Next Article in Journal
A Review of High-Intensity Focused Ultrasound
Previous Article in Journal
Assessment of Surgical Difficulty in Patients with Rectal Cancer—The Impact of Pelvimetry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancements Exploring Major Depressive Disorder: Insights on Oxidative Stress, Serotonin Metabolism, BDNF, HPA Axis Dysfunction, and Pharmacotherapy Advances

by
Ana Salomé Correia
1,2,3 and
Nuno Vale
1,2,4,*
1
PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
2
CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
3
Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
4
Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
*
Author to whom correspondence should be addressed.
Int. J. Transl. Med. 2024, 4(1), 176-196; https://doi.org/10.3390/ijtm4010010
Submission received: 19 January 2024 / Revised: 16 February 2024 / Accepted: 1 March 2024 / Published: 5 March 2024

Abstract

:
Major depressive disorder (MDD), a prevalent mental illness, is marked by a complex mixture of biological factors. This review focuses on the roles of oxidative stress, tryptophan-serotonin metabolism, brain-derived neurotrophic factor (BDNF), and the hypothalamic–pituitary–adrenal (HPA) axis in MDD’s pathophysiology. Oxidative stress, defined as an imbalance between pro-oxidants and antioxidants, is closely linked to MDD’s neurobiological changes. The tryptophan (TRP)-/serotonin (5-HT) metabolic pathway is also known to be crucial in mood regulation, with its dysregulation being a central aspect of MDD. Additionally, BDNF, key for neuronal growth and plasticity, often shows alterations in MDD patients, supporting its role in the disorder’s progression. Furthermore, the HPA axis, which manages stress response, is frequently disrupted in MDD, further contributing to its complex pathology. In addition to exploring these biological mechanisms, this review also explores the pharmacotherapy of MDD, including new advances. These advancements in treatment strategies are crucial for managing MDD effectively. Understanding these mechanisms and the latest pharmacological interventions is essential for developing more effective treatments for MDD.

1. Background

Depression is a mental health disease, manifesting in various forms that impact individuals uniquely. The World Health Organization (WHO) reports that approximately 280 million people worldwide suffer with depression [1]. MDD, commonly only referred to as depression, represents a debilitating condition characterized by at least one distinct depressive episode lasting a minimum of two weeks, marked by shifts in mood, anhedonia, and cognitive symptom disturbances [2]. This condition is heterogeneous, with potential consequences extending to suicide [3,4]. People suffering from MDD also have several social stigmas and high occurrences of physical health conditions like heart disease and type 2 diabetes [4]. The complexity of this disease presents a significant challenge for neuroscience, as no single established mechanism can fully elucidate its nature.
A challenge in dealing with this disease is the frequent recurrence, treatment ineffectiveness, and the absence of diagnosis and treatment options, particularly in low- and middle-income countries [1,2,5]. Nevertheless, there are viable treatment approaches, notably psychotherapy and/or the use of antidepressants [2]. Both demonstrate efficacy in addressing MDD. Still, approximately 30% of individuals affected by MDD do not experience remission, even after multiple treatments [2,6].
Despite the complex nature of MDD, several studies support the importance of various factors, including neurotransmitters, oxidative stress and neurotrophic factors (particularly BDNF) in contributing to this condition [7,8]. Structural alterations, in brain regions such as the prefrontal cortex (PFC) and hippocampus are also significant [9]. Indeed, previous studies have shown that the hippocampus is smaller in depressed patients [10,11].
In this review, we are going to explore the role of oxidative stress, 5-HT pathway, BDNF, and HPA axis dysfunction in MDD. Additionally, we are also going to focus on the pharmacotherapy advancements of MDD in recent years. Figure 1 represents a succinct illustration of some aspects involved in depression that will be addressed in this review.

2. Exploring the Role of Oxidative Stress, Tryptophan-Serotonin Metabolism, Brain-Derived Neurotrophic Factor, and Hypothalamic–Pituitary–Adrenal Axis Dysfunction in Major Depressive Disorder

2.1. Oxidative Stress as a Key Contributor to Major Depressive Disorder Pathogenesis

Oxidative stress forms from an imbalance in the equilibrium between the formation of free radicals, particularly reactive oxygen species (ROS), and antioxidants. Furthermore, the exposure to environmental stressors, like smoking or ultraviolet radiation, can accentuate the challenge of maintaining this equilibrium [12]. A balanced diet rich in antioxidants, regular exercise, and stress reduction techniques can help maintain oxidative balance [13].
The principal ROS presented in the human body are mainly produced in mitochondria, peroxisomes, and the endoplasmic reticulum, being generated through enzymatic reactions that encompass enzymes such as cyclooxygenases (COXs), nicotinamide adenine dinucleotide phosphate (NADPH), lipoxygenases and through the known Fenton reaction [14,15]. Oxidative stress damages cells primarily through the oxidation of lipids, proteins, and DNA, triggering a cascade of cellular mechanisms leading to cell damage [15,16].
To protect against the effects of increased ROS levels, cells have antioxidant defenses, such as superoxide dismutase (SOD) and catalase (CAT), important to maintain ROS homeostasis [12,17]. Exogenous antioxidant defenses may also be introduced by diet or nutritional supplementation, such as carotenoids, vitamins C and E [12,18].
High-oxidative stress levels accentuate various cellular mechanisms implicated in MDD, including the stress response, neuroinflammation, neurotransmitter signaling disturbance, and impaired neurogenesis/synaptic plasticity [19,20]. The brain is vulnerable to oxidative stress, due to its high oxygen consumption and lipid content. Also, neurons, astrocytes and microglia are rich in mitochondria and NADPH oxidase (NOX), generating high levels of ROS. This makes oxidative stress an important cause of neurodegeneration and a key factor in MDD. Addressing these changes with suitable antioxidants could be an effective strategy for treating MDD [15,20,21].
Depression is linked to reduced consumption of antioxidants like vitamins A, B, C, E, selenium, and zinc [22]. There is also evidence that high levels of lipid peroxidation in the brain lead to increased oxidative stress levels [20]. Analysis of lipid peroxidation markers in MDD (particularly malondialdehyde) demonstrated that this process was greater in MDD individuals than in controls, being connected with more severity [23]. When combined with a decrease in antioxidant defenses, these mechanisms support the role of oxidative stress as a key player in depression [20]. Also, research reported elevated levels of peroxidation biomarkers combined with reduced antioxidant activity in the plasma and serum of individuals with MDD [21,24,25]. Targeting hypoxia-related pathways may also be a promising tool for depressive disorders in the context of mitigating high-oxidative stress levels, enhanced by hypoxia [8,26]. In fact, recent findings from our investigation group revealed that hypoxia–ischemia triggers ROS elevation in neuron-like cell lines. Treatment with edaravone enhanced cell viability and decreased ROS levels, likely attributable to its free radical-scavenging characteristics [27]. Various studies have also observed reductions in antioxidant levels in both depressed patients and animal models of depression [21,28]. Furthermore, it has been demonstrated that antidepressants have antioxidant properties, and antioxidants exhibit antidepressant effects. For instance, fluoxetine and citalopram enhanced SOD activity and ascorbic acid levels, reducing malondialdehyde in MDD patients [21,29]. Research conducted on cell lines further supports the link between oxidative stress and depression. Notably, the introduction of H2O2 to these cells resulted in a reduction in cellular viability. However, this effect was ameliorated when serotonergic compounds such as mirtazapine and L-TRP were applied. These compounds not only enhanced cell viability in the presence of H2O2 but also led to a reduction in ROS levels and mitigated DNA damage following exposure to oxidative stress [30,31].
These studies support the significant role of oxidative stress as an important factor in the pathogenesis of depression and suggest that antioxidant activity holds promise as a potential therapeutic approach.

2.2. Unraveling the Hypothalamic–Pituitary–Adrenal Axis Dysregulation in Major Depressive Disorder

HPA axis is important in regulating functions such as energy balance, reproduction, and response to stress [32,33]. The elements of the HPA axis are the hypothalamus, pituitary gland and adrenal glands. The hippocampus contains neuroendocrine neurons that synthesize and secrete vasopressin and corticotropin-releasing hormone (CRH), mainly in the paraventricular nucleus (PVN). In response to stress, CRH and vasopressin are released into hypophysial portal vessels that access the anterior lobe of the pituitary gland to secrete adrenocorticotropic hormone (ACTH) via the activation of the cyclic adenosine monophosphate (cAMP) pathway, after binding to the CRH receptor 1 and vasopressin V1b (or V3) receptor, respectively. The adrenal cortex produces glucocorticoid hormones (mainly cortisol in humans) in response to stimulation by ACTH, which binds to the melanocortin type 2 receptor. These glucocorticoids regulate physiological responses and inhibit further HPA axis activation [34].
When the HPA axis is triggered, it leads to an increase in glucocorticoid levels, an important component in the body’s adaptation to stress [35,36]. Different reactions to stress are observed, depending on if it is of a short-term or long-term nature. The short-term is a natural physiological response, while the long-term tends to be detrimental. Under physiological settings and in a stress-free context, healthy persons release between 10 and 20 mg of cortisol on a daily basis with regular peaks [37]. Initially, there’s a rise in ACTH levels, which triggers the release of cortisol. However, when someone experiences prolonged, chronic stress, cortisol levels remain elevated due to more adrenal sensitivity. [38]. Thus, when faced with uncontrollable and prolonged stress, it can lead to a range of alterations in various aspects of the CNS, contributing to neuropsychiatric and neurodegenerative conditions [35,39]. In fact, elevated cortisol secretion during stressful situations can significantly impact the functioning of the brain. This hormone has a pronounced effect on the hippocampus due to the abundance of steroid receptors it possesses [37,40]. Recurrent exposure to stress triggers alterations in neuronal structures. When stress is acute, the hippocampal atrophy that may occur is often reversible. However, chronic stress can result in the neuronal death within the hippocampus. Numerous studies suggested that conditions like depression and post-traumatic stress disorder are associated with a reduction in the volume of the hippocampus, as well as that of the PFC and amygdala [37,41].
Glucocorticoid receptors (GRs) and mineralocorticoid receptors (MRs) are the intracellular nuclear receptors that respond to cortisol. GRs are found throughout the brain and peripheral tissues, while MRs are primarily located in cardiovascular tissues, the liver, kidneys, and certain brain regions like the corticolimbic areas [42]. These receptors can activate or repress the transcription of genes in the cell nucleus and also mediate non-genomic effects [43]. During the day, nuclear MRs are typically occupied by cortisol, remaining active. However, during the night, when cortisol levels are very low, these MRs become unoccupied. When cortisol levels rise significantly, both nuclear GRs and membrane-associated MRs and GRs become occupied by this hormone, leading to changes in synaptic plasticity at the cellular level [42,44].
Hyperactivity of the HPA axis is one of the most studied findings in MDD. This hyperactivity results in an increased level of glucocorticoids, which can disrupt the body’s ability to regulate these hormones effectively [45].
In individuals with MDD, the GR typically becomes compromised, resulting in a diminished capacity for negative feedback regulation [46]. This dysregulation leads to an increased synthesis of glucocorticoids [47]. Antidepressant drugs, on the other hand, also work by influencing the functioning of this system. These influences can take different forms, including the regulation of the expression of the GR and post-translational modifications. As a result, the therapeutic effects of antidepressants involve, at least in part, the restoration of the function of GR [48].
High stress levels have also been associated with disturbances in 5-HT pathways, modifications in the structure of brain regions like the hippocampus and PFC, and epigenetic alterations in genes such as BDNF [49,50,51]. Long-term exposure to corticosterone has been demonstrated to induce modifications in the structure of neuronal dendrites, promoting the atrophy [52,53]. Moreover, research on postmortem tissue from individuals with depression and animal models has provided insights into the modifications at the cellular level connected with this condition, particularly structural changes in the brain, including dendritic atrophy, neuronal loss, and alterations in glial elements [54].
The connection between oxidative stress and MDD’s associated stress response is also important in the context of MDD [55]. Increased production of ROS leads to the hyperactivation of the HPA axis [56]. Furthermore, the release of glucocorticoids caused by HPA axis activation increases the activity of cellular reduction-oxidation systems. When stress activates GRs, there is an increase in mitochondrial membrane potential and mitochondrial oxidation [57]. Consequently, it leads to the generation of ROS, leading to oxidative damage [15,58].
Both glucocorticoids and inflammation have been linked to the pathogenesis of depression [59]. According to the glucocorticoid resistance paradigm, less sensitivity to cortisol’s anti-inflammatory properties causes increased inflammation in depression. Animal research have indicated that repeated social defeat can produce glucocorticoid-resistant monocytes, depressive-like behaviors in animal models, and enhance neuroinflammatory signaling [60,61]. Human studies have concurrently observed reduced GR function/expression, HPA axis hyperactivity, and increased inflammation in depressed patients [46,60]. In fact, depressed people frequently show HPA axis hyperactivity and GR dysfunction, despite elevated inflammatory markers and the glucocorticoids’ anti-inflammatory properties. This seeming paradox might be attributed to the fact that the glucocorticoid and inflammatory systems naturally coexist in equilibrium. Chronic stress may alter this balance, favoring inflammatory processes and weakening glucocorticoid signaling. Persistent stress may disrupt the homeostatic balance, causing inflammation while inhibiting the anti-inflammatory effects of glucocorticoids. Also, inflammation and glucocorticoid signaling have separate effects on the same biological processes and structures, resulting in cumulative damage that contributes to depression. In this case, these two systems may not interact directly, but they do converge on shared pathways that lead to depression. Understanding the connection is critical for finding therapy targets in depression [62].
Additionally, inflammatory changes within the brain have been linked to conditions such as dementia and MDD, highlighting the implications of these interactions [63]. Stress-induced proinflammatory cytokine production also stimulates the indoleamine 2,3-dioxygenase (IDO)/ kynurenine (KYN) pathway in cells including macrophages. IDO has an important part in the catabolism of TRP, which results in lower 5-HT levels [64]. Particularly noteworthy is the link between elevated concentrations of interleukin (IL)-1 and IL-6 and the stress response [65,66]. Indeed, an elevated production of IL-6 has been observed to play a role in MDD prognosis and how individuals respond to treatment, possibly through activation of the HPA axis [67].
The study of the stress response, which is predominantly regulated by the HPA axis, provides a road to understanding the underlying biological mechanisms that are present in depression, allowing for the development of more effective therapies.

2.3. Exploring Tryptophan/Serotonin’s Role in Major Depressive Disorder

TRP is an amino acid with important implications in several physiological reactions. There are two major pathways of TRP metabolism: the KYN pathway and the 5-HT pathway [68].
The 5-HT pathway of TRP metabolization is extremely important in human physiology, influencing several physiological functions. 5-HT networks are important players in behavioral aspects, including mood regulation and memory processing. Moreover, these networks modulate motor coordination, circadian rhythm, and thermoregulation. The 5-HT pathway extends its influence on diverse physiological processes such as gastrointestinal regulation and nociception [69].
The pathway starts with the conversion of TRP to 5-hydroxytryptophan (5-HTP) by TRP hydroxylase enzymes (TPH1 or 2). 5-HTP is decarboxylated by aromatic acid decarboxylase (AADC) to produce 5-HT, which undergoes further metabolism. Specifically, 5-HT can be converted to N-acetylserotonin (NAS) by arylalkylamine N-acetyltransferase (AANAT), and then to melatonin by N-acetylserotonin O-methyltransferase (ASMT). Alternatively, monoamine oxidase (MAO) transforms 5-HT into its major metabolite, 5-hydroxyindoleacetic acid (5-HIAA) [70,71,72,73]. Interestingly, a study coordinated by our research group examining the TRP-5-HIAA axis following exposure to elevated concentrations of corticosterone and H2O2 have unveiled significant alterations in the levels of its metabolites, particularly 5-HT, within the extracellular environment of the cells. These findings support the impact of environmental modulation and exposure to stressors on this metabolic axis [74].
The activity of 5-HT has precise control mechanisms regulating its synthesis, release, and metabolism. The majority of 5-HT is intracellular, ensuring a tight regulation of its concentration. Stored in intracellular vesicles, 5-HT is released into the synaptic cleft upon neuronal depolarization, where it binds to receptors on both presynaptic and postsynaptic membranes [75]. These receptors are distributed in the CNS, PNS, and peripheral tissues, such as smooth muscles. The diverse distribution of 5-HT receptors throughout the body supports the broad influence of 5-HT on human physiology and behavior. Modulating these receptors can have significant impacts on various conditions, including mood disorders and gastrointestinal issues [76]. A summary of 5-HT receptors is presented in Table 1.
Presynaptic receptors are self-regulators, having an inhibitory influence on the release of additional 5-HT. In contrast, postsynaptic receptors modulate either excitatory or inhibitory signaling pathways, depending on the specific receptor subtype involved, through the activation of second messenger cascades. 5-HT that is reabsorbed into the originating cell via the serotonin transporter (SERT) is subsequently stored in intracellular vesicles or undergoes metabolism through the action of MAO within the cytoplasm. On the other hand, 5-HT circulating in the periphery is metabolized by the liver and lungs [75].
The involvement of monoamines in depression has been extensively researched, and 5-HT is closely connected with the depressive condition. In the present day, antidepressants designed to enhance the availability of 5-HT in the synaptic cleft, known as SSRIs, have proven to be effective and rank among the most widely prescribed medications globally [78]. Nevertheless, it is important to recognize that depression is a complex disorder, and while impaired 5-HT activity can contribute to its development, it neither represents a sole causative factor nor is it sufficient on its own [79].
Evidence from several studies supports the diverse roles played by 5-HT receptor subtypes in the development of MDD. In a general way, antagonists of 5-HT2A, 5-HT2C, 5-HT3, 5-HT6, and 5-HT7 receptors, in addition to agonists of 5-HT1A, 5-HT1B, 5-HT2C, 5-HT4, and 5-HT6 receptors, have demonstrated the ability to promote antidepressant-like responses [80]. 5-HT1A-B and 5-HT2A are the most studied 5-HT receptors in the context of depression, despite the role of other 5-HT receptors also being a target of studies in MDD, especially 5-HT3 [81]. Our research group recently hypothesized that 5-HT3 antagonists, such as scopolamine, might combat oxidative stress by 5-HT3 interaction, despite the need of further clarification [82]. Table 2 represents a short summary of the connection between these three receptors and depression.
The 5-HT1A receptor is an abundant 5-HT receptor in the brain with presynaptic and postsynaptic subtypes. When 5-HT binds to these receptors, it induces neuronal hyperpolarization and reduces its firing rate, but the response to sustained stimulation differs between these subtypes. In depression, the 5-HT1A receptor is increased presynaptically, causing a decrease in the release of 5-HT [87]. 5-HT1A autoreceptors act as a feedback mechanism to regulate 5-HT release, being located on the 5-HT neurons in the brainstem. When 5-HT1A receptor agonists are administered, it can inhibit the firing of serotonergic neurons and thus reduce the release of 5-HT in the short term. However, with chronic administration, there’s a desensitization of these presynaptic receptors, which leads to an overall increase in the release of 5-HT in the brain to bind to postsynaptic heteroreceptors, modulating the activity of the receiving neuron [88].
Stimulation of the 5-HT2A receptor in neurons can trigger effects through different signaling pathways, such as modulation of GABA and glutamate, mainly due to the high presence of this receptor in glutamatergic and GABAergic neurons [89]. In most cases, activating the 5-HT2A receptor leads to an increase in the levels of intracellular calcium. Additionally, the activation of the 5-HT2A receptor results in the phosphorylation of ERK through a variety of intracellular signaling mechanisms, mostly promoted by Src and calmodulin, a process that regulates downstream signaling components [85,90]. The ERK pathway is involved in the regulation of various cellular processes that can influence mood and behavior, playing an important role in neuroplasticity. Dysregulation of this pathway has been linked to depressive symptoms [91]. These receptors also interact with β-arrestin proteins that are multifunctional intracellular proteins that directly interact with many cellular components, contributing to multiple aspects of, for example, GPCR signaling [89,92]. Previous studies have demonstrated that these receptors coexist with β-arrestin-1 and -2 in cortical neurons. Interestingly, in mice lacking β-arrestin-2, where 5-HT2A receptors predominantly remain on the cell surface, 5-HT fails to induce behavioral responses, including head-twitching. This suggests that β-arrestin-2 mediates the internal mobility of 5-HT2A receptors within cells, which is linked to the appearance of head-twitching in response to increased 5-HT levels. DOI, a 5-HT2A-C receptor agonist, may still cause head-twitching in mice without β-arrestin-2. This suggests that β-arrestins are not necessary for DOI-mediated reactions [93,94]. These findings highlight the role of the specific ligand in determining how the receptor’s signaling pathway is triggered [89]. Additionally, the dynamic regulation of signal intensity and duration in 5-HT2A receptor signaling is known to be managed through the rapid endocytosis mediated by β-arrestins [95]. Another way 5-HT2A receptor subtypes can affect signaling is by their capacity to form stable complexes with other GPCRs, such as metabotropic glutamate receptor type 2 (mGluR2) and dopamine D2 receptors. Although the function of these receptor complexes in live creatures are still to be completely understood, this process may influence how receptors attach to molecules and interact with signaling pathways [89].
The 5-HT3 receptors have permeability to various ions, namely Na+, K+, and Ca2+. Activation of these receptors initiates the opening of ion channels, triggering rapid membrane depolarization [96,97]. The roles of 5-HT3 receptors are linked to their specific localization. The activation of nerve-terminal 5-HT3 receptors modulates the release of diverse neurotransmitters, such 5-HT, DA, or GABA. In contrast, the activation of postsynaptic 5-HT3 receptors primarily participates in rapid synaptic transmission [97,98]. Pre- and postsynaptic receptors have unique features. Notably, presynaptic 5-HT3 receptors are highly permeable to Ca2+ ions, but postsynaptic receptors are less permeable to Ca2+ in comparison to Na+ and K+. Presynaptic 5-HT3 receptors, which control neurotransmitter release, vary from postsynaptic receptors, which are engaged in fast synaptic transmission, due to their differential calcium permeability. Ca2+ influx causes neurotransmitter release into the synaptic cleft [99]. Furthermore, the 5-HT3 receptor can be composed of five identical 5-HT3A subunits (homopentameric) or a combination of 5-HT3A and one of the other four 5-HT3B, 5-HT3C, 5-HT3D, or 5-HT3E subunits (heteropentameric). The homomeric 5-HT3A receptors are equally permeable to both monovalent and divalent cations, but heteromeric 5-HT3 receptors have lower Ca2+ permeability [97]. Additionally, heteromeric receptors exhibit faster activation and deactivation kinetics compared to homomeric receptors. It is worth mentioning that the affinity and efficacy of 5-HT3 receptor agonists and antagonists are dependent on the specific receptor structure [97].
Despite all the evidence that connects 5-HT signaling pathways to MDD, it is important to note that a recent comprehensive systematic umbrella review revealed that there is no robust evidence that depression is caused by lower 5-HT concentrations or activity [100], despite the fact that another very recent review was published with the aim of contradicting this review [101]. However, in recent years, specific changes in 5-HT neurotransmission have remained a viable therapeutic option for treating mood and anxiety disorders. Indeed, the efficacy of SSRIs in relieving symptoms has been extensively documented. However, the assumption that clinical depression might be traced only to the poor functioning of a single neurotransmitter is widely recognized as unlikely [102]. Evidence reveals that depleting TRP in healthy individuals, who exhibit no apparent risk factors for depression, does not result in a significant decline in mood [103]. Thus, the concept that lowering brain 5-HT levels alone is sufficient to cause depression appears to be unviable. However, in individuals who have recovered from depression and have remained free from both pharmaceutical and psychological treatments for extended periods, TRP depletion can lead to a clinically-significant reduction in mood, implying that in individuals with a history of depression, prior episodes render them vulnerable, and reductions in brain 5-HT levels can trigger clinical relapses [103]. All the complexities of the serotonergic system and depression highlight the importance of further research to clarify the effects of different drugs on the 5-HT system in MDD [73].

2.4. Exploring Brain-Derived Neurotrophic Factor in Major Depressive Disorder

BDNF is important in several aspects of human physiology and its expression is observed in both the central nervous system (CNS) and the peripheral nervous system (PNS) [104,105].
Neurons are the principal producers of BDNF, which begins as preproBDNF and then a preprotein called proBDNF [106]. ProBDNF undergoes further changes to mature form [107]. BDNF produces dimers and biologically active BDNF is typically made up of a dimer composed of two identical mature peptide chains linked together by noncovalent interactions [108].
The human BDNF gene contains 11 exons (I–IX, Vh, and VIIIh), and the production of BDNF transcripts is cell and activity specific, with different transcripts playing different roles in biochemical processes [109,110]. Exon IV is the most extensively studied, because it holds significance in the modulation of mood, cognition, and behavior [111]. The methylation status of the promoter associated with this exon has emerged as a potential biomarker for antidepressant therapy in individuals with MDD [112].
BDNF plays an important role in many processes, including neuronal survival and differentiation, synaptic plasticity, neurogenesis, neuroprotection, learning, memory, mood control, and other cognitive processes. Low BDNF levels relate to brain shrinkage, cognitive decline, and an increased risk of mental illnesses [113]. BDNF also influences the release and activity of different neurotransmitters, affecting neuronal communication and general brain function [114]. This neurotrophin is also involved in the formation of dendrites and synaptic specializations, maturation and refinement of dendritic arbors, and axon growth and differentiation [115]. Based on the actions of this neurotrophin, several therapies and techniques have been found to boost the levels of BDNF, including physical exercise, the ingestion of omega-3 fatty acids, and several antidepressants, such as SSRIs [116,117].
The relationship between BDNF and MDD is a large topic of investigation. The neurotrophic theory of depression is based on the relationship between lower BDNF levels and an increased risk of depression [118]. Table 3 summarizes the link between BDNF and MDD.
Human studies have demonstrated a positive link between peripheral blood levels of BDNF and both hippocampal size and cognitive function [119,120]. Low BDNF levels are consistently associated with an increased prevalence of depressive symptoms, neuronal loss, and atrophy in key brain regions [121]. A study highlighted that patients experiencing their first episode of drug-free MDD and responding to SSRI treatments exhibited elevated serum BDNF levels [122]. Moreover, antidepressant-treated individuals have shown increased BDNF expression in the hippocampus when compared to their untreated counterparts [123]. Postmortem examinations of brains from patients with depression, including suicide cases, have revealed reduced BDNF mRNA levels, confirming this trend [124,125]. Animal models of depression also aligned with this evidence, showing that chronic stress and depression lead to diminished BDNF levels, increased cell death, and reduced neurogenesis in the hippocampus, alongside decreased BDNF expression in other brain areas [126,127]. A genetic variation known as Val66Met, which naturally occurs in the BDNF gene, is also linked with MDD [128]. Recent research has linked this polymorphism to MDD and its potential use as a biomarker for predicting a patient’s response to antidepressants and electroconvulsive therapy [129]. Various forms of physical exercise have also been found to stimulate the production of this neurotrophin, leading to cognitive enhancement and a reduction in symptoms associated with depression and anxiety [105,130].
Table 3. Summary of the BDNF and MDD connection. Adapted from [105].
Table 3. Summary of the BDNF and MDD connection. Adapted from [105].
FeatureExplanation
Levels of BDNFReduced BDNF levels have been observed in individuals with MDD [118].
Structure and functionDeficiencies or imbalances in BDNF levels contribute to depression by promoting structural and functioning changes [131].
5-HT BDNF is affected by 5-HT, and 5-HT stimulation can increase BDNF production and release. 5-HT receptors can also control BDNF production, which influences neuronal function and, consequently, mood modulation [132].
NeuroplasticityBDNF has a role in neuroplasticity, which is essential for synaptic connections and structural changes in the brain connected to MDD [133].
Oxidative stressOxidative stress can impair BDNF production and signaling pathways. The link between oxidative stress and BDNF levels is critical in the development and progression of depression [15].
HPA axis dysregulationStress-induced HPA axis hyperactivity and the subsequent increase in glucocorticoid levels diminish BDNF expression, which plays an important role in the development of depression [134].

3. Pharmacological Interventions in the Management of Major Depressive Disorder

3.1. An Overlook of Pharmacotherapy for Major Depressive Disorder

The global economic impact of depression is significant, and it has consistently remained the third leading contributor to the worldwide disease burden, as designated by the WHO since 2008 [135]. Thus, there is the need for advancements in research.
The first advancement in the treatment of depression was made in the early 1950s, when researchers developed iproniazid as a drug for tuberculosis which also proved to be effective in alleviating symptoms of depression, being a type of MAOI [136]. Around the same time, another class of antidepressants known as tricyclics (TCAs) appeared. Imipramine was the first TCA, approved in 1959 by the Food and Drug Administration (FDA) for the treatment of MDD. Like MAOIs, TCAs were developed in the 1950s and were found to elevate monoamine levels, primarily by blocking the reuptake of 5-HT and noradrenaline [84,137]. While these discoveries represented important advancements, the acceptance and use of these early antidepressants were impeded by public stigma and severe side effects [137]. By the late 1980s, SSRIs offered a more targeted approach to depression treatment. As previously referred, they function by inhibiting the reuptake of 5-HT into neurons within the raphe nuclei, resulting in increased 5-HT. These drugs were found to have improved side effect profiles. The introduction of SSRIs led to an increase in the use of antidepressants among adults. Despite being developed several decades ago, SSRIs remain some of the most prescribed drugs in the world [84,138]. However, approximately 33% of patients with MDD do not respond to treatment with these commonly used drugs, and 67% do not achieve remission with this first-line approach [84]. Today, several classes of antidepressants are present in the market. Besides the previously mentioned, serotonin–noradrenaline reuptake inhibitors (SNRIs) such as venlafaxine, block 5-HT and NA reuptake in the synapse, serotonin modulators (e.g., vilazodone) and atypical antidepressants (e.g., mirtazapine) are also widely used [139]. In recent years, research has shifted its focus towards potential new therapies, including noncompetitive NMDA receptor antagonists [84].
Depression treatment approaches include both pharmaceutical interventions and non-pharmaceutical options, such as psychotherapy, electroconvulsive therapy, and transcranial magnetic stimulation [135]. Psychotherapy has demonstrated its efficacy in alleviating depressive symptoms and enhancing the overall quality of life for individuals with depression [140]. Consequently, numerous clinical guidelines are progressively endorsing the use of psychotherapy either as a standalone treatment or in combination with antidepressants [135].

3.2. Antidepressant Breakthroughs: Advancements in Pharmacotherapy for Depression

While most antidepressants are considered safe and effective, they have limitations, including a delayed onset of action and side effects that can impact a patient’s adherence to treatment, such as weight gain, sexual dysfunction, dizziness, headaches, anxiety, psychosis, and cognitive impairments. Over the past few decades, several efforts in antidepressant research have been directed towards discovering drugs that act more rapidly, with greater safety [135,141].
The most common type of new drugs is based on NMDA receptor properties [135,141,142]. The interest in these receptors comes from the studied role of glutamate in depression. Glutamate is an excitatory neurotransmitter in the brain, important in synaptic plasticity, cognitive functions, and emotional and reward processes. Ionotropic receptors (NMDA, AMPA, and kainate receptors), as well as metabotropic receptor mGluR, have been demonstrated to play a role in regulating mood and related functions that are compromised in individuals with depression [143]. Clinical investigations and animal research observed dysfunction within the glutamatergic system in diverse limbic and cortical regions of the brains of individuals experiencing depression [144].
GABAergic modulators are also promising novel targets for antidepressants. GABA, the primary inhibitory neurotransmitter, is important in balancing brain function by counteracting glutamate [135]. Studies revealed that individuals with depression often exhibit GABA-related neurotransmission impairments [145]. A recent study analyzed the status and research trends of the GABAergic system in depression from 2004 to 2020, revealing that GABA levels in the PFC, anterior cingulate cortex, and occipital lobe are decreased in patients with MDD [146]. Nonetheless, while GABAergic ligands have shown effectiveness in treating depression, directly influencing the GABA pathway, they can lead to side effects like drowsiness or sedation, which can impede daily functioning. Therefore, the balance seems to be shifting in favor of glutamate receptor ligands [141].
More optimism regarding MDD pharmacotherapy appeared when researchers discovered the antidepressant properties of intravenous ketamine, a NMDA receptor antagonist [142]. This drug blocks NMDA receptors on GABA interneurons, releasing the inhibition of glutamate release, a process that activates AMPA receptors on glutamatergic cells and increases BDNF and glutamate, increasing synaptic efficiency [147]. The administration of ketamine to depressed patients resulted in a sustained three-day reduction in depressive symptoms [148]. This discovery led to the search for related medications like S-ketamine (esketamine), which was developed for intranasal use and FDA-approved as an adjunctive treatment for treatment-resistant depression in 2019. Notably, both intravenous ketamine and intranasal esketamine have shown promise in rapidly alleviating MDD symptoms, leading to esketamine’s second FDA-approved indication in 2020. Despite these promising aspects, the benefits of ketamine and esketamine must be weighed against their costs and potential side effects, such as sedation and dissociation [142,149]. Additionally, combining electroconvulsive therapy with antidepressants has been a known approach, and recent findings regarding its use with esketamine have shown great promise and high effectiveness, particularly in cases of drug-resistant depression [141,150].
The combination of bupropion and dextromethorphan also resulted in a significant reduction in depression scores. Dextromethorphan is an uncompetitive NMDA receptor antagonist and sigma-1 receptor agonist, a class of drugs that is recognized for their potential to facilitate synaptic plasticity and neuronal resilience, primarily through their capacity to upregulate BDNF secretion [151]. Bupropion inhibits the reuptake of both NA and DA, as well as CYP2D6 enzymes, increasing the bioavailability of dextromethorphan. This drug combination was approved only in the USA in August 2022 for the treatment of MDD in adults (currently, is only used off-label in Europe [152]). In clinical trials, this combination was generally well tolerated, and was not associated with a signal for increased psychotomimetic effects or weight gain [153].
Another trial explored the use of adjunctive esmethadone, another uncompetitive NMDA receptor antagonist [154]. This trial demonstrated rapid and robust reductions in depressive symptoms, although this was not consistently replicated in a subsequent phase 3 trial. Additional phase 3 studies are currently ongoing to confirm the initial findings [142,155].
Neurosteroids like brexanolone and zuranolone represent another class of potential antidepressants [142]. These drugs modulate GABA neurotransmission. Brexanolone, which was approved by the FDA in 2019 for postpartum MDD, demonstrated rapid and long-lasting reductions in depression symptoms [156]. Zuranolone is also being studied across various MDD cases, with positive results from clinical trials. Indeed, this drug demonstrated enhanced efficacy in alleviating depressive symptoms by day 15, exhibiting a rapid onset of action by day 3, and exhibiting a favorable safety profile [157].
A relatively new category of antidepressants includes orexin receptor antagonists and compounds that act through two neuropeptides (orexin-A and orexin-B) and two GPCRs (the orexin type 1 and 2 receptors). The dysfunction of the orexin system has been implicated in the pathophysiology of depression in human and animal studies, although the exact mechanism of this dysfunction remains unclear. Nevertheless, ligands for type 1 and 2 receptors can modulate various aspects such as feeding, sleep, motivated behavior, anxiety, and addiction, making them potentially influential in regulating different facets of depression. Orexin receptor antagonists are being investigated as potential treatments for MDD, with several ongoing clinical trials [158].
New drugs like ansofaxine (a potential triple reuptake inhibitor of 5-HT, NA, and DA) exhibit side effects that are quite similar to traditional antidepressants. These side effects are generally mild to moderate, with slightly different frequency patterns compared to drugs like SSRIs. Notably, this drug does not seem to cause sexual dysfunction, typically associated with conventional antidepressants [141,159]. In September of 2023, FDA also approved gepirone, for the treatment of MDD. This drug is a 5-HT1A receptor agonist, and an active metabolite of gepirone, 1-(2-pyrimidinyl)piperazine, is an α 2-adrenergic receptor antagonist. This drug has the big advantage of not causing sexual dysfunction or weight gain, as well as inducing less sedation and minimal withdrawal symptoms [160].
Psychedelic drugs have also gained attention as potential alternative antidepressants. Preliminary findings suggest that psilocybin, derived from mushrooms, may lead to sustained antidepressant effects in patients with treatment-resistant depression and terminal cancer [142]. In individuals suffering from treatment-resistant depression, the administration of a single dose of psilocybin, combined with psychological support in conjunction with SSRI treatment, exhibited a generally positive safety profile and displayed significant therapeutic effectiveness as assessed through a diverse array of clinician-conducted evaluations and self-reported measures [161]. In fact, a recent study suggests that a single, moderate dose of psilocybin significantly reduces depressive symptoms compared to a placebo, for at least two weeks [162].
The cholinergic system is increasingly recognized as having a significant role in mood regulation. Acetylcholine, a neurotransmitter, is implicated in mood disorders with depressive symptoms, although its exact role remains unclear. Recent evidence, such as the rapid and sustained antidepressant effects of scopolamine in depressed individuals, has induced the interest in the cholinergic system’s involvement in MDD and bipolar disorder, being suggested that excessive cholinergic activity can trigger MDD. This hypothesis is supported by studies showing that drugs increasing acetylcholine activity can induce depressive symptoms [163]. However, understanding of the efficacy and mechanism of treatments targeting the reversal of acetylcholinesterase increase using acetylcholinesterase inhibitors (AChEIs) remains limited [164].
Regarding other targets, previous studies have found that treatments aimed at regulating the HPA axis, such as GR antagonists, do not effectively alleviate the symptoms of depressed patients [141]. Nevertheless, this is a subject of research interest, and a recent study revealed that antalarmin, a CRH receptor 1 antagonist, alleviated LPS-induced depression-like behavior in mice [165]. Therapies regarding the direct manipulation of KYN pathways are also still not available in the common medical practice for MDD [166].
Drug repurposing, the exploration of new uses for existing market-available drugs, is gaining attention in research. These drugs have passed safety evaluations, undergone formulation development, and successfully completed preclinical and clinical testing, substantially reducing the risk of failure [167]. Table 4 represents examples of widely studied drugs with the potential to be repurposed in the context of depression, their original indication, as well as evidence in depression that supports their potential use in this disease.
Drug repurposing in the treatment of depression represents an essential approach in the world of mental health. It offers the potential for rapid and cost-effective solutions, while also reducing the risks and challenges associated with developing entirely new drugs.

Author Contributions

Conceptualization, A.S.C. and N.V.; methodology, A.S.C.; formal analysis, A.S.C. and N.V.; investigation, A.S.C.; writing—original draft preparation, A.S.C.; writing—review and editing, N.V.; supervision, N.V.; project administration, N.V.; funding acquisition, N.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financed by Fundo Europeu de Desenvolvimento Regional (FEDER) funds through the COMPETE 2020 Operational Programme for Competitiveness and Internationaliza-tion (POCI), Portugal 2020, and by Portuguese funds through Fundação para a Ciência e a Tecnologia (FCT) in the framework of projects IF/00092/2014/CP1255/CT0004 and CHAIR in On-co-Innovation from the Faculty of Medicine, University of Porto (FMUP).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

A.S.C. acknowledges FCT for her PhD grant (SFRH/BD/146093/2019).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. WHO. Depressive Disorder (Depression). Available online: https://www.who.int/news-room/fact-sheets/detail/depression (accessed on 28 June 2023).
  2. Otte, C.; Gold, S.M.; Penninx, B.W.; Pariante, C.M.; Etkin, A.; Fava, M.; Mohr, D.C.; Schatzberg, A.F. Major depressive disorder. Nat. Rev. Dis. Prim. 2016, 2, 16065. [Google Scholar] [CrossRef]
  3. Kennedy, S.H. Core symptoms of major depressive disorder: Relevance to diagnosis and treatment. Dialogues Clin. Neurosci. 2008, 10, 271–277. [Google Scholar] [CrossRef]
  4. Major depressive disorder. Nat. Rev. Dis. Prim. 2023, 9, 45. [CrossRef]
  5. Cuijpers, P. The Challenges of Improving Treatments for Depression. JAMA 2018, 320, 2529. [Google Scholar] [CrossRef] [PubMed]
  6. Maj, M.; Stein, D.J.; Parker, G.; Zimmerman, M.; Fava, G.A.; De Hert, M.; Demyttenaere, K.; McIntyre, R.S.; Widiger, T.; Wittchen, H. The clinical characterization of the adult patient with depression aimed at personalization of management. World Psychiatry 2020, 19, 269–293. [Google Scholar] [CrossRef]
  7. Jesulola, E.; Micalos, P.; Baguley, I.J. Understanding the pathophysiology of depression: From monoamines to the neurogenesis hypothesis model—Are we there yet? Behav. Brain Res. 2018, 341, 79–90. [Google Scholar] [CrossRef]
  8. Burtscher, J.; Niedermeier, M.; Hüfner, K.; van den Burg, E.; Kopp, M.; Stoop, R.; Burtscher, M.; Gatterer, H.; Millet, G.P. The interplay of hypoxic and mental stress: Implications for anxiety and depressive disorders. Neurosci. Biobehav. Rev. 2022, 138, 104718. [Google Scholar] [CrossRef] [PubMed]
  9. Zhang, F.; Peng, W.; Sweeney, J.A.; Jia, Z.; Gong, Q. Brain structure alterations in depression: Psychoradiological evidence. CNS Neurosci. Ther. 2018, 24, 994–1003. [Google Scholar] [CrossRef]
  10. Gradin, V.B.; Pomi, A. The Role of Hippocampal Atrophy in Depression: A Neurocomputational Approach. J. Biol. Phys. 2008, 34, 107–120. [Google Scholar] [CrossRef] [PubMed]
  11. Videbech, P. Hippocampal Volume and Depression: A Meta-Analysis of MRI Studies. Am. J. Psychiatry 2004, 161, 1957–1966. [Google Scholar] [CrossRef]
  12. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef] [PubMed]
  13. Carraro, E.; Schilirò, T.; Biorci, F.; Romanazzi, V.; Degan, R.; Buonocore, D.; Verri, M.; Dossena, M.; Bonetta, S.; Gilli, G. Physical Activity, Lifestyle Factors and Oxidative Stress in Middle Age Healthy Subjects. Int. J. Environ. Res. Public Health 2018, 15, 1152. [Google Scholar] [CrossRef] [PubMed]
  14. Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef]
  15. Correia, A.S.; Cardoso, A.; Vale, N. Oxidative Stress in Depression: The Link with the Stress Response, Neuroinflammation, Serotonin, Neurogenesis and Synaptic Plasticity. Antioxidants 2023, 12, 470. [Google Scholar] [CrossRef]
  16. Sies, H. Oxidative Stress: Concept and Some Practical Aspects. Antioxidants 2020, 9, 852. [Google Scholar] [CrossRef]
  17. Deponte, M. Glutathione catalysis and the reaction mechanisms of glutathione-dependent enzymes. Biochim. Biophys. Acta-Gen. Subj. 2013, 1830, 3217–3266. [Google Scholar] [CrossRef]
  18. Sindhu, R.K.; Kaur, P.; Kaur, P.; Singh, H.; Batiha, G.E.S.; Verma, I. Exploring multifunctional antioxidants as potential agents for management of neurological disorders. Environ. Sci. Pollut. Res. Int. 2022, 29, 24458–24477. [Google Scholar] [CrossRef] [PubMed]
  19. Liu, N.; Wang, Z.-Z.; Zhao, M.; Zhang, Y.; Chen, N.-H. Role of non-coding RNA in the pathogenesis of depression. Gene 2020, 735, 144276. [Google Scholar] [CrossRef]
  20. Bhatt, S.; Nagappa, A.N.; Patil, C.R. Role of oxidative stress in depression. Drug Discov. Today 2020, 25, 1270–1276. [Google Scholar] [CrossRef]
  21. Ji, N.; Lei, M.; Chen, Y.; Tian, S.; Li, C.; Zhang, B. How Oxidative Stress Induces Depression? ASN Neuro 2023, 15. [Google Scholar] [CrossRef]
  22. Ferriani, L.O.; Silva, D.A.; Molina, M.d.C.B.; Mill, J.G.; Brunoni, A.R.; da Fonseca, M.d.J.M.; Moreno, A.B.; Benseñor, I.M.; de Aguiar, O.B.; Barreto, S.M.; et al. Associations of depression and intake of antioxidants and vitamin B complex: Results of the Brazilian Longitudinal Study of Adult Health (ELSA-Brasil). J. Affect. Disord. 2022, 297, 259–268. [Google Scholar] [CrossRef]
  23. Lanctot, K.L.; Mazereeuw, G.; Herrmann, N.; Andreazza, A.; Khan, M. A meta-analysis of lipid peroxidation markers in major depression. Neuropsychiatr. Dis. Treat. 2015, 11, 2479–2491. [Google Scholar] [CrossRef]
  24. Yager, S.; Forlenza, M.J.; Miller, G.E. Depression and oxidative damage to lipids. Psychoneuroendocrinology 2010, 35, 1356–1362. [Google Scholar] [CrossRef]
  25. Camkurt, M.A.; Fındıklı, E.; İzci, F.; Kurutaş, E.B.; Tuman, T.C. Evaluation of malondialdehyde, superoxide dismutase and catalase activity and their diagnostic value in drug naïve, first episode, non-smoker major depression patients and healthy controls. Psychiatry Res. 2016, 238, 81–85. [Google Scholar] [CrossRef] [PubMed]
  26. McGarry, T.; Biniecka, M.; Veale, D.J.; Fearon, U. Hypoxia, oxidative stress and inflammation. Free Radic. Biol. Med. 2018, 125, 15–24. [Google Scholar] [CrossRef] [PubMed]
  27. Silva, D.; Rocha, R.; Correia, A.S.; Mota, B.; Madeira, M.D.; Vale, N.; Cardoso, A. Repurposed Edaravone, Metformin, and Perampanel as a Potential Treatment for Hypoxia–Ischemia Encephalopathy: An In Vitro Study. Biomedicines 2022, 10, 3043. [Google Scholar] [CrossRef] [PubMed]
  28. Liu, T.; Zhong, S.; Liao, X.; Chen, J.; He, T.; Lai, S.; Jia, Y. A Meta-Analysis of Oxidative Stress Markers in Depression. PLoS ONE 2015, 10, e0138904. [Google Scholar] [CrossRef] [PubMed]
  29. Khanzode, S.D.; Dakhale, G.N.; Khanzode, S.S.; Saoji, A.; Palasodkar, R. Oxidative damage and major depression: The potential antioxidant action of selective serotonin re-uptake inhibitors. Redox Rep. 2003, 8, 365–370. [Google Scholar] [CrossRef] [PubMed]
  30. Correia, A.S.; Fraga, S.; Teixeira, J.P.; Vale, N. Cell Model of Depression: Reduction of Cell Stress with Mirtazapine. Int. J. Mol. Sci. 2022, 23, 4942. [Google Scholar] [CrossRef] [PubMed]
  31. Correia, A.S.; Cardoso, A.; Vale, N. Significant Differences in the Reversal of Cellular Stress Induced by Hydrogen Peroxide and Corticosterone by the Application of Mirtazapine or L-Tryptophan. Int. J. Transl. Med. 2022, 2, 482–505. [Google Scholar] [CrossRef]
  32. DeMorrow, S. Role of the Hypothalamic–Pituitary–Adrenal Axis in Health and Disease. Int. J. Mol. Sci. 2018, 19, 986. [Google Scholar] [CrossRef]
  33. Frigerio, F. The HPA Axis and the Regulation of Energy Balance. In Cellular Physiology and Metabolism of Physical Exercise; Luzi, L., Ed.; Springer: Milan, Italy, 2012; pp. 109–121. ISBN 978-88-470-2418-2. [Google Scholar]
  34. Smith, S.M.; Vale, W.W. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin. Neurosci. 2006, 8, 383–395. [Google Scholar] [CrossRef] [PubMed]
  35. Hemmerle, A.M.; Herman, J.P.; Seroogy, K.B. Stress, depression and Parkinson’s disease. Exp. Neurol. 2012, 233, 79–86. [Google Scholar] [CrossRef]
  36. Su, Y.A.; Lin, J.Y.; Liu, Q.; Lv, X.Z.; Wang, G.; Wei, J.; Zhu, G.; Chen, Q.L.; Tian, H.J.; Zhang, K.R.; et al. Associations among serum markers of inflammation, life stress and suicide risk in patients with major depressive disorder. J. Psychiatr. Res. 2020, 129, 53–60. [Google Scholar] [CrossRef] [PubMed]
  37. Dziurkowska, E.; Wesolowski, M. Cortisol as a Biomarker of Mental Disorder Severity. J. Clin. Med. 2021, 10, 5204. [Google Scholar] [CrossRef]
  38. Russell, G.; Lightman, S. The human stress response. Nat. Rev. Endocrinol. 2019, 15, 525–534. [Google Scholar] [CrossRef]
  39. Vaz, R.P.; Cardoso, A.; Serrão, P.; Pereira, P.A.; Madeira, M.D. Chronic stress leads to long-lasting deficits in olfactory-guided behaviors, and to neuroplastic changes in the nucleus of the lateral olfactory tract. Horm. Behav. 2018, 98, 130–144. [Google Scholar] [CrossRef] [PubMed]
  40. Law, R.; Clow, A. Stress, the cortisol awakening response and cognitive function. Int. Rev. Neurobiol. 2020, 150, 187–217. [Google Scholar]
  41. McEwen, B.S. Physiology and Neurobiology of Stress and Adaptation: Central Role of the Brain. Physiol. Rev. 2007, 87, 873–904. [Google Scholar] [CrossRef]
  42. de Kloet, E.R.; Vreugdenhil, E.; Oitzl, M.S.; Joëls, M. Brain Corticosteroid Receptor Balance in Health and Disease. Endocr. Rev. 1998, 19, 269–301. [Google Scholar] [CrossRef]
  43. Dallman, M.F. Fast glucocorticoid actions on brain: Back to the future. Front. Neuroendocrinol. 2005, 26, 103–108. [Google Scholar] [CrossRef]
  44. Groeneweg, F.L.; Karst, H.; de Kloet, E.R.; Joëls, M. Rapid non-genomic effects of corticosteroids and their role in the central stress response. J. Endocrinol. 2011, 209, 153–167. [Google Scholar] [CrossRef]
  45. Pariante, C.M.; Lightman, S.L. The HPA axis in major depression: Classical theories and new developments. Trends Neurosci. 2008, 31, 464–468. [Google Scholar] [CrossRef]
  46. Pariante, C.M. Why are depressed patients inflamed? A reflection on 20 years of research on depression, glucocorticoid resistance and inflammation. Eur. Neuropsychopharmacol. 2017, 27, 554–559. [Google Scholar] [CrossRef]
  47. Menke, A. Is the HPA axis as target for depression outdated, or is there a new hope? Front. Psychiatry 2019, 10, 101. [Google Scholar] [CrossRef]
  48. Anacker, C.; Zunszain, P.A.; Carvalho, L.A.; Pariante, C.M. The glucocorticoid receptor: Pivot of depression and of antidepressant treatment? Psychoneuroendocrinology 2011, 36, 415–425. [Google Scholar] [CrossRef]
  49. Leonard, B.E. The concept of depression as a dysfunction of the immune system. In Depression: From Psychopathology to Pharmacotherapy; S. Karger AG: Basel, Switzerland, 2010; Volume 27, pp. 53–71. ISBN 9783805596060. [Google Scholar]
  50. Belleau, E.L.; Treadway, M.T.; Pizzagalli, D.A. The Impact of Stress and Major Depressive Disorder on Hippocampal and Medial Prefrontal Cortex Morphology. Biol. Psychiatry 2019, 85, 443–453. [Google Scholar] [CrossRef]
  51. Song, Y.; Miyaki, K.; Suzuki, T.; Sasaki, Y.; Tsutsumi, A.; Kawakami, N.; Shimazu, A.; Takahashi, M.; Inoue, A.; Kan, C.; et al. Altered DNA methylation status of human brain derived neurotrophis factor gene could be useful as biomarker of depression. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2014, 165, 357–364. [Google Scholar] [CrossRef]
  52. Sapolsky, R.; Krey, L.; McEwen, B. Prolonged glucocorticoid exposure reduces hippocampal neuron number: Implications for aging. J. Neurosci. 1985, 5, 1222–1227. [Google Scholar] [CrossRef]
  53. Tian, L.; Hui, C.W.; Bisht, K.; Tan, Y.; Sharma, K.; Chen, S.; Zhang, X.; Tremblay, M.-E. Microglia under psychosocial stressors along the aging trajectory: Consequences on neuronal circuits, behavior, and brain diseases. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2017, 79, 27–39. [Google Scholar] [CrossRef]
  54. Banasr, M.; Dwyer, J.M.; Duman, R.S. Cell atrophy and loss in depression: Reversal by antidepressant treatment. Curr. Opin. Cell Biol. 2011, 23, 730–737. [Google Scholar] [CrossRef]
  55. Ait Tayeb, A.E.K.; Poinsignon, V.; Chappell, K.; Bouligand, J.; Becquemont, L.; Verstuyft, C. Major Depressive Disorder and Oxidative Stress: A Review of Peripheral and Genetic Biomarkers According to Clinical Characteristics and Disease Stages. Antioxidants 2023, 12, 942. [Google Scholar] [CrossRef]
  56. Trifunovic, S.; Stevanovic, I.; Milosevic, A.; Ristic, N.; Janjic, M.; Bjelobaba, I.; Savic, D.; Bozic, I.; Jakovljevic, M.; Tesovic, K.; et al. The Function of the Hypothalamic–Pituitary–Adrenal Axis during Experimental Autoimmune Encephalomyelitis: Involvement of Oxidative Stress Mediators. Front. Neurosci. 2021, 15, 649485. [Google Scholar] [CrossRef]
  57. Du, J.; Wang, Y.; Hunter, R.; Wei, Y.; Blumenthal, R.; Falke, C.; Khairova, R.; Zhou, R.; Yuan, P.; Machado-Vieira, R.; et al. Dynamic regulation of mitochondrial function by glucocorticoids. Proc. Natl. Acad. Sci. USA 2009, 106, 3543–3548. [Google Scholar] [CrossRef]
  58. Spiers, J.G.; Chen, H.-J.C.; Sernia, C.; Lavidis, N.A. Activation of the hypothalamic-pituitary-adrenal stress axis induces cellular oxidative stress. Front. Neurosci. 2015, 8, 456. [Google Scholar] [CrossRef]
  59. Correia, A.S.; Cardoso, A.; Vale, N. Highlighting Immune System and Stress in Major Depressive Disorder, Parkinson’s, and Alzheimer’s Diseases, with a Connection with Serotonin. Int. J. Mol. Sci. 2021, 22, 8525. [Google Scholar] [CrossRef]
  60. Amasi-Hartoonian, N.; Sforzini, L.; Cattaneo, A.; Pariante, C.M. Cause or consequence? Understanding the role of cortisol in the increased inflammation observed in depression. Curr. Opin. Endocr. Metab. Res. 2022, 24, 100356. [Google Scholar] [CrossRef]
  61. Weber, M.D.; Godbout, J.P.; Sheridan, J.F. Repeated Social Defeat, Neuroinflammation, and Behavior: Monocytes Carry the Signal. Neuropsychopharmacology 2017, 42, 46–61. [Google Scholar] [CrossRef]
  62. Horowitz, M.A.; Zunszain, P.A.; Anacker, C.; Musaelyan, K.; Pariante, C.M. Glucocorticoids and Inflammation: A Double-Headed Sword in Depression? In Inflammation in Psychiatry; Halaris, A., Leonard, B.E., Eds.; S. Karger AG: Basel, Switzerland, 2013; Volume 28, pp. 127–143. ISBN 978-3-318-02310-7. [Google Scholar]
  63. Houwing, D.J.; Buwalda, B.; van der Zee, E.A.; de Boer, S.F.; Olivier, J.D.A. The Serotonin Transporter and Early Life Stress: Translational Perspectives. Front. Cell. Neurosci. 2017, 11, 117. [Google Scholar] [CrossRef]
  64. Dantzer, R. Cytokine, Sickness Behavior, and Depression. Immunol. Allergy Clin. N. Am. 2009, 29, 247–264. [Google Scholar] [CrossRef]
  65. Qing, H.; Desrouleaux, R.; Israni-Winger, K.; Mineur, Y.S.; Fogelman, N.; Zhang, C.; Rashed, S.; Palm, N.W.; Sinha, R.; Picciotto, M.R.; et al. Origin and Function of Stress-Induced IL-6 in Murine Models. Cell 2020, 182, 372–387.e14. [Google Scholar] [CrossRef]
  66. Goshen, I.; Yirmiya, R. Interleukin-1 (IL-1): A central regulator of stress responses. Front. Neuroendocrinol. 2009, 30, 30–45. [Google Scholar] [CrossRef]
  67. Ting, E.Y.-C.; Yang, A.C.; Tsai, S.-J. Role of Interleukin-6 in Depressive Disorder. Int. J. Mol. Sci. 2020, 21, 2194. [Google Scholar] [CrossRef]
  68. Platten, M.; Nollen, E.A.A.; Röhrig, U.F.; Fallarino, F.; Opitz, C.A. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat. Rev. Drug Discov. 2019, 18, 379–401. [Google Scholar] [CrossRef] [PubMed]
  69. Berger, M.; Gray, J.A.; Roth, B.L. The Expanded Biology of Serotonin. Annu. Rev. Med. 2009, 60, 355–366. [Google Scholar] [CrossRef]
  70. Comai, S.; Bertazzo, A.; Brughera, M.; Crotti, S. Tryptophan in health and disease. Adv. Clin. Chem. 2020, 95, 165–218. [Google Scholar]
  71. Hsu, C.-N.; Tain, Y.-L. Developmental Programming and Reprogramming of Hypertension and Kidney Disease: Impact of Tryptophan Metabolism. Int. J. Mol. Sci. 2020, 21, 8705. [Google Scholar] [CrossRef]
  72. Höglund, E.; Øverli, Ø.; Winberg, S. Tryptophan Metabolic Pathways and Brain Serotonergic Activity: A Comparative Review. Front. Endocrinol. 2019, 10, 158. [Google Scholar] [CrossRef]
  73. Correia, A.S.; Vale, N. Tryptophan Metabolism in Depression: A Narrative Review with a Focus on Serotonin and Kynurenine Pathways. Int. J. Mol. Sci. 2022, 23, 8493. [Google Scholar] [CrossRef] [PubMed]
  74. Correia, A.S.; Silva, I.; Reguengo, H.; Oliveira, J.C.; Vasques-Nóvoa, F.; Cardoso, A.; Vale, N. The Effect of the Stress Induced by Hydrogen Peroxide and Corticosterone on Tryptophan Metabolism, Using Human Neuroblastoma Cell Line (SH-SY5Y). Int. J. Mol. Sci. 2023, 24, 4389. [Google Scholar] [CrossRef]
  75. Mohammad-Zadeh, L.F.; Moses, L.; Gwaltney-Brant, S.M. Serotonin: A review. J. Vet. Pharmacol. Ther. 2008, 31, 187–199. [Google Scholar] [CrossRef]
  76. Marin, P.; Bécamel, C.; Chaumont-Dubel, S.; Vandermoere, F.; Bockaert, J.; Claeysen, S. Chapter 5—Classification and signaling characteristics of 5-HT receptors: Toward the concept of 5-HT receptosomes. Handb. Behav. Neurosci. 2020, 31, 91–120. [Google Scholar]
  77. Pytliak, M.; Vargová, V.; Mechírová, V.; Felšöci, M. Serotonin Receptors—From Molecular Biology to Clinical Applications. Physiol. Res. 2011, 60, 15–25. [Google Scholar] [CrossRef]
  78. Overview—Antidepressants—NHS. Available online: https://www.nhs.uk/mental-health/talking-therapies-medicine-treatments/medicines-and-psychiatry/antidepressants/overview/ (accessed on 11 October 2023).
  79. Cowen, P.J.; Browning, M. What has serotonin to do with depression? World Psychiatry 2015, 14, 158–160. [Google Scholar] [CrossRef]
  80. Amidfar, M.; Colic, L.; Walter, M.; Kim, Y.-K. Complex Role of the Serotonin Receptors in Depression: Implications for Treatment BT. In Understanding Depression: Volume 1. Biomedical and Neurobiological Background; Kim, Y.-K., Ed.; Springer: Singapore, 2018; pp. 83–95. ISBN 978-981-10-6580-4. [Google Scholar]
  81. Nautiyal, K.M.; Hen, R. Serotonin receptors in depression: From A to B. F1000Research 2017, 6, 123. [Google Scholar] [CrossRef]
  82. Correia, A.S.; Silva, I.; Oliveira, J.C.; Reguengo, H.; Vale, N. Serotonin Type 3 Receptor Is Potentially Involved in Cellular Stress Induced by Hydrogen Peroxide. Life 2022, 12, 1645. [Google Scholar] [CrossRef] [PubMed]
  83. Smith, A.L.W.; Harmer, C.J.; Cowen, P.J.; Murphy, S.E. The Serotonin 1A (5-HT1A) Receptor as a Pharmacological Target in Depression. CNS Drugs 2023, 37, 571–585. [Google Scholar] [CrossRef] [PubMed]
  84. Yohn, C.N.; Gergues, M.M.; Samuels, B.A. The role of 5-HT receptors in depression. Mol. Brain 2017, 10, 28. [Google Scholar] [CrossRef] [PubMed]
  85. Zhang, G.; Stackman, R.W., Jr. The role of serotonin 5-HT 2A receptors in memory and cognition. Front. Pharmacol. 2015, 6, 225. [Google Scholar] [CrossRef]
  86. Bhatt, S.; Devadoss, T.; Manjula, S.N.; Rajangam, J. 5-HT3 Receptor Antagonism: A Potential Therapeutic Approach for the Treatment of Depression and other Disorders. Curr. Neuropharmacol. 2021, 19, 1545–1559. [Google Scholar] [CrossRef]
  87. Albert, P.; Le François, B. Modifying 5-HT1A Receptor Gene Expression as a New Target for Antidepressant Therapy. Front. Neurosci. 2010, 4, 35. [Google Scholar] [CrossRef]
  88. Kaufman, J.; DeLorenzo, C.; Choudhury, S.; Parsey, R. V The 5-HT1A receptor in Major Depressive Disorder. Eur. Neuropsychopharmacol. 2016, 26, 397–410. [Google Scholar] [CrossRef] [PubMed]
  89. Guiard, B.P.; Giovanni, G. Di Central serotonin-2A (5-HT2A) receptor dysfunction in depression and epilepsy: The missing link? Front. Pharmacol. 2015, 6, 46. [Google Scholar] [CrossRef]
  90. Göőz, M.; Göőz, P.; Luttrell, L.M.; Raymond, J.R. 5-HT2A Receptor Induces ERK Phosphorylation and Proliferation through ADAM-17 Tumor Necrosis Factor-α-converting Enzyme (TACE) Activation and Heparin-bound Epidermal Growth Factor-like Growth Factor (HB-EGF) Shedding in Mesangial Cells. J. Biol. Chem. 2006, 281, 21004–21012. [Google Scholar] [CrossRef] [PubMed]
  91. Wang, J.Q.; Mao, L. The ERK Pathway: Molecular Mechanisms and Treatment of Depression. Mol. Neurobiol. 2019, 56, 6197–6205. [Google Scholar] [CrossRef] [PubMed]
  92. Shukla, A.K.; Dwivedi-Agnihotri, H. Structure and function of β-arrestins, their emerging role in breast cancer, and potential opportunities for therapeutic manipulation. Adv. Cancer Res. 2020, 145, 139–156. [Google Scholar] [PubMed]
  93. Schmid, C.L.; Raehal, K.M.; Bohn, L.M. Agonist-directed signaling of the serotonin 2A receptor depends on β-arrestin-2 interactions in vivo. Proc. Natl. Acad. Sci. USA 2008, 105, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
  94. Abbas, A.; Roth, B.L. Arresting serotonin. Proc. Natl. Acad. Sci. USA 2008, 105, 831–832. [Google Scholar] [CrossRef] [PubMed]
  95. Felsing, D.E.; Nilson, A.; Jain, M.; Raval, S.; Inoue, A.; Allen, J. β-arrestins mediate rapid 5-HT2A receptor endocytosis to regulate intensity and duration of signaling. FASEB J. 2019, 33, 502.3. [Google Scholar] [CrossRef]
  96. Peters, J.A.; Lambert, J.J. Electrophysiology of 5-HT3 receptors in neuronal cell lines. Trends Pharmacol. Sci. 1989, 10, 172–175. [Google Scholar] [CrossRef]
  97. Bétry, C.; Etiévant, A.; Oosterhof, C.; Ebert, B.; Sanchez, C.; Haddjeri, N. Role of 5-HT 3 Receptors in the Antidepressant Response. Pharmaceuticals 2011, 4, 603–629. [Google Scholar] [CrossRef]
  98. van Hooft, J. 5-HT3 receptors and neurotransmitter release in the CNS: A nerve ending story? Trends Neurosci. 2000, 23, 605–610. [Google Scholar] [CrossRef]
  99. Kawamoto, E.M.; Vivar, C.; Camandola, S. Physiology and Pathology of Calcium Signaling in the Brain. Front. Pharmacol. 2012, 3, 61. [Google Scholar] [CrossRef]
  100. Moncrieff, J.; Cooper, R.E.; Stockmann, T.; Amendola, S.; Hengartner, M.P.; Horowitz, M.A. The serotonin theory of depression: A systematic umbrella review of the evidence. Mol. Psychiatry 2022, 28, 3243–3256. [Google Scholar] [CrossRef]
  101. Jauhar, S.; Arnone, D.; Baldwin, D.S.; Bloomfield, M.; Browning, M.; Cleare, A.J.; Corlett, P.; Deakin, J.F.W.; Erritzoe, D.; Fu, C.; et al. A leaky umbrella has little value: Evidence clearly indicates the serotonin system is implicated in depression. Mol. Psychiatry 2023, 28, 3149–3152. [Google Scholar] [CrossRef] [PubMed]
  102. Jauhar, S.; Cowen, P.J.; Browning, M. Fifty years on: Serotonin and depression. J. Psychopharmacol. 2023, 37, 237–241. [Google Scholar] [CrossRef]
  103. Ruhé, H.G.; Mason, N.S.; Schene, A.H. Mood is indirectly related to serotonin, norepinephrine and dopamine levels in humans: A meta-analysis of monoamine depletion studies. Mol. Psychiatry 2007, 12, 331–359. [Google Scholar] [CrossRef] [PubMed]
  104. Schirò, G.; Iacono, S.; Ragonese, P.; Aridon, P.; Salemi, G.; Balistreri, C.R. A Brief Overview on BDNF-Trk Pathway in the Nervous System: A Potential Biomarker or Possible Target in Treatment of Multiple Sclerosis? Front. Neurol. 2022, 13, 917527. [Google Scholar] [CrossRef]
  105. Correia, A.S.; Cardoso, A.; Vale, N. BDNF Unveiled: Exploring Its Role in Major Depression Disorder Serotonergic Imbalance and Associated Stress Conditions. Pharmaceutics 2023, 15, 2081. [Google Scholar] [CrossRef]
  106. Rentería, I.; García-Suárez, P.C.; Fry, A.C.; Moncada-Jiménez, J.; Machado-Parra, J.P.; Antunes, B.M.; Jiménez-Maldonado, A. The Molecular Effects of BDNF Synthesis on Skeletal Muscle: A Mini-Review. Front. Physiol. 2022, 13, 1345. [Google Scholar] [CrossRef] [PubMed]
  107. Yang, B.; Ren, Q.; Zhang, J.C.; Chen, Q.X.; Hashimoto, K. Altered expression of BDNF, BDNF pro-peptide and their precursor proBDNF in brain and liver tissues from psychiatric disorders: Rethinking the brain–liver axis. Transl. Psychiatry 2017, 7, e1128. [Google Scholar] [CrossRef]
  108. Brigadski, T.; Leßmann, V. BDNF: A regulator of learning and memory processes with clinical potential. e-Neuroforum 2014, 5, 1–11. [Google Scholar] [CrossRef]
  109. Wang, C.S.; Kavalali, E.T.; Monteggia, L.M. BDNF signaling in context: From synaptic regulation to psychiatric disorders. Cell 2022, 185, 62–76. [Google Scholar] [CrossRef]
  110. Pruunsild, P.; Kazantseva, A.; Aid, T.; Palm, K.; Timmusk, T. Dissecting the human BDNF locus: Bidirectional transcription, complex splicing, and multiple promoters. Genomics 2007, 90, 397–406. [Google Scholar] [CrossRef]
  111. Nair, B.; Wong-Riley, M.T.T. Transcriptional Regulation of Brain-derived Neurotrophic Factor Coding Exon IX. J. Biol. Chem. 2016, 291, 22583–22593. [Google Scholar] [CrossRef]
  112. Pathak, H.; Borchert, A.; Garaali, S.; Burkert, A.; Frieling, H. BDNF exon IV promoter methylation and antidepressant action: A complex interplay. Clin. Epigenetics 2022, 14, 187. [Google Scholar] [CrossRef]
  113. Oh, H.; Lewis, D.A.; Sibille, E. The Role of BDNF in Age-Dependent Changes of Excitatory and Inhibitory Synaptic Markers in the Human Prefrontal Cortex. Neuropsychopharmacology 2016, 41, 3080–3091. [Google Scholar] [CrossRef]
  114. Bathina, S.; Das, U.N. Brain-derived neurotrophic factor and its clinical implications. Arch. Med. Sci. 2015, 11, 1164–1178. [Google Scholar] [CrossRef]
  115. Cohen-Cory, S.; Kidane, A.H.; Shirkey, N.J.; Marshak, S. Brain-derived neurotrophic factor and the development of structural neuronal connectivity. Dev. Neurobiol. 2010, 70, 271–288. [Google Scholar] [CrossRef]
  116. Miranda, M.; Morici, J.F.; Zanoni, M.B.; Bekinschtein, P. Brain-Derived Neurotrophic Factor: A Key Molecule for Memory in the Healthy and the Pathological Brain. Front. Cell. Neurosci. 2019, 13, 363. [Google Scholar] [CrossRef]
  117. Paduchová, Z.; Katrenčíková, B.; Vaváková, M.; Laubertová, L.; Nagyová, Z.; Garaiova, I.; Zdenkǎduračková, Z.Z.; Trebatická, J. The Effect of Omega-3 Fatty Acids on Thromboxane, Brain-Derived Neurotrophic Factor, Homocysteine, and Vitamin D in Depressive Children and Adolescents: Randomized Controlled Trial. Nutrients 2021, 13, 1095. [Google Scholar] [CrossRef]
  118. Martinowich, K.; Manji, H.; Lu, B. New insights into BDNF function in depression and anxiety. Nat. Neurosci. 2007, 10, 1089–1093. [Google Scholar] [CrossRef]
  119. Azman, K.F.; Zakaria, R. Recent Advances on the Role of Brain-Derived Neurotrophic Factor (BDNF) in Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 6827. [Google Scholar] [CrossRef]
  120. Erickson, K.I.; Voss, M.W.; Prakash, R.S.; Basak, C.; Szabo, A.; Chaddock, L.; Kim, J.S.; Heo, S.; Alves, H.; White, S.M.; et al. Exercise training increases size of hippocampus and improves memory. Proc. Natl. Acad. Sci. USA 2011, 108, 3017–3022. [Google Scholar] [CrossRef] [PubMed]
  121. You, H.; Lu, B. Diverse Functions of Multiple Bdnf Transcripts Driven by Distinct Bdnf Promoters. Biomolecules 2023, 13, 655. [Google Scholar] [CrossRef]
  122. Yoshimura, R.; Okamoto, N.; Chibaatar, E.; Natsuyama, T.; Ikenouchi, A. The Serum Brain-Derived Neurotrophic Factor Increases in Serotonin Reuptake Inhibitor Responders Patients with First-Episode, Drug-Naïve Major Depression. Biomedicines 2023, 11, 584. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, B.; Dowlatshahi, D.; MacQueen, G.M.; Wang, J.F.; Young, L.T. Increased hippocampal bdnf immunoreactivity in subjects treated with antidepressant medication. Biol. Psychiatry 2001, 50, 260–265. [Google Scholar] [CrossRef] [PubMed]
  124. Dwivedi, Y. Involvement of Brain-Derived Neurotrophic Factor in Late-Life Depression. Am. J. Geriatr. Psychiatry 2013, 21, 433–449. [Google Scholar] [CrossRef]
  125. Youssef, M.M.; Underwood, M.D.; Huang, Y.-Y.; Hsiung, S.; Liu, Y.; Simpson, N.R.; Bakalian, M.J.; Rosoklija, G.B.; Dwork, A.J.; Arango, V.; et al. Association of BDNF Val66Met Polymorphism and Brain BDNF Levels with Major Depression and Suicide. Int. J. Neuropsychopharmacol. 2018, 21, 528–538. [Google Scholar] [CrossRef]
  126. Kubera, M.; Obuchowicz, E.; Goehler, L.; Brzeszcz, J.; Maes, M. In animal models, psychosocial stress-induced (neuro) inflammation, apoptosis and reduced neurogenesis are associated to the onset of depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2011, 35, 744–759. [Google Scholar] [CrossRef]
  127. Jesse, C.R.; Donato, F.; Giacomeli, R.; Del Fabbro, L.; da Silva Antunes, M.; De Gomes, M.G.; Goes, A.T.R.; Boeira, S.P.; Prigol, M.; Souza, L.C. Chronic unpredictable mild stress decreases BDNF and NGF levels and Na+, K+-ATPase activity in the hippocampus and prefrontal cortex of mice: Antidepressant effect of chrysin. Neuroscience 2015, 289, 367–380. [Google Scholar] [CrossRef]
  128. Park, C.; Kim, J.; Namgung, E.; Lee, D.-W.; Kim, G.H.; Kim, M.; Kim, N.; Kim, T.D.; Kim, S.; Lyoo, I.K. The BDNF Val66Met polymorphism affects the vulnerability of the brain structural network. Front. Hum. Neurosci. 2017, 11, 400. [Google Scholar] [CrossRef]
  129. Pathak, P.; Mehra, A.; Ram, S.; Pal, A.; Grover, S. Association of serum BDNF level and Val66Met polymorphism with response to treatment in patients of major depressive disease: A step towards personalized therapy. Behav. Brain Res. 2022, 430, 113931. [Google Scholar] [CrossRef]
  130. Sleiman, S.F.; Henry, J.; Al-Haddad, R.; El Hayek, L.; Abou Haidar, E.; Stringer, T.; Ulja, D.; Karuppagounder, S.S.; Holson, E.B.; Ratan, R.R.; et al. Exercise promotes the expression of brain derived neurotrophic factor (BDNF) through the action of the ketone body β-hydroxybutyrate. Elife 2016, 5, e15092. [Google Scholar] [CrossRef]
  131. van Praag, H.; Castren, E.; Ieraci, A.; Aprahamian, I.; Arosio, B.; Rosa Guerini, F.; Oude Voshaar, R.C.; Fondazione Don Carlo Gnocchi, I. Blood Brain-Derived Neurotrophic Factor (BDNF) and Major Depression: Do We Have a Translational Perspective? Front. Behav. Neurosci. 2021, 15, 626906. [Google Scholar] [CrossRef]
  132. Homberg, J.R.; Molteni, R.; Calabrese, F.; Riva, M.A. The serotonin–BDNF duo: Developmental implications for the vulnerability to psychopathology. Neurosci. Biobehav. Rev. 2014, 43, 35–47. [Google Scholar] [CrossRef]
  133. Yang, T.; Nie, Z.; Shu, H.; Kuang, Y.; Chen, X.; Cheng, J.; Yu, S.; Liu, H. The Role of BDNF on Neural Plasticity in Depression. Front. Cell. Neurosci. 2020, 14, 82. [Google Scholar] [CrossRef] [PubMed]
  134. Kunugi, H.; Hori, H.; Adachi, N.; Numakawa, T. Interface between hypothalamic-pituitary-adrenal axis and brain-derived neurotrophic factor in depression. Psychiatry Clin. Neurosci. 2010, 64, 447–459. [Google Scholar] [CrossRef]
  135. Li, Z.; Ruan, M.; Chen, J.; Fang, Y. Major Depressive Disorder: Advances in Neuroscience Research and Translational Applications. Neurosci. Bull. 2021, 37, 863–880. [Google Scholar] [CrossRef]
  136. Lopez-Munoz, F.; Alamo, C. Monoaminergic Neurotransmission: The History of the Discovery of Antidepressants from 1950s Until Today. Curr. Pharm. Des. 2009, 15, 1563–1586. [Google Scholar] [CrossRef] [PubMed]
  137. Hillhouse, T.M.; Porter, J.H.; Clin, E.; Author, P. A brief history of the development of antidepressant drugs: From monoamines to glutamate HHS Public Access Author manuscript. Exp. Clin. Psychopharmacol. 2015, 23, 1–21. [Google Scholar] [CrossRef] [PubMed]
  138. Stahl, S.M. Mechanism of action of serotonin selective reuptake inhibitors. J. Affect. Disord. 1998, 51, 215–235. [Google Scholar] [CrossRef]
  139. Sheffler, Z.M.; Patel, P.; Abdijadid, S. Antidepressants; StatPearls: St. Petersburg, FL, USA, 26 May 2023. [Google Scholar]
  140. Kolovos, S.; Kleiboer, A.; Cuijpers, P. Effect of psychotherapy for depression on quality of life: Meta-analysis. Br. J. Psychiatry 2016, 209, 460–468. [Google Scholar] [CrossRef]
  141. Stachowicz, K.; Sowa-Kućma, M. The treatment of depression—Searching for new ideas. Front. Pharmacol. 2022, 13, 988648. [Google Scholar] [CrossRef]
  142. Thase, M.E. Have Effective Antidepressants Finally Arrived? Developments in Major Depressive Disorder Therapy. J. Clin. Psychiatry 2023, 84, 48388. [Google Scholar] [CrossRef]
  143. Khoodoruth, M.A.S.; Estudillo-Guerra, M.A.; Pacheco-Barrios, K.; Nyundo, A.; Chapa-Koloffon, G.; Ouanes, S. Glutamatergic System in Depression and Its Role in Neuromodulatory Techniques Optimization. Front. Psychiatry 2022, 13, 886918. [Google Scholar] [CrossRef]
  144. Sarawagi, A.; Soni, N.D.; Patel, A.B. Glutamate and GABA Homeostasis and Neurometabolism in Major Depressive Disorder. Front. Psychiatry 2021, 12, 637863. [Google Scholar] [CrossRef]
  145. Ghosal, S.; Hare, B.D.; Duman, R.S. Prefrontal cortex GABAergic deficits and circuit dysfunction in the pathophysiology and treatment of chronic stress and depression. Curr. Opin. Behav. Sci. 2017, 14, 1–8. [Google Scholar] [CrossRef]
  146. Lin, J.; Ling, F.; Huang, P.; Chen, M.; Song, M.; Lu, K.; Wang, W. The Development of GABAergic Network in Depression in Recent 17 Years: A Visual Analysis Based on CiteSpace and VOSviewer. Front. Psychiatry 2022, 13, 874137. [Google Scholar] [CrossRef]
  147. Aleksandrova, L.R.; Phillips, A.G.; Wang, Y.T. Antidepressant effects of ketamine and the roles of AMPA glutamate receptors and other mechanisms beyond NMDA receptor antagonism. J. Psychiatry Neurosci. 2017, 42, 222–229. [Google Scholar] [CrossRef]
  148. Berman, R.M.; Cappiello, A.; Anand, A.; Oren, D.A.; Heninger, G.R.; Charney, D.S.; Krystal, J.H. Antidepressant effects of ketamine in depressed patients. Biol. Psychiatry 2000, 47, 351–354. [Google Scholar] [CrossRef]
  149. Popova, V.; Daly, E.J.; Trivedi, M.; Cooper, K.; Lane, R.; Lim, P.; Mazzucco, C.; Hough, D.; Thase, M.E.; Shelton, R.C.; et al. Efficacy and Safety of Flexibly Dosed Esketamine Nasal Spray Combined with a Newly Initiated Oral Antidepressant in Treatment-Resistant Depression: A Randomized Double-Blind Active-Controlled Study. Am. J. Psychiatry 2019, 176, 428–438. [Google Scholar] [CrossRef] [PubMed]
  150. Kavakbasi, E.; Hassan, A.; Baune, B.T. Combination of Electroconvulsive Therapy Alternating with Intravenous Esketamine Can Lead to Rapid Remission of Treatment Resistant Depression. J. ECT 2021, 37, e20–e21. [Google Scholar] [CrossRef]
  151. Ryskamp, D.A.; Korban, S.; Zhemkov, V.; Kraskovskaya, N.; Bezprozvanny, I. Neuronal Sigma-1 Receptors: Signaling Functions and Protective Roles in Neurodegenerative Diseases. Front. Neurosci. 2019, 13, 862. [Google Scholar] [CrossRef]
  152. Pedraz-Petrozzi, B.; Deuschle, M.; Gilles, M. Improvement of depressive symptoms, after a suicide attempt, with dextromethorphan/bupropion combination treatment in a patient with treatment-resistant depression and psychiatric comorbidities. Clin. Case Rep. 2023, 11, e7045. [Google Scholar] [CrossRef] [PubMed]
  153. Blair, H.A. Dextromethorphan/bupropion in major depressive disorder: A profile of its use. Drugs Ther. Perspect. 2023, 39, 270–278. [Google Scholar] [CrossRef]
  154. Fava, M.; Stahl, S.M.; De Martin, S.; Mattarei, A.; Bettini, E.; Comai, S.; Alimonti, A.; Bifari, F.; Pani, L.; Folli, F.; et al. Esmethadone-HCl (REL-1017): A promising rapid antidepressant. Eur. Arch. Psychiatry Clin. Neurosci. 2023, 273, 1463–1476. [Google Scholar] [CrossRef] [PubMed]
  155. Fava, M.; Stahl, S.; Pani, L.; De Martin, S.; Pappagallo, M.; Guidetti, C.; Alimonti, A.; Bettini, E.; Mangano, R.M.; Wessel, T.; et al. REL-1017 (Esmethadone) as Adjunctive Treatment in Patients with Major Depressive Disorder: A Phase 2a Randomized Double-Blind Trial. Am. J. Psychiatry 2022, 179, 122–131. [Google Scholar] [CrossRef] [PubMed]
  156. Cornett, E.M.; Rando, L.; Labbé, A.M.; Perkins, W.; Kaye, A.M.; Kaye, A.D.; Viswanath, O.; Urits, I. Brexanolone to Treat Postpartum Depression in Adult Women. Psychopharmacol. Bull. 2021, 51, 115. [Google Scholar]
  157. Clayton, A.H.; Lasser, R.; Parikh, S.V.; Iosifescu, D.V.; Jung, J.; Kotecha, M.; Forrestal, F.; Jonas, J.; Kanes, S.J.; Doherty, J. Zuranolone for the Treatment of Adults with Major Depressive Disorder: A Randomized, Placebo-Controlled Phase 3 Trial. Am. J. Psychiatry 2023, 180, 676–684. [Google Scholar] [CrossRef]
  158. Fagan, H.; Jones, E.; Baldwin, D.S. Orexin Receptor Antagonists in the Treatment of Depression: A Leading Article Summarising Pre-clinical and Clinical Studies. CNS Drugs 2023, 37, 1–12. [Google Scholar] [CrossRef]
  159. Mi, W.; Di, X.; Wang, Y.; Li, H.; Xu, X.; Li, L.; Wang, H.; Wang, G.; Zhang, K.; Tian, F.; et al. A phase 3, multicenter, double-blind, randomized, placebo-controlled clinical trial to verify the efficacy and safety of ansofaxine (LY03005) for major depressive disorder. Transl. Psychiatry 2023, 13, 163. [Google Scholar] [CrossRef]
  160. Bansal, Y.; Bhandari, R.; Kaur, S.; Kaur, J.; Singh, R.; Kuhad, A. Gepirone hydrochloride: A novel antidepressant with 5-HT1A agonistic properties. Drugs Today 2019, 55, 423–437. [Google Scholar] [CrossRef]
  161. Goodwin, G.M.; Croal, M.; Feifel, D.; Kelly, J.R.; Marwood, L.; Mistry, S.; O’Keane, V.; Peck, S.K.; Simmons, H.; Sisa, C.; et al. Psilocybin for treatment resistant depression in patients taking a concomitant SSRI medication. Neuropsychopharmacology 2023, 48, 1492–1499. [Google Scholar] [CrossRef]
  162. Von Rotz, R.; Schindowski, E.M.; Jungwirth, J.; Schuldt, A.; Rieser, N.M.; Zahoranszky, K.; Seifritz, E.; Nowak, A.; Nowak, P.; Jäncke, L.; et al. Single-dose psilocybin-assisted therapy in major depressive disorder: A placebo-controlled, double-blind, randomised clinical trial. EClinicalMedicine 2022, 56, 101809. [Google Scholar] [CrossRef] [PubMed]
  163. Dulawa, S.C.; Janowsky, D.S. Cholinergic regulation of mood: From basic and clinical studies to emerging therapeutics. Mol. Psychiatry 2019, 24, 694–709. [Google Scholar] [CrossRef]
  164. Fitzgerald, P.J.; Hale, P.J.; Ghimire, A.; Watson, B.O. Repurposing Cholinesterase Inhibitors as Antidepressants? Dose and Stress-Sensitivity May Be Critical to Opening Possibilities. Front. Behav. Neurosci. 2021, 14, 620119. [Google Scholar] [CrossRef]
  165. Sun, J.; Qiu, L.; Zhang, H.; Zhou, Z.; Ju, L.; Yang, J. CRHR1 antagonist alleviates LPS-induced depression-like behaviour in mice. BMC Psychiatry 2023, 23, 17. [Google Scholar] [CrossRef] [PubMed]
  166. Brown, S.J.; Huang, X.-F.; Newell, K.A. The kynurenine pathway in major depression: What we know and where to next. Neurosci. Biobehav. Rev. 2021, 127, 917–927. [Google Scholar] [CrossRef]
  167. Mohammad Sadeghi, H.; Adeli, I.; Mousavi, T.; Daniali, M.; Nikfar, S.; Abdollahi, M. Drug Repurposing for the Management of Depression: Where Do We Stand Currently? Life 2021, 11, 774. [Google Scholar] [CrossRef]
  168. Sizar, O.; Khare, S.; Jamil, R.T.; Talati, R. Statin Medications; StatPearls: St. Petersburg, FL, USA, 2023. [Google Scholar]
  169. Gutlapalli, S.D.; Farhat, H.; Irfan, H.; Muthiah, K.; Pallipamu, N.; Taheri, S.; Thiagaraj, S.S.; Shukla, T.S.; Giva, S.; Penumetcha, S.S. The Anti-Depressant Effects of Statins in Patients with Major Depression Post-Myocardial Infarction: An Updated Review 2022. Cureus 2022, 14, e32323. [Google Scholar] [CrossRef] [PubMed]
  170. De Giorgi, R.; Pesci, N.R.; Rosso, G.; Maina, G.; Cowen, P.J.; Harmer, C.J. The pharmacological bases for repurposing statins in depression: A review of mechanistic studies. Transl. Psychiatry 2023, 13, 253. [Google Scholar] [CrossRef]
  171. Lochner, M.; Thompson, A.J. The muscarinic antagonists scopolamine and atropine are competitive antagonists at 5-HT 3 receptors. Neuropharmacology 2016, 108, 220–228. [Google Scholar] [CrossRef]
  172. Fang, Y.; Guo, P.; Lv, L.; Feng, M.; Wang, H.; Sun, G.; Wang, S.; Qian, M.; Chen, H. Scopolamine augmentation for depressive symptoms and cognitive functions in treatment-resistant depression: A case series. Asian J. Psychiatr. 2023, 82, 103484. [Google Scholar] [CrossRef]
  173. Wang, X.; Zhu, X.; Ji, X.; Yang, J.; Zhou, J. Group-Based Symptom Trajectory of Intramuscular Administration of Scopolamine Augmentation in Moderate to Severe Major Depressive Disorder: A Post-Hoc Analysis. Neuropsychiatr. Dis. Treat. 2023, 19, 1043–1053. [Google Scholar] [CrossRef] [PubMed]
  174. Home|ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ (accessed on 18 October 2023).
  175. Moćko, P.; Śladowska, K.; Kawalec, P.; Babii, Y.; Pilc, A. The Potential of Scopolamine as an Antidepressant in Major Depressive Disorder: A Systematic Review of Randomized Controlled Trials. Biomedicines 2023, 11, 2636. [Google Scholar] [CrossRef]
  176. Chateauvieux, S.; Morceau, F.; Diederich, M. Valproic Acid. In Encyclopedia of Toxicology, 3rd ed.; Springer: Berlin/Heidelberg, Germany, 2014; pp. 905–908. [Google Scholar] [CrossRef]
  177. Brian Chen, Y.-C.; Liang, C.-S.; Wang, L.-J.; Hung, K.-C.; Carvalho, A.F.; Solmi, M.; Vieta, E.; Tseng, P.-T.; Lin, P.-Y.; Tu, Y.-K.; et al. Comparative effectiveness of valproic acid in different serum concentrations for maintenance treatment of bipolar disorder: A retrospective cohort study using target trial emulation framework. eClinicalMedicine 2022, 54, 101678. [Google Scholar] [CrossRef] [PubMed]
  178. Ghabrash, M.F.; Comai, S.; Tabaka, J.; Saint-Laurent, M.; Booij, L.; Gobbi, G. Valproate augmentation in a subgroup of patients with treatment-resistant unipolar depression. World J. Biol. Psychiatry 2016, 17, 165–170. [Google Scholar] [CrossRef]
  179. Yonemoto, L. Lamotrigine. In The Essence of Analgesia and Analgesics; Cambridge University Press: Cambridge, UK, 2010; pp. 306–309. ISBN 9780511841378. [Google Scholar]
  180. Matsuzaka, Y.; Urashima, K.; Sakai, S.; Morimoto, Y.; Kanegae, S.; Kinoshita, H.; Imamura, A.; Ozawa, H. The effectiveness of lamotrigine for persistent depressive disorder: A case report. Neuropsychopharmacol. Rep. 2022, 42, 120–123. [Google Scholar] [CrossRef] [PubMed]
  181. Singh, G.; Can, A.S.; Correa, R. Pioglitazone; StatPearls: St. Petersburg, FL, USA, 2023. [Google Scholar]
  182. Colle, R.; de Larminat, D.; Rotenberg, S.; Hozer, F.; Hardy, P.; Verstuyft, C.; Fève, B.; Corruble, E. Pioglitazone could induce remission in major depression: A meta-analysis. Neuropsychiatr. Dis. Treat. 2016, 13, 9–16. [Google Scholar] [CrossRef] [PubMed]
  183. Watson, K.; Akil, H.; Rasgon, N. Toward a Precision Treatment Approach for Metabolic Depression: Integrating Epidemiology, Neuroscience, and Psychiatry. Biol. Psychiatry Glob. Open Sci. 2023, 3, 623–631. [Google Scholar] [CrossRef]
  184. Zhao, Q.; Wu, X.; Yan, S.; Xie, X.; Fan, Y.; Zhang, J.; Peng, C.; You, Z. The antidepressant-like effects of pioglitazone in a chronic mild stress mouse model are associated with PPARγ-mediated alteration of microglial activation phenotypes. J. Neuroinflamm. 2016, 13, 259. [Google Scholar] [CrossRef]
  185. Beheshti, F.; Hosseini, M.; Hashemzehi, M.; Soukhtanloo, M.; Asghari, A. The effects of PPAR-γ agonist pioglitazone on anxiety and depression-like behaviors in lipopolysaccharide injected rats. Toxin Rev. 2021, 40, 1223–1232. [Google Scholar] [CrossRef]
  186. Rague, J. N-Acetylcysteine. In History of Modern Clinical Toxicology; Elsevier: Amsterdam, The Netherlands, 2022; pp. 201–212. ISBN 9780128222188. [Google Scholar]
  187. Hans, D.; Rengel, A.; Hans, J.; Bassett, D.; Hood, S. N-Acetylcysteine as a novel rapidly acting anti-suicidal agent: A pilot naturalistic study in the emergency setting. PLoS ONE 2022, 17, e0263149. [Google Scholar] [CrossRef]
  188. Brivio, P.; Gallo, M.T.; Gruca, P.; Lason, M.; Litwa, E.; Fumagalli, F.; Papp, M.; Calabrese, F. Chronic N-Acetyl-Cysteine Treatment Enhances the Expression of the Immediate Early Gene Nr4a1 in Response to an Acute Challenge in Male Rats: Comparison with the Antidepressant Venlafaxine. Int. J. Mol. Sci. 2023, 24, 7321. [Google Scholar] [CrossRef]
  189. Nazarian, S.; Akhondi, H. Minocycline; StatPearls: St. Petersburg, FL, USA, 2022. [Google Scholar]
  190. Nettis, M.A. Minocycline in Major Depressive Disorder: And overview with considerations on treatment-resistance and comparisons with other psychiatric disorders. Brain Behav. Immun.-Health 2021, 17, 100335. [Google Scholar] [CrossRef] [PubMed]
  191. Qiu, Y.; Duan, A.; Yin, Z.; Xie, M.; Chen, Z.; Sun, X.; Wang, Z.; Zhang, X. Efficacy and tolerability of minocycline in depressive patients with or without treatment-resistant: A meta-analysis of randomized controlled trials. Front. Psychiatry 2023, 14, 1139273. [Google Scholar] [CrossRef]
  192. Rojewska, E.; Ciapała, K.; Piotrowska, A.; Makuch, W.; Mika, J. Pharmacological Inhibition of Indoleamine 2,3-Dioxygenase-2 and Kynurenine 3-Monooxygenase, Enzymes of the Kynurenine Pathway, Significantly Diminishes Neuropathic Pain in a Rat Model. Front. Pharmacol. 2018, 9, 724. [Google Scholar] [CrossRef] [PubMed]
  193. Tippens, A.S. Nimodipine. In xPharm: The Comprehensive Pharmacology Reference; Elsevier: Amsterdam, The Netherlands, 2007; pp. 1–7. ISBN 9780080552323. [Google Scholar]
  194. Zink, C.F.; Giegerich, M.; Prettyman, G.E.; Carta, K.E.; van Ginkel, M.; O’Rourke, M.P.; Singh, E.; Fuchs, E.J.; Hendrix, C.W.; Zimmerman, E.; et al. Nimodipine improves cortical efficiency during working memory in healthy subjects. Transl. Psychiatry 2020, 10, 372. [Google Scholar] [CrossRef] [PubMed]
  195. Taragano, F.E.; Allegri, R.; Vicario, A.; Bagnatti, P.; Lyketsos, C.G. A double blind, randomized clinical trial assessing the efficacy and safety of augmenting standard antidepressant therapy with nimodipine in the treatment of ‘vascular depression’. Int. J. Geriatr. Psychiatry 2001, 16, 254–260. [Google Scholar] [CrossRef]
  196. Maan, J.S.; Ershadi, M.; Khan, I.; Saadabadi, A. Quetiapine. StatPearls: St. Petersburg, FL, USA, 2023. [Google Scholar]
  197. Tran, K.; Argáez, C. Quetiapine for Major Depressive Disorder: A Review of Clinical Effectiveness, Cost-Effectiveness, and Guidelines; Canadian Agency for Drugs and Technologies in Health: Ottawa, ON, Canada, 2020. [Google Scholar]
  198. Ravindran, N.; McKay, M.; Paric, A.; Johnson, S.; Chandrasena, R.; Abraham, G.; Ravindran, A.V. Randomized, Placebo-Controlled Effectiveness Study of Quetiapine XR in Comorbid Depressive and Anxiety Disorders. J. Clin. Psychiatry 2022, 83, 40248. [Google Scholar] [CrossRef]
  199. Bauer, M.; Pretorius, H.W.; Constant, E.L.; Earley, W.R.; Szamosi, J.; Brecher, M. Extended-Release Quetiapine as Adjunct to an Antidepressant in Patients with Major Depressive Disorder: Results of a Randomized, Placebo-Controlled, Double-Blind Study. J. Clin. Psychiatry 2009, 70, 7032. [Google Scholar] [CrossRef]
  200. Marino, J. Celecoxib. In The Essence of Analgesia and Analgesics; Cambridge University Press: Cambridge, UK, 2010; pp. 238–242. ISBN 9780511841378. [Google Scholar]
  201. Wang, Z.; Wu, Q.; Wang, Q. Effect of celecoxib on improving depression: A systematic review and meta-analysis. World J. Clin. Cases 2022, 10, 7872–7882. [Google Scholar] [CrossRef] [PubMed]
  202. Gędek, A.; Szular, Z.; Antosik, A.Z.; Mierzejewski, P.; Dominiak, M. Celecoxib for Mood Disorders: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Clin. Med. 2023, 12, 3497. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Stress response, oxidative stress, BDNF signaling and 5-HT signaling are crucial to depression development and progression. These systems can be influenced through diet and lifestyle factors, being managed using approaches like pharmacotherapy and psychotherapy.
Figure 1. Stress response, oxidative stress, BDNF signaling and 5-HT signaling are crucial to depression development and progression. These systems can be influenced through diet and lifestyle factors, being managed using approaches like pharmacotherapy and psychotherapy.
Ijtm 04 00010 g001
Table 1. Summary of the types of 5-HT receptors and their inhibitory/excitatory potential [77].
Table 1. Summary of the types of 5-HT receptors and their inhibitory/excitatory potential [77].
ReceptorType/and MechanismPotential
5-HT1A-FGi/o-protein coupled; decrease cellular levels of cAMPInhibitory
5-HT2A, 5-HT2B, 5-HT2CGq/11-protein coupled; increase cellular levels of inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG)Excitatory
5-HT3Ligand-gated Na + and K + cation channel; depolarize plasma membraneExcitatory
5-HT4Gs-protein coupled; increase cellular levels of cAMPExcitatory
5-HT5A-BGi/o-protein coupled; decrease cellular levels of cAMPInhibitory
5-HT6Gs-protein coupled; increase cellular levels of cAMPExcitatory
5-HT7Gs-protein coupled; increase cellular levels of cAMPExcitatory
Table 2. Most studied 5-HT receptors in depression, as well as their connection to this disease.
Table 2. Most studied 5-HT receptors in depression, as well as their connection to this disease.
5-HT ReceptorFunction in Depression
5-HT1AAgonists of 5-HT1A receptors are frequently used in the treatment of depression because they can enhance 5-HT signaling. Stimulation of the 5-HT1A receptor is an existing therapeutic target for treating depression and anxiety, using drugs such as buspirone [83].
5-HT2AThis receptor can control neuronal excitability in most networks involved in depression through interactions with the monoaminergic, GABAergic, and glutamatergic neurotransmissions [84]. Preclinical studies show that 5-HT2A receptor antagonists have antipsychotic and antidepressant properties, whereas agonist ligands possess cognition-enhancing and hallucinogenic properties [85].
5-HT3This is an area of ongoing research. 5-HT3 receptor antagonists inhibit the binding of 5-HT to postsynaptic 5-HT3 receptor and might increase its availability to other receptors like 5-HT1A, 1B and 1D as well as 5-HT2 receptors, producing an antidepressant-like effect [86].
Table 4. Main indication, known mechanism of action and relevance in depression of potential to be repurposed in the context of this disease.
Table 4. Main indication, known mechanism of action and relevance in depression of potential to be repurposed in the context of this disease.
DrugMain Indication and Mechanism of ActionRelevance in Depression
StatinsManagement and treatment of hypercholesteremia. Selective, competitive inhibitor of hydroxymethylglutaryl-CoA (HMG-CoA) reductase [168].Demonstrated antidepressant effects, useful as add-on therapy in patients with cardiovascular disease, with MDD [169]. Beneficial effect through positive actions on 5-HT neurotransmission, neurogenesis, and neuroplasticity, HPA axis regulation and modulation of inflammation [170].
ScopolaminePostoperative nausea and vomiting and motion sickness. Competitive antagonist of 5-HT3 receptors and nonselective muscarinic antagonist [171].Evidence of antidepressant effects in patients with MDD and bipolar depression [172]. Added to antidepressants can effectively relieve the symptoms of patients with severe depression [173]. Currently, 2 clinical trials in MDD and bipolar disorder are ongoing (NCT04719663 and NCT04211961 [174]). Studies in rodents have revealed that the antidepressant-like effects are connected to mTORC1 signaling in the PFC. This activation of mTORC1 seems to be initiated by a glutamate surge in the PFC, resulting from the disinhibition of glutamatergic neurons. This increased glutamate transmission leads to the activation of AMPA receptors, that raises the levels of BDNF, which then stimulates mTORC1 signaling and promotes synaptogenesis processes [175].
Valproic AcidTreatment/management of epilepsy. Mechanism of action not fully understood: Inhibits voltage-gated sodium channels, GABA transaminase, increases the expression and activity of glutamic acid decarboxylase (GAD), inhibits the action of histone deacetylases (HDAC) enzymes, notably HDAC1, modulates the activity of various calcium channels [176].Demonstrated efficacy in preventing mood recurrence and enhancing the quality of life for individuals with bipolar disorder when used as a maintenance therapy [177]. Supplementary use of this drug resulted in significant and sustained clinical enhancement over a prolonged duration in individuals with severe treatment-resistant depression [178].
LamotrigineTreatment/management of epilepsy. Mechanism of action for lamotrigine is not entirely understood; selectively binds and inhibits voltage-gated sodium channels, stabilizing presynaptic membranes and inhibiting presynaptic glutamate release [179].Used off-label for bipolar disorder [179]. Could potentially offer an effective option in addressing individuals with treatment-resistant persistent depressive disorder, being a viable substitute for the combination of antidepressant and benzodiazepine therapies in this disorder [180].
PioglitazoneTreatment of type 2 diabetes mellitus. Peroxisome proliferator-activated receptor (PPAR)-gamma and PPAR-alpha agonist [181].Pioglitazone, alone or as add-on therapy to conventional treatments, could induce remission of depressive episodes [182]. Evidence of enhancing antidepressant response among people with comorbid MDD and type 2 diabetes [183].
Induced the neuroprotective phenotype of microglia in chronic mild stress-treated mice, mediated by PPARγ [184], and antidepressant effect in LPS injected rats [185].
N-acetyl cysteineTherapy for acetaminophen toxicity. Serves as a prodrug to L-cysteine, a precursor to glutathione [186].Evidence as an adjunctive therapy to reduce symptoms of Bipolar Affective Disorder, MDD, and Schizophrenia [187].
Enhanced coping mechanisms, not only for addressing acute stressors but possibly also for mitigating the impact of persistent stress-inducing factors [188].
MinocyclineTetracycline antibiotic, anti-infectious activity against both Gram-positive and Gram-negative bacteria. Bind to the 30S ribosomal subunit of bacteria, preventing protein synthesis [189].Potential novel treatment for treatment-resistant depression [190]. May improve depressive symptoms and augment response to treatment in patients with treatment-resistant depression [191]. Inhibits both the IDO and the p-38 components of inflammation-induced depression [192].
NimodipinePrevent vasospasm secondary to subarachnoid hemorrhage. Blocks voltage-gated L-type calcium channels [193].This drug has been shown to be effective in treating mood symptoms for bipolar and unipolar depression [194]. An old clinical trial revealed that this drug in the context of vascular depression, augmentation of fluoxetine with nimodipine led to better treatment results and lower rates of recurrence [195].
QuetiapineSchizophrenia and acute manic episodes. Antagonist for D2 receptors and 5-HT2A receptors [196].Quetiapine monotherapy in older adults with MDD was found to be effective [197]. Quetiapine augmentation may be a useful intervention for MDD with comorbid anxiety [198]. Adjunctive quetiapine was effective in patients with MDD who had shown an inadequate response to antidepressant treatment [199].
CelecoxibAnalgesic for patients with osteoarthritis and rheumatoid arthritis. Selective inhibition of COX-2 [200].A recent meta-analysis demonstrated that celecoxib could be effective for improving depressive symptoms [201]. Antidepressant efficacy was demonstrated when used as an add-on treatment for MDD and mania, possibly by reducing inflammatory markers [202].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Correia, A.S.; Vale, N. Advancements Exploring Major Depressive Disorder: Insights on Oxidative Stress, Serotonin Metabolism, BDNF, HPA Axis Dysfunction, and Pharmacotherapy Advances. Int. J. Transl. Med. 2024, 4, 176-196. https://doi.org/10.3390/ijtm4010010

AMA Style

Correia AS, Vale N. Advancements Exploring Major Depressive Disorder: Insights on Oxidative Stress, Serotonin Metabolism, BDNF, HPA Axis Dysfunction, and Pharmacotherapy Advances. International Journal of Translational Medicine. 2024; 4(1):176-196. https://doi.org/10.3390/ijtm4010010

Chicago/Turabian Style

Correia, Ana Salomé, and Nuno Vale. 2024. "Advancements Exploring Major Depressive Disorder: Insights on Oxidative Stress, Serotonin Metabolism, BDNF, HPA Axis Dysfunction, and Pharmacotherapy Advances" International Journal of Translational Medicine 4, no. 1: 176-196. https://doi.org/10.3390/ijtm4010010

Article Metrics

Back to TopTop