Skip to main content
Chronic Stress logoLink to Chronic Stress
. 2019 Jun 26;3:2470547019858083. doi: 10.1177/2470547019858083

Novel Targets for Fast Antidepressant Responses: Possible Role of Endogenous Neuromodulators

Anderson Camargo 1, Ana Lúcia S Rodrigues 2,
PMCID: PMC7219953  PMID: 32440595

Abstract

The available medications for the treatment of major depressive disorder have limitations, particularly their limited efficacy, delayed therapeutic effects, and the side effects associated with treatment. These issues highlight the need for better therapeutic agents that provide more efficacious and faster effects for the management of this disorder. Ketamine, an N-methyl-D-aspartate receptor antagonist, is the prototype for novel glutamate-based antidepressants that has been shown to cause a rapid and sustained antidepressant effect even in severe refractory depressive patients. Considering the importance of these findings, several studies have been conducted to elucidate the molecular targets for ketamine’s effect. In addition, efforts are under way to characterize ketamine-like drugs. This review focuses particularly on evidence that endogenous glutamatergic neuromodulators may be able to modulate mood and to elicit fast antidepressant responses. Among these molecules, agmatine and creatine stand out as those with more published evidence of similarities with ketamine, but guanosine and ascorbic acid have also provided promising results. The possibility that these neuromodulators and ketamine have common neurobiological mechanisms, mainly the ability to activate mechanistic target of rapamycin and brain-derived neurotrophic factor signaling, and synthesis of synaptic proteins in the prefrontal cortex and/or hippocampus is presented and discussed.

Keywords: agmatine, ascorbic acid, creatine, fast-acting antidepressant, guanosine, ketamine

Introduction

Major depressive disorder (MDD) is a common and chronic neuropsychiatric condition, characterized by affective and physiological impairments that cause a profound impact on the health of the affected individuals worldwide and a great economic burden.1 The World Health Organization estimates that more than 300 million individuals are affected by MDD at present, and the number of individuals affected by this disorder increased by almost 20% in the last 10 years.2 Given this scenario, MDD is now the leading cause of disability worldwide.

Despite the high prevalence of MDD, and the advances obtained in the last years in the comprehension of the neurobiological basis of this disorder, its treatment still represents a challenge. The limitations of the currently available antidepressants are related to their limited efficacy (only approximately 50% of the patients fail to achieve remission), the delayed therapeutic effects and a great number of adverse/side effects, which includes headaches, constipation, weight changes, and mainly sexual dysfunction.1,3,4 These limitations are particularly problematic for patients with elevated risk for suicide. Noteworthy, it is estimated that up to 50% of the 800,000 suicides that occur per year worldwide are associated with MDD, and patients affected by this disorder are almost 20-fold more likely to die by suicide than the general population.1,2,5 Therefore, appropriate and effective treatments are necessary to be established for a better management of this disorder. The most promising therapeutic strategy for this challenge emerged at the beginning of the 21st century, when Berman et al. demonstrated for the first time that the N-methyl-D-aspartate (NMDA) receptor antagonist ketamine caused fast and long-lasting antidepressant effects.6 This study represents the onset of a series of other studies that aimed at investigating the ability of ketamine to provide fast antidepressant responses, even in refractory patients, as well as those that have been focused on the investigation of the mechanisms underlying the fast antidepressant responses of ketamine.711 Despite the promising effects of ketamine, its prolonged use has some limitations, mainly related to side effects and the possibility of neurotoxic effects upon chronic use. In addition to these drawbacks associated with ketamine’s pharmacological/toxicological properties, the oral bioavailability of ketamine is slow.12 Thereby, ketamine is generally administered by intravenous route in hospitalized patients.13

Novel drugs that may afford fast antidepressant responses have been extensively investigated. Here, we provide a brief history and overview of the development of antidepressant drugs, the discovery of ketamine, and novel targets for fast antidepressant responses, particularly the potential role of endogenous neuromodulators.

Beyond Monoamine-Based Therapies

The first hypothesis formulated to explain the neurobiology of MDD postulated that depressive symptoms occur as a consequence of reduced levels of monoamines in the synaptic cleft.14 This assumption was based on serendipitous discoveries. Reserpine, an antihypertensive drug that causes noradrenaline depletion, was reported to cause depressive symptoms.15,16 In parallel to this finding, the role of monoamines in MDD was further supported by discovery of the first antidepressant agents, tricyclic antidepressants (TCAs) and monoamine oxidase inhibitors (MAOIs), which have robust effects on monoaminergic transmission.1 TCAs such as imipramine act by inhibiting the serotonin and noradrenaline reuptake, while MAOIs such as iproniazid inhibit MAO, an enzyme responsible for catabolizing the monoamines serotonin, noradrenaline, and dopamine. These events increase monoamine levels in the synaptic cleft, ultimately resulting in mood improvement in patients with MDD generally three to four weeks after the onset of the treatment.1,17,18 The discovery of the mechanisms of imipramine and iproniazid was a crucial breakthrough for the development of monoaminergic hypothesis of MDD, which initially postulated that MDD could be due to low levels of noradrenaline in the synaptic cleft.14,19 This hypothesis was extended to acknowledge that depressive symptoms may also be related to a deficiency of serotonin in the synaptic cleft in central nervous system (CNS).20,21 These theories were reformulated, and subsequently, the monoaminergic theory was postulated suggesting that patients with MDD present a reduction of monoaminergic neurotransmitters (basically serotonin, noradrenaline, and dopamine) in the synaptic cleft.22

Fluoxetine, a selective serotonin reuptake inhibitors (SSRI) was discovered in 1984 in the Eli Lilly pharmaceutical company and went on sale in 1988 after some clinical reports confirming its efficacy in the MDD, along with the advantage of having fewer adverse/side effects when compared to TCAs and MAOIs.23 In view of the growing need for agents to treat MDD and considering fluoxetine as prototype drug, other SSRIs were developed, but the delayed therapeutic effect is a key limitation of all of these drugs.

Although monoamine-based antidepressant agents reestablish monoamine levels within a few hours after administration, their therapeutic response only occurs lately, rendering the monoaminergic hypothesis of MDD overly simplistic.1,3,24 It has recently been reported that serotonin may be co-released with glutamate in serotonergic neurons, and antidepressant agents appear to affect this mechanism.25 This event is especially pronounced within the raphe nuclei, but not restricted to them. Particularly, acute administration of SSRIs blocks the serotonin transporter, increasing extracellular serotonin levels, which results in the activation of serotonin-1 A autoreceptors (5-HT1A). As a consequence, the release of serotonin and glutamate is decreased in nerve terminals on the presynaptic neuron.26,27 When autoreceptors desensitize, approximately two to three weeks after the onset of SSRIs intake, firing rates are restored, reestablishing the glutamatergic component.28 Interestingly, the restoration of adequate synaptic levels of glutamate may contribute to the strengthening of excitatory synapses29 and may result in antidepressant responses.25

According to the assumption that the monoaminergic system does not fully explain the pathophysiology of MDD and considering that the administration of NMDA receptor antagonists produces an antidepressant-like effect in rodents,30 in 1999, Skolnick proposed that the antidepressants for the new millennium would be based on the glutamatergic system modulation.31 At the beginning of 2000, Berman et al. published a groundbreaking study, which for the first time showed that it was possible to obtain fast (within 4 h) and long-lasting (for up to three days) antidepressant effects.6 Following these findings, the rapid and efficacious antidepressant actions of ketamine were confirmed in a larger double-blind, placebo-controlled study by Zarate et al. that demonstrated a single subanesthetic dose of ketamine produced improvement of depressive symptoms in refractory depressive patients.7 This effect was observed within 110 min and was sustained for up to seven days in most of the patients. A great body of clinical evidence3234 and experimental studies8,9,11,35 have demonstrated the rapid, robust, and sustained antidepressant-like effect elicited by ketamine, largely erasing any doubts on the antidepressant actions of this compound. The pronounced and extremely rapid antidepressant effect of ketamine contrasted with classical monoamine-based pharmacotherapy, which might take until four weeks to present the therapeutic effect.

Mechanisms Underlying the Fast-Acting Antidepressant Effect of Ketamine

The mechanisms of action by which ketamine exerts its rapid effects have been the subject of interest by many research groups, which have prospectively shown that the molecular targets for ketamine’s effects go beyond the antagonism of NMDA receptors.36 Notably, ketamine’s rapid action seems to be triggered by antagonism of NMDA receptors in GABAergic interneurons, preventing the inhibitory action of this system on glutamatergic tonus.37 In turn, glutamatergic neurons release glutamate in the synaptic cleft, which preferentially activates AMPA (alpha-amino-3-hydroxy-methyl-5-4-isoxazole propionic acid) receptors. Subsequently, AMPA receptors stimulation promotes a transient sodium influx that depolarizes the cell and activates the voltage-dependent calcium channels (VDCC).10,37 The calcium entry by VDCC promotes exocytosis of synaptic vesicles containing the brain-derived neurotrophic factor (BDNF) in the synaptic cleft, which in turn, activates tropomyosin receptor kinase B (TrkB).10 Upon activation, TrkB stimulates phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt), which is followed by several events that culminate on the mechanistic target of rapamycin (mTOR) activation.8,9,11,3840

mTOR regulates the initial steps for translation of proteins involved in the formation of new dendritic spines and synaptogenesis. Particularly, mTOR phosphorylates and activates 70-kDa ribosomal protein S6 kinase (p70S6K) and also phosphorylates and inactivates the eukaryotic initiation factor 4E-binding protein (4E-BP) facilitating translation initiation.41,42 Among the proteins that have been functionally linked to the activation of mTOR signaling, stand out postsynaptic density protein-95 kDa (PSD-95), AMPA receptor subunit 1 (GluA1), and synapsin, which are required for the formation, maturation, and function of new synapses.8,9 Given this background, the mTOR-mediated signaling pathway underpins the mechanism of action of fast-acting antidepressants responses. The pharmacological mechanisms that underlie the fast-acting antidepressant effect of ketamine are depicted in Figure 1.

Figure 1.

Figure 1.

The proposed mechanism of action underlying the fast and sustained antidepressant effects of ketamine. It is postulated that ketamine acts antagonizing NMDAR in GABAergic interneurons (a), thereby decreasing inhibitory action of this system on glutamatergic tonus (b). Thus, glutamatergic neurons release glutamate-containing vesicles in the synaptic cleft, which preferentially activates AMPAR, since NMDAR is antagonized by ketamine (c). Upon activation, AMPAR induces a transient sodium influx that depolarizes the cell and activates VDCC, which induces exocytosis of BDNF-containing vesicles. Released BDNF, in turn, activates TrkB receptors. Upon activation, TrkB stimulates signaling pathways, particularly PI3K/Akt/mTOR-mediated pathway. This signaling pathway culminates in the synthesis of synaptic proteins such as synapsin, PSD-95 (which anchors AMPAR), and AMPAR subunit 1 (GluA1), which are inserted to the cell membrane, contributing to synaptogenesis and rapid antidepressant effect of ketamine (d). 4E-BP: eukaryotic initiation factor 4E-binding protein; Akt: protein kinase B; AMPAR: alpha-amino-3-hydroxy-methyl-5-4-isoxazole propionic acid receptor; BDNF: brain-derived neurotrophic factor; GluA1: AMPA receptor subunit 1; GSK-3β: glycogen synthase kinase 3β; mTOR: mechanistic target of rapamycin; NMDAR: N-methyl-D-aspartate receptor; p70S6K: 70 kDa ribosomal protein S6 kinase; PI3K: phosphatidylinositol 3-kinase; PSD-95: postsynaptic density protein-95 kDa; TrkB: tropomyosin receptor kinase B; VDCC: voltage-dependent calcium channels. Figure designed using images from Servier Medical Art and Mind the Graph.

Importantly, a completely different class of glutamate-based rapid-acting antidepressant agents, including NMDA receptor antagonists (CP-101,606/Traxoprodil, MK-0657/Rislenemdaz), glycine-binding site ligands (GLYX-13/Rapastinel), metabotropic glutamate receptor modulators (AZD2066), and other glutamatergic modulators (Riluzole) could have convergent effects on pro-synaptogenesis signaling pathway, like ketamine.43

The Potential Role of Endogenous Neuromodulators as Fast-Acting Antidepressants

Considering the limitation of widespread clinical use of ketamine, the search of compounds that might share similar mechanisms of action to ketamine emerges as a promising therapeutic strategy. Regarding this issue, our research group has focused on the investigation of the possible role of endogenous glutamatergic neuromodulators for fast antidepressant responses, namely agmatine, creatine, guanosine, and ascorbic acid.

Agmatine

Agmatine, an endogenous polyamine, is synthesized from L-arginine in a reaction catalyzed by the enzyme arginine decarboxylase and is catabolized by the enzyme agmatinase that converts agmatine into urea and putrescine.44 Agmatine is an intermediary in the biosynthesis of polyamines, a pathway also related to the synthesis of important neurotransmitters, such as glutamate and GABA. Agmatine is widely distributed in mammalian tissues.45 In the CNS, it is especially present in the cytoplasm in a network of neurons in the rostral brainstem and forebrain.46 Noteworthy, agmatine is postulated to be a neuromodulator47 that is taken up by presynaptic axon terminals, stored in synaptic vesicles (even as with other neurotransmitters such glutamate), and released upon membrane depolarization,4850 similar to classical neurotransmitters. Despite these features, no well-characterized receptor for agmatine was reported yet. The neuroprotective effects of agmatine were shown in a mouse 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinson’s disease51,52 and in cultured cerebellar granule cells and hippocampal cells submitted to glutamate and/or NMDA-induced neurotoxicity53,54 and corticosterone.55 Agmatine also provides neuroprotective effects in a rat model of Alzheimer’s disease induced by β-amyloid peptide (Aβ fragment 25-35)56 and protected against memory impairment induced by streptozotocin57,58 and lipopolysaccharide (LPS).59 The mechanims underlying these neuroprotective effects of agmatine include antiexcitotoxic, antioxidant, antiapoptotic, and pro-survival properties,5159 which may be relevant for the ability of agmatine to afford protection against neurodegenerative and psychiatric diseases.

Notably, increased agmatine concentrations are evident in the serum of MDD patients60 and reduced concentrations of agmatine were shown in the cerebral cortex of suicides,61 suggesting a role of this neuromodulator in the pathophysiology of MDD. In line with this finding, a clinical study showed the antidepressant effect of agmatine, but this study was carried out with only three patients.62 Our research group and others have provided clear evidence that this compound has an antidepressant-like effect.6375 Zomkowski et al. showed the antidepressant-like effects of agmatine in behavioral tests predictive of antidepressant activity in mice, the forced swim test (FST) and tail suspension test (TST).63 Subsequent studies reinforced the antidepressant-like effect of agmatine6468 involving interaction with nitrergic,63,69 serotonergic,65 and opioidergic systems.66 In addition, the antidepressant-like effect elicited by the subchronic treatment with agmatine is dependent on the phosphorylation of protein kinase A (PKA), Akt, glycogen synthase kinase 3β (GSK-3β), and extracellular signal-regulated kinase (ERK1/2), with the subsequent activation of cyclic-AMP responsive-element binding protein (CREB), and transcription of BDNF.70 The antidepressant-like response elicited by agmatine following its subchronic administration was also associated with synaptic proteins expression as well as the maintenance of the astrocytes and microglia integrity.71

The antidepressant-like effect of agmatine appears to involve inhibition of NMDA receptors,69 since agmatine was able to enhance the antidepressant potency of the NMDA receptor antagonist MK-801 for up to 100 fold.72 Subsequent studies investigated whether this compound could present fast-acting antidepressant response. Regarding this issue, Neis et al. demonstrated that the antidepressant-like effect of agmatine in the TST is dependent on the activation of AMPA and TrkB receptors, PI3K/mTOR signaling, and upregulation of synaptic proteins, in a way similar to ketamine.73 Moreover, the acute administration of agmatine at a very low dose by oral route was able to reverse the behavioral alterations induced by chronic unpredictable mild stress74 and by chronic administration of corticosterone75 in mice, suggesting that this compound may have fast-acting antidepressant properties. It is important to mention that these stress-induced models of depression are sensitive only to chronic, but not acute administration of conventional antidepressants. However, a single ketamine administration has been reported to be effective in these models of depression.9,35 Reinforcing the notion that agmatine would have properties similar to ketamine, a single administration of agmatine or ketamine counteracted the depressive-like phenotype of CREB-regulated transcription coactivator 1 (Crtc1) knockout mice in the FST, suggesting that agmatine has a rapid antidepressant-like effect.76 In HT22 hippocampal cell line, the combination of subthreshold concentrations of agmatine and ketamine produced cytoprotective effects against corticosterone-induced cell death by a mechanism dependent on Akt and mTOR/p70S6 kinase signaling pathway activation and increased expression of synaptic proteins.55 Altogether, these findings suggest that agmatine may act as a ketamine-like compound, and further studies are important to investigate whether agmatine is able to afford rapid antidepressant effects in depressive patients. It is of particular interest considering that agmatine may be used even chronically at high doses without producing overt signs of toxicity.77,78

Creatine

Creatine, a supplement frequently used for ergogenic purpose, is widely distributed in mammalian tissues and has the potential to treat or mitigate a broad range of CNS diseases.79 The creatine stores are found mainly in skeletal muscle, although substantial concentrations are also found in the brain.80 Peripherally, the synthesis of this compound occurs initially in kidneys, from the amino acids glycine and L-arginine that undergo a reaction catalyzed by the enzyme L-arginine glycine amidinotransferase, resulting in ornithine and guanidinoacetate.80 Subsequently, guanidinoacetate is transported to the liver where a methyl group from S-adenosyl-L-methionine is transferred to guanidinoacetate, forming creatine in a reaction catalyzed by guanidinoacetate-methyltransferase.80,81 However, creatine is also synthesized in the CNS.82 Noteworthy, creatine has been postulated to act as a neuromodulator synthesized and taken up by central neurons and released in an action-potential dependent manner, modulating various neurotransmitter systems and signaling pathways.35,79,8386 Notably, the neuroprotective effect of creatine was demonstrated by several lines of evidence, particularly in Parkinson’s disease model induced by 6-hydroxydopamine and MPP+,87,88 in neurotoxicity induced by hyperammonemia,89 as well as in glutamate-induced excitotoxicity and Alzheimer’s disease model induced by β-amyloid peptide.90 Furthermore, a growing body of clinical studies have also shown alterations in creatine levels in the brain of patients that exhibit depressive symptoms,9193 suggesting that this neuromodulator could exert an important role in the pathophysiology of MDD.

Remarkably, several preclinical studies reported the antidepressant-like effect of creatine in mice subjected to TST and FST.35,85,86,9497 This response is dependent on the modulation of dopaminergic,94 serotonergic,96 and noradrenergic systems 98 and also on the activation of PKA, protein kinase C (PKC), and mitogen-activated protein kinases (MAPKs).86 Moreover, the coadministration of subeffective doses of creatine and NMDA antagonists MK-801 or ketamine elicited an antidepressant-like response in mice, suggesting a possible modulation of glutamatergic system.95 The antidepressant effect of creatine on β-amyloid-treated mice was also demonstrated, a response associated with GSK-3β/Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway.99

In view of the results which show a similar effect between creatine and ketamine,95,97 subsequent studies investigated whether creatine could be able to stimulate Akt/mTOR-mediated signaling pathway. Notably, creatine was able to increase Akt and mTOR phosphorylation in the hippocampus of mice.85 Reinforcing the notion that creatine modulates PI3K/Akt/mTOR signaling, the antidepressant-like effect of creatine in mice submitted to TST was abolished by PI3K and mTOR inhibitors.35,85 Interestingly, a single creatine administration was able to reverse corticosterone-induced depressive-like behavior, as well as increased mTOR and p70S6K phosphorylation, ultimately leading to increase on PSD-95 immunocontent in the hippocampus.35 These effects were comparable to those results previously reported in the prefrontal cortex of rats administered with ketamine.8,9 This evidence suggests that creatine could share with ketamine the ability to promote a fast antidepressant-like response. However, the antidepressant-like effect of creatine in the TST was not dependent on AMPA receptor activation, as opposed to ketamine.10,95

No evidence until now reports the ability of creatine to enhance the number and function of dendritic spines, an event that has been shown to be crucial for the rapid antidepressant responses. Therefore, future studies are necessary to ascertain the similarities between creatine and ketamine. Noteworthy, several clinical studies have also demonstrated the beneficial effects of creatine in patients with MDD.100105 Particularly, Lyoo et al. showed an improvement in depressive symptoms in 52 patients with MDD that received creatine combined with escitalopram for eight weeks, as early as week 2 of treatment.104 These findings were later reinforced by another study performed by Hellem et al., which showed that 14 patients with MDD in an eight-week open-label trial of daily creatine treatment presented a significant reduction in depressive symptomatology as early as week 2 when compared to baseline scores.105 These studies suggest that creatine could present a faster antidepressant effect when compared to conventional antidepressants, but it remains to be established whether creatine could be able to afford a rapid-onset antidepressant response.

Guanosine

Guanosine is a guanine-based purine that has been recently proposed to be not only an intracellular signaling component but also an extracellular signaling molecule, which regulates important functions in the CNS.106,107 In such a way, the guanosinergic system was postulated as a system in which guanosine could be the molecule with main biological activity.106,108,109 These assumptions were supported by the fact that guanosine is released in the brain under physiological conditions and even more during pathological events, triggering widespread actions in several brain regions.110 The main source of guanosine comes from astrocytes and neurons, which release nucleotides into the extracellular space that are rapidly catabolized by ecto-5′-nucleotidases forming this nucleoside.109,111 Notably, while extracellular adenine-based purines are rapidly metabolized following an insult, guanosine concentration increases progressively, suggesting that it may be an endogenous neuroprotective agent.110

Indeed, a vast number of reports have shown the neuroprotective effect of guanosine against several injuries, including ischemia,112119 sepsis-induced cognitive impairment,120 ammonia intoxication,121 hepatic encephalopathy,122 cytotoxicity induced by MPP+ and 6-hydroxydopamine,123,124 glutamate,125 azide-induced oxidative damage,126 methylmercury,127 and LPS-induced inflammation.128 Furthermore, the neuroprotective effect of guanosine was also demonstrated in animal models of Alzheimer’s 129 and Parkinson’s disease.130 The mechanisms that underlie the neuroprotective effects of guanosine are associated with its ability to attenuate neuroinflammation and oxidative stress as well as to stimulate glutamate uptake.124,125,131133 Moreover, guanosine is supposed to exert its neuroprotective effects by synchronizing distinct signaling pathways such as PI3K/Akt and MAPKs signaling.114,125,134 The neurotrophic effects of guanosine were also reported by several lines of evidence, which have demonstrated that this nucleoside is able to induce proliferation and differentiation, as well as stimulate the neurite arborization and outgrowth.111,135137 These effects may be underpinned by the guanosine’s ability to promote synthesis and release of neurotrophic factors such as nerve growth factor (NGF), transforming growth factor beta (TGF-β), fibroblast growth factor 2 (FGF-2), and BDNF.135142 Despite the abovementioned studies, no receptor for guanosine was characterized until now.143145 However, guanosine is recognized as a glutamatergic neuromodulator.146

Noteworthy, plasma levels of guanosine were reduced in patients with MDD, suggesting that this nucleoside could play a role in the pathophysiology of MDD.147 Accordingly, our research group demonstrated that guanosine produces an antidepressant-like effect in mice submitted to TST and FST.137,148,149 Of note, the PI3K/Akt signaling pathway and its downstream target mTOR seem to be required for the behavioral response of guanosine.148 In view of these findings, one may suppose that the mechanisms underlying the antidepressant-like response of guanosine are, at least in part, similar to those displayed by ketamine, which includes neurotrophic properties, the modulation of glutamatergic transmission, and the ability to stimulate the PI3K/Akt and mTOR-mediated signaling pathways. Noteworthy, reinforcing the notion that guanosine could share the mechanism of action of ketamine, a previous study reported that a single administration of a subeffective dose of guanosine combined with a subeffective dose of ketamine produced an antidepressant-like effect in the TST.148 In addition, a recent study reported the augmentation effect of ketamine by guanosine in the novelty-suppressed feeding test by a mechanism dependent on mTOR signaling pathway.150 Given this scenario, further studies are crucial to understand whether guanosine shares with ketamine a common mechanism of action and could present a fast-acting antidepressant effect.

Ascorbic Acid

Ascorbic acid, also known as vitamin C, is a water-soluble vitamin that occurs physiologically as the ascorbate anion. It exerts antioxidant activity and participates as a coenzyme in the production of proteins such as collagen, as well as in the synthesis of norepinephrine, serotonin, and carnitine.151 This compound is synthetized in the majority of mammals, but humans are not able to synthetize it due to the absence of L-gulono-γ-lactone oxidase, a key enzyme for ascorbate biosynthesis. Therefore, humans should obtain ascorbic acid from foods and dietary supplements.152

Besides its function as a vitamin, ascorbic acid is a neuromodulator that modulates the glutamatergic and dopaminergic systems.153 The first indication that ascorbic acid may exert an antidepressant effect was a case report published in 1980 that showed a reduction in the severity of depressive symptoms in a child with chronic hepatitis and under adrenocorticotropic hormone therapy that received high doses of ascorbic acid.154 Some subsequent clinical studies reinforce its potential as an antidepressant agent. Brody et al. reported that ascorbic acid was effective in decreasing scores on Beck Depression Inventories in healthy young adults.155 In addition, lower depressive symptoms were observed in an elderly population on high dietary intake of vitamin C.156 Pediatric patients treated for six months with fluoxetine and ascorbic acid presented a significant decrease in depressive symptoms when compared to the fluoxetine plus placebo group, further suggesting that ascorbic acid may afford beneficial effects on mood.157 In line with this study, the administration of ascorbic acid with antidepressants decreased depression scores in 22 patients.158 Preclinical studies have also supported the assumption that ascorbic acid may be effective as an antidepressant agent. The first preclinical study that showed that ascorbic acid might elicit antidepressant-like effects was published in 2009 by our research group. The administration of ascorbic acid to mice caused an antidepressant-like effect in TST by a mechanism dependent on the monoaminergic systems.158 In addition, a synergistic antidepressant-like effect was found when ascorbic acid was administered in combination with conventional antidepressants.159 Subsequent studies from our group indicated several targets for the antidepressant-like effects of ascorbic acid: (a) inhibition of NMDA receptors and the L-arginine-NO-cyclic guanosine 3,5-monophosphate pathway;160 (b) inhibition of potassium channels;161 (c) activation of phosphatidylinositol-3 kinase (PI3K) and mTOR signaling pathway, inhibition of GSK-3β, and induction of heme oxygenase-1;162 (d) modulation of GABAA and GABAB receptors;163 (e) activation of the opioid system.164 Moreover, the administration of ascorbic acid elicited antidepressant-like effects in mice subjected to several models of depression, namely chronic unpredictable stress,165 acute restraint stress,166 and administration of the proinflammatory cytokine TNF-α.167 Considering that the antidepressant-like effect of ascorbic acid is associated with the modulation of mTOR signaling pathway,162 the possibility of ascorbic acid exerts a fast antidepressant-like effect in a way similar to ketamine deserves further investigation. Therefore, the characterization of the antidepressant behavioral response provided by ascorbic acid and its ability to modulate hippocampal synaptic plasticity is under investigation in our laboratory.

Conclusions and Future Directions

Although ketamine is able to produce fast-onset responses following a single administration even to severely depressed individuals, its side effects and the possibility of neurotoxicity upon chronic administration have led to the investigation of novel fast-acting antidepressant agents. Our research group has focused on the investigation of endogenous mood modulators that may act as ketamine-like compounds. We provide evidence from preclinical studies that the endogenous glutamatergic neuromodulators agmatine and creatine have antidepressant behavioral profile similar to ketamine, besides presenting the ability to elicit antidepressant response by activating mTOR signaling pathway and/or acutely increasing synaptic proteins and BDNF levels in the hippocampus (creatine and agmatine) and prefrontal cortex (agmatine). Moreover, there is evidence under way in our laboratory indicating that guanosine and ascorbic acid also have the potential to afford antidepressant responses similar to ketamine. Considering that all of these compounds are safe even upon chronic use and exert these effects at very low doses, we consider that they are promising compounds to be tested in clinical studies. A particular interesting approach would be the investigation of the augmentation effect of low doses of ketamine by these compounds in order to provide efficacious fast-acting antidepressant response with lesser side effects.

Acknowledgments

The authors thank Servier Medical Arts and Mind the Graph for providing images for Figures.

Declaration of Conflicting Interests

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding

The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: The authors acknowledge funding from Conselho Nacional de Desenvolvimento Científico e Tecnológico (#310113/2017-2) and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior. Ana Lúcia S. Rodrigues is a recipient of CNPq Research Productivity Fellowship.

References

  • 1.Otte C, Gold SM, Penninx BW, et al. Major depressive disorder. Nat Publ Gr 2016; 2: 1–21. [DOI] [PubMed] [Google Scholar]
  • 2.World Health Organization. Depression and other common mental disorders: Global health estimates. 1st ed. Geneva: World Health Organization, 2017, p. 24.
  • 3.Papakostas GI, Ionescu DF. Towards new mechanisms: an update on therapeutics for treatment-resistant major depressive disorder. Mol Psychiatry 2015; 20(10): 1142–1150. [DOI] [PubMed] [Google Scholar]
  • 4.Wang S-M, Han C, Bahk W-M, et al. Addressing the side effects of contemporary antidepressant drugs: a comprehensive review. Chonnam Med J 2018; 54(2): 101–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Chesney E, Goodwin GM, Fazel S. Risks of all-cause and suicide mortality in mental disorders: a meta-review. World Psychiatry 2014; 13(2): 153–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 2000; 47(4): 351–354. [DOI] [PubMed] [Google Scholar]
  • 7.Zarate CA, Singh JB, Carlson PJ, et al. A randomized trial of an n-methyl-d-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry 2006; 63(8): 856–864. [DOI] [PubMed] [Google Scholar]
  • 8.Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science 2010; 329(5994): 959–964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Li N, Liu RJ, Dwyer JM, et al. Glutamate N-methyl-D-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits caused by chronic stress exposure. Biol Psychiatry 2011; 69(8): 754–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lepack AE, Fuchikami M, Dwyer JM, Banasr M, Duman RS. BDNF release is required for the behavioral actions of ketamine. Int J Neuropsychopharmacol 2014; 18(1): 1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zhou W, Wang N, Yang C, Li XM, Zhou ZQ, Yang JJ. Ketamine-induced antidepressant effects are associated with AMPA receptors-mediated upregulation of mTOR and BDNF in rat hippocampus and prefrontal cortex. Eur Psychiatry 2014; 29(7): 419–423. [DOI] [PubMed] [Google Scholar]
  • 12.Gao M, Rejaei D, Liu H. Ketamine use in current clinical practice. Acta Pharmacol Sin 2016; 37(7): 865–872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zanos P, Moaddel R, Morris P, et al. Ketamine and ketamine metabolite pharmacology: insights into therapeutic mechanisms. Pharmacol Rev 2018; 70(3): 621–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Schildkraut JJ. The catecholamine hypothesis of affective disorders: a review of supporting evidence. Am J Psychiatry 1965; 122(5): 509–522. [DOI] [PubMed] [Google Scholar]
  • 15.Freis ED. Mental depression in hypertensive patients treated for long periods with large doses of reserpine. N Engl J Med 1954; 252(25): 1006–1008. [DOI] [PubMed] [Google Scholar]
  • 16.Celano CM, Freudenreich O, Fernandez-Robles C, Stern TA, Caro MA, Huffman JC. Depressogenic effects of medications: a review. Dialogues Clin Neurosci 2011; 13(1): 109–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Berton O, Nestler EJ. New approaches to antidepressant drug discovery: beyond monoamines. Nat Rev Neurosci 2006; 7(2): 137–151. [DOI] [PubMed] [Google Scholar]
  • 18.Finberg JPM. Update on the pharmacology of selective inhibitors of MAO-A and MAO-B: focus on modulation of CNS monoamine neurotransmitter release. Pharmacol Ther 2014; 143(2): 133–152. [DOI] [PubMed] [Google Scholar]
  • 19.Schildkraut JJ, Gordon EK, Durell J. Catecholamine metabolism in affective disorders. I. Normetanephrine and VMA excretion in depressed patients treated with imipramine. J Psychiatr Res 1965; 3(4): 213–228. [DOI] [PubMed] [Google Scholar]
  • 20.Coppen A. The biochemistry of affective disorders. Br J Psychiatry 1967; 113(504): 1237–1264. [DOI] [PubMed] [Google Scholar]
  • 21.Lapin IP, Oxenkrug GF. Intensification of the central serotoninergic processes as a possible determinant of the thymoleptic effect. Lancet 1969; 7586(1): 132–136. [DOI] [PubMed] [Google Scholar]
  • 22.Delgado PL. Depression: the case for a monoamine deficiency. J Clin Psychiatry 2000; 61(6): 7–11. [PubMed] [Google Scholar]
  • 23.Montgomery AM. The efficacy of fluoxetine as an antidepressant in the short and long term. Int Clin Psychopharmacol 1989; 4(1): 113–119. [PubMed] [Google Scholar]
  • 24.Crisafulli C, Fabbri C, Porcelli S, et al. Pharmacogenetics of antidepressants. Front Pharmacol 2011; 2: 1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Fischer AG, Jocham G, Ullsperger M. Dual serotonergic signals: a key to understanding paradoxical effects? Trends Cogn Sci 2015; 19(1): 21–26. [DOI] [PubMed] [Google Scholar]
  • 26.Scuvée-Moreau JJ, Dresse AE. Effect of various antidepressant drugs on the spontaneous firing rate of locu coeruleus and dorsal raphe neurons of the rat. Eur J Pharmacol 1979; 57(2–3): 219–225. [DOI] [PubMed] [Google Scholar]
  • 27.Hajós M, Gartside SE, Sharp T. Inhibition of median and dorsal raphe neurones following administration of the selective serotonin reuptake inhibitor paroxetine. Naunyn Schmiedebergs Arch Pharmacol 1995; 351(6): 624–629. [DOI] [PubMed] [Google Scholar]
  • 28.El Mansari M, Sánchez C, Chouvet G, Renaud B, Haddjeri N. Effects of acute and long-term administration of escitalopram and citalopram on serotonin neurotransmission: an in vivo electrophysiological study in rat brain. Neuropsychopharmacology 2005; 30(7): 1269–1277. [DOI] [PubMed] [Google Scholar]
  • 29.Thompson SM, Kallarackal AJ, Kvarta MD, Van Dyke AM, LeGates TA, Cai X. An excitatory synapse hypothesis of depression. Trends Neurosci 2015; 38(5): 279–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Trullas R, Skolnick P. Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. Eur J Pharmacol 1990; 185(1): 1–10. [DOI] [PubMed] [Google Scholar]
  • 31.Skolnick P. Antidepressants for the new millennium. Eur J Pharmacol 1999; 375: 31–40. [DOI] [PubMed] [Google Scholar]
  • 32.Price RB, Nock MK, Charney DS, Mathew SJ. Effects of intravenous ketamine on explicit and implicit measures of suicidality in treatment-resistant depression. Biol Psychiatry 2009; 66(5): 522–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.DiazGranados N, Ibrahim L, Brutsche N, et al. Rapid resolution of suicidal ideation after a single infusion of an NMDA antagonist in patients with treatment-resistant major depressive disorder. J Clin Psychiatry 2010; 71(12): 1605–1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Abdallah CG, Dutta A, Averill CL, et al. Ketamine, but not the NMDAR antagonist lanicemine, increases prefrontal global connectivity in depressed patients. Chronic Stress 2018; 2: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Pazini FL, Cunha MP, Rosa JM, et al. Creatine, similar to ketamine, counteracts depressive-like behavior induced by corticosterone via PI3K/Akt/mTOR pathway. Mol Neurobiol 2016; 53(10): 6818–6834. [DOI] [PubMed] [Google Scholar]
  • 36.Pereira VS, Hiroaki-Sato A. A brief history of antidepressant drug development: from tricyclics to beyond ketamine. Acta Neuropsychiatr 2018; 30: 307–322. [DOI] [PubMed] [Google Scholar]
  • 37.Abdallah CG, Adams TG, Kelmendi B, Esterlis I, Sanacora G, Krystal JH. Ketamine’s mechanism of action: a path to rapid-acting antidepressants. Depress Anxiety 2016; 33(8): 689–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Duman RS, Voleti B. Signaling pathways underlying the pathophysiology and treatment of depression: novel mechanisms for rapid-acting agents. Trends Neurosci 2012; 35(1): 47–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Liu WX, Wang J, Xie ZM, et al. Regulation of glutamate transporter 1 via BDNF-TrkB signaling plays a role in the anti-apoptotic and antidepressant effects of ketamine in chronic unpredictable stress model of depression. Psychopharmacology 2016; 233(3): 405–415. [DOI] [PubMed] [Google Scholar]
  • 40.Zhang WJ, Wang HH, Lv YD, Liu CC, Sun WY, Tian LJ. Downregulation of Egr-1 expression level via GluN2B underlies the antidepressant effects of ketamine in a chronic unpredictable stress animal model of depression. Neuroscience 2018; 372: 38–45. [DOI] [PubMed] [Google Scholar]
  • 41.Fingar DC, Richardson CJ, Tee AR, Cheatham L, Tsou C, Blenis J. mTOR controls cell cycle progression through its cell growth effectors S6K1 and 4E-BP1 / eukaryotic translation initiation factor 4E. Mol Cell Biol 2004; 24(1): 200–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hoeffer CA, Klann E. mTOR signaling: at the crossroads of plasticity, memory and disease. Trends Neurosci 2010; 33(2): 67–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kadriu B, Musazzi L, Henter ID, Graves M, Popoli M, Zarate CA. Glutamatergic neurotransmission: pathway to developing novel rapid-acting antidepressant treatments. Int J Neuropsychopharmacol 2019; 22(2): 119–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Reis DJ, Regunathan S. Is agmatine a novel neurotransmitter in brain? Trends Pharmacol Sci 2000; 21(5): 187–193. [DOI] [PubMed] [Google Scholar]
  • 45.Raasch W, Regunathan S, Li G, Reis DJ. Agmatine, the bacterial amine, is widely distributed in mammalian tissues. Life Sci 1995; 56(26): 2319–2330. [DOI] [PubMed] [Google Scholar]
  • 46.Otake K, Ruggiero DA, Regunathan S, Wang H, Milner TA, Reis DJ. Regional localization of agmatine in the rat brain: an immunocytochemical study. Brain Res 1998; 787(1): 1–14. [DOI] [PubMed] [Google Scholar]
  • 47.Freitas AE, Neis VB, Rodrigues ALS. Agmatine, a potential novel therapeutic strategy for depression. Eur Neuropsychopharmacol 2016; 26(12): 1885–1899. [DOI] [PubMed] [Google Scholar]
  • 48.Goracke-Postle CJ, Overland AC, Riedl MS, Stone LS, Fairbanks CA. Potassium- and capsaicin-induced release of agmatine from spinal nerve terminals. J Neurochem 2007; 102(6): 1738–1748. [DOI] [PubMed] [Google Scholar]
  • 49.Reis DJ, Regunathan S. Agmatine: an endogenous ligand at imidazoline receptors is a novel neurotransmittera. Ann N Y Acad Sci Title 1999; 21: 65–80. [DOI] [PubMed] [Google Scholar]
  • 50.Seo S, Liu P, Leitch B. Spatial learning-induced accumulation of agmatine and glutamate at hippocampal CA1 synaptic terminals. Neuroscience 2011; 192: 28–36. [DOI] [PubMed] [Google Scholar]
  • 51.Gilad GM, Gilad VH, Finberg JPM, Rabey JM. Neurochemical evidence for agmatine modulation of 1-methyl-4-phenyl-1,2,3, 6-tetrahydropyridine (MPTP) neurotoxicity. Neurochem Res 2005; 30(6–7): 713–719. [DOI] [PubMed] [Google Scholar]
  • 52.Matheus FC, Aguiar AS, Castro AA, et al. Neuroprotective effects of agmatine in mice infused with a single intranasal administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Behav Brain Res 2012; 235(2): 263–272. [DOI] [PubMed] [Google Scholar]
  • 53.Olmos G, DeGregorio-Rocasolano N, Regalado MP, et al. Protection by-imidazol(ine) drugs and agmatine of glutamate-induced neurotoxicity in cultured cerebellar granule cells through blockade of NMDA receptor. Br J Pharmacol 1999; 127(6): 1317–1326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wang WP, Iyo AH, Miguel-Hidalgo J, Regunathan S, Zhu MY. Agmatine protects against cell damage induced by NMDA and glutamate in cultured hippocampal neurons. Brain Res. 2006; 1084(1): 210–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Tavares MK, dos Reis S, Platt N, et al. Agmatine potentiates neuroprotective effects of subthreshold concentrations of ketamine via mTOR/S6 kinase signaling pathway. Neurochem Int 2018; 118: 275–285. [DOI] [PubMed] [Google Scholar]
  • 56.Bergin DH, Liu P. Agmatine protects against β-amyloid 25-35-induced memory impairments in the rat. Neuroscience 2010; 169(2): 794–811. [DOI] [PubMed] [Google Scholar]
  • 57.Bhutada P, Mundhada Y, Humane V, et al. Agmatine, an endogenous ligand of imidazoline receptor protects against memory impairment and biochemical alterations in streptozotocin-induced diabetic rats. Prog Neuro-Psychopharmacology Biol Psychiatry 2012; 37(1): 96–105. [DOI] [PubMed] [Google Scholar]
  • 58.Moosavi M, Zarifkar AH, Farbood Y, Dianat M, Sarkaki A, Ghasemi R. Agmatine protects against intracerebroventricular streptozotocin-induced water maze memory deficit, hippocampal apoptosis and Akt/GSK3β signaling disruption. Eur J Pharmacol 2014; 736: 107–114. [DOI] [PubMed] [Google Scholar]
  • 59.Zarifkar A, Choopani S, Ghasemi R, et al. Agmatine prevents LPS-induced spatial memory impairment and hippocampal apoptosis. Eur J Pharmacol 2010; 634(1–3): 84–88. [DOI] [PubMed] [Google Scholar]
  • 60.Halaris A, Zhu H, Feng Y, Piletz JE. Plasma agmatine and platelet imidazoline receptors in depression. Ann. N. Y. Acad. Sci 1999; 881: 445–451. [DOI] [PubMed] [Google Scholar]
  • 61.Chen GG, Almeida D, Fiori L, Turecki G. Evidence of reduced agmatine concentrations in the cerebral cortex of suicides. Int J Neuropsychopharmacol 2018; 21(10): 895–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Shopsin B. The clinical antidepressant effect of exogenous agmatine is not reversed by parachlorophenylalanine: a pilot study. Acta Neuropsychiatr 2013; 25(2): 113–118. [DOI] [PubMed] [Google Scholar]
  • 63.Zomkowski ADE, Hammes L, Lin J, Calixto JB, Santos ARS, Rodrigues ALS. Agmatine produces antidepressant-like effects in two models of depression in mice. Neuroreport 2002; 13(4): 387–391. [DOI] [PubMed] [Google Scholar]
  • 64.Li YF, Gong ZH, Cao JB, Wang HL, Luo ZP, Li J. Antidepressant-like effect of agmatine and its possible mechanism. Eur J Pharmacol 2003; 469(1–3): 81–88. [DOI] [PubMed] [Google Scholar]
  • 65.Zomkowski ADE, Rosa AO, Lin J, Santos ARS, Calixto JB, Rodrigues ALS. Evidence for serotonin receptor subtypes involvement in agmatine antidepressant like-effect in the mouse forced swimming test. Brain Res 2004; 1023(2): 253–263. [DOI] [PubMed] [Google Scholar]
  • 66.Zomkowski ADE, Santos ARS, Rodrigues ALS. Evidence for the involvement of the opioid system in the agmatine antidepressant-like effect in the forced swimming test. Neurosci Lett 2005; 381(3): 279–283. [DOI] [PubMed] [Google Scholar]
  • 67.Zeidan MP, Zomkowski ADE, Rosa AO, Rodrigues ALS, Gabilan NH. Evidence for imidazoline receptors involvement in the agmatine antidepressant-like effect in the forced swimming test. Eur J Pharmacol 2007; 565(1–3): 125–131. [DOI] [PubMed] [Google Scholar]
  • 68.Budni J, Gadotti VM, Kaster MP, Santos ARS, Rodrigues ALS. Involvement of different types of potassium channels in the antidepressant-like effect of tramadol in the mouse forced swimming test. Eur J Pharmacol 2007; 575: 87–93. [DOI] [PubMed] [Google Scholar]
  • 69.Neis VB, Manosso LM, Moretti M, Freitas AE, Daufenbach J, Rodrigues ALS. Depressive-like behavior induced by tumor necrosis factor-α is abolished by agmatine administration. Behav Brain Res 2014; 261: 336–344. [DOI] [PubMed] [Google Scholar]
  • 70.Freitas AE, Bettio LEB, Neis VB, et al. Sub-chronic agmatine treatment modulates hippocampal neuroplasticity and cell survival signaling pathways in mice. J Psychiatr Res 2014; 58: 137–146. [DOI] [PubMed] [Google Scholar]
  • 71.Freitas AE, Egea J, Buendia I, et al. Agmatine, by improving neuroplasticity markers and inducing Nrf2, prevents corticosterone-induced depressive-like behavior in mice. Mol Neurobiol 2016; 53(5): 3030–3045. [DOI] [PubMed] [Google Scholar]
  • 72.Neis VB, Moretti M, Manosso LM, Lopes MW, Leal RB, Rodrigues ALS. Agmatine enhances antidepressant potency of MK-801 and conventional antidepressants in mice. Pharmacol Biochem Behav 2015; 130: 9–14. [DOI] [PubMed] [Google Scholar]
  • 73.Neis VB, Moretti M, Bettio LEB, et al. Agmatine produces antidepressant-like effects by activating AMPA receptors and mTOR signaling. Eur Neuropsychopharmacol 2016; 26(6): 959–971. [DOI] [PubMed] [Google Scholar]
  • 74.Neis VB, Bettio LEB, Moretti M, et al. Acute agmatine administration, similar to ketamine, reverses depressive-like behavior induced by chronic unpredictable stress in mice. Pharmacol Biochem Behav 2016; 150–151: 108–114. [DOI] [PubMed] [Google Scholar]
  • 75.Neis VB, Bettio LB, Moretti M, et al. Single administration of agmatine reverses the depressive-like behavior induced by corticosterone in mice: comparison with ketamine and fluoxetine. Pharmacol Biochem Behav 2018; 173(April): 44–50. [DOI] [PubMed] [Google Scholar]
  • 76.Meylan EM, Breuillaud L, Seredenina T, et al. Involvement of the agmatinergic system in the depressive-like phenotype of the Crtc1 knockout mouse model of depression. Transl Psychiatry 2016; 6(7): e852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Keynan O, Mirovsky Y, Dekel S, Gilad VH, Gilad GM. Safety and efficacy of dietary agmatine sulfate in lumbar disc-associated radiculopathy. An open-label, dose-escalating study followed by a randomized, double-blind, placebo-controlled trial. Pain Med 2010; 11(3): 356–368. [DOI] [PubMed] [Google Scholar]
  • 78.Neis VB, Rosa PB, Olescowicz G, Rodrigues ALS. Therapeutic potential of agmatine for CNS disorders. Neurochem Int 2017; 108: 318–331. [DOI] [PubMed] [Google Scholar]
  • 79.Pazini FL, Cunha MP, Rodrigues ALS. The possible beneficial effects of creatine for the management of depression. Prog Neuro-Psychopharmacology Biol Psychiatry 2019; 89: 193–206. [DOI] [PubMed] [Google Scholar]
  • 80.Wallimann T, Harris R. Creatine: a miserable life without it. Amino Acids 2016; 48(8): 1739–1750. [DOI] [PubMed] [Google Scholar]
  • 81.Defalco AJ, Davies RK. The synthesis of creatine by the brain of the intact rat. J Neurochem 1961; 7(4): 308–312. [DOI] [PubMed] [Google Scholar]
  • 82.Van Pilsum JF, Stephens GC, Taylor D. Distribution of creatine, guanidinoacetate and the enzymes for their biosynthesis in the animal kingdom. Implications for phylogeny. Biochem J 1972; 126(2): 325–345. [PubMed] [Google Scholar]
  • 83.Almeida LS, Salomons GS, Hogenboom F, Jakobs C, Schoffelmer ANM. Exocytotic release of creatine in rat brain. Synapse 2006; 60: 118–123. [DOI] [PubMed] [Google Scholar]
  • 84.Bothwell JH, Styles P, Bhakoo KK. Swelling-activated taurine and creatine effluxes from rat cortical astrocytes are pharmacologically distinct. J Membr Biol 2002; 185(2): 157–164. [DOI] [PubMed] [Google Scholar]
  • 85.Cunha MP, Budni J, Ludka FK, et al. Involvement of PI3K/Akt signaling pathway and its downstream intracellular targets in the antidepressant-like effect of creatine. Mol Neurobiol 2016; 53(5): 2954–2968. [DOI] [PubMed] [Google Scholar]
  • 86.Cunha MP, Budni J, Pazini FL, et al. Involvement of PKA, PKC, CAMK-II and MEK1/2 in the acute antidepressant-like effect of creatine in mice. Pharmacol Rep 2014; 66(4): 653–659. [DOI] [PubMed] [Google Scholar]
  • 87.Cunha MP, Martín-de-Saavedra MD, Romero A, et al. Protective effect of creatine against 6-hydroxydopamine-induced cell death in human neuroblastoma SH-SY5Y cells: involvement of intracellular signaling pathways. Neuroscience 2013; 238: 185–194. [DOI] [PubMed] [Google Scholar]
  • 88.Cunha MP, Pazini FL, Lieberknecht V, et al. MPP + -Lesioned mice: an experimental model of motor, emotional, memory/learning, and striatal neurochemical dysfunctions. Mol Neurobiol 2017; 54(8): 6356–6377. [DOI] [PubMed] [Google Scholar]
  • 89.Braissant O. Ammonia toxicity to the brain: effects on creatine metabolism and transport and protective roles of creatine. Mol Genet Metab 2010; 100: S53–S58. [DOI] [PubMed] [Google Scholar]
  • 90.Brewer GJ, Wallimann TW. Protective effect of the energy precursor creatine against toxicity of glutamate and β-amyloid in rat hippocampal neurons. J Neurochem 2000; 74(5): 1968–1978. [DOI] [PubMed] [Google Scholar]
  • 91.O’Neill J, Lai TM, Sheen C, et al. Cingulate and thalamic metabolites in obsessive-compulsive disorder. Psychiatry Res Neuroimaging 2016; 254: 34–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Venkatraman TN, Krishnan RR, Steffens DC, Song AW, Taylor WD. Biochemical abnormalities of the medial temporal lobe and medial prefrontal cortex in late-life depression. Psychiatry Res Neuroimaging 2009; 172(1): 49–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Nery FG, Stanley JA, Chen HH, et al. Normal metabolite levels in the left dorsolateral prefrontal cortex of unmedicated major depressive disorder patients: a single voxel 1H spectroscopy study. Psychiatry Res Neuroimaging 2009; 174(3): 177–183. [DOI] [PubMed] [Google Scholar]
  • 94.Cunha MP, MacHado DG, Capra JC, Jacinto J, Bettio LEB, Rodrigues ALS. Antidepressant-like effect of creatine in mice involves dopaminergic activation. J Psychopharmacol 2012; 26(11): 1489–1501. [DOI] [PubMed] [Google Scholar]
  • 95.Cunha MP, Pazini FL, Ludka FK, et al. The modulation of NMDA receptors and l-arginine/nitric oxide pathway is implicated in the anti-immobility effect of creatine in the tail suspension test. Amino Acids 2015; 47(4): 795–811. [DOI] [PubMed] [Google Scholar]
  • 96.Cunha MP, Pazini FL, Oliveira A, Machado DG, Rodrigues ALS. Evidence for the involvement of 5-HT1A receptor in the acute antidepressant-like effect of creatine in mice. Brain Res Bull 2013; 95: 61–69. [DOI] [PubMed] [Google Scholar]
  • 97.Cunha MP, Pazini FL, Rosa JM, et al. Creatine, similarly to ketamine, affords antidepressant-like effects in the tail suspension test via adenosine A1and A2Areceptor activation. Purinergic Signal 2015; 11(2): 215–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Cunha MP, Pazini FL, Oliveira Á, et al. The activation of α1-adrenoceptors is implicated in the antidepressant-like effect of creatine in the tail suspension test. Prog Neuro-Psychopharmacology Biol Psychiatry 2013; 44: 39–50. [DOI] [PubMed] [Google Scholar]
  • 99.Rosa JM, Pazini FL, Cunha MP, et al. Antidepressant effects of creatine on amyloid β1–40-treated mice: the role of GSK-3β/Nrf2 pathway. Prog Neuro-Psychopharmacol Biol Psychiatry 2018; 86: 270–278. [DOI] [PubMed] [Google Scholar]
  • 100.Amital D, Vishne T, Rubinow A, Levine J. Observed effects of creatine monohydrate in a patient with depression and fibromyalgia. Am J Psychiatry 2006; 163(10): 1840–1841. [DOI] [PubMed] [Google Scholar]
  • 101.Roitman S, Green T, Sdher Y, Karni N, Levine J. Creatine monohydrate in resistant depression: a preliminary study. Bipolar Disord 2007; 9: 754–758. [DOI] [PubMed] [Google Scholar]
  • 102.Kondo DG, Sung YH, Hellem TL, et al. Open-label adjunctive creatine for female adolescents with SSRI-resistant major depressive disorder: a 31-phosphorus magnetic resonance spectroscopy study. J Affect Disord 2011; 135(1–3): 354–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Kondo DG, Forrest LN, Shi X, et al. Creatine target engagement with brain bioenergetics: a dose-ranging phosphorus-31 magnetic resonance spectroscopy study of adolescent females with SSRI-resistant depression. Amino Acids 2016; 48(8): 1941–1954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lyoo IK, Yoon S, Kim TS, et al. A randomized, double-blind placebo-controlled trial of oral creatine monohydrate augmentation for enhanced response to a selective serotonin reuptake inhibitor in women with major depressive disorder. Am J Psychiatry 2012; 169(9): 937–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hellem TL, Sung YH, Shi XF, et al. Creatine as a novel treatment for depression in females using methamphetamine: a pilot study. J Dual Diagn 2015; 11: 189–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Bettio LEB, Gil-Mohapel J, Rodrigues ALS. Guanosine and its role in neuropathologies. Purinergic Signal 2016; 12(3): 411–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Bettio LEB, Gil-Mohapel J, Rodrigues ALS. Current perspectives on the antidepressant-like effects of guanosine. Neural Regen Res 2016; 11(9): 1411–1413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lanznaster D, Dal-Cim T, Piermartiri TCB, Tasca CI. Guanosine: a neuromodulator with therapeutic potential in brain disorders. Aging Dis 2016; 7(5): 657–679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Schmidt AP, Lara DR, Souza DO. Proposal of a guanine-based purinergic system in the mammalian central nervous system. Pharmacol Ther 2007; 116(3): 401–416. [DOI] [PubMed] [Google Scholar]
  • 110.Ciccarelli R, Di Iorio P, Giuliani P, et al. Rat cultured astrocytes release guanine-based purines in basal conditions and after hypoxia/hypoglycemia. Glia 1999; 25(1): 93–98. [PubMed] [Google Scholar]
  • 111.Ciccarelli R, Ballerini P, Sabatino G, et al. Involvement of astrocytes in purine-mediated reparative processes in the brain. Int J Dev Neurosci 2001; 19(4): 395–414. [DOI] [PubMed] [Google Scholar]
  • 112.Oleskovicz SPB, Martins WC, Leal RB, Tasca CI. Mechanism of guanosine-induced neuroprotection in rat hippocampal slices submitted to oxygen-glucose deprivation. Neurochem Int 2008; 52(3): 411–418. [DOI] [PubMed] [Google Scholar]
  • 113.Dal-Cim T, Martins WC, Santos ARS, Tasca CI. Guanosine is neuroprotective against oxygen/glucose deprivation in hippocampal slices via large conductance Ca2+-activated K+channels, phosphatidilinositol-3 kinase/protein kinase B pathway activation and glutamate uptake. Neuroscience 2011; 183: 212–220. [DOI] [PubMed] [Google Scholar]
  • 114.Dal-Cim T, Ludka FK, Martins WC, et al. Guanosine controls inflammatory pathways to afford neuroprotection of hippocampal slices under oxygen and glucose deprivation conditions. J Neurochem 2013; 126(4): 437–450. [DOI] [PubMed] [Google Scholar]
  • 115.Dal-Cim T, Martins WC, Thomaz DT, et al. Neuroprotection promoted by guanosine depends on glutamine synthetase and glutamate transporters activity in hippocampal slices subjected to oxygen/glucose deprivation. Neurotox Res 2016; 29(4): 460–468. [DOI] [PubMed] [Google Scholar]
  • 116.Hansel G, Ramos DB, Delgado CA, et al. The potential therapeutic effect of guanosine after cortical focal ischemia in rats. PLoS One 2014; 9(2): 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Hansel G, Tonon AC, Guella FL, et al. Guanosine protects against cortical focal ischemia. Involvement of inflammatory response. Mol Neurobiol. 2015; 52(3): 1791–1803. [DOI] [PubMed] [Google Scholar]
  • 118.Ramos DB, Muller GC, Rocha GBM, et al. Intranasal guanosine administration presents a wide therapeutic time window to reduce brain damage induced by permanent ischemia in rats. Purinergic Signal 2016; 12(1): 149–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Thomaz DT, Dal-Cim TA, Martins WC, et al. Guanosine prevents nitroxidative stress and recovers mitochondrial membrane potential disruption in hippocampal slices subjected to oxygen/glucose deprivation. Purinergic Signal 2016; 12(4): 707–718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Petronilho F, Périco SR, Vuolo F, et al. Protective effects of guanosine against sepsis-induced damage in rat brain and cognitive impairment. Brain Behav Immun 2012; 26(6): 904–910. [DOI] [PubMed] [Google Scholar]
  • 121.Cittolin-Santos GF, de Assis AM, Guazzelli PA, et al. Guanosine exerts neuroprotective effect in an experimental model of acute ammonia intoxication. Mol Neurobiol 2017; 54(5): 3137–3148. [DOI] [PubMed] [Google Scholar]
  • 122.Paniz LG, Calcagnotto ME, Pandolfo P, et al. Neuroprotective effects of guanosine administration on behavioral, brain activity, neurochemical and redox parameters in a rat model of chronic hepatic encephalopathy. Metab Brain Dis 2014; 29(3): 645–654. [DOI] [PubMed] [Google Scholar]
  • 123.Pettifer KM, Jiang S, Bau C, et al. MPP+-induced cytotoxicity in neuroblastoma cells: antagonism and reversal by guanosine. Purinergic Signal 2007; 3(4): 399–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Marques NF, Massari CM, Tasca CI. Guanosine protects striatal slices against 6-OHDA-induced oxidative damage, mitochondrial dysfunction, and ATP depletion. Neurotox Res 2019; 35(2): 475–483. [DOI] [PubMed] [Google Scholar]
  • 125.Molz S, Dal-Cim T, Budni J, et al. Neuroprotective effect of guanosine against glutamate-induced cell death in rat hippocampal slices is mediated by the phosphatidylinositol-3 kinase/Akt/ glycogen synthase kinase 3β pathway activation and inducible nitric oxide synthase inhibition. J Neurosci Res 2011; 89(9): 1400–1408. [DOI] [PubMed] [Google Scholar]
  • 126.Quincozes-Santos A, Bobermin LD, Souza DG, Bellaver B, Gonçalves CA, Souza DO. Guanosine protects C6 astroglial cells against azide-induced oxidative damage: a putative role of heme oxygenase 1. J Neurochem 2014; 130(1): 61–74. [DOI] [PubMed] [Google Scholar]
  • 127.Roos DH, Puntel RL, Santos MM, et al. Guanosine and synthetic organoselenium compounds modulate methylmercury-induced oxidative stress in rat brain cortical slices: involvement of oxidative stress and glutamatergic system. Toxicol Vitr 2009; 23(2): 302–307. [DOI] [PubMed] [Google Scholar]
  • 128.Bellaver B, Souza DG, Bobermin LD, Gonçalves CA, Souza DO, Quincozes-Santos A. Guanosine inhibits LPS-induced pro-inflammatory response and oxidative stress in hippocampal astrocytes through the heme oxygenase-1 pathway. Purinergic Signal 2015; 11(4): 571–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lanznaster D, Mack JM, Coelho V, et al. Guanosine prevents anhedonic-like behavior and impairment in hippocampal glutamate transport following amyloid-β1–40 administration in mice. Mol Neurobiol 2017; 54: 5482–5496. [DOI] [PubMed] [Google Scholar]
  • 130.Massari CM, López-Cano M, Núñez F, Fernández-Dueñas V, Tasca CI, Ciruela F. Antiparkinsonian efficacy of guanosine in rodent models of movement disorder. Front Pharmacol 2017; 8: 4–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Frizzo ME, Schwalm FD, Frizzo JK, Soares FA, Souza DO. Guanosine enhances glutamate transport capacity in brain cortical slices. Cell Mol Neurobiol 2005; 25(5): 913–921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Moretto MB, Boff B, Lavinsky D, et al. Importance of schedule of administration in the therapeutic efficacy of guanosine: early intervention after injury enhances glutamate uptake in model of hypoxia-ischemia. J Mol Neurosci 2009; 38(2): 216–219. [DOI] [PubMed] [Google Scholar]
  • 133.Vinadé ER, Schmidt AP, Frizzo MES, et al. Effects of chronic administered guanosine on behavioral parameters and brain glutamate uptake in rats. J Neurosci Res 2005; 79(1–2): 248–253. [DOI] [PubMed] [Google Scholar]
  • 134.Dal-Cim T, Molz S, Egea J, et al. Guanosine protects human neuroblastoma SH-SY5Y cells against mitochondrial oxidative stress by inducing heme oxigenase-1 via PI3K/Akt/GSK-3β pathway. Neurochem Int 2012; 61(3): 397–404. [DOI] [PubMed] [Google Scholar]
  • 135.Bau C, Middlemiss PJ, Hindley S, et al. Guanosine stimulates neurite outgrowth in PC12 cells via activation of heme oxygenase and cyclic GMP. Purinergic Signal 2005; 1(2): 161–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Rathbone M, Pilutti L, Caciagli F, Jiang S. Neurotrophic effects of extracellular guanosine. Nucleosides, Nucleotides and Nucleic Acids 2008; 27(6–7): 666–672. [DOI] [PubMed] [Google Scholar]
  • 137.Bettio LEB, Neis VB, Pazini FL, et al. The antidepressant-like effect of chronic guanosine treatment is associated with increased hippocampal neuronal differentiation. Eur J Neurosci 2016; 43(8): 1006–1015. [DOI] [PubMed] [Google Scholar]
  • 138.Middlemiss PJ, Gysbers JW, Rathbone MP. Extracellular guanosine and guanosine-5′-triphosphate increase: NGF synthesis and release from cultured mouse neopallial astrocytes. Brain Res 1995; 677(1): 152–156. [DOI] [PubMed] [Google Scholar]
  • 139.Gysbers J, Rathbone M. Neurite outgrowth in PC12 cells is enhanced by guanosine through both cAMP-dependent and -independent mechanisms. Neurosci Lett 1996; 220(3): 175–178. [DOI] [PubMed] [Google Scholar]
  • 140.Gysbers J, Rathbone M. GTP and guanosine synergistically enhance NGF-induced neurite outgrowth from PC12 cells. Int J Dev Neurosci 1996; 14(1): 19–34. [DOI] [PubMed] [Google Scholar]
  • 141.Su C, Elfeki N, Ballerini P, et al. Guanosine improves motor behavior, reduces apoptosis, and stimulates neurogenesis in rats with parkinsonism. J Neurosci Res 2009; 87(3): 617–625. [DOI] [PubMed] [Google Scholar]
  • 142.Su C, Jiang C, Jacobs S, Ullal S, Rathbone MP, Jiang S. Guanosine stimulates neural stem cell proliferation through cAMP-CREB pathway. J Biol Regul Homeost Agents 2013; 27(3): 673–680. [PubMed] [Google Scholar]
  • 143.Tasca CI, Cardoso LF, Souza DO. Effects of guanine nucleotides on adenosine and glutamate modulation of cAMP levels in optic tectum slices from chicks. Neurochem Int 1999; 34(3): 213–220. [DOI] [PubMed] [Google Scholar]
  • 144.Traversa U, Bombi G, Di Iorio P, Ciccarelli R, Werstiuk ES, Rathbone MP. Specific [(3)H]-guanosine binding sites in rat brain membranes. Br J Pharmacol 2002; 135(4): 969–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Traversa U, Bombi G, Camaioni E, et al. Rat brain guanosine binding site: biological studies and pseudo-Receptor construction. Bioorganic Med Chem 2003; 11(24): 5417–5425. [DOI] [PubMed] [Google Scholar]
  • 146.Lanznaster D, Tasca CI, Ciruela F, Oliveira KA, Fernández-Dueñas V. Neuromodulatory effects of guanine-based purines in health and disease. Front Cell Neurosci 2018; 12: 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ali-Sisto T, Tolmunen T, Toffol E, et al. Purine metabolism is dysregulated in patients with major depressive disorder. Psychoneuroendocrinology 2016; 70: 25–32. [DOI] [PubMed] [Google Scholar]
  • 148.Bettio LEB, Cunha MP, Budni J, et al. Guanosine produces an antidepressant-like effect through the modulation of NMDA receptors, nitric oxide-cGMP and PI3K/mTOR pathways. Behav Brain Res 2012; 234(2): 137–148. [DOI] [PubMed] [Google Scholar]
  • 149.Bettio LEB, Freitas AE, Neis VB, et al. Guanosine prevents behavioral alterations in the forced swimming test and hippocampal oxidative damage induced by acute restraint stress. Pharmacol Biochem Behav 2014; 127: 7–14. [DOI] [PubMed] [Google Scholar]
  • 150.Camargo A, Pazini FL, Rosa JM, et al. Augmentation effect of ketamine by guanosine in the novelty-suppressed feeding test is dependent on mTOR signaling pathway. J Psychiatr Res 2019; 115: 103–112. [DOI] [PubMed] [Google Scholar]
  • 151.Rice ME. Ascorbate regulation and its neuroprotective role in the brain. Trends Neurosci 2000; 23(5): 209–216. [DOI] [PubMed] [Google Scholar]
  • 152.Moretti M, Fraga DB, Rodrigues ALS. Ascorbic acid to manage psychiatric disorders. CNS Drugs 2017; 31(7): 571–583. [DOI] [PubMed] [Google Scholar]
  • 153.Rebec G V, Pierce C. A vitamin as neuromodulator: ascorbate release into the extracellular fluid of the brain regulates dopaminergic and glutamatergic transmission. Prog Neurobiol. 1994; 43(6): 537–565. [DOI] [PubMed] [Google Scholar]
  • 154.Cocchi P, Silenzi M, Calabri G, Salvi G. Medical treatment of the ductus arteriosus. Pediatrics 1980; 65(4): 862–863. [PubMed] [Google Scholar]
  • 155.Brody S. High-dose ascorbic acid increases intercourse frequency and improves mood: a randomized controlled clinical trial. Biol Psychiatry 2002; 52(4): 371–374. [DOI] [PubMed] [Google Scholar]
  • 156.Oishi J, Doi H, Kawakami N. Nutrition and depressive symptoms in community-dwelling elderly persons in Japan. Acta Med Okayama 2009; 63(1): 9–17. [DOI] [PubMed] [Google Scholar]
  • 157.Amr M, El-Mogy A, Shams T, Vieira K, Lakhan S. Efficacy of vitamin C as an adjunct to fluoxetine therapy in pediatric major depressive disorder. Clin Nutr 2013, pp. 33–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Aburawi SM, Ghambirlou FA, Attumi AA, Altubuly RA, Kara AA. Effect of ascorbic acid on mental depression drug therapy: clinical study. J Psychol Psychother 2014; 4(1): 1–8. [Google Scholar]
  • 159.Binfaré RW, Rosa AO, Lobato KR, Santos ARS, Rodrigues ALS. Ascorbic acid administration produces an antidepressant-like effect: evidence for the involvement of monoaminergic neurotransmission. Prog Neuro-Psychopharmacology Biol Psychiatry 2009; 33(3): 530–540. [DOI] [PubMed] [Google Scholar]
  • 160.Moretti M, Freitas AE, Budni J, Fernandes SC, Balen GO, Rodrigues ALS. Involvement of nitric oxide-cGMP pathway in the antidepressant-like effect of ascorbic acid in the tail suspension test. Behav Brain Res 2011; 225(1): 328–333. [DOI] [PubMed] [Google Scholar]
  • 161.Moretti M, Budni J, Ribeiro CM, Rodrigues ALS. Involvement of different types of potassium channels in the antidepressant-like effect of ascorbic acid in the mouse tail suspension test. Eur J Pharmacol 2012; 687(3): 21–27. [DOI] [PubMed] [Google Scholar]
  • 162.Moretti M, Budni J, Freitas AE, Rosa PB, Rodrigues ALS. Antidepressant-like effect of ascorbic acid is associated with the modulation of mammalian target of rapamycin pathway. J Psychiatr Res 2014; 48(1): 16–24. [DOI] [PubMed] [Google Scholar]
  • 163.Rosa PB, Neis VB, Ribeiro CM, Moretti M, Rodrigues ALS. Antidepressant-like effects of ascorbic acid and ketamine involve modulation of GABAA and GABAB receptors. Pharmacol Reports 2016; 68(5): 996–1001. [DOI] [PubMed] [Google Scholar]
  • 164.Moretti M, Ribeiro CM, Neis VB, Bettio LEB, Rosa PB, Rodrigues ALS. Evidence for the involvement of opioid system in the antidepressant-like effect of ascorbic acid. Naunyn Schmiedebergs Arch Pharmacol 2018; 391(2): 169–176. [DOI] [PubMed] [Google Scholar]
  • 165.Moretti M, Colla A, De Oliveira Balen G, et al. Ascorbic acid treatment, similarly to fluoxetine, reverses depressive-like behavior and brain oxidative damage induced by chronic unpredictable stress. J Psychiatr Res. 2012; 46(3): 331–340. [DOI] [PubMed] [Google Scholar]
  • 166.Moretti M, Budni J, Dos Santos DB, et al. Protective effects of ascorbic acid on behavior and oxidative status of restraint-stressed mice. J Mol Neurosci 2013; 49(1): 68–79. [DOI] [PubMed] [Google Scholar]
  • 167.Moretti M, Budni J, Freitas AE, et al. TNF-α-induced depressive-like phenotype and p38MAPK activation are abolished by ascorbic acid treatment. Eur Neuropsychopharmacol 2015; 25(6): 902–912. [DOI] [PubMed] [Google Scholar]

Articles from Chronic Stress are provided here courtesy of SAGE Publications

RESOURCES