The administration of VRT increased the serum BDNF levels and reduced the oxidative stress in rats subjected to physical effort. The association of magnesium potentiated the effects of VRT on physical performances, the antioxidant activity and the decreasing in serum stress markers in treadmill test in rats.


Brain-Derived Neurotrophic Factor , Magnesium , Rats , Animals , Vortioxetine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Antioxidants , Magnesium Chloride , Hydrocortisone , Superoxides , Glutathione Peroxidase , Malondialdehyde , Oxidative Stress , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Biomarkers , Glutathione , Physical Functional Performance , Glucose , Lactate Dehydrogenases , Water
19.
Zh Nevrol Psikhiatr Im S S Korsakova ; 122(6. Vyp. 2): 84-90, 2022.
Article Ru | MEDLINE | ID: mdl-35797201

The opinion article is devoted to the analysis of a large-scale comprehensive systematic review of relevant and practically oriented studies of vortioxetine use in depression for the period to 2020. The mechanisms of multimodal action, issues of efficacy and safety of the drug, including direct procognitive effects, effects on anhedonia, anxiety, sleep disorders, analgesic and anti-inflammatory effects are highlighted. An increase in the efficacy of vortioxetine when the dose is increased to 20 mg/day is emphasized. Prospective directions for antidepressant research are outlined.


Depressive Disorder, Major , Vortioxetine , Depressive Disorder, Major/drug therapy , Humans , Prospective Studies , Systematic Reviews as Topic , Vortioxetine/pharmacology , Vortioxetine/therapeutic use
20.
Biochem Pharmacol ; 202: 115097, 2022 08.
Article En | MEDLINE | ID: mdl-35609645

Depression is a very prevalent psychiatric disorder which threats nearly one in six of the population in this world. To date, the pathogenesis of depression remains elusive and is thought to depend on multiple factors in which chronic stress is critical. Currently, it has been demonstrated that besides monoaminergic dysfunction, depression is accompanied by several other important pathological phenomena such as impaired neurogenesis and decreased brain-derived neurotrophic factor (BDNF)-cAMP response element binding protein (CREB) signaling cascade in the hippocampus. F3/Contactin is a cell-adhesion molecule which has been reported to correlate with hippocampal neurogenesis and BDNF-CREB signaling. Here we assumed that F3/Contactin may be implicated in depression, and various methods including western blotting, immunofluorescence, virus-mediated gene transfer and chronic stress models of depression were adopted together. It was found that both chronic restraint stress (CRS) and chronic social defeat stress (CSDS) significantly decreased the expression of F3/Contactin in the hippocampus. Adeno-associated virus (AAV)-mediated over-expression of hippocampal F3/Contactin notably prevented the CRS-induced and CSDS-induced depressive-like behaviors in mice. Moreover, hippocampal F3/Contactin over-expression also fully reversed the CRS-induced and CSDS-induced dysfunction in the hippocampal BDNF-CREB signaling and neurogenesis of mice. Furthermore, administration of vortioxetine, a multimodal-acting antidepressant, fully ameliorated the inhibitory actions of both CRS and CSDS on the hippocampal F3/Contactin expression. In contrast, AAV-mediated knockdown of hippocampal F3/Contactin significantly abolished the protecting effects of vortioxetine against CRS and CSDS. Collectively, hippocampal F3/Contactin is implicated in depression and could be a novel antidepressant target.


Antidepressive Agents , Brain-Derived Neurotrophic Factor , Vortioxetine , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Contactins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Depression/drug therapy , Depression/etiology , Disease Models, Animal , Hippocampus , Humans , Mice , Mice, Inbred C57BL , Stress, Psychological/complications , Stress, Psychological/metabolism , Vortioxetine/pharmacology
...