- Review
- Open access
- Published:
Altered synaptic homeostasis: a key factor in the pathophysiology of depression
Cell & Bioscience volume 15, Article number: 29 (2025)
Abstract
Depression, a widespread psychiatric disorder, is characterized by a diverse array of symptoms such as melancholic mood and anhedonia, imposing a significant burden on both society and individuals. Despite extensive research into the neurobiological foundations of depression, a complete understanding of its complex mechanisms is yet to be attained, and targeted therapeutic interventions remain under development. Synaptic homeostasis, a compensatory feedback mechanism, involves neurons adjusting synaptic strength by regulating pre- or postsynaptic processes. Recent advancements in depression research reveal a crucial association between the disorder and disruptions in synaptic homeostasis within neural regions and circuits pivotal for emotional and cognitive functions. This paper explores the mechanisms governing synaptic homeostasis in depression, focusing on the role of ion channels, the regulation of presynaptic neurotransmitter release, synaptic scaling processes, and essential signaling molecules. By mapping new pathways in the study of synaptic homeostasis as it pertains to depression, this research aims to provide valuable insights for identifying novel therapeutic targets for more effective antidepressant treatments.
Introduction
Depressive disorder, a pervasive mental health condition, is primarily characterized by low mood, anhedonia, lack of motivation, and social deficits. Affecting over 280 million people worldwide [1, 2], it imposes a significant burden on society and individuals alike. Although pharmacological treatments are widely used for managing depressive disorder, they often come with side effects and limitations, such as delayed onset of action, high relapse rates, headaches, sleep disturbances, gastrointestinal issues, sexual dysfunction, withdrawal symptoms, and an increased risk of suicidal ideation. Moreover, these treatments are ineffective for a subset of patients [3,4,5].
Synaptic homeostasis, a crucial compensatory negative feedback mechanism, involves neurons adjusting synaptic strength by regulating presynaptic neurotransmitter release and the expression or localization of ion channels or neurotransmitter receptors on the postsynaptic membrane, thereby counteracting excessive excitation or inhibition [6,7,8,9]. This process operates over hours to days, restoring neurons to their set point and maintaining neural network stability. Recent research has increasingly linked synaptic homeostasis to a range of neuropsychiatric disorders, including autism spectrum disorders, Parkinson’s disease, Alzheimer’s disease, epilepsy, and schizophrenia [10,11,12,13,14].
It has been proposed that depression may stem from disruptions in the homeostatic mechanisms regulating synaptic plasticity [15]. However, the exact pathways linking synaptic homeostasis to depression are not yet fully understood. As research continues to explore the neurobiological underpinnings of depression, the complex relationship between the disorder and synaptic homeostasis is becoming clearer. This review aims to examine the correlation between depression and imbalances in synaptic homeostasis, as well as the potential antidepressant mechanisms of medications that modulate synaptic homeostasis, with the goal of advancing both research and therapeutic strategies for depression.
Synaptic homeostasis as a critical factor in depression
Synapses, the specialized intercellular junctions between neurons or between neurons and other cells, are essential for transmitting information via electrical and chemical signals, thereby constituting the fundamental unit of communication within neural networks [16, 17]. As critical components of the central nervous system (CNS), synapses are particularly susceptible to various stimuli. Stress and depression can result in the reduction of brain region volumes, such as the prefrontal cortex (PFC) and hippocampus, which are pivotal for mood and cognition [18,19,20,21,22], and can also lead to a decline in the number and function of dendritic spines [23,24,25,26,27]. Antidepressant treatments have demonstrated the ability to reverse these adverse changes [28,29,30,31], underscoring a significant link between synaptic dysfunction and depressive states. The preservation of normal synaptic function is dependent on homeostatic mechanisms that stabilize synaptic transmission amidst fluctuating conditions. These mechanisms typically encompass (1) ion channels [32,33,34], (2) presynaptic neurotransmitter release [8, 35], (3) synaptic scaling of the postsynaptic density [36], and (4) related signaling molecules [37, 38]. Mounting evidence indicates that disturbances in synaptic homeostasis are integral to the mood-related circuitry disruptions observed in depression (Fig. 1).
Ion channels in synaptic homeostasis: implications for the pathophysiology of depression
Alterations in intrinsic neuronal excitability, a hallmark of various CNS diseases [39], are regulated by the properties, distribution, and abundance of ion channels embedded in the cell membrane. These channels are crucial for converting synaptic inputs into specific neuronal outputs [40]. Ion channels, including voltage-gated calcium [41, 42], potassium [43, 44], and sodium channels, facilitate ion exchange across the cell membrane, thereby maintaining the balance between neuronal excitation and inhibition. They directly modulate neurotransmitter release and synaptic efficacy, and indirectly influence neuronal excitability. The interplay among these channels generates action potentials, regulates neuronal firing frequency, and affects synaptic homeostasis by modulating synaptic transmission. Disruptions in ion channel function can significantly impact the CNS, and such imbalances are implicated in the development of psychiatric disorders such as bipolar affective disorder, autism, epilepsy, schizophrenia, and depression [45].
Ion channel alterations within the CNS have been documented in both depressive patients and animal models, as summarized in Table 1. For instance, in the striatal-nucleus accumbens of patients with major depressive disorder (MDD), there is an upregulation of genes encoding voltage-gated potassium, calcium, and sodium channels, except for the voltage-gated potassium channel (KV) 9.3 [46]. Conversely, down-regulation of KCNJ10 mRNA (encoding inwardly rectifying potassium channel (Kir)2q14.1 in the hippocampus) and SCN1A mRNA (encoding voltage-gated sodium channel (NaV) 1.1 in the PFC) has been observed in MDD patients [47, 48]. Enhanced expression of Kir4.1 and γ-aminobutyric acid (GABA) B receptor subunit 1 proteins is detected in the parietal cortex of MDD patients [49], along with elevated transient receptor potential melastatin 2 (TRPM2) protein expression in the hippocampus [50].
Animal studies reveal that KCNB1 mRNA, encoding KV2.1, is upregulated in the lateral habenula (LHb) of mice exhibiting acute learned helplessness [51]. Chronic mild stress (CMS) induces changes in the expression of KV2.1 and KV4.2 in the frontal cortex and hippocampus, with fluoxetine treatment reversing only the KV2.1 changes [52]. Chronic social defeat stress (CSDS) decreases the expression of KV4.2 in the lateral hypothalamic area GABAergic neurons and KV7.4 in the ventral tegmental area (VTA) dopaminergic neurons [53, 54], with functional degradation of KV4.2 also observed in the nucleus accumbens (NAc) medium spiny neurons of mice subjected to chronic unpredictable mild stress (CUMS) [55]. Kv4.2 knockout (KO) mice show increased immobility during forced swimming, and medial prefrontal cortex (mPFC) layer 5 pyramidal neurons receiving 5-hydroxytryptamine (5-HT) have a reduced increase in spontaneous excitatory postsynaptic currents (sEPSCs) frequency after a single swimming stress compared to wild-type mice [56].
Elevated levels of Kir4.1 protein in astrocytes of the LHb and hippocampus are found in congenitally learned helpless and lipopolysaccharide (LPS) models [57, 58]. Increased expression of KCNJ9 (Kir3.3) and KCNJ5 (Kir3.4) is observed in the medial habenula and LHb of Wistar-Kyoto rats [59]. Chronic stress raises Kir6.1 and Kir6.2 expression in the hippocampus [60], but not in the mPFC [61]. Astrocyte conditional knockout (cKO) of Kir4.1 impairs the dynamic balance of extracellular potassium ions and glutamate (Glu), and reduces the amplitude and frequency of sEPSCs in CA1 pyramidal neurons of Kir4.1 cKO mice [62]. Genetic deletion of Kir6.1 increases the frequency of sEPSCs in hippocampal CA3 pyramidal neurons [63].
Chronic stress and LPS enhance TWIK-related potassium channel (TREK) 1 expression in the hippocampus and frontal cortex [52, 64, 65]. TREK1 deficient mice exhibit resistance to depression, enhancing 5-HT neurotransmission efficacy and reducing corticosterone levels under stress [66]. Specific knockdown of TREK1 in mouse hippocampal neurons increases the amplitude of miniature excitatory postsynaptic currents (mEPSCs) in hippocampal CA1 pyramidal neurons and attenuates CUMS-induced reduction in mEPSCs amplitude and depressive-like behavior. Conversely, specific overexpression of TREK1 in hippocampal neurons promotes CUMS-induced decreases in CA1 pyramidal mEPSCs amplitude and exacerbates depressive-like behavior in mice [64].
In rats subjected to chronic restraint stress (CRS), hippocampal pyramidal neurons exhibit elevated expression of the L-type voltage-gated calcium channel subunit alpha-1C (CaV1.2) at both the mRNA and protein levels, accompanied by an enhanced amplitude of L-type calcium currents [67]. Conversely, chronic unpredictable stress (CUS) induces a delayed upregulation of CaV1.2 protein expression specifically within the PFC, a phenomenon not observed in other stress-responsive brain regions such as the hippocampus or amygdala [68]. The CACNA1C gene, which encodes CaV1.2, is pivotal in regulating the intracellular second messenger system, thereby influencing synaptic plasticity, gene expression, and neurotransmitter release. Studies have demonstrated that hippocampal synaptic plasticity is compromised in mice with a conditional knockout of CACNA1C in the hippocampus [69]. In these CaV1.2 cKO mice, the frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) in lateral amygdala principal neurons increases, while the frequency and amplitude of sEPSCs decrease, indicating a shift in the balance of inhibitory and excitatory activity within the lateral amygdala [70].
The expression of the hyperpolarization-activated cyclic nucleotide-gated channel (HCN) 2 is reduced in the VTA of CMS mice and in the NAc Shell of SDS mice [71, 72]. Altered HCN2 expression has been shown to affect neuronal firing frequency, with HCN2 knockdown leading to a significant increase in GABAergic output from reticular thalamic nucleus neurons to ventrobasal neurons [71, 73]. Additionally, the expression of the transient potential receptor vanilloid (TRPV) 2 is diminished in the hippocampus of CUMS rats [74], while TRPV4 and TRPM2 levels are elevated in the hippocampus of LPS and CUS mice [50, 75]. Notably, TRPM2 KO mice exhibit a significant increase in the amplitude and frequency of mEPSCs in hippocampal dentate gyrus neurons, alongside antidepressant-like behavior [50]. These findings collectively suggest that ion channel alterations significantly impact synaptic homeostasis within neural circuits, positioning them as promising targets for the development of novel antidepressant therapies.
Modulation of ion channels influences depression-like behaviors
The modulation of ion channels has been demonstrated to significantly influence depression-like behaviors in both clinical populations and animal models. For instance, in vitro and in vivo studies have shown that KCNQ-type K + channel openers, when applied to the VTA of mice subjected to social frustration stress, can reduce depressive-like behaviors and mitigate the overactivation of VTA dopamine (DA) neurons [79, 80]. Moreover, the conditional knockout of the multifunctional protein p11 (also known as S100A10) in parvalbumin neurons results in decreased hippocampal KV3.1 expression, impairing the high-frequency firing capacity of these neurons and increasing susceptibility to depression. This decrease in presynaptic KV3.1 expression leads to enhanced GABAergic synaptic responses, uncontrolled synaptic vesicle release, and disruption of short-term synaptic plasticity in the parvalbumin-granule cell synapses of the dentate gyrus [81]. Clinical research has indicated that Ezogabine, a KCNQ2/3 channel opener, is effective in treating depressive disorders [82]. Additionally, Lys05, a Kir4.1 inhibitor, has shown rapid antidepressant effects in Kir4.1-driven depressive-like phenotypes and various animal models of depression, highlighting Kir4.1 as a potential target for rapid-acting antidepressants [83]. The peptide spadin, which blocks TREK1 channels, has been found to exert antidepressant effects within a short timeframe [84], and N-[4]-N-(2-(3,4-dihydroisoquinolin-2(1H)-yl)−2-oxoethyl)methanesulfonamide (TKDC), an inhibitor of TREK1 channels, exhibits antidepressant-like actions following both acute and chronic treatment [85]. Furthermore, intermediate states of the TREK1 channel are being considered as potential targets for antidepressant therapy [86]. Escitalopram has been reported to inhibit the current of Nav1.2 and alter its activation and inactivation states [87]. In rats, chronic stress reduces hippocampal TRPV2 expression, and the TRPV2 agonist probenecid can alleviate depressive-like behaviors and increase hippocampal levels of 5-HT, norepinephrine (NE), and DA [74]. The deletion of transient receptor potential channel 5 (Trpc5) in oxytocin neurons of the hypothalamic paraventricular nucleus leads to obesity and postnatal depressive behaviors in female mice, whereas overexpression of Trpc5 reverses these phenotypes [88]. Chronic social isolation has been found to upregulate the small-conductance Ca2+ activated K + channel 3 (SK3) in the dorsal raphe nucleus, resulting in reduced 5-HT neuronal activity. Inhibitors of SK channels can ameliorate the behavioral deficits caused by chronic social isolation [76]. In CUS-exposed rats, there is an increase in the protein expression of HCN1 and perisomatic Ih currents in neurons of the dorsal CA1 region. Administration of shRNA-HCN1 to reduce Ih in dorsal CA1 neurons has been shown to mitigate the depressive-like behavioral deficits induced by CUS [77].
Presynaptic neurotransmitter release in the context of depression
Presynaptic homeostatic plasticity plays a critical role in counterbalancing impaired postsynaptic neurotransmitter receptor function by rapidly and precisely modulating neurotransmitter release [89]. Neurotransmitters, essential endogenous signaling molecules, facilitate communication within the central and peripheral nervous systems [90]. The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex is central to the presynaptic release of neurotransmitters. This complex is formed through the interaction of synaptic vesicle fusion proteins and synaptic membrane fusion proteins, mediating the fusion of synaptic vesicles with the presynaptic membrane and the subsequent release of neurotransmitters into the synaptic cleft [91,92,93].
Stress and depression have been shown to influence the expression of the SNARE complex in the brain. Elevated levels of SNARE complex proteins have been observed in the frontal cortex of individuals with suicidal schizophrenia and depression [94]. Additionally, acute foot shock stress in rats has been found to cause the accumulation of presynaptic SNARE complexes in the prefrontal/frontal cortex [95]. Studies on antidepressants have demonstrated that prolonged treatment can reduce depolarization-induced Glu release from hippocampal synaptic terminals, alter protein–protein interactions, modulate the assembly of presynaptic SNARE complexes, and decrease synaptic vesicle fusion and the number of complexes in the presynaptic membrane [96, 97]. For instance, fluoxetine has been shown to impair SNARE complex function, thereby decreasing the release of both Glu and GABA [98].
Depression and stress disrupt the function of both excitatory glutamatergic and inhibitory GABAergic circuits in the brain, compromising the efficiency and integrity of neural networks involved in emotion and cognition [99]. There is increasing evidence that chronic stress-induced anxiety and depression are associated with an imbalance in excitation/inhibition within the PFC [100, 101]. Emerging research indicates that synaptic transmission of both excitatory and inhibitory presynaptic neurotransmitters is altered during depressive states (see Table 2), particularly in terms of the probability of presynaptic neurotransmitter release and changes in synaptic vesicle volume and quantity. For example, CSDS significantly increases the paired-pulse ratio (PPR) of excitatory postsynaptic currents in basolateral amygdala (BLA) synapses projecting to the mPFC and the ventral hippocampus, indicating reduced Glu release probability [102]. In a learned helplessness model, the frequency of mEPSCs was decreased, and the PPR was elevated in the ventrolateral periaqueductal gray region of the midbrain in rats [103]. Chronic restraint stress in mice reduced the frequency of miniature inhibitory postsynaptic currents (mIPSCs) in layers II/III of the anterior cingulate cortex (ACC) and increased the PPRs of field excitatory postsynaptic potentials and evoked excitatory postsynaptic currents [104]. Conversely, chronic stress decreased the frequency of mIPSCs in parvocellular neurons of the hypothalamic paraventricular nucleus without affecting the PPR, suggesting a reduction in the number of presynaptic GABAergic synapses [105]. Acute stress enhances both the readily releasable pool of vesicles and depolarization-evoked Glu release [106], while chronic stress alters the distribution pattern of synaptic vesicles and increases vesicle density in the CA3 region of the hippocampus [107]. Furthermore, chronic stress has been proposed to reduce the number of synaptic vesicles in the inner molecular layer of the hippocampal dentate gyrus [108]. In summary, presynaptic neurotransmitter release in neural circuits is significantly altered during stress and depression, primarily through changes in the probability of neurotransmitter release and modifications to the synaptic vesicle pool.
Neurotransmitter release involves several pivotal processes: (1) metabolism by enzymes, (2) reuptake by the presynaptic neuron, and (3) binding to receptors on postsynaptic neurons or target cells, thereby triggering a physiological response in the postsynaptic or adjacent cell. Disruptions in neurotransmitter release can alter local neurotransmitter concentrations, leading to impairments in brain function that contribute to a range of physical, psychiatric, and neurodegenerative disorders [129]. Extensive research has established a strong correlation between neurotransmitter levels and the incidence of depression [130, 131]. Studies have demonstrated that specific neurotransmitters—such as 5-HT, Glu, GABA, NE, and DA—are essential for maintaining normal mood and are implicated in the pathogenesis of depression [132,133,134,135]. Notably, patients with MDD exhibit significantly elevated serum glutamate levels compared to controls, suggesting a potential link between Glu alterations and MDD [136]. Cerebrospinal fluid analyses in depressed individuals have revealed that 5-HT levels are associated with the onset of depressive symptoms [137], and pre-treatment cerebrospinal fluid Glu levels in depressed patients have been found to correlate positively with suicidal ideation [138]. Stress has been shown to influence DA levels in the PFC and NAc [139, 140]. Moreover, various antidepressant treatments achieve their therapeutic effects by modulating neurotransmitter levels [141, 142].
Synaptic scaling of the postsynaptic density in the context of depression
The postsynaptic density (PSD) is a specialized region of the cytoskeleton at the synaptic junction, serving as the structural foundation for postsynaptic signaling and integration [143,144,145]. The outer surface of the PSD is rich in neurotransmitter receptors involved in homeostatic synaptic scaling, as well as trans-synaptic adhesion molecules embedded in the plasma membrane [146]. Autopsy studies have revealed significantly decreased expression of the PSD marker protein PSD-95 in the PFC of individuals with depression [147]. Animal studies have shown that six weeks of exposure to CUMS results in reduced levels of brain-derived neurotrophic factor (BDNF), synaptophysin, and PSD-95, along with ultrastructural changes such as decreased synaptic number density, surface density, and PSD thickness in lateral amygdala neurons [148].
Homeostatic regulation of postsynaptic neurotransmitter receptors, termed synaptic scaling, involves the bi-directional regulation of the amplitude of mEPSCs to counterbalance chronic alterations in neuronal activity [149, 150]. An increase in firing rate is met with a proportional decrease in excitatory synaptic strength, while a decrease in firing rate leads to a proportional increase in synaptic strength [151]. These changes in synaptic strength are mediated by adjusting the quantity of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) in the postsynaptic membrane [152]. Synaptic scaling continuously modulates the number of receptors in the postsynaptic membrane through mechanisms involving receptor trafficking, expression of scaffolding proteins, gene transcription, and neural activity modulation [149, 153].Both retinoic acid receptor α (RARα) agonists, such as AM580, and ketamine modulate synaptic scaling by affecting the translation of AMPAR proteins in the hippocampus, thereby exerting antidepressant effects through distinct pathways [154, 155] (Fig. 2). The eukaryotic elongation factor 2 (eEF2) signaling pathway is a key regulator of protein synthesis, synaptic plasticity, learning, and memory [156]. Ketamine induces rapid antidepressant-like effects by inhibiting spontaneous N-methyl-D-aspartate receptor (NMDAR)-mediated mEPSCs, leading to acute inhibition of eEF2 kinase activity and a subsequent rapid increase in BDNF translation. Ketamine’s synaptic scaling is associated with upregulation of the AMPAR subunits GluA1 and GluA2 [157]. The RARα agonist AM580 can elicit a ketamine-like rapid antidepressant response through RARα activation, with its synaptic scaling achieved by the insertion of AMPARs containing the GluA1 subunit [158, 159].
Ketamine and RARα receptor agonists can regulate synaptic scaling through different pathways and exert antidepressant effects. A simplified overview of the antidepressant effects of ketamine and RARα receptor agonists through the regulation of synaptic scaling. Ketamine enhances the recruitment of postsynaptic GluA1/2, facilitating synaptic scaling and driving rapid antidepressant effects. Similarly, RARα receptor agonists directly activate retinoic acid receptors, which also leads to an increased recruitment of postsynaptic GluA1, thereby promoting synaptic scaling. This mechanism produces antidepressant effects comparable to those of ketamine. eEF2K elongation factor 2 kinase, eEF2 elongation factor 2, FMRP fragile X mental retardation protein, CaM calmodulin, CaN calcineurin, RALDH retinal dehydrogenase, BDNF brain-derived neurotrophic factor, GluA1/2 AMPA receptor subunits 1/2
Related signaling molecules in the regulation of synaptic homeostasis and their relevance to depression
In the field of depression research, the importance of various signaling molecules in maintaining synaptic homeostasis has gained significant attention. This section highlights the roles of three key molecules: a neurotrophic factor that enhances neuronal communication, a cytokine released by glial cells with profound neuronal effects, and a small molecule derived from Vitamin A metabolism.
Brain-derived neurotrophic factor (BDNF)
BDNF is a vital neurotrophic factor essential for the development and regulation of synaptic plasticity [160]. Upon release, BDNF binds to its high-affinity receptor, tropomyosin receptor kinase B (TrkB), initiating a series of synaptic modulation events. BDNF plays a central role in synaptic homeostasis by influencing neurotransmitter release from presynaptic terminals. For instance, a study using viral-mediated gene targeting in the CA3-CA1 hippocampal circuit demonstrated that the absence of presynaptic TrkB reduced the probability of neurotransmitter release, underscoring the critical role of presynaptic TrkB receptors in BDNF-mediated presynaptic function [161]. Additionally, BDNF modulates the density of postsynaptic receptors. Research on the antimanic effects of lithium revealed that chronic lithium treatment decreased the surface expression of the GluA1 subunit in hippocampal neurons, significantly reducing AMPAR-mediated mEPSCs. This effect was mediated by BDNF and TrkB, highlighting their importance in postsynaptic homeostatic plasticity [162]. The link between BDNF and depression is well-documented. Duman and Monteggia proposed the neurotrophic hypothesis of depression, which posits that stress and depression lead to a deficiency in neurotrophic factors, contributing to cellular atrophy and loss in key brain regions of individuals with MDD [163]. Studies have consistently shown that MDD patients exhibit lower levels of central and peripheral BDNF compared to non-depressed individuals, with peripheral BDNF levels inversely correlated with symptom severity and directly associated with symptom improvement [164, 165]. Innovative approaches to BDNF delivery have shown promise. For example, a study utilizing quercetin nanogels to deliver BDNF demonstrated that BDNF-quercetin alginate nanogels could effectively cross the blood–brain barrier via the nasal-brain route, providing sustained and controlled BDNF release in the brain. Treatment with these nanogels significantly increased plasma and hippocampal BDNF levels in rats subjected to CUMS, potentially exerting antidepressant effects through the regulation of the glutamatergic system, the phosphoinositide 3-kinase-Protein kinase B (PI3K-Akt) pathway, and the BDNF-TrkB signaling pathway [166].Furthermore, ketamine has been shown to rapidly induce BDNF protein synthesis by inhibiting eukaryotic elongation factor 2 (eEF2) kinase, leading to increased surface expression of the GluA1 and GluA2 subunits. This regulation of synaptic homeostasis contributes to ketamine's rapid antidepressant-like effects [155, 157]. These findings collectively suggest that the rapid upregulation of proteins such as BDNF can modulate synaptic homeostasis to mediate antidepressant responses.
Tumor necrosis factor-alpha (TNF-α)
TNF-α, a key cytokine in the CNS, plays a dual role in maintaining synaptic homeostasis and contributing to the pathogenesis of depression [167]. Glia-derived TNF-α is crucial for the synaptic scaling of both excitatory and inhibitory synapses. Research has demonstrated that prolonged neuronal inactivity triggers the release of soluble TNF-α from glial cells. This accumulation of TNF-α enhances AMPA receptor levels at excitatory synapses while downregulating GABA receptor levels at inhibitory synapses in a homeostatic manner that depends on TNF-α receptors. Notably, Hebbian forms of synaptic plasticity do not require TNF-α [168]. However, some studies suggest that glia-derived TNF-α may not directly drive synaptic scaling but is essential for maintaining synaptic plasticity in a stable state [169].
Clinical evidence supports the involvement of TNF-α in depression. A meta-analysis of 24 studies revealed that plasma TNF-α levels are significantly elevated in individuals with depression compared to healthy controls [170]. Additionally, the protein and mRNA levels of TNF-α in the PFC of depressed individuals who died by suicide were markedly higher than in control groups [171]. Clinical studies have also shown that reductions in peripheral TNF-α levels correlate with improvements in depressive symptoms, and effective treatment for MDD normalizes TNF-α levels [172, 173]. The surge in cytokine release (including TNF-α, interleukin-1 beta and interleukin-6) during early life inflammation leads to dysregulation of microglial phagocytic capacity. During adolescence, unpredictable stressors exacerbate microglial phagocytosis around the spines of glutamatergic neurons in the ACC, promoting depressive-like behaviors [174]. In animal models, wild-type mice treated with antidepressants fluoxetine and desipramine showed reduced immobility in the forced swim test and tail suspension test. In contrast, TNF-α KO mice exhibited a diminished response to these medications, requiring higher doses to achieve an antidepressant effect. Furthermore, selective ablation of TNF-α in astrocytes confirmed that astrocytic TNF-α is essential for the antidepressant effects of chronic fluoxetine treatment [175].
Retinoic acid (RA)
RA, a metabolite of vitamin A, is well-known for its role in embryonic development but also significantly influences synaptic homeostasis and plasticity in the adult brain [176]. In hippocampal cultures, suppression of neuronal activity with tetrodotoxin (TTX) and APV induced synaptic scaling, which was associated with increased RA synthesis. Inhibition of RA synthesis abolished this synaptic scaling. This process is mediated by RARα signaling, which promotes local synthesis of the GluA1 subunit. Knockdown of RARα blocks RA-induced synaptic scaling, while activation of RARα receptors replicates the effects of RA on synaptic scaling [158].RA has been implicated in depressive disorders [177]. Preclinical and epidemiological studies indicate that serum retinol levels are significantly elevated in MDD patients compared to healthy individuals, and the synthesis of the vitamin A metabolite all-trans RA is enhanced, suggesting a critical role for the RA system in depression [178]. Chronic RA treatment can induce depressive- and anxiety-like behaviors by stimulating GABA synthesis, increasing GABA receptor expression, and downregulating glutamate receptor expression. These changes reduce hippocampal neuronal excitability and may impair hippocampal homeostatic synaptic plasticity by lowering GluA1 mRNA levels [179]. Conversely, acute activation of the RA signaling pathway has been shown to induce synaptic scaling in hippocampal neurons and mediate antidepressant-like effects in mice [155]. Thus, RA is a key regulator of synaptic homeostasis and plays a significant role in depression.
The network of signaling molecules involved in synaptic homeostasis is extensive and not yet fully understood. For example, molecules such as Endostatin, Presenilin 1, and Arc/Arg3.1 have been reported to regulate synaptic homeostasis [180,181,182], but their precise links to synaptic homeostasis in depression remain to be fully elucidated.
Conclusion
The array of mechanisms involved in synaptic homeostasis is extensive, including ion channels, presynaptic neurotransmitter release, postsynaptic receptors, and related signaling molecules. However, their precise connections to synaptic homeostasis in the context of depression remain to be fully understood. Therefore, a deeper understanding of the molecular mechanisms governing synaptic homeostasis is essential for clarifying the links between synaptic homeostasis and depression. Moreover, the etiology and therapeutic response in depression are intricately linked to disruptions in synaptic homeostasis within mood-regulating brain regions and circuits. Current investigations into the impact of stress and depression, as well as the synaptic effects of antidepressant therapies, have primarily focused on the PFC and hippocampus. However, emerging evidence indicates that other brain regions and circuits, such as those involved in reward and anti-reward processing (e.g., VTA, NAc, and LHb), also play significant roles in the pathophysiology of depression. Future research is essential to elucidate the effects of antidepressant treatments on synaptic homeostasis across additional circuits and neuronal populations. Together, this review contributes to understanding the implications of altered synaptic homeostasis on depression, which paves the way for the development of more accessible, safer, and efficacious antidepressant interventions.
Availability of data and materials
Not applicable.
Abbreviations
- CNS:
-
Central nervous system
- SNARE:
-
Soluble N-ethylmaleimide-sensitive factor attachment protein receptor
- PFC:
-
Prefrontal cortex
- LHb:
-
Lateral habenula
- VTA:
-
Ventral tegmental area
- NAc:
-
Nucleus accumbens
- mPFC:
-
Medial prefrontal cortex
- BLA:
-
Basolateral amygdala
- ACC:
-
Anterior cingulate cortex
- Glu:
-
Glutamate
- GABA:
-
γ-Aminobutyric acid
- NE:
-
Norepinephrine
- 5-HT:
-
5-Hydroxytryptamine
- DA:
-
Dopamine
- AMPARs:
-
α-Amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors
- GluA1/2:
-
AMPA receptor subunits 1/2
- RA:
-
Retinoic acid
- RARα:
-
Retinoic acid receptor α
- NMDAR:
-
N-methyl-d-aspartate receptor
- eEF2:
-
Eukaryotic elongation factor 2
- TrkB:
-
Tropomyosin receptor kinase B
- PI3K:
-
Phosphoinositide 3-kinase
- Akt:
-
Protein kinase B
- TNF-α:
-
Tumor necrosis factor-alpha
- MDD:
-
Major depressive disorder
- KV :
-
Voltage-gated potassium channel
- Kir:
-
Inwardly rectifying potassium channel
- TRPM2:
-
Transient receptor potential melastatin 2
- TREK:
-
TWIK-related potassium channel
- SK3:
-
Small-conductance Ca2+ activated K+ channel 3
- CaV1.2:
-
L-type voltage-gated calcium channel subunit alpha-1C
- HCN:
-
Hyperpolarization-activated cyclic nucleotide-gated channel
- TRPV:
-
Transient potential receptor vanilloid
- Trpc5:
-
Transient receptor potential channel 5
- NaV :
-
Voltage-gated sodium channel
- PSD:
-
Postsynaptic density
- BDNF:
-
Brain-derived neurotrophic factor
- CMS:
-
Chronic mild stress
- CSDS:
-
Chronic social defeat stress
- CUMS:
-
Chronic unpredictable mild stress
- LPS:
-
Lipopolysaccharide
- CRS:
-
Chronic restraint stress
- CUS:
-
Chronic unpredictable stress
- PPR:
-
Paired-pulse ratio
- mEPSCs:
-
Miniature excitatory postsynaptic currents
- mIPSCs:
-
Miniature inhibitory postsynaptic currents
- sEPSCs:
-
Spontaneous excitatory postsynaptic currents
- sIPSCs:
-
Spontaneous inhibitory postsynaptic currents
- cKO:
-
Conditional knockout
- KO:
-
Knockout
References
McCarron RM, Shapiro B, Rawles J, Luo J. Depression. Ann Intern Med. 2021;174(5):65-ITC80.
Herrman H, Patel V, Kieling C, Berk M, Buchweitz C, Cuijpers P, et al. Time for united action on depression: a Lancet-World Psychiatric Association Commission. Lancet. 2022;399(10328):957–1022.
Cipriani A, Furukawa TA, Salanti G, Chaimani A, Atkinson LZ, Ogawa Y, et al. Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: a systematic review and network meta-analysis. Focus (Am Psychiatr Publ). 2018;16(4):420–9.
Solmi M, Fornaro M, Ostinelli EG, Zangani C, Croatto G, Monaco F, et al. Safety of 80 antidepressants, antipsychotics, anti-attention-deficit/hyperactivity medications and mood stabilizers in children and adolescents with psychiatric disorders: a large scale systematic meta-review of 78 adverse effects. World Psychiatry. 2020;19(2):214–32.
Conroy SK, Holtzheimer PE. Neuromodulation Strategies for the Treatment of Depression. Am J Psychiatry. 2021;178(12):1082–8.
Chan ES, Ge Y, So YW, Bai YF, Liu L, Wang YT. Allosteric potentiation of GABAA receptor single-channel conductance by netrin-1 during neuronal-excitation-induced inhibitory synaptic homeostasis. Cell Rep. 2022;41(5): 111584.
Li B, Suutari BS, Sun SD, Luo Z, Wei C, Chenouard N, et al. Neuronal inactivity co-opts LTP machinery to drive potassium channel splicing and homeostatic spike widening. Cell. 2020;181(7):1547-1565.e15.
Davis GW, Müller M. Homeostatic control of presynaptic neurotransmitter release. Annu Rev Physiol. 2015;77:251–70.
Ancona Esselmann SG, DÃaz-Alonso J, Levy JM, Bemben MA, Nicoll RA. Synaptic homeostasis requires the membrane-proximal carboxy tail of GluA2. Proc Natl Acad Sci U S A. 2017;114(50):13266–71.
Mullins C, Fishell G, Tsien RW. Unifying views of autism spectrum disorders: a consideration of autoregulatory feedback loops. Neuron. 2016;89(6):1131–56.
Soukup SF, Vanhauwaert R, Verstreken P. Parkinson’s disease: convergence on synaptic homeostasis. EMBO J. 2018;37(18): e98960.
Knopman DS, Amieva H, Petersen RC, Chételat G, Holtzman DM, Hyman BT, et al. Alzheimer disease. Nat Rev Dis Primers. 2021;7(1):33.
Tewari BP, Woo AM, Prim CE, Chaunsali L, Patel DC, Kimbrough IF, et al. Astrocytes require perineuronal nets to maintain synaptic homeostasis in mice. Nat Neurosci. 2024;27(8):1475–88.
Dickman DK, Davis GW. The schizophrenia susceptibility gene dysbindin controls synaptic homeostasis. Science. 2009;326(5956):1127–30.
Duman RS, Aghajanian GK. Synaptic dysfunction in depression: potential therapeutic targets. Science. 2012;338(6103):68–72.
Eccles JC. The synapse: from electrical to chemical transmission. Annu Rev Neurosci. 1982;5:325–39.
Pereda AE. Electrical synapses and their functional interactions with chemical synapses. Nat Rev Neurosci. 2014;15(4):250–63.
Savitz J, Drevets WC. Bipolar and major depressive disorder: neuroimaging the developmental-degenerative divide. Neurosci Biobehav Rev. 2009;33(5):699–771.
Koolschijn PCMP, van Haren NEM, Lensvelt-Mulders GJLM, Hulshoff Pol HE, Kahn RS. Brain volume abnormalities in major depressive disorder: a meta-analysis of magnetic resonance imaging studies. Hum Brain Mapp. 2009;30(11):3719–35.
MacQueen GM, Yucel K, Taylor VH, Macdonald K, Joffe R. Posterior hippocampal volumes are associated with remission rates in patients with major depressive disorder. Biol Psychiatry. 2008;64(10):880–3.
Neumeister A, Wood S, Bonne O, Nugent AC, Luckenbaugh DA, Young T, et al. Reduced hippocampal volume in unmedicated, remitted patients with major depression versus control subjects. Biol Psychiatry. 2005;57(8):935–7.
MacQueen G, Frodl T. The hippocampus in major depression: evidence for the convergence of the bench and bedside in psychiatric research? Mol Psychiatry. 2011;16(3):252–64.
McEwen BS, Nasca C, Gray JD. Stress effects on neuronal structure: hippocampus, amygdala, and prefrontal cortex. Neuropsychopharmacology. 2016;41(1):3–23.
Radley JJ, Rocher AB, Miller M, Janssen WGM, Liston C, Hof PR, et al. Repeated stress induces dendritic spine loss in the rat medial prefrontal cortex. Cereb Cortex. 2006;16(3):313–20.
Hains AB, Vu MAT, Maciejewski PK, van Dyck CH, Gottron M, Arnsten AFT. Inhibition of protein kinase C signaling protects prefrontal cortex dendritic spines and cognition from the effects of chronic stress. Proc Natl Acad Sci U S A. 2009;106(42):17957–62.
Kang HJ, Voleti B, Hajszan T, Rajkowska G, Stockmeier CA, Licznerski P, et al. Decreased expression of synapse-related genes and loss of synapses in major depressive disorder. Nat Med. 2012;18(9):1413–7.
Csabai D, Wiborg O, Czéh B. Reduced synapse and axon numbers in the prefrontal cortex of rats subjected to a chronic stress model for depression. Front Cell Neurosci. 2018;12:24.
Zhao JL, Jiang WT, Wang X, Cai ZD, Liu ZH, Liu GR. Exercise, brain plasticity, and depression. CNS Neurosci Ther. 2020;26(9):885–95.
Liang X, Tang J, Qi YQ, Luo YM, Yang CM, Dou XY, et al. Exercise more efficiently regulates the maturation of newborn neurons and synaptic plasticity than fluoxetine in a CUS-induced depression mouse model. Exp Neurol. 2022;354: 114103.
Seo MK, Lee CH, Cho HY, Lee JG, Lee BJ, Kim JE, et al. Effects of antidepressant drugs on synaptic protein levels and dendritic outgrowth in hippocampal neuronal cultures. Neuropharmacology. 2014;79:222–33.
Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329(5994):959–64.
Morgan PJ, Bourboulou R, Filippi C, Koenig-Gambini J, Epsztein J. Kv11 contributes to a rapid homeostatic plasticity of intrinsic excitability in CA1 pyramidal neurons in vivo. Elife. 2019;27(8):e49915.
Marder E, Prinz AA. Current compensation in neuronal homeostasis. Neuron. 2003;37(1):2–4.
Marder E, Prinz AA. Modeling stability in neuron and network function: the role of activity in homeostasis. BioEssays. 2002;24(12):1145–54.
Rozenfeld E, Ehmann N, Manoim JE, Kittel RJ, Parnas M. Homeostatic synaptic plasticity rescues neural coding reliability. Nat Commun. 2023;14(1):2993.
Turrigiano GG. AMPA receptors unbound: membrane cycling and synaptic plasticity. Neuron. 2000;26(1):5–8.
Davis GW. Homeostatic control of neural activity: from phenomenology to molecular design. Annu Rev Neurosci. 2006;29:307–23.
Fernandes D, Carvalho AL. Mechanisms of homeostatic plasticity in the excitatory synapse. J Neurochem. 2016;139(6):973–96.
Beck H, Yaari Y. Plasticity of intrinsic neuronal properties in CNS disorders. Nat Rev Neurosci. 2008;9(5):357–69.
Schulz DJ. Plasticity and stability in neuronal output via changes in intrinsic excitability: it’s what’s inside that counts. J Exp Biol. 2006;209(Pt 24):4821–7.
Catterall WA, Perez-Reyes E, Snutch TP, Striessnig J. International Union of Pharmacology. XLVIII. Nomenclature and structure-function relationships of voltage-gated calcium channels. Pharmacol Rev. 2005;57(4):411–25.
Mortensen OV. MKP3 eliminates depolarization-dependent neurotransmitter release through downregulation of L-type calcium channel Cav1.2 expression. Cell Calcium. 2013;53(3):224–30.
Lambe EK, Aghajanian GK. The role of Kv1.2-containing potassium channels in serotonin-induced glutamate release from thalamocortical terminals in rat frontal cortex. J Neurosci. 2001;21(24):9955–63.
Kaczmarek LK, Zhang Y. Kv3 channels: enablers of rapid firing, neurotransmitter release, and neuronal endurance. Physiol Rev. 2017;97(4):1431–68.
Imbrici P, Camerino DC, Tricarico D. Major channels involved in neuropsychiatric disorders and therapeutic perspectives. Front Genet. 2013;4:76.
Smolin B, Karry R, Gal-Ben-Ari S, Ben-Shachar D. Differential expression of genes encoding neuronal ion-channel subunits in major depression, bipolar disorder and schizophrenia: implications for pathophysiology. Int J Neuropsychopharmacol. 2012;15(7):869–82.
Medina A, Watson SJ, Bunney W, Myers RM, Schatzberg A, Barchas J, et al. Evidence for alterations of the glial syncytial function in major depressive disorder. J Psychiatr Res. 2016;72:15–21.
Riga MS, Pérez-Fernández M, Miquel-Rio L, Paz V, Campa L, MartÃnez-Losa M, et al. Scn1a haploinsufficiency in the prefrontal cortex engages to cognitive impairment and depressive phenotype. Brain. 2024;awae167.
Xiong Z, Zhang K, Ren Q, Chang L, Chen J, Hashimoto K. Increased expression of inwardly rectifying Kir4.1 channel in the parietal cortex from patients with major depressive disorder. J Affect Disord. 2019;245:265–9.
Ko SY, Wang SE, Lee HK, Jo S, Han J, Lee SH, et al. Transient receptor potential melastatin 2 governs stress-induced depressive-like behaviors. Proc Natl Acad Sci U S A. 2019;116(5):1770–5.
Ryu H, Kim M, Park H, Choi HK, Chung C. Stress-induced translation of KCNB1 contributes to the enhanced synaptic transmission of the lateral habenula. Front Cell Neurosci. 2023;17:1278847.
Chen C, Wang L, Rong X, Wang W, Wang X. Effects of fluoxetine on protein expression of potassium ion channels in the brain of chronic mild stress rats. Acta Pharm Sin B. 2015;5(1):55–61.
Li XY, Zhang SY, Hong YZ, Chen ZG, Long Y, Yuan DH, et al. TGR5-mediated lateral hypothalamus-dCA3-dorsolateral septum circuit regulates depressive-like behavior in male mice. Neuron. 2024;112(11):1795-1814.e10.
Li L, Sun H, Ding J, Niu C, Su M, Zhang L, et al. Selective targeting of M-type potassium Kv 74 channels demonstrates their key role in the regulation of dopaminergic neuronal excitability and depression-like behaviour. Br J Pharmacol. 2017;174(23):4277–94.
Aceto G, Colussi C, Leone L, Fusco S, Rinaudo M, Scala F, et al. Chronic mild stress alters synaptic plasticity in the nucleus accumbens through GSK3β-dependent modulation of Kv4.2 channels. Proc Natl Acad Sci U S A. 2020;117(14):8143–53.
Lockridge A, Su J, Yuan LL. Abnormal 5-HT modulation of stress behaviors in the Kv4.2 knockout mouse. Neuroscience. 2010;170(4):1086–97.
Cui Y, Yang Y, Ni Z, Dong Y, Cai G, Foncelle A, et al. Astroglial Kir41 in the lateral habenula drives neuronal bursts in depression. Nature. 2018;554(7692):323–7.
Song Z, Bian Z, Zhang Z, Wang X, Zhu A, Zhu G. Astrocytic Kir41 regulates NMDAR/calpain signaling axis in lipopolysaccharide-induced depression-like behaviors in mice. Toxicol Appl Pharmacol. 2021;429:115711.
Korlatowicz A, Pabian P, Solich J, Kolasa M, Latocha K, Dziedzicka-Wasylewska M, et al. Habenula as a possible target for treatment-resistant depression phenotype in wistar kyoto rats. Mol Neurobiol. 2023;60(2):643–54.
Fan Y, Kong H, Ye X, Ding J, Hu G. ATP-sensitive potassium channels: uncovering novel targets for treating depression. Brain Struct Funct. 2016;221(6):3111–22.
Li F, Jiang SY, Tian T, Li WJ, Xue Y, Du RH, et al. Kir61/K-ATP channel in astrocytes is an essential negative modulator of astrocytic pyroptosis in mouse model of depression. Theranostics. 2022;12(15):6611–25.
Djukic B, Casper KB, Philpot BD, Chin LS, McCarthy KD. Conditional knock-out of Kir41 leads to glial membrane depolarization, inhibition of potassium and glutamate uptake, and enhanced short-term synaptic potentiation. J Neurosci. 2007;27(42):11354–65.
Soundarapandian MM, Wu D, Zhong X, Petralia RS, Peng L, Tu W, et al. Expression of functional Kir61 channels regulates glutamate release at CA3 synapses in generation of epileptic form of seizures. J Neurochem. 2007;103(5):1982–8.
Wu F, Sun H, Gong W, Li X, Pan Z, Shan H, et al. Genetic and pharmacological inhibition of two-pore domain potassium channel TREK-1 alters depression-related behaviors and neuronal plasticity in the hippocampus in mice. CNS Neurosci Ther. 2021;27(2):220–32.
Kim A, Jung HG, Kim YE, Kim SC, Park JY, Lee SG, et al. The knockdown of TREK-1 in hippocampal neurons attenuate lipopolysaccharide-induced depressive-like behavior in mice. Int J Mol Sci. 2019;20(23):5902.
Heurteaux C, Lucas G, Guy N, El Yacoubi M, Thümmler S, Peng XD, et al. Deletion of the background potassium channel TREK-1 results in a depression-resistant phenotype. Nat Neurosci. 2006;9(9):1134–41.
Moreno C, Hermosilla T, Hardy P, Aballai V, Rojas P, Varela D. Cav12 activity and downstream signaling pathways in the hippocampus of an animal model of depression. Cells. 2020;9(12):2609.
Bavley CC, Fischer DK, Rizzo BK, Rajadhyaksha AM. Cav1.2 channels mediate persistent chronic stress-induced behavioral deficits that are associated with prefrontal cortex activation of the p25/Cdk5-glucocorticoid receptor pathway. Neurobiol Stress. 2017;7:27–37.
Moosmang S, Haider N, Klugbauer N, Adelsberger H, Langwieser N, Müller J, et al. Role of hippocampal Cav1.2 Ca2+ channels in NMDA receptor-independent synaptic plasticity and spatial memory. J Neurosci. 2005;25(43):9883–92.
Temme SJ, Murphy GG. The L-type voltage-gated calcium channel CaV1.2 mediates fear extinction and modulates synaptic tone in the lateral amygdala. Learn Mem. 2017;24(11):580–8.
Zhong P, Vickstrom CR, Liu X, Hu Y, Yu L, Yu HG, et al. HCN2 channels in the ventral tegmental area regulate behavioral responses to chronic stress. Elife. 2018;2(7): e32420.
Cheng J, Umschweif G, Leung J, Sagi Y, Greengard P. HCN2 channels in cholinergic interneurons of nucleus accumbens shell regulate depressive behaviors. Neuron. 2019;101(4):662-672.e5.
Ying SW, Jia F, Abbas SY, Hofmann F, Ludwig A, Goldstein PA. Dendritic HCN2 channels constrain glutamate-driven excitability in reticular thalamic neurons. J Neurosci. 2007;27(32):8719–32.
Zhou Y, Cong T, Chen J, Chu Z, Sun Y, Zhao D, et al. Protective role of TRPV2 in synaptic plasticity through the ERK1/2-CREB-BDNF pathway in chronic unpredictable mild stress rats. Biochem Biophys Res Commun. 2024;20(721): 150128.
Li W, Xu Y, Liu Z, Shi M, Zhang Y, Deng Y, et al. TRPV4 inhibitor HC067047 produces antidepressant-like effect in LPS-induced depression mouse model. Neuropharmacology. 2021;15(201): 108834.
Sargin D, Oliver DK, Lambe EK. Chronic social isolation reduces 5-HT neuronal activity via upregulated SK3 calcium-activated potassium channels. Elife. 2016;22(5): e21416.
Kim CS, Brager DH, Johnston D. Perisomatic changes in h-channels regulate depressive behaviors following chronic unpredictable stress. Mol Psychiatry. 2018;23(4):892–903.
Cao XZ, Zhu MY, Xu G, Li F, Yan Y, Zhang JJ, et al. HCN channels in the lateral habenula regulate pain and comorbid depressive-like behaviors in mice. CNS Neurosci Ther. 2024;30(7): e14831.
Krishnan V, Han MH, Graham DL, Berton O, Renthal W, Russo SJ, et al. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell. 2007;131(2):391–404.
Friedman AK, Juarez B, Ku SM, Zhang H, Calizo RC, Walsh JJ, et al. KCNQ channel openers reverse depressive symptoms via an active resilience mechanism. Nat Commun. 2016;24(7):11671.
Medrihan L, Umschweif G, Sinha A, Reed S, Lee J, Gindinova K, et al. Reduced Kv3.1 activity in dentate gyrus parvalbumin cells induces vulnerability to depression. Biol Psychiatry. 2020;88(5):405–14.
Costi S, Morris LS, Kirkwood KA, Hoch M, Corniquel M, Vo-Le B, et al. Impact of the KCNQ2/3 channel opener ezogabine on reward circuit activity and clinical symptoms in depression: results from a randomized controlled trial. Am J Psychiatry. 2021;178(5):437–46.
Zhou X, Zhao C, Xu H, Xu Y, Zhan L, Wang P, et al. Pharmacological inhibition of Kir4.1 evokes rapid-onset antidepressant responses. Nat Chem Biol. 2024;20(7):857–66.
Djillani A, Pietri M, Mazella J, Heurteaux C, Borsotto M. Fighting against depression with TREK-1 blockers: Past and future. A focus on spadin Pharmacol Ther. 2019;194:185–98.
Luo Q, Chen L, Cheng X, Ma Y, Li X, Zhang B, et al. An allosteric ligand-binding site in the extracellular cap of K2P channels. Nat Commun. 2017;8(1):378.
Ma Y, Luo Q, Fu J, Che Y, Guo F, Mei L, et al. Discovery of an inhibitor for the TREK-1 channel targeting an intermediate transition state of channel gating. J Med Chem. 2020;63(19):10972–83.
Nakatani Y, Amano T. Functional modulation of Nav1.2 voltage-gated sodium channels induced by escitalopram. Biol Pharm Bull. 2018;41(9):1471–4.
Li Y, Cacciottolo TM, Yin N, He Y, Liu H, Liu H, et al. Loss of transient receptor potential channel 5 causes obesity and postpartum depression. Cell. 2024;187(16):4176-4192.e17.
Ortega JM, Genç Ö, Davis GW. Molecular mechanisms that stabilize short term synaptic plasticity during presynaptic homeostatic plasticity. Elife. 2018;13(7): e40385.
Hyman SE. Neurotransmitters. Curr Biol. 2005;15(5):R154–8.
Molecular RJ, Release MUN. Annu Rev Biophys. 2022;9(51):377–408.
Stepien KP, Rizo J. Synaptotagmin-1-, Munc18-1-, and Munc13-1-dependent liposome fusion with a few neuronal SNAREs. Proc Natl Acad Sci U S A. 2021;118(4): e2019314118.
Rizo J, Xu J. The synaptic vesicle release machinery. Annu Rev Biophys. 2015;44:339–67.
Honer WG, Falkai P, Bayer TA, Xie J, Hu L, Li HY, et al. Abnormalities of SNARE mechanism proteins in anterior frontal cortex in severe mental illness. Cereb Cortex. 2002;12(4):349–56.
Musazzi L, Milanese M, Farisello P, Zappettini S, Tardito D, Barbiero VS, et al. Acute stress increases depolarization-evoked glutamate release in the rat prefrontal/frontal cortex: the dampening action of antidepressants. PLoS ONE. 2010;5(1): e8566.
Bonanno G, Giambelli R, Raiteri L, Tiraboschi E, Zappettini S, Musazzi L, et al. Chronic antidepressants reduce depolarization-evoked glutamate release and protein interactions favoring formation of SNARE complex in hippocampus. J Neurosci. 2005;25(13):3270–9.
Milanese M, Tardito D, Musazzi L, Treccani G, Mallei A, Bonifacino T, et al. Chronic treatment with agomelatine or venlafaxine reduces depolarization-evoked glutamate release from hippocampal synaptosomes. BMC Neurosci. 2013;29(14):75.
Lazarevic V, Mantas I, Flais I, Svenningsson P. Fluoxetine suppresses glutamate- and GABA-mediated neurotransmission by altering SNARE complex. Int J Mol Sci. 2019;20(17):4247.
Duman RS, Sanacora G, Krystal JH. Altered connectivity in depression: GABA and glutamate neurotransmitter deficits and reversal by novel treatments. Neuron. 2019;102(1):75–90.
Page CE, Coutellier L. Prefrontal excitatory/inhibitory balance in stress and emotional disorders: evidence for over-inhibition. Neurosci Biobehav Rev. 2019;105:39–51.
Chen Y, Zheng Y, Yan J, Zhu C, Zeng X, Zheng S, et al. Early life stress induces different behaviors in adolescence and adulthood may related with abnormal medial prefrontal cortex excitation/inhibition balance. Front Neurosci. 2021;15: 720286.
Kim J, Kang S, Choi TY, Chang KA, Koo JW. Metabotropic glutamate receptor 5 in amygdala target neurons regulates susceptibility to chronic social stress. Biol Psychiatry. 2022;92(2):104–15.
Chou D, Peng HY, Lin TB, Lai CY, Hsieh MC, Wen YC, et al. (2R,6R)-hydroxynorketamine rescues chronic stress-induced depression-like behavior through its actions in the midbrain periaqueductal gray. Neuropharmacology. 2018;1(139):1–12.
Ito H, Nagano M, Suzuki H, Murakoshi T. Chronic stress enhances synaptic plasticity due to disinhibition in the anterior cingulate cortex and induces hyper-locomotion in mice. Neuropharmacology. 2010;58(4–5):746–57.
Verkuyl JM, Hemby SE, Joëls M. Chronic stress attenuates GABAergic inhibition and alters gene expression of parvocellular neurons in rat hypothalamus. Eur J Neurosci. 2004;20(6):1665–73.
Treccani G, Musazzi L, Perego C, Milanese M, Nava N, Bonifacino T, et al. Stress and corticosterone increase the readily releasable pool of glutamate vesicles in synaptic terminals of prefrontal and frontal cortex. Mol Psychiatry. 2014;19(4):433–43.
Magariños AM, Verdugo JM, McEwen BS. Chronic stress alters synaptic terminal structure in hippocampus. Proc Natl Acad Sci U S A. 1997;94(25):14002–8.
Hei M, Chen P, Wang S, Li X, Xu M, Zhu X, et al. Effects of chronic mild stress induced depression on synaptic plasticity in mouse hippocampus. Behav Brain Res. 2019;3(365):26–35.
BÄ…k J, Bobula B, Hess G. Restraint stress and repeated corticosterone administration differentially affect neuronal excitability, synaptic transmission and 5-HT7 receptor reactivity in the dorsal raphe nucleus of young adult male rats. Int J Mol Sci. 2022;23(22):14303.
Yu G, Cao F, Hou T, Cheng Y, Jia B, Yu L, et al. Astrocyte reactivation in medial prefrontal cortex contributes to obesity-promoted depressive-like behaviors. J Neuroinflammation. 2022;19(1):166.
Salter EW, Sunstrum JK, Matovic S, Inoue W. Chronic stress dampens excitatory synaptic gain in the paraventricular nucleus of the hypothalamus. J Physiol. 2018;596(17):4157–72.
Son H, Baek JH, Go BS, Jung DH, Sontakke SB, Chung HJ, et al. Glutamine has antidepressive effects through increments of glutamate and glutamine levels and glutamatergic activity in the medial prefrontal cortex. Neuropharmacology. 2018;143:143–52.
Anderson EM, Gomez D, Caccamise A, McPhail D, Hearing M. Chronic unpredictable stress promotes cell-specific plasticity in prefrontal cortex D1 and D2 pyramidal neurons. Neurobiol Stress. 2019;10: 100152.
Karst H, Sarabdjitsingh RA, van der Weerd N, Feenstra E, Damsteegt R, Joëls M. Age-dependent shift in spontaneous excitation-inhibition balance of infralimbic prefrontal layer II/III neurons is accelerated by early life stress, independent of forebrain mineralocorticoid receptor expression. Neuropharmacology. 2020;1(180): 108294.
Yan W, Liu JF, Han Y, Zhang W, Luo YX, Xue YX, et al. Protein kinase Mζ in medial prefrontal cortex mediates depressive-like behavior and antidepressant response. Mol Psychiatry. 2018;23(9):1878–91.
Li Z, Lee CS, Peng HY, Lin TB, Hsieh MC, Lai CY, et al. Lights at night mediate depression-like behavioral and molecular phenotypes in a glucocorticoid-dependent manner in male rats. Neuropharmacology. 2024;1(248): 109888.
Karst H, Droogers WJ, van der Weerd N, Damsteegt R, van Kronenburg N, Sarabdjitsingh RA, et al. Acceleration of GABA-switch after early life stress changes mouse prefrontal glutamatergic transmission. Neuropharmacology. 2023;15(234): 109543.
Lin S, Huang L, Luo ZC, Li X, Jin SY, Du ZJ, et al. The ATP level in the medial prefrontal cortex regulates depressive-like behavior via the medial prefrontal cortex-lateral habenula pathway. Biol Psychiatry. 2022;92(3):179–92.
Zhu Z, Wang G, Ma K, Cui S, Wang JH. GABAergic neurons in nucleus accumbens are correlated to resilience and vulnerability to chronic stress for major depression. Oncotarget. 2017;8(22):35933–45.
Xu A, Cui S, Wang JH. Incoordination among subcellular compartments is associated with depression-like behavior induced by chronic mild stress. Int J Neuropsychopharmacol. 2016;19(5):122.
Riga D, Kramvis I, Koskinen MK, van Bokhoven P, van der Harst JE, Heistek TS, et al. Hippocampal extracellular matrix alterations contribute to cognitive impairment associated with a chronic depressive-like state in rats. Sci Transl Med. 2017;9(421):eaai8753.
Heshmati M, Christoffel DJ, LeClair K, Cathomas F, Golden SA, Aleyasin H, et al. Depression and social defeat stress are associated with inhibitory synaptic changes in the nucleus accumbens. J Neurosci. 2020;40(32):6228–33.
Han RT, Kim YB, Park EH, Kim JY, Ryu C, Kim HY, et al. Long-term isolation elicits depression and anxiety-related behaviors by reducing oxytocin-induced GABAergic transmission in central amygdala. Front Mol Neurosci. 2018;11:246.
Li M, Sun X, Wang Z, Li Y. Caspase-1 affects chronic restraint stress-induced depression-like behaviors by modifying GABAergic dysfunction in the hippocampus. Transl Psychiatry. 2023;13(1):229.
McKlveen JM, Morano RL, Fitzgerald M, Zoubovsky S, Cassella SN, Scheimann JR, et al. Chronic Stress increases prefrontal inhibition: a mechanism for stress-induced prefrontal dysfunction. Biol Psychiatry. 2016;80(10):754–64.
Yang L, Xu T, Zhang K, Wei Z, Li X, Huang M, et al. The essential role of hippocampal alpha6 subunit-containing GABAA receptors in maternal separation stress-induced adolescent depressive behaviors. Behav Brain Res. 2016;15(313):135–43.
Mitchell SJ, Maguire EP, Cunningham L, Gunn BG, Linke M, Zechner U, et al. Early-life adversity selectively impairs α2-GABAA receptor expression in the mouse nucleus accumbens and influences the behavioral effects of cocaine. Neuropharmacology. 2018;141:98–112.
Talani G, Biggio F, Gorule AA, Licheri V, Saolini E, Colombo D, et al. Sex-dependent changes of hippocampal synaptic plasticity and cognitive performance in C57BL/6J mice exposed to neonatal repeated maternal separation. Neuropharmacology. 2023;1(222): 109301.
Teleanu RI, Niculescu AG, Roza E, Vladâcenco O, Grumezescu AM, Teleanu DM. Neurotransmitters-key factors in neurological and neurodegenerative disorders of the central nervous system. Int J Mol Sci. 2022;23(11):5954.
Werner FM, Coveñas R. Classical neurotransmitters and neuropeptides involved in major depression: a review. Int J Neurosci. 2010;120(7):455–70.
Nemeroff CB. Recent advances in the neurobiology of depression. Psychopharmacol Bull. 2002;36(Suppl 2):6–23.
Silić A, Vukojević J, Peitl V, De Hert M, Karlović D. Major depressive disorder: a possible typisation according to serotonin, inflammation, and metabolic syndrome. Acta Neuropsychiatr. 2022;34(1):15–23.
Ironside M, Moser AD, Holsen LM, Zuo CS, Du F, Perlo S, et al. Reductions in rostral anterior cingulate GABA are associated with stress circuitry in females with major depression: a multimodal imaging investigation. Neuropsychopharmacology. 2021;46(12):2188–96.
Blier P, El Mansari M. Serotonin and beyond: therapeutics for major depression. Philos Trans R Soc Lond B Biol Sci. 2013;368(1615):20120536.
Benson KL, Bottary R, Schoerning L, Baer L, Gonenc A, Eric Jensen J, et al. 1H MRS measurement of cortical GABA and glutamate in primary insomnia and major depressive disorder: relationship to sleep quality and depression severity. J Affect Disord. 2020;1(274):624–31.
Inoshita M, Umehara H, Watanabe SY, Nakataki M, Kinoshita M, Tomioka Y, et al. Elevated peripheral blood glutamate levels in major depressive disorder. Neuropsychiatr Dis Treat. 2018;14:945–53.
Gjerris A, Sørensen AS, Rafaelsen OJ, Werdelin L, Alling C, Linnoila M. 5-HT and 5-HIAA in cerebrospinal fluid in depression. J Affect Disord. 1987;12(1):13–22.
Garakani A, Martinez JM, Yehuda R, Gorman JM. Cerebrospinal fluid levels of glutamate and corticotropin releasing hormone in major depression before and after treatment. J Affect Disord. 2013;146(2):262–5.
Valenti O, Lodge DJ, Grace AA. Aversive stimuli alter ventral tegmental area dopamine neuron activity via a common action in the ventral hippocampus. J Neurosci. 2011;31(11):4280–9.
Valenti O, Gill KM, Grace AA. Different stressors produce excitation or inhibition of mesolimbic dopamine neuron activity: response alteration by stress pre-exposure. Eur J Neurosci. 2012;35(8):1312–21.
Medrihan L, Sagi Y, Inde Z, Krupa O, Daniels C, Peyrache A, et al. Initiation of behavioral response to antidepressants by cholecystokinin neurons of the dentate gyrus. Neuron. 2017;95(3):564-576.e4.
Fawcett J, Barkin RL. Review of the results from clinical studies on the efficacy, safety and tolerability of mirtazapine for the treatment of patients with major depression. J Affect Disord. 1998;51(3):267–85.
Ziff EB. Enlightening the postsynaptic density. Neuron. 1997;19(6):1163–74.
Sheng M, Kim E. The postsynaptic organization of synapses. Cold Spring Harb Perspect Biol. 2011;3(12): a005678.
Carlin RK, Grab DJ, Cohen RS, Siekevitz P. Isolation and characterization of postsynaptic densities from various brain regions: enrichment of different types of postsynaptic densities. J Cell Biol. 1980;86(3):831–45.
Harris KM, Weinberg RJ. Ultrastructure of synapses in the mammalian brain. Cold Spring Harb Perspect Biol. 2012;4(5): a005587.
Feyissa AM, Chandran A, Stockmeier CA, Karolewicz B. Reduced levels of NR2A and NR2B subunits of NMDA receptor and PSD-95 in the prefrontal cortex in major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33(1):70–5.
Zhang L, Luo J, Zhang M, Yao W, Ma X, Yu SY. Effects of curcumin on chronic, unpredictable, mild, stress-induced depressive-like behaviour and structural plasticity in the lateral amygdala of rats. Int J Neuropsychopharmacol. 2014;17(5):793–806.
Turrigiano GG. The self-tuning neuron: synaptic scaling of excitatory synapses. Cell. 2008;135(3):422–35.
Turrigiano G. Homeostatic synaptic plasticity: local and global mechanisms for stabilizing neuronal function. Cold Spring Harb Perspect Biol. 2012;4(1): a005736.
Turrigiano GG. Homeostatic plasticity in neuronal networks: the more things change, the more they stay the same. Trends Neurosci. 1999;22(5):221–7.
O’Brien RJ, Kamboj S, Ehlers MD, Rosen KR, Fischbach GD, Huganir RL. Activity-dependent modulation of synaptic AMPA receptor accumulation. Neuron. 1998;21(5):1067–78.
Siddoway B, Hou H, Xia H. Molecular mechanisms of homeostatic synaptic downscaling. Neuropharmacology. 2014;78:38–44.
Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng P, et al. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature. 2011;475(7354):91–5.
Suzuki K, Kim JW, Nosyreva E, Kavalali ET, Monteggia LM. Convergence of distinct signaling pathways on synaptic scaling to trigger rapid antidepressant action. Cell Rep. 2021;37(5): 109918.
Taha E, Gildish I, Gal-Ben-Ari S, Rosenblum K. The role of eEF2 pathway in learning and synaptic plasticity. Neurobiol Learn Mem. 2013;105:100–6.
Nosyreva E, Szabla K, Autry AE, Ryazanov AG, Monteggia LM, Kavalali ET. Acute suppression of spontaneous neurotransmission drives synaptic potentiation. J Neurosci. 2013;33(16):6990–7002.
Aoto J, Nam CI, Poon MM, Ting P, Chen L. Synaptic signaling by all-trans retinoic acid in homeostatic synaptic plasticity. Neuron. 2008;60(2):308–20.
Wang HL, Zhang Z, Hintze M, Chen L. Decrease in calcium concentration triggers neuronal retinoic acid synthesis during homeostatic synaptic plasticity. J Neurosci. 2011;31(49):17764–71.
McAllister AK, Katz LC, Lo DC. Neurotrophins and synaptic plasticity. Annu Rev Neurosci. 1999;22:295–318.
Lin PY, Kavalali ET, Monteggia LM. Genetic dissection of presynaptic and postsynaptic BDNF-TrkB signaling in synaptic efficacy of CA3-CA1 synapses. Cell Rep. 2018;24(6):1550–61.
Gideons ES, Lin PY, Mahgoub M, Kavalali ET, Monteggia LM. Chronic lithium treatment elicits its antimanic effects via BDNF-TrkB dependent synaptic downscaling. Elife. 2017;16(6): e25480.
Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiatry. 2006;59(12):1116–27.
Cavaleri D, Moretti F, Bartoccetti A, Mauro S, Crocamo C, Carrà G, et al. The role of BDNF in major depressive disorder, related clinical features, and antidepressant treatment: Insight from meta-analyses. Neurosci Biobehav Rev. 2023;149: 105159.
Zelada MI, Garrido V, Liberona A, Jones N, Zúñiga K, Silva H, et al. Brain-derived neurotrophic factor (BDNF) as a predictor of treatment response in major depressive disorder (MDD): a systematic review. Int J Mol Sci. 2023;24(19):14810.
Xu D, Gao LN, Song XJ, Dong QW, Chen YB, Cui YL, et al. Enhanced antidepressant effects of BDNF-quercetin alginate nanogels for depression therapy. J Nanobiotechnology. 2023;21(1):379.
Montgomery SL, Bowers WJ. Tumor necrosis factor-alpha and the roles it plays in homeostatic and degenerative processes within the central nervous system. J Neuroimmune Pharmacol. 2012;7(1):42–59.
Stellwagen D, Malenka RC. Synaptic scaling mediated by glial TNF-alpha. Nature. 2006;440(7087):1054–9.
Steinmetz CC, Turrigiano GG. Tumor necrosis factor-α signaling maintains the ability of cortical synapses to express synaptic scaling. J Neurosci. 2010;30(44):14685–90.
Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67(5):446–57.
Pandey GN, Rizavi HS, Zhang H, Bhaumik R, Ren X. Abnormal protein and mRNA expression of inflammatory cytokines in the prefrontal cortex of depressed individuals who died by suicide. J Psychiatry Neurosci. 2018;43(6):376–85.
Del Grande da Silva G, Wiener CD, Barbosa LP, Gonçalves Araujo JM, Molina ML, San Martin P, et al. Pro-inflammatory cytokines and psychotherapy in depression: results from a randomized clinical trial. J Psychiatr Res. 2016;75:57–64.
Liu JJ, Wei YB, Strawbridge R, Bao Y, Chang S, Shi L, et al. Peripheral cytokine levels and response to antidepressant treatment in depression: a systematic review and meta-analysis. Mol Psychiatry. 2020;25(2):339–50.
Cao P, Chen C, Liu A, Shan Q, Zhu X, Jia C, et al. Early-life inflammation promotes depressive symptoms in adolescence via microglial engulfment of dendritic spines. Neuron. 2021;109(16):2573-2589.e9.
Duseja R, Heir R, Lewitus GM, Altimimi HF, Stellwagen D. Astrocytic TNFα regulates the behavioral response to antidepressants. Brain Behav Immun. 2015;44:187–94.
Shearer KD, Stoney PN, Morgan PJ, McCaffery PJ. A vitamin for the brain. Trends Neurosci. 2012;35(12):733–41.
Hu P, van Dam AM, Wang Y, Lucassen PJ, Zhou JN. Retinoic acid and depressive disorders: Evidence and possible neurobiological mechanisms. Neurosci Biobehav Rev. 2020;112:376–91.
Otto LR, Clemens V, Üsekes B, Cosma NC, Regen F, Hellmann-Regen J. Retinoid homeostasis in major depressive disorder. Transl Psychiatry. 2023;13(1):67.
Huang C, Chen JT. Chronic retinoic acid treatment induces affective disorders by impairing the synaptic plasticity of the hippocampus. J Affect Disord. 2020;1(274):678–89.
Wang T, Hauswirth AG, Tong A, Dickman DK, Davis GW. Endostatin is a trans-synaptic signal for homeostatic synaptic plasticity. Neuron. 2014;83(3):616–29.
Pratt KG, Zimmerman EC, Cook DG, Sullivan JM. Presenilin 1 regulates homeostatic synaptic scaling through Akt signaling. Nat Neurosci. 2011;14(9):1112–4.
Shepherd JD, Rumbaugh G, Wu J, Chowdhury S, Plath N, Kuhl D, et al. Arc/Arg3.1 mediates homeostatic synaptic scaling of AMPA receptors. Neuron. 2006;52(3):475–84.
Acknowledgements
Not applicable.
Funding
This work was partially supported by the National Science Foundation of China (Grant No. 82374586), the National Key R&D Program of China (Project No. 2022YFC3500405), the Natural Science Foundation of Shandong Province (Grant No. ZR2022QH083), and the China Postdoctoral Science Foundation (Grants No. 2021M702045 and 2024M751906), the National Comprehensive Traditional Chinese Medicine Reform Demonstration Zone Science and Technology Collaborative Development Project(GZY-KJS-SD-2024-046), and the Shanghai nstitute of Traditional chinese Medicine for mental health (SZB2024101). We express our sincere thanks to the Shandong Key Laboratory of Innovation and Application Research in Basic Theory of Traditional Chinese Medicine and Key Laboratory of Traditional Chinese Medicine Classical Theory for their invaluable support and commitment to this research.
Author information
Authors and Affiliations
Contributions
B. Wang, and Y. Chen conceived and designed project. B. Wang, T. He, G. Qiu, C. Li, S. Xue prepared the reference. T. Wang prepared the figures. B. Wang and J. Yan wrote the manuscript. Y. Xia, L. Yao and Y. Chen helped revise the manuscript. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
With the submission of this manuscript, we would like to undertake that all authors of this paper have read and approved the final version submitted.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
About this article
Cite this article
Wang, B., He, T., Qiu, G. et al. Altered synaptic homeostasis: a key factor in the pathophysiology of depression. Cell Biosci 15, 29 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13578-025-01369-y
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13578-025-01369-y