Abstract
Agomelatine is a selective agonist of melatonin receptor 1A/melatonin receptor 1B (MT1/MT2) and antagonist of 5-hydroxytryptamine 2C receptors. It is used clinically to treat major depressive episodes in adults. The pro-chronobiological activity of agomelatine reconstructs sleep-wake rhythms and normalizes circadian disturbances via its agonistic effect of melatonin receptor 1A/melatonin receptor 1B, which work simultaneously to counteract depression and anxiety disorder. Moreover, by antagonizing neocortical postsynaptic 5-hydroxytryptamine 2C receptors, agomelatine enhances the release of dopamine and noradrenaline in the prefrontal cortex, increases the activity of dopamine and noradrenaline, and thereby reduces depression and anxiety disorder. The combination of these two effects means that agomelatine exhibits a unique pharmacological role in the treatment of depression, anxiety, and disturbance of the circadian rhythm. Emotion and sleep are closely related to memory and cognitive function. Memory disorder is defined as any forms of memory abnormality, which is typically evident in a broad range of neurodegenerative diseases, including Alzheimer’s disease. Memory impairment and cognitive impairment are common symptoms of neurodegenerative and psychiatric diseases. Therefore, whether agomelatine can improve memory and cognitive behaviors if used for alleviating depression and circadian-rhythm sleep disorders has become a research “hotspot”. This review presents the latest findings on the effects of agomelatine in the treatment of psychologic and circadian-rhythm sleep disorders in clinical trials and animal experiments. Our review evaluates recent studies on treatment of memory impairment and cognitive impairment in neurodegenerative and psychiatric diseases.
Keywords: agomelatine, antidepressant, anxiety, apathy, circadian-rhythm sleep disorder, cognitive impairment, depression, melatonergic, memory disorder, mood disorder, neurodegenerative disease
Introduction
Agomelatine (AGO) is N-(2-[7-methoxy-1-naphthalenyl]ethyl) acetamide (S20098). It was discovered and developed by the European Pharmaceutical Company Servier Laboratories Limited in 1992 (Yous et al., 1992; Armstrong et al., 1993). As the first melatonergic antidepressant, AGO was approved by the European Medicines Agency in the European Union in 2009 and Therapeutic Goods Administration in Australia in 2010 for the treatment of major depression. AGO alleviates circadian-rhythm sleep disorders in patients suffering from depression with synergistic agonism at melatonin receptor 1A/melatonin receptor 1B (MT1/MT2) and antagonism at 5-hydroxytryptamine 2C (5-HT2C) receptors. AGO provides a useful alternative pharmacological strategy to existing antidepressant drugs (Norman and Olver, 2019).
Commonly used drugs for depression are selective serotonin reuptake inhibitors (SSRIs) and selective serotonin-norepinephrine reuptake inhibitors (SSNRIs), which ameliorate depression by increasing the 5-hydroxytryptamine (5-HT) level. However, SSRIs and SSNRIs produce adverse effects, such as withdrawal syndrome, sexual dysfunction, difficulties in sleeping, and agitation (Erdoğan et al., 2020). Compared with SSRIs and SSNRIs, AGO exerts an antidepressant effect by binding directly to 5-HT2C receptors in postsynaptic membranes without affecting 5-HT concentrations in synaptic clefts. Furthermore, compared with natural endogenous melatonin, the replacement of an indole ring with a naphthalene ring in AGO (Table 1) enhances the metabolic stability of AGO and prolongs its biological half-life, which leads to more effective correction of circadian-rhythm disorders and alleviation of sleep disorders by activating MT1 and MT2. With synergistic agonism at MT1/MT2 and antagonism at 5-HT receptors, AGO has obvious positive effects on the sleep-wake cycle and depression while lacking serious side effects (including sexual side effects) (Kennedy et al., 2008).
Table 1.
Comparison of drug characteristics between agomelatine and melatonin
| Drug name | Agomelatine | Melatonin |
|---|---|---|
| Source | Chemical synthesis | Endogenous hormone |
| Weight (kDa) | 243 | 232 |
| Half-life | < 2 h | 35–50 min |
| Chemical formula | C15 H17 NO2 | C13 H16 N2 O2 |
| Chemical structure |
|
|
| Pharmacological properties | Antagonist of 5-HT2C receptors; agonist of MT1 and MT2 | Agonist of MT1 and MT2 |
5-HT2C: 5-Hydroxytryptamine 2C; MT1: melatonin receptor 1A; MT2: melatonin receptor 1B.
Neurodegenerative disease refers to the principal pathology associated with disorders such as Alzheimer’s disease, Huntington’s disease and Parkinson’s disease. The patients with these diseases exhibit diverse patterns of sleep disturbance, memory disorder and cognitive impairment. Many researchers have reported a strong association among sleep disturbance, mood changes, memory complaints, and reduced cognitive performance (Tempesta et al., 2018; Guan et al., 2020; Gutierrez et al., 2021; Xie et al., 2021; Hernandez and Shukla, 2022). Sleep disorders, emotional abnormalities, and cognitive decline are usually present in the same individual and influence each other. An improvement in sleep and mood disorders often alleviates memory deficit and cognitive decline. Therefore, based on the peculiar characteristic of AGO integrating melatonergic agonism and 5-HT antagonism and the efficacy of AGO in improving sleep and mood, it is intriguing and meaningful to ascertain if AGO can also ameliorate the deficits in memory and cognitive behaviors while attenuating depression and sleep-rhythm disorders.
Here, we summarize the latest research progress of AGO in the treatment of depression, sleep disorders, and cognitive impairments. We aim to provide new insights into the pharmacological action and mechanisms of AGO in the clinical setting.
Search Strategy and Selection Criteria
The studies cited in this narrative review were published from 2000 to 2021. They were searched by QS and TL on 31 December 2021 using the PubMed database and met the inclusion criteria, search terms include at least one of the following the keywords “agomelatine” and “S20098”, and at least one of the following keywords: “mood disorder”, “depression”, “anxiety”, “apathy”, “anhedonia”, “sleep-rhythm disorder”, “memory”, “cognition”, “dementia”, and “neurodegenerative disorder”.
Agomelatine and Mood Disorders
Mood disorders are a group of mental or psychiatric disorders, such as depression, anxiety and apathy, and are characterized by abnormalities of emotional state (Marshall, 2020). These emotional disorders have a negative impact on the physical and mental health of patients, and also impose a heavy burden on their families and society. According to Chisholm et al. (2016), depression and anxiety disorders cost the global economy USD1.15 trillion each year. AGO can improve depression, anxiety, apathy, and other mood disorders, especially major depressive disorder (MDD).
Depression
Depression (also known as depressive disorder) is one of the most common mood disorders, and is characterized by a persistent low mood state. The accompanying symptoms of depression include inactivity, loss of concentration, social withdrawal, sleep disturbances, and cognitive impairments, such as memory deficit (LeMoult and Gotlib, 2019; Price and Duman, 2020).
AGO is used mainly for the treatment of depression, especially MDD. Heun et al. (2013) undertook a study on 222 older patients with MDD. They found that AGO treatment for 8 weeks relieved depressive symptoms efficiently and was well tolerated in older patients suffering from depression (Heun et al., 2013). Similarly, Robillard et al. (2018) reported that AGO reduced depressive symptoms significantly in 24 young adults with depression. Moreover, they found that the timing of dim light melatonin onset (DLMO) was shifted 3.6-hour earlier after treatment with AGO, which indicated a strong correlation between the improvement of depression and the phase shift of DLMO (Robillard et al., 2018). In a network meta-analysis, Cipriani et al. (2018) reported that AGO was more efficacious and acceptable in adult patients with major depression than other antidepressants. Recently, a 1-year multicenter observational study in France showed that AGO improved the quality of life and daily functioning of MDD patients and alleviated depression-related functional disability, with good efficacy and tolerability noted during the treatment period (Gorwood et al., 2020). In an open evaluation, AGO treatment for over 14 weeks in 37 patients with acute depression and seasonal affective disorder led to 76% having a response and 70% achieving remission (Pjrek et al., 2007).
Bipolar disorder is a chronic episodic mental disorder characterized by intermittent episodes of depressive and manic symptoms (Post, 2005; Tondo et al., 2017). Long-term use of antidepressants can increase the risk of patients with depression suffering mania or hypomania. Hence, a combination of antidepressants and mood stabilizers (e.g., lithium or sodium valproate) is, in general, the main approach for treating bipolar disorder (Grunze et al., 2018). AGO, as an adjunctive treatment with lithium or sodium valproate, was evaluated in bipolar disorder and was found to be efficacious in alleviating the symptoms of the patients after 6-week treatment (Calabrese et al., 2007; Fornaro et al., 2013).
The mechanism of action of AGO in the treatment of depression is focused on the synergistic effects of AGO on activation of MT1/MT2 and antagonism of 5-HT2C receptors. It has been reported that 90% of patients suffering from depression showed different degrees of sleep disorders (Tsuno et al., 2005). The quality of sleep and the outcome of depression had a mutually causal relationship. Thus, improvement in sleep quality can directly alleviate the emotional status of patients suffering from depression. Activation of MT1/MT2 by AGO can improve the sleep quality and daytime wakefulness of patients which, in turn, attenuates depressive symptoms.
5-HT2C receptors are the only known G protein-coupled receptors subjected to “RNA editing”, a mechanism which generates multiple variants of a particular gene product, thereby producing isoforms of 5-HT2C receptors with various properties (i.e., affinity, coupling, and constitutive activity) (Werry et al., 2008; Schmauss et al., 2010). Weissmann et al. (2016) documented a marked increase in RNA editing on 5-HT2C receptors in specific brain regions of patients who felt suicidal and depressed. Hence, region-specific changes in editing of the messenger-RNA of 5-HT2C receptors and deficient receptor function likely contribute to the etiology of depressive disorder or suicidal ideation (Weissmann et al., 2016). Usually, 5-HT2C receptors inhibit downstream release of dopamine and norepinephrine from some brain regions (e.g., prefrontal cortex) involved in regulation of emotion (Alex and Pehek, 2007). Compared with SSRI antidepressants, the antidepressant and anti-anxiety effects of antidepressants are stronger if 5-HT2C receptors are antagonized (Demireva et al., 2020). AGO can antagonize 5-HT2C receptors effectively without influencing the activity of G protein-coupled receptors, and it can normalize signaling at 5-HT2C sites by blocking the actions of agonists and inverse agonists, thereby returning activity to a baseline value (Werry et al., 2008). By antagonizing 5-HT2C receptors, AGO can also disinhibit release of dopamine and norepinephrine in the prefrontal cortex, thereby contributing to the effects of antidepressants (Millan et al., 2003; Stahl, 2014). In addition, AGO has neuroprotective effects because it has been shown to improve neuronal plasticity in the rat brain (Calabrese et al., 2011; Dagyte et al., 2011), upregulate brain-derived neurotrophic factor (BDNF) expression in the prefrontal cortex and hippocampus (Yucel et al., 2016; Lu et al., 2018), and activate the extracellular signal-regulated kinase-protein kinase B-glycogen synthase kinase 3β (ERK-AKT-GSK3β) signaling pathway (Duda et al., 2020), which can help to ameliorate the depressive disorder.
The development of antidepressants is slow due to the complexity of the etiology and unclear pathogenesis of depression. However, converging evidence from clinical trials and animal experiments has shown that the unique antidepressant effects of AGO breakthrough the conventional-treatment concept of SSRI and SSNRI antidepressants to provide a new strategy for the treatment of depression. Nonetheless, the antidepressant pharmacological mechanism of AGO has not been elucidated.
Anxiety disorder
Anxiety disorder is a common clinical neurosis characterized by excessive and persistent worry, with vegetative symptoms such as headaches and gastrointestinal complaints. Studies have demonstrated that AGO exhibits a good anxiolytic effect and tolerability (Stein et al., 2017; Slee et al., 2019), with a stronger clinical response and earlier improvement of symptoms than those elicited by SSRIs or SSNRIs (Buoli et al., 2017). AGO alleviated social isolation-induced anxiety in rats effectively and reversed increased plasma levels of vasopressin (Harvey et al., 2019). Similar to depression, the pathogenesis of anxiety disorder is complex, and the anxiolytic mechanism of AGO is incompletely understood. Several studies have suggested that the anxiolytic effects of AGO are associated mainly with its antagonism of 5-HT2C receptors (Sant’Ana et al., 2019; Demireva et al., 2020). The modulation of glutamate neurotransmission, anti-inflammatory action, antioxidant action, and correction of melatonin rhythms are also involved in the anxiolytic-like effects of AGO (Tchekalarova et al., 2018; Santos et al., 2019). Moreover, anxiety disorder and depression often accompany each other, and a similar pathogenesis may exist between them. Therefore, AGO may relieve anxiety disorder by improving depression, and vice versa.
Other emotional disorders
Apart from its antidepressant and anxiolytic properties, it has been suggested that AGO may exhibit special curative effects on particular neuropsychiatric symptoms, such as apathy, anhedonia, and abulia. Apathy is one of the most prevalent behavioral and psychological symptoms of dementia. Apathy is characterized by an insidious decline in motivation and goal-directed actions, which leads to reduced interest in social, recreational, occupational, and creative pursuits. Callegari and colleagues showed that AGO (but not melatonin) improved apathy in patients with frontotemporal dementia and was well-tolerated (Callegari et al., 2016). Moreover, an analysis of the literature showed that AGO had an obviously positive effect on the treatment of apathy in people suffering from dementia (Harrison et al., 2016). De Berardis et al. (2013) showed that AGO reversed escitalopram-induced apathy markedly in a patient with MDD. Whether used alone or in combination with acetyl-L-carnitine, AGO can alleviate apathy in older patients with mild or moderate depression (Gavrilova et al., 2015). Clinical studies suggest that AGO has potential for the treatment of apathy, but how it improves apathy merits further exploration.
Another prominent symptom of many neuropsychiatric disorders is anhedonia (loss of interest and pleasurable feelings in response to previously rewarding stimuli) (Husain and Roiser, 2018). Anhedonia is most notable in MDD and schizophrenia. A pooled analysis of an open clinical trial showed that AGO could improve depression and anhedonia in a broad range of patients suffering from depression (di Giannantonio et al., 2019). AGO has been shown to reverse anhedonia-like deficits in rats exposed to chronic constant light (Tchekalarova et al., 2018). How AGO improves anhedonia is not well understood, but it has been reported that treatment with AGO or an antagonist of 5-HT2C receptors, SB242084, reversed the anhedonia-like state in mice with knockout of glutamate ionotropic receptor NMDA type subunit 2D (Yamamoto et al., 2017). Hence, antagonizing 5-HT2C receptors may be a strategy for treating anhedonia. Importantly, anhedonia has also been linked to dysfunctions in the dopamine system, which plays a part in reward prediction, motivational arousal, and responsiveness to conditioned incentive stimuli (Tye et al., 2013). Chenu and collaborators (Chenu et al., 2013) reported that chronic administration of AGO for 14 days increased the number of spontaneously active dopamine neurons, the “burst” activity of dopamine neurons, the firing rate of 5-HT neurons in the dorsal-raphe nucleus, and tonic activation of postsynaptic 5-HT1A receptors located in the hippocampus. Those findings suggest that AGO may be involved in modulation of dopamine release in anhedonia. Nevertheless, the therapeutic mechanism of action of AGO in apathy and anhedonia must be elucidated.
Agomelatine and Sleep Disorders
Sleep disorders are a group of conditions in which the normal sleep pattern or sleep behaviors are disturbed. Primary sleep disorders include insomnia, hypersomnia, early waking, circadian-rhythm disorders, parasomnias, sleep-related movement disorders, and sleep-related breathing disorders (Pavlova and Latreille, 2019). Sleep disorders bring heavy economic burdens to patients’ families and society. Annual insomnia-related expenses have been estimated to be USD150–175 billion worldwide in 2016 (Reynolds and Ebben, 2017). Patients with psychiatric disorders and dementia have more severe sleep disorders which, in turn, contribute to earlier onset and more rapid progression of neurodegenerative disorders (Benca et al., 1992; Shi et al., 2018). Froböse et al. (2012) showed that a young patient with fatal familial insomnia had improved sleep efficiency, enhanced slow-wave sleep, and fewer awakenings during sleep periods after AGO treatment. In another study, AGO treatment for 6 months decreased the Depression Scale score significantly, and improved limb movements, sleep, and awakening significantly in patients suffering from depression and Parkinson’s disease (Avila et al., 2015). Quera-Salva et al. (2010) demonstrated that AGO ameliorated all aspects of sleep-wake abnormalities in patients with depression (particularly falling sleep and the quality of sleep) with an improvement in daytime alertness. The effects of AGO on sleep architecture in MDD have been measured using polysomnography: significant improvements in sleep efficiency, slow-wave sleep, and the distribution of delta activity throughout the night have been documented, but with no change in the amount or latency of rapid eye movement (REM) sleep. Moreover, after AGO treatment, the depressive symptoms of patients suffering from depression were reduced significantly and, on average, the timing of DLMO shifted 3.6-hour earlier, sleep onset was phase-shifted 28-minute earlier, and total sleep time increased by 24 minutes (Robillard et al., 2018).
In adults with autism spectrum disorder with intellectual disability, AGO treatment alleviated circadian-rhythm sleep-wake abnormalities, corrected rhythm disorders of the M5 sleep phase, and had good tolerability (Ballester et al., 2019). O’Neill et al. (2014) revealed AGO administration to result in an immediate and sustained improvement in sleep and the indices of challenging behavior in a man with severe brain damage suffering from a non-24-hour sleep-wake disorder. Armstrong et al. (1993) demonstrated that AGO advanced sleep onset in rats with delayed sleep-phase syndrome, which lays the foundation for further studies of AGO on circadian-rhythm disorders and sleep disorders. Descamps and colleagues showed that AGO could attenuate impairments of sleep-wake architecture, reverse the beta-1 electroencephalogram power band, and improve stress-related rebound of REM sleep in older rats (Descamps et al., 2014). Furthermore, Tchekalarova et al. (2020) demonstrated that AGO could adjust circadian homeostasis of motor activity and the sleep-wake cycle in a rat model of chronic constant light, including enhancing the latency of RME sleep and non-REM sleep and upregulating expression of MT1 and BDNF protein. The researchers previously demonstrated AGO to be effective in restoring impaired circadian patterns and plasma melatonin levels as well as improving depression and anxiety-like behavior in rats exposed to chronic constant light (Tchekalarova et al., 2018).
In mammals, melatonin is a neuroendocrine hormone. It is synthesized and secreted principally by the pineal gland at night (Lerner et al., 1960). Its primary physiological function is to convey information concerning the daily cycle of light and darkness to body structures, to regulate circadian rhythms, and to synchronize rhythms. Usually, the rhythmic secretion of melatonin is driven by the circadian clock in the suprachiasmatic nucleus of the hypothalamus. However, light can suppress or synchronize melatonin production according to the light schedule, suggesting that melatonin secretion from the pineal gland is closely related to the duration of darkness (Kennaway, 2019). Melatonin acts via melatonin receptors distributed widely in various tissues to respond to periodic changes of light, such as the sleep-wake cycle. Exposure to light activates the suprachiasmatic nucleus and suppresses melatonin production, which then transmits the light information from the circadian clock and induces awakening in daytime. At night, the synthesis and secretion of melatonin remain high, which promotes sleep (Zisapel, 2018). Therefore, if the rhythmic secretion of melatonin is disrupted, then circadian rhythms are also disrupted (e.g., poor sleep quality and circadian-rhythm sleep-wake disorders). 5-HT and its receptors have been reported to be involved in sleep and wakefulness, as well as cognition and mood. Antagonism of 5-HT2A/5-HT2C receptors prolongs the duration of slow-wave sleep and enhances low-frequency (< 7 Hz) activity in the sleep electroencephalogram (Landolt and Wehrle, 2009). Moreover, antagonism of 5-HT2C receptors stimulates dopaminergic and adrenergic pathways and exerts antidepressant and anxiolytic actions in behavioral paradigms, which favors sleep (Millan, 2005). AGO activates MT1/MT2 and antagonizes 5-HT2C receptors synchronously, so it has a unique role in regulating the sleep-wake cycle and correcting sleep structure.
Agomelatine and Impairment of Cognition and Memory
A series of studies demonstrated that emotional disorders and sleep disorders can impact the function of memory and cognition negatively (McHutchison et al., 2020; Xu et al., 2020). Neurodegenerative diseases characterized by memory and cognitive impairments (e.g., Alzheimer’s disease and dementia) are characterized by abnormal emotional performance and sleep performance. About 50 million people worldwide have dementia (mainly Alzheimer’s disease), and the number is expected to increase to 152 million by 2050 (Alzheimer’s Disease International and Patterson, 2018). The estimated total global societal cost of dementia is $1.3 trillion, which is expected to surpass $2.8 trillion by 2030 as the number of people living with dementia and care costs increase (World Health Organization, 2021). However, efficacious disease-modifying therapeutics for dementia management are lacking. Interestingly, AGO has been reported to be efficacious in the treatment of emotional disorders, and also in improving cognitive deficits (Bogolepova et al., 2011; Altınyazar and Kiylioglu, 2016; Callegari et al., 2016).
Additional Table 1 summarizes the effects of AGO on memory impairment and cognitive impairment in clinical trials. Most AGO studies have focused on the treatment of patients with emotional disorders, but some studies have also shown that AGO improves cognitive function in patients with Alzheimer’s disease (Altınyazar and Kiylioglu, 2016), frontotemporal dementia (Callegari et al., 2016), stroke (Bogolepova et al., 2011; Antonen et al., 2015), schizophrenia, or severe depression. Through evaluation of cognitive ability, Englisch et al. (2018) and Bruno et al. (2014) found that AGO treatment could efficiently ameliorate the reasoning/problem-solving ability and correct “perseverative errors” in the Wisconsin Card Sorting Test in patients with schizophrenia (Additional Table 1). Moreover, studies have indicated that patients suffering from depression with cognitive impairments (especially those with major depression) had different degrees of improvements in cognitive ability after AGO treatment (Gavrilova et al., 2014; Gorwood et al., 2014, 2015; Kalyn et al., 2015; Cléry-Melin and Gorwood, 2017; Medvedev et al., 2018).
Additional Table 1.
Characteristics of AGO for the treatment of memory impairment and cognitive impairment in clinical studies
| Study design | Patients | Agent and dose (route) | Primary measure and results | Conclusions | Study |
|---|---|---|---|---|---|
| 1 month, case report | 1 AD patient | AGO (25 mg/d, p.o.) | MMSE; MMSE score: 19 (before AGO treatment) vs. 23 (after 1-month AGO treatment) |
Significant improvement in cognitive function | Altınyazar and Kiylioglu, 2016 |
| 10 wk, double-blind | 24 FTD patients melatonin (10 mg/d, p.o.) | AGO (50 mg/d, p.o.) or | AES-C; 50.6 ± 2.4 (AGO) vs. 42.7 ± 2.4 (melatonin), P = 0.006 | Significant reduction of apathy and improvement in cognitive function by AGO, but not melatonin | Callegari et al., 2016 |
| 6–8 wk, randomized | 2048 MDD patients | AGO (25 or 50 mg/d, p.o.) | TMT-A, TMT-B, d2 test; 67.68% of patients were responders and 43.31% were in remission |
Significant improvement in cognitive function | Gorwood et al., 2014 |
| 8 wk, single-blind | 42 patients with mild-to-moderate depression | Antidepressant monotherapy (fluvoxamine, venlafaxine or AGO, 25 mg/d) or use the same antidepressant in combination with actovegin (p.o.) | MMSE; 26.23 ± 2.32 (day 0) vs. 28.44 ± 1.49 (day56), P < 0.01 | Significant improvement in cognitive function, with faster improvement in cognitive function by the antidepressant in combination with actovegin | Safarova et al., 2018 |
| 10-word memory test; 6.42 ± 0.97 (day 0) vs. 7.00 ± 0.89 (day 56), P < 0.01 | |||||
| Drawing of clock; 8.04 ± 1.24 (day 0) vs. 8.81 ± 1.08 (day 56), P < 0.01 | |||||
| 7 d, double-blind | 48 healthy volunteers | AGO (25–50 mg/d, p.o.) | Facial expression recognition task; AGO (25 mg) vs. placebo (group ×emotion F(6, 174) = 2.6, P = 0.018), AGO (50 mg) vs. placebo (group ×emotion F(6, 180) = 0.8, P = 0.6) |
Significantly reduced subjective ratings of sadness, reduced recognition of sad facial expressions, improved positive affective memory and reduced emotion-potentiated startle response with 25 mg of AGO, but not 50 mg of AGO | Harmer et al., 2011 |
| 12 wk, proof-of-concept study | 27 patients with schizophrenia and comorbid depression (p.o.) | Antipsychotic drugs in combination with AGO | MCCB; composite score (baseline vs. after treatment, P = 0.01), reasoning/problem-solving subscore (baseline vs. after treatment, P = 0.001) | Significant improvement in the MCCB composite score and the reasoning/problem-solving subscore | Englisch et al., 2018 |
| 2–8 wk, randomized study | 33 patients with CBI and comorbid mild depression | AGO (25 mg/d, p.o.) | MMSE, MoCA; baseline vs. after treatment, P < 0.001 | Significant improvement in cognitive function | Antonen et al., 2015 |
| 6 wk, randomized study | 35 patients with depression and marked cognitive impairment | AGO (25 mg/d, p.o.) | MMSE, Rey test for auditory-speech learning, RAVLT; 74.3% of patients were responders and 51.4% were in remission | Significant improvement in cognitive function | Medvedev et al., 2018 |
| 16 wk, open-label uncontrolled trial | 20 patients with schizophrenia | AGO (50 mg/d, p.o.) | Wisconsin Card Sorting Test; perseverative errors: 14.75 ± 10.05 (baseline) vs. 11.25 ± 10.46 (week 16), P= 0.033, Cohen’s d = 0.3 | Significant improvement in performances of the Stroop task and “perseverative errors” in the Wisconsin Card Sorting Test | Bruno et al.,2014 |
| 12 wk, open-label,preliminary study | 15 FMS patients | AGO (25 mg/d, p.o.) | Wisconsin Card Sorting Test; perseverative errors: 26.87 ± 30.963 (baseline) vs. 25.13 ± 30.223 (week 16), P = 0.301, Cohen’s d = 0.1 | Improvement in cognitive function | Bruno et al., 2013 |
| 6–8 wk, multicenter study | 2048 outpatients with depression | AGO (25 mg/d, p.o.) | TMT-A, TMT-B, d2 test; TMT-A (time) and TMT-B (time): P < 0.0001, TMT-A (mistakes): P = 0.0004, TMT-B tests TMT-B (mistakes): P < 0.0001, F1 (number of omission mistakes):P < 0.05 |
Significant improvement in d2, TMT-A, and | Cléry-Melin and Gorwood, 2017 |
| 42 d, randomized study | 18 patients with depression | AGO (25–50 mg/d, p.o.) | MMSE; 26.76 ± 3.30 (day 0) vs. 28.17 ± 2.0 (day 42) | Improvement in cognitive function | Kalyn et al., 2015 |
| 6 wk, multicenter, observational, phase-IV study |
1565 patients with depression | AGO (25–50 mg/d, p.o.) | MAThyS; 4.05 ± 1.11 (baseline) vs. 4.87 ± 1.16 (week 6), P < 0.001 | Significant improvement in cognitive function | Gorwood et al., 2015 |
| 8 wk, randomized study | 40 patients with mild-to-moderate depression | Antidepressant monotherapy (fluvoxamine, venlafaxine or AGO, 25–50 mg/d) or use the same antidepressant in combination with carnitine (p.o.) | MMSE; 26.90 ± 2.10 (day 0) vs. 28.15 ± 1.53 (day 56), P < 0.05; 10-word memory test; 6.68 ± 0.86 (day 0) vs. 6.92 ± 0.96 (day 56) | Significant improvement in cognitive function, but faster improvement using the antidepressant in combination with carnitine | Gavrilova et al., 2015 |
| Drawing of clock; 7.92 ± 1.18 (day 0) vs. 8.33 ± 1.23 (day 56) | |||||
| 3 months, randomized study | 40 patients with post-stroke depression | AGO (25 mg/d, p.o.) | MMSE; 25.2 ± 1.1 (0 day) vs. 27.2 ± 0.6 (month 3) | Improvement in cognitive function | Bogolepova et al., 2011 |
| 6 wk, randomized study | 20 older patients with mild-to-moderate depression | AGO (25–50 mg/d, p.o.) | MMSE; median MMSE score before treatment = 28.5 and at study termination = 29.5 (P > 0.05) | AGO had no effect on cognitive function and did not elicit pronounced or serious adverse events, but the cognition of patients showed an increase | Gavrilova et al., 2014 |
AD: Alzheimer’s disease; AES-C: apathy evaluation scale-clinician version; AGO: agomelatine; CBI: chronic brain ischemia; FMS: fibromyalgia syndrome; FTD: frontotemporal dementia; MAThyS: multidimensional assessment of thymic states; MCCB: measurement and treatment research to improve cognition in schizophrenia consensus cognitive battery; MDD: major depressive disorder; MMSE: mini-mental state examination; MoCA: Montreal cognitive assessment; p.o.: per os; RAVLT: Rey auditory verbal learning test; TMT-A: trail-making test-A; TMT-B: trail-making test-B.
Safarova et al. (2018) and Gavrilova et al. (2015) showed that a combination of AGO and carnicetine or AGO and actovegin allowed for a more rapid and pronounced therapeutic effect for cognitive dysfunction in older patients with mild or moderate depression. In a trial involving 20 older patients with mild or moderate depression, AGO reduced anxiety disorder and depressive symptoms effectively as well as improving the health-related quality of life of patients significantly, with a slight increase in the Mini-Mental State Examination score and without pronounced or serious adverse events (Gavrilova et al., 2014). In another trial of 15 patients with primary fibromyalgia, treatment with AGO improved depression, anxiety disorder, and pain significantly in patients; the authors reported a trend towards the improvement of performances in executive/cognitive symptoms (Bruno et al., 2013). Through a double-blind parallel-group design in healthy volunteers, Harmer et al. (2011) observed that AGO (25 mg) decreased the subjective rating of sadness, reduced recognition of sad facial expressions, improved affective memory, and reduced the emotion-potentiated startle response, whereas AGO (50 mg) reduced only the emotion-potentiated startle response without affecting other types of emotional processing. The researchers indicated that AGO is also beneficial for memory improvement in healthy cohorts. Nevertheless, how AGO improves the cognitive dysfunction accompanied by these diseases is not known.
Additional Table 2 summarizes the characteristics of AGO on memory and cognition in healthy animals and pathological animal models. Chronic AGO treatment reduced the error percentage of streptozotocin-treated rats in the eight-arm radial arm maze test, thereby suggesting that AGO could improve the spatial working memory of rats with Alzheimer’s disease (Bergamini et al., 2016; Ilieva et al., 2019). Furthermore, Gupta et al. (2015) demonstrated that AGO treatment reduced the escape latency and residence time in the target quadrant in the Morris water maze test of mice with chronic cerebral hypoperfusion, thereby implying that the long-term learning and memory of mice were improved. Moreover, the cognitive function of rats with renovascular hypertension-induced vascular dementia was ameliorated obviously by AGO treatment (Singh et al., 2015). It has also been demonstrated that AGO significantly attenuates 3-nitropropionic acid-induced learning-memory deficits in rats with Huntington’s disease (Gupta and Sharma, 2014), and improves the restraint stress-induced impairments of short-term recognition memory and long-term spatial learning and memory in mice and rats (Conboy et al., 2009; Gumuslu et al., 2014; Lapmanee et al., 2017).
Additional Table 2.
Characteristics of AGO for the treatment of memory impairment and cognitive impairment in animal studies
| Study model | Animal | Agent and dose (route) | Main results and conclusion | Study |
|---|---|---|---|---|
| CCH | Mouse | AGO (2–4 mg/kg per day, i.p.),24 d | In the Morris water maze test, AGO treatment decreased escape latency time in CCH-induced mice (P < 0.05). AGO improved long-term memory. |
Gupta et al., 2015 |
| Renovascular hypertension induced-vascular dementia | Rat | AGO (2–4 mg/kg per day, i.p.), 24 d | In the Morris water maze test, the escape latency time was reduced significantly on day 4 (40.1 ± 2.6 (2-mg AGO) and 39.8 ± 2.59 (4-mg AGO), P < 0.001). AGO improved long-term memory. |
Singh et al., 2015 |
| 3-Nitropropionic acid-induced Huntington’s disease | Rat | AGO (2–4 mg/kg per day, p.o.), 28 d | In the Morris water maze test, AGO treatment reduced the escape latency time significantly on day 4 (P < 0.001) as compared with that in the 3-3-nitropropionic acid-treated group. AGO improved long-term memory. |
Gupta and Sharma, 2014 |
| Streptozotocin-induced model of AD |
Rat | AGO (40 mg/kg per day, i.p.), 30 d | In the radial arm maze test, AGO treatment decreased working memory error (P < 0.01). AGO improved working memory. |
Ilieva et al., 2019 |
| PRS | Rat | AGO (40 mg/kg per day, i.p.), 21 d | In the social memory test, PRS rats treated with AGO displayed a reduction in sniffing behavior compared with vehicle-treated PRS rats (P < 0.05). AGO improved social memory performance. |
Marrocco et al., 2014 |
| Restraint-induced stress | Rat | AGO (10 mg/kg per day, i.p.), 4 wk | In the Morris water maze test, AGO-treated stressed rats exhibited a reduction in escape latency on days 1 (P < 0.01), 2 (P < 0.001), and 3 (P < 0.001) and correct quadrant time (P < 0.001) compared with vehicle-treated stressed rats. AGO improved long-term memory. In the novel object recognition test, a higher discrimination ratio was evidenced in AGO-treated rats than in vehicle-treated stressed rats (P < 0.05). AGO improved recognition memory. |
Lapmanee et al., 2017 |
| Chronic social defeat stress | Mouse | AGO (10 mg/kg per day, i.p.), 3 wk | In the novel object recognition test, stressed-AGO mice displayed a significantly increase in the recognition index compared with stressed (hydroxyethylcellulose-treated) mice (P < 0.05). AGO improved recognition memory. |
Martin et al., 2017 |
| Memory deficit induced by scopolamine | Mouse | AGO (1, 10, or 30 mg/kg per day, i.p.), 30 min before testing | In the passive avoidance task, AGO (30 mg/kg) significantly increased the retention time as compared with scopolamine (P < 0.05). AGO alleviated episodic memory deficit. |
İlkaya et al., 2015 |
| Pentylenetetrazol-induced kindling | Mouse | AGO (10 mg/kg per day, p.o.), 3 wk | In the Y maze test, AGO treatment significantly enhanced percentage alternation and the number of arm entries (P < 0.001) following PTZ kindling-induced seizures. AGO improved working memory. |
Azim et al., 2017 |
| Chronic mild stress-induced cognitive deterioration | Mouse | AGO (10 mg/kg per day, i.p.), 5 wk | In the Morris water maze test, AGO shortened the escape latency significantly in the familiarization session (P < 0.001) and increased the time spent in the escape platform quadrant (P < 0.01). AGO improved long-term memory. In the novel object recognition test, AGO increased the ratio index significantly compared with stress-exposed control mice (P < 0.001). AGO improved recognition memory |
Gumuslu et al., 2014 |
| Normal | Rat | Single intraperitoneal administration of AGO (10–40 mg/kg) in the evening or AGO (2.5, 10, or 40 mg/kg) in the morning | In the novel object recognition test, AGO (10 and 40 mg/kg) increased the recognition index significantly (P < 0.01). AGO improved recognition memory in the morning or evening. |
Bertaina-Anglade et al., 2011 |
| Stress | Rat | AGO (10 mg/kg per day, i.p.), 22 d | In the radial-arm water maze test, AGO treatment reduced arm-entry errors significantly in non-stressed and stressed rats (P < 0.001) AGO blocked the predator stress-induced impairment of spatial memory and improved spatial memory. |
Conboy et al., 2009 |
| Normal | Rat | AGO (40 mg/kg per day, i.p.), 20 wk | In the Morris water maze test, AGO-treated rats located the hidden platform significantly faster than did control rats on days 2 (P < 0.01), 3 (P < 0.001), and 4 (P < 0.01). During the probe trial, AGO-treated rats spent significantly more time (P < 0.001) in the target quadrant compared with that of control rats. AGO enhanced spatial memory. |
Demir Özkay et al., 2015 |
| Normal | Rat | AGO (40 mg/kg per day, i.p.), 22 d | In the novel object recognition test, AGO increased the D2 index (ability to discriminate between a familiar object and novel object) (P < 0.05). AGO improved recognition memory. |
Ladurelle et al., 2012 |
| KA status epilepticus | Rat | AGO (40 mg/kg per day, i.p.), 10 wk | In the radial arm maze test, KA-vehicle and KA-AGO groups exhibited more working memory errors compared with naive rats treated with vehicle after the first session (P < 0.05). AGO had no effect on kainate acid-induced memory impairment. |
Tchekalarova et al., 2017 |
| Streptozotocin-induced model of type-2 diabetes mellitus | Rat | AGO (40–80 mg/kg per day, i.p.), 2 wk | In the Morris water maze test, diabetic rats treated with AGO (40 mg/kg) exhibited a significant reduction of escape latency on day 4 (P < 0.001). Administration of AGO at 40 (P < 0.05) or 80 mg/kg (P < 0.01) induced a significant increase in the target quadrant time of diabetic rats. AGO treatment reversed the impaired long-term learning and memory performance of diabetic rats effectively. In the passive avoidance task, treating diabetic rats with 40 (P < 0.05) or 80 mg/kg (P < 0.001) of AGO prolonged the entrance latency to the dark compartment. AGO improved episodic memory deficit. |
Can Ö et al., 2018 |
| Chronic psychosocial stress | Mouse | AGO (10–25 mg/kg per day, p.o.), acute treatment or 12 d | Although there was no stress × drug interaction or main effect of drug in the simple reversal learning test, mice completed more reversals after AGO (25 mg/kg) (P < 0.05) treatment relative to vehicle-treated naive mice in the complex reversal learning test. AGO reversed cognitive deficits partially. |
Bergamini et al., 2016 |
| “Pessimistic” and “optimistic” traits | Rat | AGO (5, 10, or 40 mg/kg per day, p.o.), acute treatment | Following acute treatment with AGO, the proportion of lose-shift behavior in the probabilistic reversal-learning test was reduced significantly in pessimistic rats compared with optimistic rats (P < 0.05). | Drozd et al., 2019 |
| Intracerebroventricular Aβ1-42 model of AD | Rat | AGO (40 mg/kg per day, i.p.), 30 d | AGO treatment did not improve spatial memory of the Aβ group in the radial arm maze test, but AGO enhanced the α-secretase concentration (P < 0.05) and decreased the γ-secretase concentration (P < 0.05 or P < 0.01) in the hippocampus, with decreased Aβ1–42 levels in the frontal cortex and hippocampus (P < 0.01 or P < 0.001). | Ilieva et al., 2021 |
| Cisplatin-induced model | Rat | AGO (20–40 mg/kg, p.o.), 7 d | In the passive avoidance test, AGO (20 and 40 mg/kg) improved the cisplatin-induced decrease in the step-through latency significantly (P <0.001). In the novel object recognition test, AGO (20 and 40 mg/kg) attenuated the cisplatin-induced decrease in the discrimination index significantly (P < 0.05). AGO improved memory retention and recognition. |
Cankara et al., 2021 |
AD: Alzheimer’s disease; AGO: agomelatine; Aβ: amyloid β; CCH: chronic cerebral hypoperfusion; i.p.: intraperitoneal; KA: kainate acid; p.o.: per os; PRS: prenatal restraint stress.
Marrocco et al. (2014) found that AGO treatment markedly corrected abnormalities in social-memory performance in adult rats with unstressed or prenatal restraint stress, and Martin et al. (2017) simultaneously observed that effectively alleviated episodic memory deficits of mice with chronic social defeat stress in the novel object recognition test (NORT). Hence, AGO might improve the interactive behaviors of animals by enhancing their recognition memory. In addition, it has been demonstrated that AGO improves the memory deficit induced by scopolamine in mice (İlkaya et al., 2015), and alleviates the short-term memory despair of mice injected with pentylenetetrazol to induce kindling (Azim et al., 2017), but did not correct spatial-memory impairment in rats suffering from epilepsy induced by kainate acid (Tchekalarova et al., 2017). Those findings suggest that the effects of AGO treatment in various animal models differ because of different epilepsy-triggering treatments, which provides a direction for study on the pharmacological mechanism of AGO. In the probabilistic reversal-learning test, Drozd et al. (2019) found that AGO treatment improved the ability of cognitive judgment in pessimistic rats. Notably, single administration of AGO (10 and 40 mg/kg) in the evening or morning improved the recognition memory of normal rats significantly in the NORT (Bertaina-Anglade et al., 2011). Moreover, studies in normal rats revealed that long-term AGO treatment enhanced spatial memory obviously in the Morris water maze test (Demir Özkay et al., 2015), and improved recognition memory in the NORT (Ladurelle et al., 2012). How AGO improves memory and cognitive function is not known, probably owing to the perplexing causes of memory impairment and cognitive impairment in particular diseases.
It has been demonstrated that AGO reduced amyloid β-42 (Aβ42) accumulation in the frontal cortex and hippocampus of male rats with streptozotocin-induced Alzheimer’s disease (Ilieva et al., 2019), activated melatonin receptors, and prevented Aβ-induced tau phosphorylation by activation of GSK3β and oxidative damage in PC12 cells (Yao et al., 2019). Furthermore, chronic administration of AGO completely prevented the stress-induced increase in glutamate release in the prefrontal/frontal cortex of rats by the synergistic effect of melatonergic and 5-HT2C receptors in one study (Tardito et al., 2010). Recently, Ilieva et al. (2021) suggested that chronic treatment with AGO alleviated anxiety disorder and depressive-like behavior and decreased the Aβ level in the hippocampus by enhancing α-secretase and decreasing γ-secretase in a rat model of Alzheimer’s disease. Cankara et al. (2021) showed that AGO attenuated cisplatin-induced neurotoxicity in a mouse hippocampal neuronal cell line (HT22), and improved cisplatin-induced deficits in memory and recognition. In addition, in rat models with cognitive impairments, AGO also reversed neuronal loss in the hippocampus (Can Ö et al., 2018; Ilieva et al., 2021) and caused significant enhancement in the volume of hippocampal CA1–3 subfields and the total number of pyramidal neurons in this region (Demir Özkay et al., 2015), thereby implying that AGO had neuroprotective effects.
BDNF is a neurotrophin distributed widely in the mammalian brain. BDNF has prominent functions in plastic regulation, including control of neuronal and glial development, neuroprotection, and modulation of short- and long-lasting synaptic interactions, which are critical for cognition and memory (von Bohlen Und Halbach and von Bohlen Und Halbach, 2018). It has been reported that AGO alleviates depressive symptoms effectively in patients suffering from depression, with restoration of the plasma level of BDNF (Martinotti et al., 2016). AGO also increases the hippocampal BDNF level and the number of BDNF-positive neurons in rats with chronic unpredictable mild stress (Lu et al., 2018). Notably, Ladurelle et al. (2012) demonstrated that chronic administration of AGO increased the level of mature BDNF in the hippocampus significantly, and promoted rapid, sustained, enhanced cognitive activity in normal rats.
Cyclic adenosine monophosphate-responsive element binding protein (CREB) is a transcription factor. CREB regulates expression of several genes involved in the control of neuroplasticity, circadian rhythms, cell survival, and cognition (Carlezon et al., 2005). Moreover, CREB is a pivotal component of the “molecular switch” that converts short-term memory to long-term memory (Lisman et al., 2018). Furthermore, the CREB family is a major regulator of BDNF expression after tropomyosin receptor kinase B (TrkB; a BDNF receptor) signaling (Esvald et al., 2020). Recent evidence suggests that chronic administration of AGO leads to improvements in memory deterioration and upregulation of hippocampal expression of CREB and BDNF in mice exposed to unpredictable, chronic, mild stress (Gumuslu et al., 2014). It has been revealed that activation of MT1 and MT2 activates ERK-90 kDa ribosomal S6 kinase-CREB-BDNF signaling (Sung et al., 2018) and BDNF-TrkB signaling in hippocampal neurons (Li et al., 2018). Moreover, mice with deletion of the 5-HT2C receptor show increased levels of the mature form of BDNF in the hippocampus (Hill et al., 2011). In naïve rats, Musazzi et al. (2014) found that chronic administration of AGO—contrary to traditional antidepressants—did not increase CREB phosphorylation, but instead modulated the mitogen-activated protein kinase (MAPK)-ERK1/2 and AKT-GSK3β pathways. ERK1/2 is one of the best-characterized members of the MAPK family. ERK1/2 mediates the proliferation, differentiation, apoptosis, inflammation, and synaptogenesis of cells (Albert-Gascó et al., 2020).
In neurons, an important function of ERK-MAPK signaling is regulation of synaptic plasticity, which relates to learning and memory processes (Alkadhi and Dao, 2019). Several studies have indicated that phosphatidylinositol 3-kinase can activate AKT in brain cells and, by activation of this protein, GSK3β, which inhibits tau hyperphosphorylation (Chen et al., 2004; Endo et al., 2006). AKT shows high expression in some brain areas that are known to regulate cognition and neuroprotection, thereby having a key role in synaptic and neural survival, neuroprotection, and neural plasticity, as well as supporting the growth and survival of neurons (Beaulieu et al., 2009; Kitagishi et al., 2012). Studies have reported that activation of melatonin receptors (MT1 and MT2) can activate MAPK-ERK1/2 and AKT-GSK3 signaling pathways (Werry et al., 2005; Hadj Ayed Tka et al., 2015; Chagraoui et al., 2016; Li et al., 2020). Therefore, AGO may protect hippocampal neurons and improve memory and cognition by regulation of MAPK-ERK and AKT-GSK3β signaling pathways, but this concept has not been demonstrated definitively.
It has been shown that 5-HT2C receptors mediate Ca2+ release from endoplasmic reticula via the phospholipase C-inositol 1,4,5-trisphosphate-Ca2+ pathway (Watson et al., 1995; Wada et al., 2006). 5-HT2C receptors show high expression and are upregulated in the brains of mice with acute infusion of Aβ (Bonn et al., 2013) and in rats with pilocarpine-induced epilepsy with memory impairment (Krishnakumar et al., 2009). Those studies indicate that intracellular Ca2+ dysregulation might (at least in part) result from 5-HT2C overexpression. Ca2+ dysregulation leads to neuronal apoptosis and cell death, which result in memory impairment and cognitive impairment (Kumar, 2020). Scholars have applied various sequencing methods and databases and analyzed “omics” data to discover new uses for existing drugs. Such studies have demonstrated AGO to also be involved in regulation of axon development, glutamatergic activity, netrin signaling, synaptic long-term potentiation, and Rho-GTPases-related pathways (an important regulator of morphological neuroplasticity) in hippocampal neurons (Patrício et al., 2015), as well as a neurotrophin signaling pathway and insulin signaling pathway in patients suffering from depression (Dmitrzak-Weglarz et al., 2021), which are closely related to memory and cognitive function.
Although the findings mentioned above are not entirely consistent (possibly caused by different animal models and interventions), they show that AGO improves memory and cognitive function by activating multiple signal-transduction pathways (Figure 1). Clinical and basic-science studies investigating the effect of AGO on memory and cognition suggest that AGO might be a novel strategy for the treatment of memory impairment and cognitive impairment. With the increase in age of populations worldwide, the dementia observed in degenerative diseases has become a growing public-health problem. However, prevention of the memory impairment and cognitive impairment of patients with dementia is not possible, and few drugs can be used for treatment. With its unique pharmacological effects, AGO provides a new idea for the treatment of dementia.
Figure 1.

Pathways involving the neuroprotective effects of AGO (schematic).
Through MT1/MT2, AGO can activate PI3K-AKT-GSK3β and MAPK-ERK1/2-RSK90-CREB cascades, which control gene transcription or protein modification. Activation of GSK3β inhibits tau phosphorylation. CREB can control BDNF transcription. By inhibiting the 5-HT2C receptor (5-HT2CR), AGO can also inhibit IP3 from releasing Ca2+ from the endoplasmic reticulum, and promote expression of BDNF which combined with TrkB and activate the MAPK-ERK1/2-RSK90-CREB signaling pathway. 5-HT2C: 5-Hydroxytryptamine 2C; AGO: agomelatine; AKT: protein kinase B; BDNF: brain-derived neurotrophic factor; CREB: cyclic adenosine monophosphate-response element binding protein; ERK: extracellular signal-related kinase; GSK3β: glycogen synthase kinase 3β; IP3: inositol 1,4,5-trisphosphate; MAPK: mitogen-activated protein kinases; MT1: melatonin receptor 1A; MT2: melatonin receptor 1B; PI3K: phosphatidylinositol-3-kinase; RSK90: 90 kDa ribosomal s6 kinase; TrkB: tropomyosin-related kinase B.
Limitations of Agomelatine
Although AGO possesses some distinct advantages, it also has some disadvantages. The commonly reported adverse events in clinical treatment using AGO are headache, nausea and fatigue, which are of mild-to-moderate severity (Stein et al., 2018). Moreover, due to its propensity to increase the level of liver enzymes, AGO is contraindicated in patients with impaired liver function (Štuhec, 2013; Friedrich et al., 2016). Hence, monitoring of liver function is recommended before AGO initiation and periodically during treatment. Nevertheless, developing AGO analogs with low toxicity and few side effects for the treatment of chronic neurodegenerative diseases is important.
Conclusions
The most remarkable feature of AGO is a synergistic action between its agonism at MT1/MT2 and antagonism at 5-HT2C receptors. Given its innovative mechanism of action and favorable safety profile, AGO is beneficial for the treatment of mood disorders and sleep disorders, and most reviews have focused on these aspects. Unlike those reviews, in light of the neuroprotective effects of AGO, we have summarized research progress regarding the improvement of memory and cognitive function using AGO. However, the few studies undertaken so far on the clinical treatment of cognition and dementia by AGO (particularly on the specific treatment of neurodegenerative diseases) need to be bolstered with additional studies. Nevertheless, clinical studies and animal experiments have strongly suggested that AGO could be a promising treatment option for improving memory, sleep, and mental activity simultaneously.
Additional files:
Additional Table 1: Characteristics of AGO for the treatment of memory and cognitive impairments in clinical studies.
Additional Table 2: Characteristics of AGO for the treatment of memory and cognitive impairments in animal studies.
Footnotes
Funding: The work was supported by Shanxi “1331 Project” Key Subjects Construction, No. 1331KSC (to JSQ); Science Research Start-up Fund for Doctors of Shanxi Province, No. SD2011 (to TL); and Science Research Start-Up Fund for Doctors of Shanxi Medical University, No. XD2017 (to TL).
Conflicts of interest: The authors declare no conflicts of interest.
Availability of data and materials: All data generated or analyzed during this study are included in this published article and its supplementary information files.
Open peer reviewers: Melinda Barkhuizen, Philip Morris International, Netherlands; Nemil N. Bhatt, The University of Texas, USA.
P-Reviewers: Barkhuizen M, Bhatt NN; C-Editor: Zhao M; S-Editors: Yu J, Li CH; L-Editors: Yu J, Song LP; T-Editor: Jia Y
References
- 1.Albert-Gascó H, Ros-Bernal F, Castillo-Gómez E, Olucha-Bordonau FE. MAP/ERK signaling in developing cognitive and emotional function and its effect on pathological and neurodegenerative processes. Int J Mol Sci. 2020;21:4471. doi: 10.3390/ijms21124471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Alex KD, Pehek EA. Pharmacologic mechanisms of serotonergic regulation of dopamine neurotransmission. Pharmacol Ther. 2007;113:296–320. doi: 10.1016/j.pharmthera.2006.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Alkadhi KA, Dao AT. Effect of exercise and Aβprotein infusion on long-term memory-related signaling molecules in hippocampal areas. Mol Neurobiol. 2019;56:4980–4987. doi: 10.1007/s12035-018-1425-x. [DOI] [PubMed] [Google Scholar]
- 4.Altınyazar V, Kiylioglu N. Insomnia and dementia:is agomelatine treatment helpful?Case report and review of the literature. Ther Adv Psychopharmacol. 2016;6:263–268. doi: 10.1177/2045125316646064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Patterson C Alzheimer's Disease International. World Alzheimer Report 2018:The state of the art of dementia research:New frontiers. London: Alzheimer's Disease International (ADI); 2018. [Google Scholar]
- 6.Antonen EG, Nikitina MV, Kruchek MM. Clinical experience of the use of agomelatine in the treatment of patients with depression and chronic brain ischemia. Zh Nevrol Psikhiatr Im S S Korsakova. 2015;115:79–85. doi: 10.17116/jnevro201511511279-85. [DOI] [PubMed] [Google Scholar]
- 7.Armstrong SM, McNulty OM, Guardiola-Lemaitre B, Redman JR. Successful use of S20098 and melatonin in an animal model of delayed sleep-phase syndrome (DSPS) Pharmacol Biochem Behav. 1993;46:45–49. doi: 10.1016/0091-3057(93)90315-k. [DOI] [PubMed] [Google Scholar]
- 8.Avila A, Cardona X, Martin-Baranera M, Leon L, Caballol N, Millet P, Bello J. Agomelatine for depression in parkinson disease:additional effect on sleep and motor dysfunction. J Clin Psychopharmacol. 2015;35:719–723. doi: 10.1097/JCP.0000000000000404. [DOI] [PubMed] [Google Scholar]
- 9.Azim MS, Agarwal NB, Vohora D. Effects of agomelatine on pentylenetetrazole-induced kindling kindling-associated oxidative stress and behavioral despair in mice and modulation of its actions by luzindole and 1-(m-chlorophenyl) piperazine. Epilepsy Behav. 2017;72:140–144. doi: 10.1016/j.yebeh.2017.03.019. [DOI] [PubMed] [Google Scholar]
- 10.Ballester P, Martínez MJ, Inda MD, Javaloyes A, Richdale AL, Muriel J, Belda C, Toral N, Morales D, Fernández E, Peiró AM. Evaluation of agomelatine for the treatment of sleep problems in adults with autism spectrum disorder and co-morbid intellectual disability. J Psychopharmacol. 2019;33:1395–1406. doi: 10.1177/0269881119864968. [DOI] [PubMed] [Google Scholar]
- 11.Beaulieu JM, Gainetdinov RR, Caron MG. Akt/GSK3 signaling in the action of psychotropic drugs. Annu Rev Pharmacol Toxicol. 2009;49:327–347. doi: 10.1146/annurev.pharmtox.011008.145634. [DOI] [PubMed] [Google Scholar]
- 12.Benca RM, Obermeyer WH, Thisted RA, Gillin JC. Sleep and psychiatric disorders. A meta-analysis. Arch Gen Psychiatry. 1992;49:651–670. doi: 10.1001/archpsyc.1992.01820080059010. [DOI] [PubMed] [Google Scholar]
- 13.Bergamini G, Cathomas F, Auer S, Sigrist H, Seifritz E, Patterson M, Gabriel C, Pryce CR. Mouse psychosocial stress reduces motivation and cognitive function in operant reward tests:a model for reward pathology with effects of agomelatine. Eur Neuropsychopharmacol. 2016;26:1448–1464. doi: 10.1016/j.euroneuro.2016.06.009. [DOI] [PubMed] [Google Scholar]
- 14.Bertaina-Anglade V, Drieu-La-Rochelle C, Mocaër E, Seguin L. Memory facilitating effects of agomelatine in the novel object recognition memory paradigm in the rat. Pharmacol Biochem Behav. 2011;98:511–517. doi: 10.1016/j.pbb.2011.02.015. [DOI] [PubMed] [Google Scholar]
- 15.Bogolepova AN, Chukanova EI, Smirnova M, Chukanova AS, Gracheva I, Semushkina EG. The use of valdoxan in the treatment of post-stroke depression. Zh Nevrol Psikhiatr Im S S Korsakova. 2011;111:42–46. [PubMed] [Google Scholar]
- 16.Bonn M, Schmitt A, Lesch KP, Van Bockstaele EJ, Asan E. Serotonergic innervation and serotonin receptor expression of NPY-producing neurons in the rat lateral and basolateral amygdaloid nuclei. Brain Struct Funct. 2013;218:421–435. doi: 10.1007/s00429-012-0406-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bruno A, Zoccali RA, Abenavoli E, Pandolfo G, Scimeca G, Spina E, Muscatello MR. Augmentation of clozapine with agomelatine in partial-responder schizophrenia:a 16-week open-label uncontrolled pilot study. J Clin Psychopharmacol. 2014;34:491–494. doi: 10.1097/JCP.0000000000000157. [DOI] [PubMed] [Google Scholar]
- 18.Bruno A, Micò U, Lorusso S, Cogliandro N, Pandolfo G, Caminiti M, Zoccali RA, Muscatello MR. Agomelatine in the treatment of fibromyalgia:a 12-week open-label uncontrolled preliminary study. J Clin Psychopharmacol. 2013;33:507–511. doi: 10.1097/JCP.0b013e31829057ae. [DOI] [PubMed] [Google Scholar]
- 19.Buoli M, Grassi S, Serati M, Altamura AC. Agomelatine for the treatment of generalized anxiety disorder. Expert Opin Pharmacother. 2017;18:1373–1379. doi: 10.1080/14656566.2017.1359257. [DOI] [PubMed] [Google Scholar]
- 20.Calabrese F, Molteni R, Gabriel C, Mocaer E, Racagni G, Riva MA. Modulation of neuroplastic molecules in selected brain regions after chronic administration of the novel antidepressant agomelatine. Psychopharmacology (Berl) 2011;215:267–275. doi: 10.1007/s00213-010-2129-8. [DOI] [PubMed] [Google Scholar]
- 21.Calabrese JR, Guelfi JD, Perdrizet-Chevallier C. Agomelatine adjunctive therapy for acute bipolar depression:preliminary open data. Bipolar Disord. 2007;9:628–635. doi: 10.1111/j.1399-5618.2007.00507.x. [DOI] [PubMed] [Google Scholar]
- 22.Callegari I, Mattei C, Benassi F, Krueger F, Grafman J, Yaldizli Ö, Sassos D, Massucco D, Scialò C, Nobili F, Serrati C, Amore M, Cocito L, Emberti Gialloreti L, Pardini M. Agomelatine improves apathy in frontotemporal dementia. Neurodegener Dis. 2016;16:352–356. doi: 10.1159/000445873. [DOI] [PubMed] [Google Scholar]
- 23.Can ÖD, Üçel U, Demir Özkay Ü, Ulupınar E. The effect of agomelatine treatment on diabetes-induced cognitive impairments in rats:concomitant alterations in the hippocampal neuron numbers. Int J Mol Sci. 2018;19:2461. doi: 10.3390/ijms19082461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cankara FN, Günaydın C, Çelik ZB, Şahin Y, Pekgöz Ş, Erzurumlu Y, Gülle K. Agomelatine confers neuroprotection against cisplatin-induced hippocampal neurotoxicity. Metab Brain Dis. 2021;36:339–349. doi: 10.1007/s11011-020-00634-y. [DOI] [PubMed] [Google Scholar]
- 25.Carlezon WA, Jr, Duman RS, Nestler EJ. The many faces of CREB. Trends Neurosci. 2005;28:436–445. doi: 10.1016/j.tins.2005.06.005. [DOI] [PubMed] [Google Scholar]
- 26.Chagraoui A, Thibaut F, Skiba M, Thuillez C, Bourin M. 5-HT2C receptors in psychiatric disorders:a review. Prog Neuropsychopharmacol Biol Psychiatry. 2016;66:120–135. doi: 10.1016/j.pnpbp.2015.12.006. [DOI] [PubMed] [Google Scholar]
- 27.Chen G, Bower KA, Ma C, Fang S, Thiele CJ, Luo J. Glycogen synthase kinase 3beta (GSK3beta) mediates 6-hydroxydopamine-induced neuronal death. FASEB J. 2004;18:1162–1164. doi: 10.1096/fj.04-1551fje. [DOI] [PubMed] [Google Scholar]
- 28.Chenu F, El Mansari M, Blier P. Electrophysiological effects of repeated administration of agomelatine on the dopamine norepinephrine and serotonin systems in the rat brain. Neuropsychopharmacology. 2013;38:275–284. doi: 10.1038/npp.2012.140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chisholm D, Sweeny K, Sheehan P, Rasmussen B, Smit F, Cuijpers P, Saxena S. Scaling-up treatment of depression and anxiety:a global return on investment analysis. Lancet Psychiatry. 2016;3:415–424. doi: 10.1016/S2215-0366(16)30024-4. [DOI] [PubMed] [Google Scholar]
- 30.Cipriani A, Furukawa TA, Salanti G, Chaimani A, Atkinson LZ, Ogawa Y, Leucht S, Ruhe HG, Turner EH, Higgins JPT, Egger M, Takeshima N, Hayasaka Y, Imai H, Shinohara K, Tajika A, Ioannidis JPA, Geddes JR. Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder:a systematic review and network meta-analysis. Lancet. 2018;391:1357–1366. doi: 10.1016/S0140-6736(17)32802-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Cléry-Melin ML, Gorwood P. A simple attention test in the acute phase of a major depressive episode is predictive of later functional remission. Depress Anxiety. 2017;34:159–170. doi: 10.1002/da.22575. [DOI] [PubMed] [Google Scholar]
- 32.Conboy L, Tanrikut C, Zoladz PR, Campbell AM, Park CR, Gabriel C, Mocaer E, Sandi C, Diamond DM. The antidepressant agomelatine blocks the adverse effects of stress on memory and enables spatial learning to rapidly increase neural cell adhesion molecule (NCAM) expression in the hippocampus of rats. Int J Neuropsychopharmacol. 2009;12:329–341. doi: 10.1017/S1461145708009255. [DOI] [PubMed] [Google Scholar]
- 33.Dagyte G, Luiten PG, De Jager T, Gabriel C, Mocaër E, Den Boer JA, Van der Zee EA. Chronic stress and antidepressant agomelatine induce region-specific changes in synapsin I expression in the rat brain. J Neurosci Res. 2011;89:1646–1657. doi: 10.1002/jnr.22697. [DOI] [PubMed] [Google Scholar]
- 34.De Berardis D, Valchera A, Fornaro M, Serroni N, Marini S, Moschetta FS, Martinotti G, Di Giannantonio M. Agomelatine reversal of escitalopram-induced apathy:a case report. Psychiatry Clin Neurosci. 2013;67:190–191. doi: 10.1111/pcn.12032. [DOI] [PubMed] [Google Scholar]
- 35.Demir Özkay Ü, Söztutar E, Can ÖD, Üçel U, Öztürk Y, Ulupinar E. Effects of long-term agomelatine treatment on the cognitive performance and hippocampal plasticity of adult rats. Behav Pharmacol. 2015;26:469–480. doi: 10.1097/FBP.0000000000000153. [DOI] [PubMed] [Google Scholar]
- 36.Demireva EY, Suri D, Morelli E, Mahadevia D, Chuhma N, Teixeira CM, Ziolkowski A, Hersh M, Fifer J, Bagchi S, Chemiakine A, Moore H, Gingrich JA, Balsam P, Rayport S, Ansorge MS. 5-HT2C receptor blockade reverses SSRI-associated basal ganglia dysfunction and potentiates therapeutic efficacy. Mol Psychiatry. 2020;25:3304–3321. doi: 10.1038/s41380-018-0227-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Descamps A, Rousset C, Dugua H, Debilly G, Delagrange P, Cespuglio R. Agomelatine restores a physiological response to stress in the aged rat. Neurosci Lett. 2014;566:257–262. doi: 10.1016/j.neulet.2014.02.044. [DOI] [PubMed] [Google Scholar]
- 38.di Giannantonio M, Montemitro C, Sepede G, Brunetti M, Baroni G, Corbo M, Anders M, Tavčar R, Martinotti G, Manzoli L. Agomelatine effectiveness tolerability and impact on anhedonia in major depression:a pooled analysis. J Clin Psychopharmacol. 2019;39:288–290. doi: 10.1097/JCP.0000000000001038. [DOI] [PubMed] [Google Scholar]
- 39.Dmitrzak-Weglarz M, Banach E, Bilska K, Narozna B, Szczepankiewicz A, Reszka E, Jablonska E, Kapelski P, Skibinska M, Pawlak J. Molecular regulation of the melatonin biosynthesis pathway in unipolar and bipolar depression. Front Pharmacol. 2021;12:666541. doi: 10.3389/fphar.2021.666541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Drozd R, Rychlik M, Fijalkowska A, Rygula R. Effects of cognitive judgement bias and acute antidepressant treatment on sensitivity to feedback and cognitive flexibility in the rat version of the probabilistic reversal-learning test. Behav Brain Res. 2019;359:619–629. doi: 10.1016/j.bbr.2018.10.003. [DOI] [PubMed] [Google Scholar]
- 41.Duda P, Hajka D, Wójcicka O, Rakus D, Gizak A. GSK3β:a master player in depressive disorder pathogenesis and treatment responsiveness. Cells. 2020;9:727. doi: 10.3390/cells9030727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Endo H, Nito C, Kamada H, Yu F, Chan PH. Akt/GSK3beta survival signaling is involved in acute brain injury after subarachnoid hemorrhage in rats. Stroke. 2006;37:2140–2146. doi: 10.1161/01.STR.0000229888.55078.72. [DOI] [PubMed] [Google Scholar]
- 43.Englisch S, Jung HS, Eisenacher S, Lewien A, Becker A, Nowak U, Braun H, Thiem J, Meyer-Lindenberg A, Zink M. Neurocognitive effects of agomelatine treatment in schizophrenia patients suffering from comorbid depression:results from the AGOPSYCH study. J Clin Psychopharmacol. 2018;38:357–361. doi: 10.1097/JCP.0000000000000909. [DOI] [PubMed] [Google Scholar]
- 44.Erdoğan Ç, Özdemir Rezaki H, Koçak OM, Buturak ŞV. Comparison of the effects of antidepressants with different mechanisms of action on efficacy cognitive functions and side effects. Turk Psikiyatri Derg. 2020;31:90–98. doi: 10.5080/u23704. [DOI] [PubMed] [Google Scholar]
- 45.Esvald EE, Tuvikene J, Sirp A, Patil S, Bramham CR, Timmusk T. CREB family transcription factors are major mediators of BDNF transcriptional autoregulation in cortical neurons. J Neurosci. 2020;40:1405–1426. doi: 10.1523/JNEUROSCI.0367-19.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Fornaro M, McCarthy MJ, De Berardis D, De Pasquale C, Tabaton M, Martino M, Colicchio S, Cattaneo CI, D'Angelo E, Fornaro P. Adjunctive agomelatine therapy in the treatment of acute bipolar II depression:a preliminary open label study. Neuropsychiatr Dis Treat. 2013;9:243–251. doi: 10.2147/NDT.S41557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Friedrich ME, Akimova E, Huf W, Konstantinidis A, Papageorgiou K, Winkler D, Toto S, Greil W, Grohmann R, Kasper S. Drug-induced liver injury during antidepressant treatment:results of AMSP a drug surveillance program. Int J Neuropsychopharmacol. 2016;19:pyv126. doi: 10.1093/ijnp/pyv126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Froböse T, Slawik H, Schreiner R, Veselý Z, Wiegand M, Bäuml J, Förstl H. Agomelatine improves sleep in a patient with fatal familial insomnia. Pharmacopsychiatry. 2012;45:34–36. doi: 10.1055/s-0031-1287778. [DOI] [PubMed] [Google Scholar]
- 49.Gavrilova SI, Kolykhalov IV, Ponomareva EV, Selezneva ND. Clinical experience with agomelatine for the treatment of depression in elderly patients in outpatient practice. Zh Nevrol Psikhiatr Im S S Korsakova. 2014;114:43–48. [PubMed] [Google Scholar]
- 50.Gavrilova SI, Kalyn YB, Safarova TP, Yakovleva OB, Sheshenin VS, Kornilov VV, Shipilova ES. Optimization of the efficacy and safety of antidepressant therapy in patients of a geriatric psychiatric unit. Zh Nevrol Psikhiatr Im S S Korsakova. 2015;115:24–32. doi: 10.17116/jnevro20151156124-32. [DOI] [PubMed] [Google Scholar]
- 51.Gorwood P, Richard-Devantoy S, Baylé F, Cléry-Melin ML. Psychomotor retardation is a scar of past depressive episodes revealed by simple cognitive tests. Eur Neuropsychopharmacol. 2014;24:1630–1640. doi: 10.1016/j.euroneuro.2014.07.013. [DOI] [PubMed] [Google Scholar]
- 52.Gorwood P, Vaiva G, Corruble E, Llorca PM, BayléFJ , Courtet P. The ability of early changes in motivation to predict later antidepressant treatment response. Neuropsychiatr Dis Treat. 2015;11:2875–2882. doi: 10.2147/NDT.S92795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gorwood P, Benichou J, Moore N, Wattez M, Secouard MC, Desobry X, Picarel-Blanchot F, de Bodinat C. Agomelatine in standard medical practice in depressed patients:results of a 1-year multicentre observational study in France. Clin Drug Investig. 2020;40:1009–1020. doi: 10.1007/s40261-020-00957-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Grunze H, Vieta E, Goodwin GM, Bowden C, Licht RW, Azorin JM, Yatham L, Mosolov S, Möller HJ, Kasper S. The World Federation of Societies of Biological Psychiatry (WFSBP) Guidelines for the biological treatment of bipolar disorders:acute and long-term treatment of mixed states in bipolar disorder. World J Biol Psychiatry. 2018;19:2–58. doi: 10.1080/15622975.2017.1384850. [DOI] [PubMed] [Google Scholar]
- 55.Guan Q, Hu X, Ma N, He H, Duan F, Li X, Luo Y, Zhang H. Sleep quality depression and cognitive function in non-demented older adults. J Alzheimers Dis. 2020;76:1637–1650. doi: 10.3233/JAD-190990. [DOI] [PubMed] [Google Scholar]
- 56.Gumuslu E, Mutlu O, Sunnetci D, Ulak G, Celikyurt IK, Cine N, Akar F, Savlı H, Erden F. The antidepressant agomelatine improves memory deterioration and upregulates CREB and BDNF gene expression levels in unpredictable chronic mild stress (UCMS)-exposed mice. Drug target insights. 2014;8:11–21. doi: 10.4137/DTI.S13870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Gupta S, Sharma B. Pharmacological benefits of agomelatine and vanillin in experimental model of Huntington's disease. Pharmacol Biochem Behav. 2014;122:122–135. doi: 10.1016/j.pbb.2014.03.022. [DOI] [PubMed] [Google Scholar]
- 58.Gupta S, Singh P, Sharma BM, Sharma B. Neuroprotective effects of agomelatine and vinpocetine against chronic cerebral hypoperfusion induced vascular dementia. Curr Neurovasc Res. 2015;12:240–252. doi: 10.2174/1567202612666150603130235. [DOI] [PubMed] [Google Scholar]
- 59.Gutierrez MEZ, Savall ASP, da Luz Abreu E, Nakama KA, Dos Santos RB, Guedes MCM, Ávila DS, Luchese C, Haas SE, Quines CB, Pinton S. Co-nanoencapsulated meloxicam and curcumin improves cognitive impairment induced by amyloid-beta through modulation of cyclooxygenase-2 in mice. Neural Regen Res. 2021;16:783–789. doi: 10.4103/1673-5374.295339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hadj Ayed Tka K, Mahfoudh Boussaid A, Zaouali MA, Kammoun R, Bejaoui M, Ghoul Mazgar S, Rosello Catafau J, Ben Abdennebi H. Melatonin modulates endoplasmic reticulum stress and Akt/GSK3-beta signaling pathway in a rat model of renal warm ischemia reperfusion. Anal Cell Pathol (Amst) 2015;2015:635172. doi: 10.1155/2015/635172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Harmer CJ, de Bodinat C, Dawson GR, Dourish CT, Waldenmaier L, Adams S, Cowen PJ, Goodwin GM. Agomelatine facilitates positive versus negative affective processing in healthy volunteer models. J Psychopharmacol. 2011;25:1159–1167. doi: 10.1177/0269881110376689. [DOI] [PubMed] [Google Scholar]
- 62.Harrison F, Aerts L, Brodaty H. Apathy in dementia:systematic review of recent evidence on pharmacological treatments. Curr Psychiatry Rep. 2016;18:103. doi: 10.1007/s11920-016-0737-7. [DOI] [PubMed] [Google Scholar]
- 63.Harvey BH, Regenass W, Dreyer W, Möller M. Social isolation rearing-induced anxiety and response to agomelatine in male and female rats:Role of corticosterone oxytocin and vasopressin. J Psychopharmacol. 2019;33:640–646. doi: 10.1177/0269881119826783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Hernandez C, Shukla S. Liposome based drug delivery as a potential treatment option for Alzheimer's disease. Neural Regen Res. 2022;17:1190–1198. doi: 10.4103/1673-5374.327328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Heun R, Ahokas A, Boyer P, Giménez-Montesinos N, Pontes-Soares F, Olivier V. The efficacy of agomelatine in elderly patients with recurrent major depressive disorder:a placebo-controlled study. J Clin Psychiatry. 2013;74:587–594. doi: 10.4088/JCP.12m08250. [DOI] [PubMed] [Google Scholar]
- 66.Hill RA, Murray SS, Halley PG, Binder MD, Martin SJ, van den Buuse M. Brain-derived neurotrophic factor expression is increased in the hippocampus of 5-HT(2C) receptor knockout mice. Hippocampus. 2011;21:434–445. doi: 10.1002/hipo.20759. [DOI] [PubMed] [Google Scholar]
- 67.Husain M, Roiser JP. Neuroscience of apathy and anhedonia:a transdiagnostic approach. Nat Rev Neurosci. 2018;19:470–484. doi: 10.1038/s41583-018-0029-9. [DOI] [PubMed] [Google Scholar]
- 68.Ilieva K, Tchekalarova J, Atanasova D, Kortenska L, Atanasova M. Antidepressant agomelatine attenuates behavioral deficits and concomitant pathology observed in streptozotocin-induced model of Alzheimer's disease in male rats. Horm Behav. 2019;107:11–19. doi: 10.1016/j.yhbeh.2018.11.007. [DOI] [PubMed] [Google Scholar]
- 69.Ilieva K, Atanasova M, Atanasova D, Kortenska L, Tchekalarova J. Chronic agomelatine treatment alleviates icvAβ-induced anxiety and depressive-like behavior through affecting Aβmetabolism in the hippocampus in a rat model of Alzheimer's disease. Physiol Behav. 2021;239:113525. doi: 10.1016/j.physbeh.2021.113525. [DOI] [PubMed] [Google Scholar]
- 70.İlkaya F, Yüce M, Ağrı AE, Güzel H, Balcı H, Uçar F, Babadağı Z, Müjdeci M, Mutlu E. The combination of agomelatine and ritanserin exerts a synergistic interaction in passive avoidance task. Hum Exp Toxicol. 2015;34:787–795. doi: 10.1177/0960327114559613. [DOI] [PubMed] [Google Scholar]
- 71.Kalyn YB, Safarova TP, Yakovleva OB, Sheshenin VS, Kornilov VV, Shipilova ES, Gavrilova SI. Experience of the antidepressive therapy with valdoxan (agomelatine) in a psychogeriatric unit of the psychiatric hospital. Zh Nevrol Psikhiatr Im S S Korsakova. 2015;115:55–62. doi: 10.17116/jnevro201511511155-62. [DOI] [PubMed] [Google Scholar]
- 72.Kennaway DJ. A critical review of melatonin assays:Past and present. J Pineal Res. 2019;67:e12572. doi: 10.1111/jpi.12572. [DOI] [PubMed] [Google Scholar]
- 73.Kennedy SH, Rizvi S, Fulton K, Rasmussen J. A double-blind comparison of sexual functioning antidepressant efficacy and tolerability between agomelatine and venlafaxine XR. J Clin Psychopharmacol. 2008;28:329–333. doi: 10.1097/JCP.0b013e318172b48c. [DOI] [PubMed] [Google Scholar]
- 74.Kitagishi Y, Kobayashi M, Kikuta K, Matsuda S. Roles of PI3K/AKT/GSK3/mTOR pathway in cell signaling of mental illnesses. Depress Res Treat. 2012;2012:752563. doi: 10.1155/2012/752563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Krishnakumar A, Nandhu MS, Paulose CS. Upregulation of 5-HT2C receptors in hippocampus of pilocarpine-induced epileptic rats:antagonism by Bacopa monnieri. Epilepsy Behav. 2009;16:225–230. doi: 10.1016/j.yebeh.2009.07.031. [DOI] [PubMed] [Google Scholar]
- 76.Kumar A. Calcium signaling during brain aging and its influence on the hippocampal synaptic plasticity. Adv Exp Med Biol. 2020;1131:985–1012. doi: 10.1007/978-3-030-12457-1_39. [DOI] [PubMed] [Google Scholar]
- 77.Ladurelle N, Gabriel C, Viggiano A, Mocaër E, Baulieu EE, Bianchi M. Agomelatine (S20098) modulates the expression of cytoskeletal microtubular proteins synaptic markers and BDNF in the rat hippocampus amygdala and PFC. Psychopharmacology (Berl) 2012;221:493–509. doi: 10.1007/s00213-011-2597-5. [DOI] [PubMed] [Google Scholar]
- 78.Landolt HP, Wehrle R. Antagonism of serotonergic 5-HT2A/2C receptors:mutual improvement of sleep cognition and mood? Eur J Neurosci. 2009;29:1795–1809. doi: 10.1111/j.1460-9568.2009.06718.x. [DOI] [PubMed] [Google Scholar]
- 79.Lapmanee S, Charoenphandhu J, Teerapornpuntakit J, Krishnamra N, Charoenphandhu N. Agomelatine venlafaxine and running exercise effectively prevent anxiety- and depression-like behaviors and memory impairment in restraint stressed rats. PLoS One. 2017;12:e0187671. doi: 10.1371/journal.pone.0187671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.LeMoult J, Gotlib IH. Depression:a cognitive perspective. Clin Psychol Rev. 2019;69:51–66. doi: 10.1016/j.cpr.2018.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lerner AB, Case JD, Takahashi Y. Isolation of melatonin and 5-methoxyindole-3-acetic acid from bovine pineal glands. J Biol Chem. 1960;235:1992–1997. [PubMed] [Google Scholar]
- 82.Li K, Shen S, Ji YT, Li XY, Zhang LS, Wang XD. Melatonin augments the effects of fluoxetine on depression-like behavior and hippocampal BDNF-TrkB signaling. Neurosci Bull. 2018;34:303–311. doi: 10.1007/s12264-017-0189-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Li P, Hu F, Cao X, Luo L, Tu Q. Melatonin receptor protects cardiomyocyte against oxidative stress-induced apoptosis through the MAPK-ERK signaling pathway. J Recept Signal Transduct Res. 2020;40:117–125. doi: 10.1080/10799893.2020.1719151. [DOI] [PubMed] [Google Scholar]
- 84.Lisman J, Cooper K, Sehgal M, Silva AJ. Memory formation depends on both synapse-specific modifications of synaptic strength and cell-specific increases in excitability. Nat Neurosci. 2018;21:309–314. doi: 10.1038/s41593-018-0076-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Lu Y, Ho CS, McIntyre RS, Wang W, Ho RC. Agomelatine-induced modulation of brain-derived neurotrophic factor (BDNF) in the rat hippocampus. Life Sci. 2018;210:177–184. doi: 10.1016/j.lfs.2018.09.003. [DOI] [PubMed] [Google Scholar]
- 86.Marrocco J, Reynaert ML, Gatta E, Gabriel C, Mocaër E, Di Prisco S, Merega E, Pittaluga A, Nicoletti F, Maccari S, Morley-Fletcher S, Mairesse J. The effects of antidepressant treatment in prenatally stressed rats support the glutamatergic hypothesis of stress-related disorders. J Neurosci. 2014;34:2015–2024. doi: 10.1523/JNEUROSCI.4131-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Marshall M. The hidden links between mental disorders. Nature. 2020;581:19–21. doi: 10.1038/d41586-020-00922-8. [DOI] [PubMed] [Google Scholar]
- 88.Martin V, Allaïli N, Euvrard M, Marday T, Riffaud A, Franc B, Mocaër E, Gabriel C, Fossati P, Lehericy S, Lanfumey L. Effect of agomelatine on memory deficits and hippocampal gene expression induced by chronic social defeat stress in mice. Sci Rep. 2017;8:45907. doi: 10.1038/srep45907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Martinotti G, Pettorruso M, De Berardis D, Varasano PA, Lucidi Pressanti G, De Remigis V, Valchera A, Ricci V, Di Nicola M, Janiri L, Biggio G, Di Giannantonio M. Agomelatine increases BDNF serum levels in depressed patients in correlation with the improvement of depressive symptoms. Int J Neuropsychopharmacol. 2016;19:pyw003. doi: 10.1093/ijnp/pyw003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.McHutchison CA, Leighton DJ, McIntosh A, Cleary E, Warner J, Porteous M, Chandran S, Pal S, Abrahams S. Relationship between neuropsychiatric disorders and cognitive and behavioural change in MND. J Neurol Neurosurg Psychiatry. 2020;91:245–253. doi: 10.1136/jnnp-2019-321737. [DOI] [PubMed] [Google Scholar]
- 91.Medvedev VE, Ter-Israelyan AY, Frolova VI, Korovyakova EA, Gushanskaya EV. Treatment of depression with cognitive impairment. Zh Nevrol Psikhiatr Im S S Korsakova. 2018;118:77–80. doi: 10.17116/jnevro20181182177-80. [DOI] [PubMed] [Google Scholar]
- 92.Millan MJ. Serotonin 5-HT2C receptors as a target for the treatment of depressive and anxious states:focus on novel therapeutic strategies. Therapie. 2005;60:441–460. doi: 10.2515/therapie:2005065. [DOI] [PubMed] [Google Scholar]
- 93.Millan MJ, Gobert A, Lejeune F, Dekeyne A, Newman-Tancredi A, Pasteau V, Rivet JM, Cussac D. The novel melatonin agonist agomelatine (S20098) is an antagonist at 5-hydroxytryptamin|ne2C receptors blockade of which enhances the activity of frontocortical dopaminergic and adrenergic pathways. J Pharmacol Exp Ther. 2003;306:954–964. doi: 10.1124/jpet.103.051797. [DOI] [PubMed] [Google Scholar]
- 94.Musazzi L, Seguini M, Mallei A, Treccani G, Pelizzari M, Tornese P, Racagni G, Tardito D. Time-dependent activation of MAPK/Erk1/2 and Akt/GSK3 cascades:modulation by agomelatine. BMC Neurosci. 2014;15:119. doi: 10.1186/s12868-014-0119-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Norman TR, Olver JS. Agomelatine for depression:expanding the horizons? Expert Opin Pharmacother. 2019;20:647–656. doi: 10.1080/14656566.2019.1574747. [DOI] [PubMed] [Google Scholar]
- 96.O'Neill B, Gardani M, Findlay G, Whyte T, Cullen T. Challenging behaviour and sleep cycle disorder following brain injury:a preliminary response to agomelatine treatment. Brain Inj. 2014;28:378–381. doi: 10.3109/02699052.2013.865264. [DOI] [PubMed] [Google Scholar]
- 97.Patrício P, Mateus-Pinheiro A, Irmler M, Alves ND, Machado-Santos AR, Morais M, Correia JS, Korostynski M, Piechota M, Stoffel R, Beckers J, Bessa JM, Almeida OF, Sousa N, Pinto L. Differential and converging molecular mechanisms of antidepressants'action in the hippocampal dentate gyrus. Neuropsychopharmacology. 2015;40:338–349. doi: 10.1038/npp.2014.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Pavlova MK, Latreille V. Sleep disorders. Am J Med. 2019;132:292–299. doi: 10.1016/j.amjmed.2018.09.021. [DOI] [PubMed] [Google Scholar]
- 99.Pjrek E, Winkler D, Konstantinidis A, Willeit M, Praschak-Rieder N, Kasper S. Agomelatine in the treatment of seasonal affective disorder. Psychopharmacology (Berl) 2007;190:575–579. doi: 10.1007/s00213-006-0645-3. [DOI] [PubMed] [Google Scholar]
- 100.Post RM. The impact of bipolar depression. J Clin Psychiatry 66 Suppl. 2005;5:5–10. [PubMed] [Google Scholar]
- 101.Price RB, Duman R. Neuroplasticity in cognitive and psychological mechanisms of depression:an integrative model. Mol Psychiatry. 2020;25:530–543. doi: 10.1038/s41380-019-0615-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Quera-Salva MA, Lemoine P, Guilleminault C. Impact of the novel antidepressant agomelatine on disturbed sleep-wake cycles in depressed patients. Human psychopharmacology. 2010;25:222–229. doi: 10.1002/hup.1112. [DOI] [PubMed] [Google Scholar]
- 103.Reynolds SA, Ebben MR. The cost of insomnia and the benefit of increased access to evidence-based treatment:cognitive behavioral therapy for insomnia. Sleep Med Clin. 2017;12:39–46. doi: 10.1016/j.jsmc.2016.10.011. [DOI] [PubMed] [Google Scholar]
- 104.Robillard R, Carpenter JS, Feilds KL, Hermens DF, White D, Naismith SL, Bartlett D, Whitwell B, Southan J, Scott EM, Hickie IB. Parallel changes in mood and melatonin rhythm following an adjunctive multimodal chronobiological intervention with agomelatine in people with depression:a proof of concept open label study. Front Psychiatry. 2018;9:624. doi: 10.3389/fpsyt.2018.00624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Safarova TP, Yakovleva OB, Sheshenin VS, Gavrilova SI. Methods of augmentation of antidepressant therapy (on the model of complex therapy with the inclusion of actovegin) in gerontopsychiatric hospital. Zh Nevrol Psikhiatr Im S S Korsakova. 2018;118:55–63. doi: 10.17116/jnevro201811806255. [DOI] [PubMed] [Google Scholar]
- 106.Sant'Ana AB, Vilela-Costa HH, Vicente MA, Hernandes PM, de Andrade T, Zangrossi H., Jr Role of 5-HT2C receptors of the dorsal hippocampus in the modulation of anxiety- and panic-related defensive responses in rats. Neuropharmacology. 2019;148:311–319. doi: 10.1016/j.neuropharm.2019.01.026. [DOI] [PubMed] [Google Scholar]
- 107.Santos P, Herrmann AP, Elisabetsky E, Piato A. Anxiolytic properties of compounds that counteract oxidative stress neuroinflammation and glutamatergic dysfunction:a review. Braz J Psychiatry. 2019;41:168–178. doi: 10.1590/1516-4446-2018-0005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Schmauss C, Zimnisky R, Mehta M, Shapiro LP. The roles of phospholipase C activation and alternative ADAR1 and ADAR2 pre-mRNA splicing in modulating serotonin 2C-receptor editing in vivo. RNA. 2010;16:1779–1785. doi: 10.1261/rna.2188110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Shi L, Chen SJ, Ma MY, Bao YP, Han Y, Wang YM, Shi J, Vitiello MV, Lu L. Sleep disturbances increase the risk of dementia:A systematic review and meta-analysis. Sleep Med Rev. 2018;40:4–16. doi: 10.1016/j.smrv.2017.06.010. [DOI] [PubMed] [Google Scholar]
- 110.Singh P, Gupta S, Sharma B. Melatonin receptor and KATP channel modulation in experimental vascular dementia. Physiol Behav. 2015;142:66–78. doi: 10.1016/j.physbeh.2015.02.009. [DOI] [PubMed] [Google Scholar]
- 111.Slee A, Nazareth I, Bondaronek P, Liu Y, Cheng Z, Freemantle N. Pharmacological treatments for generalised anxiety disorder:a systematic review and network meta-analysis. Lancet. 2019;393:768–777. doi: 10.1016/S0140-6736(18)31793-8. [DOI] [PubMed] [Google Scholar]
- 112.Stahl SM. Mechanism of action of agomelatine:a novel antidepressant exploiting synergy between monoaminergic and melatonergic properties. CNS Spectr. 2014;19:207–212. doi: 10.1017/S1092852914000248. [DOI] [PubMed] [Google Scholar]
- 113.Stein DJ, Ahokas A, Jarema M, Avedisova AS, Vavrusova L, Chaban O, Gruget C, Olivier V, Picarel-Blanchot F, de Bodinat C. Efficacy and safety of agomelatine (10 or 25 mg/day) in non-depressed out-patients with generalized anxiety disorder:A 12-week double-blind placebo-controlled study. Eur Neuropsychopharmacol. 2017;27:526–537. doi: 10.1016/j.euroneuro.2017.02.007. [DOI] [PubMed] [Google Scholar]
- 114.Stein DJ, Khoo JP, Ahokas A, Jarema M, Van Ameringen M, Vavrusova L, H?schl C, Bauer M, Bitter I, Mosolov SN, Olivier V, Matharan S, Picarel-Blanchot F, de Bodinat C. 12-week double-blind randomized multicenter study of efficacy and safety of agomelatine (25-50 mg/day) versus escitalopram (10-20 mg/day) in out-patients with severe generalized anxiety disorder. Eur Neuropsychopharmacol. 2018;28:970–979. doi: 10.1016/j.euroneuro.2018.05.006. [DOI] [PubMed] [Google Scholar]
- 115.Štuhec M. Agomelatine-induced hepatotoxicity. Wien Klin Wochenschr. 2013;125:225–226. doi: 10.1007/s00508-013-0344-0. [DOI] [PubMed] [Google Scholar]
- 116.Sung JY, Bae JH, Lee JH, Kim YN, Kim DK. The melatonin signaling pathway in a long-term memory in vitro study. Molecules. 2018;23:737. doi: 10.3390/molecules23040737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Tardito D, Milanese M, Bonifacino T, Musazzi L, Grilli M, Mallei A, Mocaer E, Gabriel-Gracia C, Racagni G, Popoli M, Bonanno G. Blockade of stress-induced increase of glutamate release in the rat prefrontal/frontal cortex by agomelatine involves synergy between melatonergic and 5-HT2C receptor-dependent pathways. BMC Neurosci. 2010;11:68. doi: 10.1186/1471-2202-11-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Tchekalarova J, Stoynova T, Ilieva K, Mitreva R, Atanasova M. Agomelatine treatment corrects symptoms of depression and anxiety by restoring the disrupted melatonin circadian rhythms of rats exposed to chronic constant light. Pharmacol Biochem Behav. 2018;171:1–9. doi: 10.1016/j.pbb.2018.05.016. [DOI] [PubMed] [Google Scholar]
- 119.Tchekalarova J, Kortenska L, Ivanova N, Atanasova M, Marinov P. Agomelatine treatment corrects impaired sleep-wake cycle and sleep architecture and increases MT(1) receptor as well as BDNF expression in the hippocampus during the subjective light phase of rats exposed to chronic constant light. Psychopharmacology (Berl) 2020;237:503–518. doi: 10.1007/s00213-019-05385-y. [DOI] [PubMed] [Google Scholar]
- 120.Tchekalarova J, Atanasova D, Nenchovska Z, Atanasova M, Kortenska L, Gesheva R, Lazarov N. Agomelatine protects against neuronal damage without preventing epileptogenesis in the kainate model of temporal lobe epilepsy. Neurobiol Dis. 2017;104:1–14. doi: 10.1016/j.nbd.2017.04.017. [DOI] [PubMed] [Google Scholar]
- 121.Tempesta D, Socci V, De Gennaro L, Ferrara M. Sleep and emotional processing. Sleep Med Rev. 2018;40:183–195. doi: 10.1016/j.smrv.2017.12.005. [DOI] [PubMed] [Google Scholar]
- 122.Tondo L, Vázquez GH, Baldessarini RJ. Depression and mania in bipolar disorder. Curr Neuropharmacol. 2017;15:353–358. doi: 10.2174/1570159X14666160606210811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Tsuno N, Besset A, Ritchie K. Sleep and depression. J Clin Psychiatry. 2005;66:1254–1269. doi: 10.4088/jcp.v66n1008. [DOI] [PubMed] [Google Scholar]
- 124.Tye KM, Mirzabekov JJ, Warden MR, Ferenczi EA, Tsai HC, Finkelstein J, Kim SY, Adhikari A, Thompson KR, Andalman AS, Gunaydin LA, Witten IB, Deisseroth K. Dopamine neurons modulate neural encoding and expression of depression-related behaviour. Nature. 2013;493:537–541. doi: 10.1038/nature11740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.von Bohlen Und Halbach O, von Bohlen Und Halbach V. BDNF effects on dendritic spine morphology and hippocampal function. Cell Tissue Res. 2018;373:729–741. doi: 10.1007/s00441-017-2782-x. [DOI] [PubMed] [Google Scholar]
- 126.Wada K, Hu L, Mores N, Navarro CE, Fuda H, Krsmanovic LZ, Catt KJ. Serotonin (5-HT) receptor subtypes mediate specific modes of 5-HT-induced signaling and regulation of neurosecretion in gonadotropin-releasing hormone neurons. Mol Endocrinol. 2006;20:125–135. doi: 10.1210/me.2005-0109. [DOI] [PubMed] [Google Scholar]
- 127.Watson JA, Elliott AC, Brown PD. Serotonin elevates intracellular Ca2+in rat choroid plexus epithelial cells by acting on 5-HT2C receptors. Cell Calcium. 1995;17:120–128. doi: 10.1016/0143-4160(95)90081-0. [DOI] [PubMed] [Google Scholar]
- 128.Weissmann D, van der Laan S, Underwood MD, Salvetat N, Cavarec L, Vincent L, Molina F, Mann JJ, Arango V, Pujol JF. Region-specific alterations of A-to-I RNA editing of serotonin 2c receptor in the cortex of suicides with major depression. Transl Psychiatry. 2016;6:e878. doi: 10.1038/tp.2016.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Werry TD, Gregory KJ, Sexton PM, Christopoulos A. Characterization of serotonin 5-HT2C receptor signaling to extracellular signal-regulated kinases 1 and 2. J Neurochem. 2005;93:1603–1615. doi: 10.1111/j.1471-4159.2005.03161.x. [DOI] [PubMed] [Google Scholar]
- 130.Werry TD, Loiacono R, Sexton PM, Christopoulos A. RNA editing of the serotonin 5HT2C receptor and its effects on cell signalling, pharmacology and brain function. Pharmacol Ther. 2008;119:7–23. doi: 10.1016/j.pharmthera.2008.03.012. [DOI] [PubMed] [Google Scholar]
- 131.World Health Organization. Dementia: Key Facts; 2021. [Accessed November 8, 2021]. https://www.who.int/newsroom/ fact-sheets/detail/dementia. [Google Scholar]
- 132.Xie WJ, Xia TJ, Zhou QY, Liu YJ, Gu XP. Role of microglia-mediated neuronal injury in neurodegenerative diseases. Zhongguo Zuzhi Gongcheng Yanjiu. 2021;25:1109–1115. [Google Scholar]
- 133.Xu W, Tan CC, Zou JJ, Cao XP, Tan L. Sleep problems and risk of all-cause cognitive decline or dementia:an updated systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. 2020;91:236–244. doi: 10.1136/jnnp-2019-321896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Yamamoto H, Kamegaya E, Hagino Y, Takamatsu Y, Sawada W, Matsuzawa M, Ide S, Yamamoto T, Mishina M, Ikeda K. Loss of GluN2D subunit results in social recognition deficit social stress 5-HT(2C) receptor dysfunction and anhedonia in mice. Neuropharmacology. 2017;112:188–197. doi: 10.1016/j.neuropharm.2016.07.036. [DOI] [PubMed] [Google Scholar]
- 135.Yao K, Zhao YF, Zu HB. Melatonin receptor stimulation by agomelatine prevents Aβ-induced tau phosphorylation and oxidative damage in PC12 cells. Drug Des Devel Ther. 2019;13:387–396. doi: 10.2147/DDDT.S182684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Yous S, Andrieux J, Howell HE, Morgan PJ, Renard P, Pfeiffer B, Lesieur D, Guardiola-Lemaitre B. Novel naphthalenic ligands with high affinity for the melatonin receptor. J Med Chem. 1992;35:1484–1486. doi: 10.1021/jm00086a018. [DOI] [PubMed] [Google Scholar]
- 137.Yucel A, Yucel N, Ozkanlar S, Polat E, Kara A, Ozcan H, Gulec M. Effect of agomelatine on adult hippocampus apoptosis and neurogenesis using the stress model of rats. Acta Histochem. 2016;118:299–304. doi: 10.1016/j.acthis.2016.02.007. [DOI] [PubMed] [Google Scholar]
- 138.Zisapel N. New perspectives on the role of melatonin in human sleep circadian rhythms and their regulation. Br J Pharmacol. 2018;175:3190–3199. doi: 10.1111/bph.14116. [DOI] [PMC free article] [PubMed] [Google Scholar]
