Skip to main content

Potassium channels in depression: emerging roles and potential targets

Abstract

Potassium ion channels play a fundamental role in regulating cell membrane repolarization, modulating the frequency and shape of action potentials, and maintaining the resting membrane potential. A growing number of studies have indicated that dysfunction in potassium channels associates with the pathogenesis and treatment of depression. However, the involvement of potassium channels in the onset and treatment of depression has not been thoroughly summarized. In this review, we performed a comprehensive analysis of the association between multiple potassium channels and their roles in depression, and compiles the SNP loci of potassium channels associated with depression, as well as antidepressant drugs that target these channels. We discussed the pivotal role of potassium channels in the treatment of depression, provide valuable insights into new therapeutic targets for antidepressant treatment and critical clues to future drug discovery.

Introduction

Depression is a prevalent mood disorder that imposes considerable economic strain on both families and society [1]. Globally, more than 300 million individuals are affected by depression [2], yet upwards of 45% of patients do not experience a significant response to antidepressant medication treatments [3]. Despite its widespread occurrence, the etiology of depression is multifaceted, its manifestations are profound, and its pathogenesis remains enigmatic [4]. This underscores the critical need for continued investigation into its underlying mechanisms and the identification of novel therapeutic targets. Ion channels are protein structures located on cell membranes that permit specific ions, these include cation channels, such as potassium channels, sodium channels, and calcium channels, as well as anion channels such as chloride channels. Potassium channels significantly influence neural activity by regulating the duration of action potentials and neurotransmitter release [5,6,7]. Previous studies highlight that potassium channels are closely linked to the occurrence and progression of various diseases, such as arrhythmias [8], epilepsy [9], Alzheimer's disease [10], cerebral ischemia [11], and diabetes [12], etc. Intriguingly, an increasing number of recent studies indicate that changes in neural activity can contribute to the development of mental disorders, including depression [13,14,15]. Potassium channels, which are essential for neuronal excitability and the conduction of signals, play a pivotal role in modulating depressive states [16]. Therefore, understanding the relationship between ion channels and depression is crucial for elucidating pathogenesis and developing effective treatments for depression.

Classification and functions of potassium channels

Potassium channels represent the most diverse subtype among all ion channels and are distributed in both excitatory and inhibitory neurons. They play essential roles in both excitable and non-excitable cells [17]. These channels consist of tetrameric integral membrane proteins that form transmembrane water pores. Based on the primary amino acid sequence of the pore-containing α-subunits as well as their physiological and pharmacological characteristics [18], potassium (K⁺) channels can be categorized into four main classes: (1) voltage-gated K+ channels (KV); (2) inward rectifier K+ channels (Kir); (3) calcium-activated K+ channels (KCa); and (4) two-pore domain K⁺ channels (K2P) (Fig. 1). They play an essential role in regulating activities such as heart rate, muscle contraction, neurotransmitter release, neuronal excitability, insulin secretion, electrolyte transport in epithelial cells, cell volume regulation, and apoptosis [19].

Fig. 1
figure 1

Structure and Classification of Potassium Channels. Based on subunit structure, potassium channels can be divided into four families: KV, Kir, KCa, and K2P

The KV family represents the most diverse and functionally complex group of potassium channels [20]. The KV channel structure consists of four subunits, each arranged around a central pore and containing six transmembrane helices (TMs) [21]. This family encompasses twelve distinct members, designated as KV1 to KV12 [22]. They regulate neuronal excitability by modulating cell membrane repolarization [23], influencing neuronal excitability and various electrophysiological properties such as spike membrane potential, action potential waveform, and frequency [24]. Most KV channels open during membrane depolarization and close during hyperpolarization, which helps return the cell to its resting state [25].

The primary structures of Kir channels feature a conserved motif comprising two putative membrane-spanning domains (TM1 and TM2), and they comprise the subfamilies Kir1 to Kir7 [26]. Kir channels allow K+ to move more easily into rather than out of the cell. The primary physiological roles of Kir channels are to stabilize the resting membrane potential (RMP) and regulate the excitability of excitable cells [27]. Kir channels coordinate both passive and active electrical properties of cells and participate in G protein-coupled receptor signaling, thereby linking cellular metabolism with membrane excitability [28].

KCa channels are a type of potassium channel whose activity is modulated by fluctuations in intracellular calcium ion concentration. KCa channels are classified into three primary categories: big conductance K + channels (BK), intermediate conductance K + channels (IK), and small conductance K + channels (SK) [29]. BK channels are composed of tetrameric structures, with each assembly consisting of four pore-forming α-subunits, each containing seven transmembrane domains (TMs). Similarly, IK and SK channels are also tetrameric, with each subunit exhibiting a Shaker-like membrane topology comprising six TMs [30]. These channels open or close in response to fluctuations in calcium levels, affecting the amplitude and duration of calcium transients. This modulation influences downstream signaling pathways triggered by changes in intracellular calcium concentration [31]. In central neurons, KCa channels regulate neuronal excitability, firing patterns, and synaptic signal transduction. They also contribute to action potential repolarization, mediate the rapid phase of afterhyperpolarization (AHP), and influence neurotransmitter release [32].

K2P channels are characterized by two pore-forming domains and four transmembrane domains. K2P channels can be subdivided into various types based on amino acid homology, including TREK, TALK, TWIK, TRESK, THIK and TASK [33]. They can be activated across the entire physiological voltage range and are crucial components of background potassium currents [34]. K2P channels are essential for neuronal excitability [35]. They play critical roles in regulating and maintaining the RMP, modulating cellular excitability, and balancing intracellular and extracellular potassium ions[36].

Genetic links between potassium channels and depression

Extensive research has underscored the profound influence of genetic factors on the psychopathology of depression and other mental health disorders [37]. One such study examined frozen striatal samples from the Stanley Foundation Brain Collection, utilizing Reverse Transcription-Polymerase Chain Reaction (RT-PCR) techniques. The analysis included 60 brain samples, drawn from both individuals with depression and those without the disorder. The findings indicated a significant upregulation of the KCNJ4 and KCNJ1 subunits of the Kir2.3 potassium channel, as well as the KCNN3 subunit of the SK3 channel, in the striatal samples from individuals with major depression. In contrast, the expression of the KCNA1 subunit of the KV1.1 channel and the KCNS3 subunit of the KV9.3 channel was downregulated in the striatum of these subjects. These alterations in potassium channel gene expression within the striatum-nucleus accumbens region suggest a decrease in electrical activity in this brain area, which may be associated with the symptoms and therapeutic responses observed in depression [38].Single nucleotide polymorphisms (SNPs), a key form of genetic variation, have significant implications for genetic research and clinical diagnosis [39]. A study involving 449 patients with MDD and 421 healthy controls to examine the association between polymorphisms in the KCNK2 gene and MDD, as well as the efficacy of antidepressant treatment. They found that the genotype frequency of rs6686529 differed significantly between MDD patients and controls. Individuals with the homozygous genotype showed greater susceptibility to MDD compared to those with the heterozygous genotype, suggesting a potential dominant effect of this polymorphism on MDD [40]. Furthermore, this polymorphism affected the efficacy of antidepressant treatment: homozygous individuals were more likely to achieve remission after 8 weeks of treatment compared to G allele carriers. These findings indicate that KCNK2 is involved in both susceptibility to MDD and response to antidepressant therapy.

After investigating six SNPs in the KCNK2 gene among 590 patients with MDD and 441 controls, it was found that the A homozygote of rs10779646 was significantly more prevalent in patients than in controls. Additionally, the G allele of rs7549184 was associated with the presence of psychotic symptoms and disease severity [41]. A study examined SNPs in the KCNK2 gene in 565 patients with major depression and found that variations in SNPs (rs12136349, rs2841616, rs2841608) correlated with the response to citalopram treatment. This suggests that genetic variations in KCNK2 could help identify individuals at risk of treatment resistance [42].

Recent research indicates that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are essential for regulating neuronal excitability and are widely expressed in the brain. The HCN4 gene, which is expressed in brain regions linked to mood and depression, has two SNPs (rs3859014 and rs12905212) significantly associated with major depression [43]. A meta-analysis identified a novel SNP, rs79878474, located on chr11p15, which is likely associated with depression. The KV3 and Kir-6.2 potassium channels, encoded by the KCNC1, KCNJ11, and ABCC8 genes, show a close relationship with depression in children and adolescents [44]. Additionally, SNPs rs9394578 and rs2815095, associated with the KCNK5 gene, are significantly linked to depression [45]. Current research on potassium channel genes and depression primarily focuses on the TREK-1 type potassium channels (Table 1), with less extensive studies on other potassium channel types, underscoring the need for further investigation in this area.

Table 1 SNPs of Potassium Channels Associated with Depression

Pathological roles of potassium channels for depression

The pathogenesis of depression is complex. Current research suggests that the onset of depression is closely related to monoamine neurotransmitters [46], the neuroendocrine system [47], immune cytokines [48], neural plasticity [49], and gut microbiota [50], etc. However, these mechanisms and hypotheses do not fully explain the pathogenesis of depression. Increasing studies indicate that potassium channels, which are fundamental to neuronal excitability and signal transmission, are closely associated with both the onset and treatment of depression channels[51](Table 2).

Table 2 Functions of Different Subtypes of Potassium Channels and Their Roles in Depression

Potassium channels in the mPFC region

The pathogenesis of depression involve several brain regions, including the frontal cortex (PFC), hippocampus, nucleus accumbens (NAc), ventral tegmental area (VTA), lateral habenula (LHb) and dorsal raphe nucleus [52]. Among these regions, the PFC is essential for regulating emotions, cognition, and decision-making [53]. It plays a significant role in emotional regulation and social interaction [54], making PFC activity a predictor of treatment outcomes for depression [55]. In individuals with major depressive disorder (MDD), the protein expression of inward-rectifying Kir4.1 channels in the parietal cortex is notably increased [56]. Additionally, studies conducted in vitro, in vivo, and post-mortem have suggested that the heightened activity of Kir4.1 channels may contribute to the development of depression [57]. Moreover, the excitability of pyramidal neurons in the cingulate cortex is enhanced, as indicated by alterations in membrane potential and spike frequency in response to current injection, which is associated with increased activity of SK channels [58] and the inhibition of BK channels [59].

A growing body of evidence indicates that potassium channel activity in the medial prefrontal cortex (mPFC) is closely linked to the onset and treatment of depression. Research suggests that KV7/KCNQ/M currents modulate the excitability and synaptic transmission of neurons in the PFC of rats. The inhibition of muscarinic potassium currents has been shown to significantly enhance the amplitude of excitatory postsynaptic potentials and depolarize the resting membrane potential (RMP), thereby influencing the pathophysiology of depression [60]. Furthermore, other studies have revealed an increase in protein expression of the KV2.1 channel in the mPFC of depressive animal models [61]. Pharmacological treatments or loss of function have demonstrated improvements in depressive-like behaviors in these animals [62]. These findings suggest that the neurobehavioral pathophysiology associated with depression in the mPFC region is linked to abnormal potassium channel function, underscoring the critical role of potassium channels in the antidepressant process.

Potassium channels in the hippocampal region

Damage or dysfunction of the hippocampus is also linked to depression and other emotional disorders [63]. Potassium channels in the hippocampal region are critical for neuronal activity and the development of depression. In mice, a reduction in KV3.1 levels in parvalbumin-positive (PV⁺) cells within the hippocampus leads to decreased high-frequency firing capacity of dentate gyrus PV+ cells, and the emergence of depressive-like behaviors. Conversely, upregulating KV3.1 or acutely activating KV3.1 with specific agonists alleviates depressive-like behaviors in mice [64]. The anesthetic ketamine exerts its sustained antidepressant effects by specifically modulating the KCNQ2 gene in glutamatergic neurons of the ventral hippocampus in mice [65]. Additionally, in lipopolysaccharide (LPS) model mice, Kir4.1 protein expression in hippocampal astrocytes is elevated, and the NLRP3 inflammasome is activated. Decreasing Kir4.1 protein levels specifically in astrocytes can decrease LPS-induced NLRP3 and calpain-1 expression and improve depressive-like behaviors in these mice [66]. Studies have shown that in depressive animal model, the activity of ATP-sensitive potassium (K-ATP) channels in the hippocampus is elevated, accompanied by increased expression of Kir6.1 and Kir6.2 subunits [67]. Administration of the N-methyl-D-aspartic(NMDA) receptor antagonist memantine inhibits K-ATP channels in the hippocampus and improves depressive-like behaviors in mice [68]. These findings suggest that Potassium channels in the hippocampus contribute to the pathogenesis of depression and may represent a promising target for treatment.

Elevated expression of TREK-1 in the hippocampus of mice subjected to chronic unpredictable mild stress (CUMS) has been documented. Targeted knockdown of TREK-1 in the hippocampal region has been shown to prevent impairment of glutamatergic synaptic transmission and ameliorate depressive-like behaviors in these mice [69]. TREK-1 knockout mice exhibit increased serotonergic neuronal activity and neurotransmission, reduced immobility time in the forced swim and tail suspension tests, and demonstrate improved outcomes in various depression-related behavioral models [70]. Moreover, the knockdown of the TASK-3 channel in monoaminergic neurons within the hippocampus leads to significant antidepressant-like effects [71]. The pivotal role of ion channels in the hippocampus underscores their substantial involvement in the pathophysiology of depression and other psychiatric conditions [72].

Potassium channels in reward-related brain regions

The reward and anti-reward circuitry encompasses several brain regions, including the NAc, the VTA and the LHb [73]. Neuronal activity in these regions is strongly linked to the onset and treatment of depression [74, 75], with potassium channels playing a crucial role. Research has demonstrated that, in the NAc of rats, accelerated inactivation of KV1.4 channels in striatal projection neurons results in diminished reward-seeking behaviors. This effect inhibits reward motivation and contributes to the development of depressive disorders [76]. Moreover, studies using the CUMS model in mice have revealed enhanced theta-frequency long-term potentiation (tLTP) in the NAc, accompanied by reduced activity of KV4.2-type potassium currents. Direct phosphorylation at Ser-616 can mitigate the alterations in tLTP mediated by KV4.2 channels in these mice [77].

Additionally, in the social defeat model of mice, reduced activity of Kv7.4 channels in dopaminergic neurons of the VTA has been observed. Activation of Kv7.4 channels can reverse depressive-like behaviors in these mice [78]. Experimental studies have shown that in depression model rats, Kir4.1 channel activity increases in the lateral habenula (LHb) region. Furthermore, interactions between astrocytes and neurons in the LHb play a role in determining neuronal firing patterns [79], which may be relevant to the mechanism of depression. Studies have shown that activation of SK channels leads to age-dependent depressive-like behaviors and cognitive deficits. Inhibition of SK channels in dopaminergic neurons within the VTA improves depressive-like behaviors in post-stroke depression models in rats [80]. Collectively, these findings underscore the significant role of potassium channels in the reward circuitry and their involvement in both the pathogenesis and treatment of depression.

Potassium channels as potential targets for antidepressant action

Potassium channels participated in the pathogenesis and treatment of depression

Recent research suggests that potassium channel openers and blockers can influence the development of depression. KCNQ2/3 potassium channel openers have shown antidepressant effects. Clinical trials with the KCNQ2/3 channel opener ezogabine in patients with MDD have demonstrated significant improvements in depressive symptoms and anhedonia [95, 99]. The human studies on KCNQ channel activator ezogabine also indicate that modulating KCNQ channels can regulate neuronal excitability within the reward circuits, offering a potential target for alleviating depressive symptoms [114]. Specific inhibition of TREK-1 channel activity has also demonstrated distinct antidepressant properties [113]. Medications targeting the TREK-1 channel, such as spadin, have shown promising therapeutic effects in patients with depression [115].

Potassium channels play a crucial role in regulating immobility time in the forced swim test in mice. Blockers of potassium channels, such as TEA and charybdotoxin, significantly reduce immobility time. Conversely, openers of potassium channels, such as pinacidil, minoxidil, and cromakalim, significantly increase immobility time in mice [116]. For inward-rectifying potassium channels, the Kir4.1 channel inhibitor Lys05 induces a rapid antidepressant response in mice [117]. Inhibitors of ATP-sensitive potassium channels, such as glibenclamide, reduce the hypothalamic–pituitary–adrenal axis hyperactivity, depression, and anxiety-related symptoms in Alzheimer's disease rat models [118]. Moreover, administering the K-ATP channel opener iptakalim significantly increases sucrose preference and reduces immobility time in the forced swim test, thereby improving depressive-like behaviors in chronic mild stress-induced depression model mice [119].

The neuroprotective agent sipatrigine, which significantly inhibits the TREK-1 channel at therapeutic concentrations, exhibits notable antidepressant effects in the forced swim and other depression models [120]. Additionally, TASK-3 channel knockout mice show significantly reduced immobility time in the tail suspension and forced swim tests compared to wild-type controls. They also exhibit enhanced locomotor activity in the novel object recognition test [112]. Collectively, these studies suggest that directly influencing potassium channel activity can alleviate depressive symptoms, highlighting the critical role of potassium channels in both the pathogenesis and treatment of depression.

Potassium ion channels can serve as therapeutic targets for antidepressant drugs

Recent studies increasingly suggest that potassium channels may play a significant role as targets for antidepressant drugs. Studies have shown that the antidepressant fluoxetine markedly enhances delayed rectifier potassium currents in cerebellar granule neurons and transient outward potassium currents in the hippocampus of hamsters [121]. Additionally, KV4.2 knockout rats display notable depressive tendencies in forced swimming and other depression tests, and fluoxetine does not reverse this depressive phenotype. This finding implies that KV4.2 might be a target of fluoxetine in the treatment of depression [90]. Fluoxetine increases the inactivation rate of KV4.3 currents in a concentration-dependent manner [122]. Various voltage-dependent potassium channel subtypes, such as KV3.1 and KV1.1, which are highly expressed in the brain, also exhibit significant inhibitory effects [123, 124]. Furthermore, fluoxetine effectively inhibits GIRK2 channels expressed in Xenopus oocytes and significantly reduces cell death in cerebellar and pontine neurons, thereby enhancing motor abilities in mice [99].

Other studies have shown that the antidepressant citalopram can decrease KV1.5 currents [84], while escitalopram inhibits TREK-1 currents in the hippocampus and prefrontal cortex of post-stroke depression model rats [125]. Chlorpromazine also treats depression and other psychiatric disorders by activating BK channels at the whole-cell level [105]. Additionally, ketamine reduces the surface density of Kir4.1 channels in astrocytes, which modulates neuronal excitability and alleviates depressive symptoms [126, 127]. These findings collectively suggest that potassium channels are potential targets for antidepressants and play a role in both the pathogenesis and treatment of depression.

Conclusion and future perspectives

This review delineates the role of potassium channels in depression. The available research evidence suggests multiple potassium channels have emerged as potential targets for antidepressant drugs. Meanwhile, we summarized several potassium channel gene polymorphisms associated with depression, which may have predictive value for susceptibility to depression and response to treatment. As far as is known from the existing studies, the complex relationship between potassium channels and depression involves not only the different brain areas (mPFC, hippocampal, NAc, etc.), but also the various cells(dopaminergic neurons, PV+ cells, astrocytes, etc.) involved, forming a sophisticated network to affect onset of depression or produce antidepressant effect (Fig. 2). But the connections between potassium channels and their relationships with depression across multiple brain regions and the specific signaling pathways involved remain unclear. Therefore, depth research into the role of potassium channels in depression is crucial. Uncovering the roles of these key channels in the pathogenesis of depression will advance our understanding and may reveal new therapeutic targets for this debilitating disorder.

Fig. 2
figure 2

Role of various potassium channels in the pathogenesis and treatment of depression. During the onset and treatment of depression, various types of potassium channels play significant roles in different brain regions

Availability of data and materials

Not applicable.

Abbreviations

KV:

Voltage-gated K+ channels

Kir:

Inward rectifier K+ channels

KCa:

Calcium-activated K+ channels

K2P:

Two-pore domain K⁺ channels

K-ATP:

ATP-sensitive potassium

TMs:

Transmembrane helices

BK:

Big conductance K+ channels

IK:

Intermediate conductance K+ channels

SK:

Small conductance K+ channels

PFC:

Prefrontal cortex

mPFC:

Medial prefrontal cortex

CUMS:

Chronic unpredictable mild stress

RMP:

Resting membrane potential

NAc:

Nucleus accumbens

VTA:

Ventral tegmental area

tLTP:

Theta-frequency long-term potentiation

MDD:

Major depressive disorder

LHb:

Lateral habenula

LPS:

Lipopolysaccharide

SNPs:

Single nucleotide polymorphisms

HCN:

Hyperpolarization-activated cyclic nucleotide-gated

NMDA:

N-methyl-d-aspartic

References

  1. 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 Lond Engl. 2022;399(10328):957–1022.

    Article  Google Scholar 

  2. Quintanilla B, Zarate CA, Pillai A. Ketamine’s mechanism of action with an emphasis on neuroimmune regulation: can the complement system complement ketamine’s antidepressant effects? Mol Psychiatry. 2024;29(9):2849–58.

    Article  CAS  PubMed  Google Scholar 

  3. 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 Off J World Psychiatr Assoc WPA. 2020;19(2):214–32.

    Google Scholar 

  4. Howes OD, Thase ME, Pillinger T. Treatment resistance in psychiatry: state of the art and new directions. Mol Psychiatry. 2022;27(1):58–72.

    Article  PubMed  Google Scholar 

  5. Imbrici P, Camerino DC, Tricarico D. Major channels involved in neuropsychiatric disorders and therapeutic perspectives. Front Genet. 2013;4:76.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Moiseenkova-Bell V, Delemotte L, Minor DL. Ion channels: intersection of structure, function, and pharmacology. J Mol Biol. 2021;433(17):167102.

    Article  CAS  PubMed  Google Scholar 

  7. Krupp JJ. Role of ion channels in neurological disorders. CNS Neurol Disord Drug Targets. 2008;7(2):120–1.

    Article  CAS  PubMed  Google Scholar 

  8. Tian C, Zhu R, Zhu L, Qiu T, Cao Z, Kang T. Potassium channels: structures, diseases, and modulators. Chem Biol Drug Des. 2014;83(1):1–26.

    Article  CAS  PubMed  Google Scholar 

  9. French JA, Porter RJ, Perucca E, Brodie MJ, Rogawski MA, Pimstone S, et al. Efficacy and safety of XEN1101, a novel potassium channel opener, in adults with focal epilepsy: a phase 2b randomized clinical trial. JAMA Neurol. 2023;80(11):1145–54.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Etcheberrigaray R, Ito E, Oka K, Tofel-Grehl B, Gibson GE, Alkon DL. Potassium channel dysfunction in fibroblasts identifies patients with Alzheimer disease. Proc Natl Acad Sci USA. 1993;90(17):8209–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sun HS, Feng ZP. Neuroprotective role of ATP-sensitive potassium channels in cerebral ischemia. Acta Pharmacol Sin. 2013;34(1):24–32.

    Article  PubMed  Google Scholar 

  12. Catterall W, Epstein PN. Ion channels. Diabetologia. 1992;35(Suppl 2):S23-33.

    Article  PubMed  Google Scholar 

  13. Duman RS, Aghajanian GK. Synaptic dysfunction in depression: potential therapeutic targets. Science. 2012;338(6103):68–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Baert T, Vande Casteele T, Bouckaert F, Vandenbulcke M. Neural networks in late life depression. Tijdschr Voor Psychiatr. 2023;65(10):624–8.

    CAS  Google Scholar 

  15. Zhang B, Qi S, Liu S, Liu X, Wei X, Ming D. Altered spontaneous neural activity in the precuneus, middle and superior frontal gyri, and hippocampus in college students with subclinical depression. BMC Psychiatry. 2021;21(1):280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Pongs O. Regulation of excitability by potassium channels. Results Probl Cell Differ. 2008;44:145–61.

    Article  CAS  PubMed  Google Scholar 

  17. Kuang Q, Purhonen P, Hebert H. Structure of potassium channels. Cell Mol Life Sci CMLS. 2015;72(19):3677–93.

    Article  CAS  PubMed  Google Scholar 

  18. Maljevic S, Lerche H. Potassium channels: a review of broadening therapeutic possibilities for neurological diseases. J Neurol. 2013;260(9):2201–11.

    Article  CAS  PubMed  Google Scholar 

  19. MacKinnon R. Potassium channels. FEBS Lett. 2003;555(1):62–5.

    Article  CAS  PubMed  Google Scholar 

  20. Christie MJ, North RA, Osborne PB, Douglass J, Adelman JP. Heteropolymeric potassium channels expressed in Xenopus oocytes from cloned subunits. Neuron. 1990;4(3):405–11.

    Article  CAS  PubMed  Google Scholar 

  21. Choe S, Kreusch A, Pfaffinger PJ. Towards the three-dimensional structure of voltage-gated potassium channels. Trends Biochem Sci. 1999;24(9):345–9.

    Article  CAS  PubMed  Google Scholar 

  22. González C, Baez-Nieto D, Valencia I, Oyarzún I, Rojas P, Naranjo D, et al. K(+) channels: function-structural overview. Compr Physiol. 2012;2(3):2087–149.

    Article  PubMed  Google Scholar 

  23. Sands Z, Grottesi A, Sansom MSP. Voltage-gated ion channels. Curr Biol CB. 2005;15(2):R44-47.

    Article  CAS  PubMed  Google Scholar 

  24. Hille B. Ionic selectivity of Na and K channels of nerve membranes. Membranes. 1975;3:255–323.

    CAS  PubMed  Google Scholar 

  25. Hu JH, Malloy C, Tabor GT, Gutzmann JJ, Liu Y, Abebe D, et al. Activity-dependent isomerization of Kv4.2 by Pin1 regulates cognitive flexibility. Nat Commun. 2020;11(1):1567.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hibino H, Inanobe A, Furutani K, Murakami S, Findlay I, Kurachi Y. Inwardly rectifying potassium channels: their structure, function, and physiological roles. Physiol Rev. 2010;90(1):291–366.

    Article  CAS  PubMed  Google Scholar 

  27. Li WP, Cui XD, Cheng M. Function and expression of inwardly rectifying potassium channel Kir2.1. Sheng Li Ke Xue Jin Zhan. 2015;46(4):287–9.

    CAS  PubMed  Google Scholar 

  28. Mayfield J, Blednov YA, Harris RA. Behavioral and genetic evidence for girk channels in the CNS: role in physiology, pathophysiology, and drug addiction. Int Rev Neurobiol. 2015;123:279–313.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wei AD, Gutman GA, Aldrich R, Chandy KG, Grissmer S, Wulff H. International union of pharmacology. LII. Nomenclature and molecular relationships of calcium-activated potassium channels. Pharmacol Rev. 2005;57(4):463–72.

    Article  CAS  PubMed  Google Scholar 

  30. Weatherall KL, Goodchild SJ, Jane DE, Marrion NV. Small conductance calcium-activated potassium channels: from structure to function. Prog Neurobiol. 2010;91(3):242–55.

    Article  CAS  PubMed  Google Scholar 

  31. Kuiper EFE, Nelemans A, Luiten P, Nijholt I, Dolga A, Eisel U. K(Ca)2 and k(ca)3 channels in learning and memory processes, and neurodegeneration. Front Pharmacol. 2012;3:107.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Berkefeld H, Fakler B, Schulte U. Ca2+-activated K+ channels: from protein complexes to function. Physiol Rev. 2010;90(4):1437–59.

    Article  CAS  PubMed  Google Scholar 

  33. Kim D. Physiology and pharmacology of two-pore domain potassium channels. Curr Pharm Des. 2005;11(21):2717–36.

    Article  CAS  PubMed  Google Scholar 

  34. Ilan N, Goldstein SA. Kcnkø: single, cloned potassium leak channels are multi-ion pores. Biophys J. 2001;80(1):241–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lesage F, Lazdunski M. Molecular and functional properties of two-pore-domain potassium channels. Am J Physiol Renal Physiol. 2000;279(5):F793-801.

    Article  CAS  PubMed  Google Scholar 

  36. Mathie A, Al-Moubarak E, Veale EL. Gating of two pore domain potassium channels. J Physiol. 2010;588(Pt 17):3149–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Vendruscolo LF, Terenina-Rigaldie E, Raba F, Ramos A, Takahashi RN, Mormède P. Evidence for a female-specific effect of a chromosome 4 locus on anxiety-related behaviors and ethanol drinking in rats. Genes Brain Behav. 2006;5(6):441–50.

    Article  CAS  PubMed  Google Scholar 

  38. 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.

    Article  CAS  PubMed  Google Scholar 

  39. Carlton VEH, Ireland JS, Useche F, Faham M. Functional single nucleotide polymorphism-based association studies. Hum Genomics. 2006;2(6):391–402.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Liou YJ, Chen TJ, Tsai SJ, Yu YWY, Cheng CY, Hong CJ. Support for the involvement of the KCNK2 gene in major depressive disorder and response to antidepressant treatment. Pharmacogenet Genomics. 2009;19(10):735–41.

    Article  CAS  PubMed  Google Scholar 

  41. Congiu C, Minelli A, Bonvicini C, Bortolomasi M, Sartori R, Maj C, et al. The role of the potassium channel gene KCNK2 in major depressive disorder. Psychiatry Res. 2015;225(3):489–92.

    Article  CAS  PubMed  Google Scholar 

  42. Perlis RH, Moorjani P, Fagerness J, Purcell S, Trivedi MH, Fava M, et al. Pharmacogenetic analysis of genes implicated in rodent models of antidepressant response: association of TREK1 and treatment resistance in the STAR(*)D study. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2008;33(12):2810–9.

    Article  CAS  Google Scholar 

  43. Kelmendi B, Holsbach-Beltrame M, McIntosh AM, Hilt L, George ED, Kitchen RR, et al. Association of polymorphisms in HCN4 with mood disorders and obsessive compulsive disorder. Neurosci Lett. 2011;496(3):195–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Thapaliya B, Ray B, Farahdel B, Suresh P, Sapkota R, Holla B, et al. Cross-continental environmental and genome-wide association study on children and adolescent anxiety and depression. Res Sq. 2023.

  45. Yang Y, Zhao H, Boomsma DI, Ligthart L, Belin AC, Smith GD, et al. Molecular genetic overlap between migraine and major depressive disorder. Eur J Hum Genet EJHG. 2018;26(8):1202–16.

    Article  CAS  PubMed  Google Scholar 

  46. Kimura M, Yamanishi Y, Hanada T, Kagaya T, Kuwada M, Watanabe T, et al. Involvement of P-type calcium channels in high potassium-elicited release of neurotransmitters from rat brain slices. Neuroscience. 1995;66(3):609–15.

    Article  CAS  PubMed  Google Scholar 

  47. Duncan PJ, Shipston MJ. BK channels and the control of the pituitary. Int Rev Neurobiol. 2016;128:343–68.

    Article  CAS  PubMed  Google Scholar 

  48. Di A, Xiong S, Ye Z, Malireddi RKS, Kometani S, Zhong M, et al. The TWIK2 potassium efflux channel in macrophages mediates NLRP3 inflammasome-induced inflammation. Immunity. 2018;49(1):56-65.e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Fernández-Fernández D, Lamas JA. Metabotropic modulation of potassium channels during synaptic plasticity. Neuroscience. 2021;21(456):4–16.

    Article  Google Scholar 

  50. Maiuolo J, Gliozzi M, Musolino V, Carresi C, Scarano F, Nucera S, et al. The contribution of gut microbiota-brain axis in the development of brain disorders. Front Neurosci. 2021;15: 616883.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Qiu Q, Yang M, Gong D, Liang H, Chen T. Potassium and calcium channels in different nerve cells act as therapeutic targets in neurological disorders. Neural Regen Res. 2025;20(5):1258–76.

    Article  PubMed  Google Scholar 

  52. Pandya M, Altinay M, Malone DA, Anand A. Where in the brain is depression? Curr Psychiatry Rep. 2012;14(6):634–42.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Chen YH, Wu JL, Hu NY, Zhuang JP, Li WP, Zhang SR, et al. Distinct projections from the infralimbic cortex exert opposing effects in modulating anxiety and fear. J Clin Invest. 2021;131(14):e145692.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Hiser J, Koenigs M. The multifaceted role of the ventromedial prefrontal cortex in emotion, decision making, social cognition, and psychopathology. Biol Psychiatry. 2018;83(8):638–47.

    Article  PubMed  Google Scholar 

  55. Ghosal S, Hare B, Duman RS. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  56. 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.

    Article  CAS  PubMed  Google Scholar 

  57. Della Vecchia S, Marchese M, Santorelli FM, Sicca F. Kir4.1 dysfunction in the pathophysiology of depression: a systematic review. Cells. 2021;10(10):2628.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nashed MG, Waye S, Hasan SMN, Nguyen D, Wiseman M, Zhang J, et al. Antidepressant activity of pharmacological and genetic deactivation of the small-conductance calcium-activated potassium channel subtype-3. Psychopharmacology. 2022;239(1):253–66.

    Article  CAS  PubMed  Google Scholar 

  59. Sun P, Wang F, Wang L, Zhang Y, Yamamoto R, Sugai T, et al. Increase in cortical pyramidal cell excitability accompanies depression-like behavior in mice: a transcranial magnetic stimulation study. J Neurosci Off J Soc Neurosci. 2011;31(45):16464–72.

    Article  CAS  Google Scholar 

  60. Peng H, Bian XL, Ma FC, Wang KW. Pharmacological modulation of the voltage-gated neuronal Kv7/KCNQ/M-channel alters the intrinsic excitability and synaptic responses of pyramidal neurons in rat prefrontal cortex slices. Acta Pharmacol Sin. 2017;38(9):1248–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. 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.

    Article  PubMed  Google Scholar 

  62. Bambico FR, Li Z, Creed M, De Gregorio D, Diwan M, Li J, et al. A key role for prefrontocortical small conductance calcium-activated potassium channels in stress adaptation and rapid antidepressant response. Cereb Cortex. 2020;30(3):1559–72.

    Article  PubMed  Google Scholar 

  63. McEwen BS. Physiology and neurobiology of stress and adaptation: central role of the brain. Physiol Rev. 2007;87(3):873–904.

    Article  PubMed  Google Scholar 

  64. 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.

    Article  CAS  PubMed  Google Scholar 

  65. Lopez JP, Lücken MD, Brivio E, Karamihalev S, Kos A, De Donno C, et al. Ketamine exerts its sustained antidepressant effects via cell-type-specific regulation of Kcnq2. Neuron. 2022;110(14):2283-2298.e9.

    Article  CAS  PubMed  Google Scholar 

  66. Song Z, Bian Z, Zhang Z, Wang X, Zhu A, Zhu G. Astrocytic Kir4.1 regulates NMDAR/calpain signaling axis in lipopolysaccharide-induced depression-like behaviors in mice. Toxicol Appl Pharmacol. 2021;429:115711.

    Article  CAS  PubMed  Google Scholar 

  67. 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.

    Article  CAS  PubMed  Google Scholar 

  68. Moriguchi S, Inagaki R, Shimojo H, Sugimura Y, Fukunaga K. memantine improves depressive-like behaviors via Kir6.1 channel inhibition in olfactory bulbectomized mice. Neuroscience. 2020;442:264–73.

    Article  CAS  PubMed  Google Scholar 

  69. 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.

    Article  CAS  PubMed  Google Scholar 

  70. 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.

    Article  CAS  PubMed  Google Scholar 

  71. Fullana MN, Ferrés-Coy A, Ortega JE, Ruiz-Bronchal E, Paz V, Meana JJ, et al. Selective knockdown of TASK3 potassium channel in monoamine neurons: a new therapeutic approach for depression. Mol Neurobiol. 2019;56(4):3038–52.

    Article  CAS  PubMed  Google Scholar 

  72. O’Roak BJ, Vives L, Girirajan S, Karakoc E, Krumm N, Coe BP, et al. Sporadic autism exomes reveal a highly interconnected protein network of de novo mutations. Nature. 2012;485(7397):246–50.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Franklin TB, Silva BA, Perova Z, Marrone L, Masferrer ME, Zhan Y, et al. Prefrontal cortical control of a brainstem social behavior circuit. Nat Neurosci. 2017;20(2):260–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. 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.

    Article  CAS  PubMed  Google Scholar 

  75. Li K, Zhou T, Liao L, Yang Z, Wong C, Henn F, et al. βCaMKII in lateral habenula mediates core symptoms of depression. Science. 2013;341(6149):1016–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. O’Donovan B, Adeluyi A, Anderson EL, Cole RD, Turner JR, Ortinski PI. Altered gating of Kv14 in the nucleus accumbens suppresses motivation for reward. eLife. 2019;5(8):e47870.

    Article  Google Scholar 

  77. 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 USA. 2020;117(14):8143–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Li L, Sun H, Ding J, Niu C, Su M, Zhang L, et al. Selective targeting of M-type potassium Kv 7.4 channels demonstrates their key role in the regulation of dopaminergic neuronal excitability and depression-like behaviour. Br J Pharmacol. 2017;174(23):4277–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Cui Y, Yang Y, Ni Z, Dong Y, Cai G, Foncelle A, et al. Astroglial Kir4.1 in the lateral habenula drives neuronal bursts in depression. Nature. 2018;554(7692):323–7.

    Article  CAS  PubMed  Google Scholar 

  80. Wang A, Zhou Y, Chen H, Jin J, Mao Y, Tao S, et al. Inhibition of SK channels in VTA affects dopaminergic neurons to improve the depression-like behaviors of post-stroke depression rats. Neuropsychiatr Dis Treat. 2023;19:2127–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Stühmer W, Ruppersberg JP, Schröter KH, Sakmann B, Stocker M, Giese KP, et al. Molecular basis of functional diversity of voltage-gated potassium channels in mammalian brain. EMBO J. 1989;8(11):3235–44.

    Article  PubMed  PubMed Central  Google Scholar 

  82. D’Adamo MC, Liantonio A, Rolland JF, Pessia M, Imbrici P. Kv1.1 channelopathies: pathophysiological mechanisms and therapeutic approaches. Int J Mol Sci. 2020;21(8):2935.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Morgan PJ, Bourboulou R, Filippi C, Koenig-Gambini J, Epsztein J. Kv1.1 contributes to a rapid homeostatic plasticity of intrinsic excitability in CA1 pyramidal neurons in vivo. eLife. 2019;27(8):e49915.

    Article  Google Scholar 

  84. Lee HM, Hahn SJ, Choi BH. Open channel block of Kv1.5 currents by citalopram. Acta Pharmacol Sin. 2010;31(4):429–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Richardson A, Ciampani V, Stancu M, Bondarenko K, Newton S, Steinert JR, et al. Kv33 subunits control presynaptic action potential waveform and neurotransmitter release at a central excitatory synapse. eLife. 2022;11:e75219.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Jerng HH, Covarrubias M. K+ channel inactivation mediated by the concerted action of the cytoplasmic N- and C-terminal domains. Biophys J. 1997;72(1):163–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kise Y, Kasuya G, Okamoto HH, Yamanouchi D, Kobayashi K, Kusakizako T, et al. Structural basis of gating modulation of Kv4 channel complexes. Nature. 2021;599(7883):158–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Birnbaum SG, Varga AW, Yuan LL, Anderson AE, Sweatt JD, Schrader LA. Structure and function of Kv4-family transient potassium channels. Physiol Rev. 2004;84(3):803–33.

    Article  CAS  PubMed  Google Scholar 

  89. 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;S0896–6273(24):00152–61.

    Google Scholar 

  90. 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.

    Article  CAS  PubMed  Google Scholar 

  91. Barrese V, Stott JB, Greenwood IA. KCNQ-encoded potassium channels as therapeutic targets. Annu Rev Pharmacol Toxicol. 2018;6(58):625–48.

    Article  Google Scholar 

  92. Tzingounis AV, Heidenreich M, Kharkovets T, Spitzmaul G, Jensen HS, Nicoll RA, et al. The KCNQ5 potassium channel mediates a component of the afterhyperpolarization current in mouse hippocampus. Proc Natl Acad Sci USA. 2010;107(22):10232–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Lewis V, Rurak G, Salmaso N, Aguilar-Valles A. An integrative view on the cell-type-specific mechanisms of ketamine’s antidepressant actions. Trends Neurosci. 2024;47(3):195–208.

    Article  CAS  PubMed  Google Scholar 

  94. Costi S, Han MH, Murrough JW. The potential of KCNQ potassium channel openers as novel antidepressants. CNS Drugs. 2022;36(3):207–16.

    Article  CAS  PubMed  Google Scholar 

  95. 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.

    Article  Google Scholar 

  96. Luján R, Marron Fernandez de E, Aguado C, Wickman K. New insights into the therapeutic potential of Girk channels. Trends Neurosci. 2014;37[1]:20–9.

  97. Jeremic D, Sanchez-Rodriguez I, Jimenez-Diaz L, Navarro-Lopez JD. Therapeutic potential of targeting G protein-gated inwardly rectifying potassium (GIRK) channels in the central nervous system. Pharmacol Ther. 2021;223: 107808.

    Article  CAS  PubMed  Google Scholar 

  98. Kawaura K, Honda S, Soeda F, Shirasaki T, Takahama K. Novel antidepressant-like action of drugs possessing GIRK channel blocking action in rats. Yakugaku Zasshi. 2010;130(5):699–705.

    Article  CAS  PubMed  Google Scholar 

  99. Takahashi T, Kobayashi T, Ozaki M, Takamatsu Y, Ogai Y, Ohta M, et al. G protein-activated inwardly rectifying K+ channel inhibition and rescue of weaver mouse motor functions by antidepressants. Neurosci Res. 2006;54(2):104–11.

    Article  CAS  PubMed  Google Scholar 

  100. Chever O, Djukic B, Mcarthy KD, Amzica F. Implication of Kir4.1 channel in excess potassium clearance: an in vivo study on anesthetized glial-conditional Kir4.1 knock-out mice. J Neurosci Off J Soc Neurosci. 2010;30(47):15769–77.

    Article  CAS  Google Scholar 

  101. Nwaobi SE, Cuddapah VA, Patterson KC, Randolph AC, Olsen ML. The role of glial-specific Kir4.1 in normal and pathological states of the CNS. Acta Neuropathol. 2016;132(1):1–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Maqoud F, Scala R, Hoxha M, Zappacosta B, Tricarico D. ATP-sensitive potassium channel subunits in neuroinflammation: novel drug targets in neurodegenerative disorders. CNS Neurol Disord Drug Targets. 2022;21(2):130–49.

    Article  CAS  PubMed  Google Scholar 

  103. Li F, Jiang SY, Tian T, Li WJ, Xue Y, Du RH, et al. Kir6.1/K-ATP channel in astrocytes is an essential negative modulator of astrocytic pyroptosis in mouse model of depression. Theranostics. 2022;12(15):6611–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Lancaster B, Nicoll RA. Properties of two calcium-activated hyperpolarizations in rat hippocampal neurones. J Physiol. 1987;389:187–203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Li HJ, Zhang YJ, Zhou L, Han F, Wang MY, Xue MQ, et al. Chlorpromazine confers neuroprotection against brain ischemia by activating BKCa channel. Eur J Pharmacol. 2014;15(735):38–43.

    Article  Google Scholar 

  106. Maylie J, Bond CT, Herson PS, Lee WS, Adelman JP. Small conductance Ca2+-activated K+ channels and calmodulin. J Physiol. 2004;554(Pt 2):255–61.

    Article  CAS  PubMed  Google Scholar 

  107. Bond CT, Maylie J, Adelman JP. SK channels in excitability, pacemaking and synaptic integration. Curr Opin Neurobiol. 2005;15(3):305–11.

    Article  CAS  PubMed  Google Scholar 

  108. Honoré E. The neuronal background K2P channels: focus on TREK1. Nat Rev Neurosci. 2007;8(4):251–61.

    Article  PubMed  Google Scholar 

  109. Prieto ML, Firouzi K, Khuri-Yakub BT, Madison DV, Maduke M. Spike frequency-dependent inhibition and excitation of neural activity by high-frequency ultrasound. J Gen Physiol. 2020;152(11):e202012672.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Zandio M, Ferrín M, Cuesta MJ. Neurobiology of depression. An Sist Sanit Navar. 2002;25(Suppl 3):43–62.

    PubMed  Google Scholar 

  111. Talley EM, Solorzano G, Lei Q, Kim D, Bayliss DA. Cns distribution of members of the two-pore-domain (KCNK) potassium channel family. J Neurosci Off J Soc Neurosci. 2001;21(19):7491–505.

    Article  CAS  Google Scholar 

  112. Gotter AL, Santarelli VP, Doran SM, Tannenbaum PL, Kraus RL, Rosahl TW, et al. TASK-3 as a potential antidepressant target. Brain Res. 2011;6(1416):69–79.

    Article  Google Scholar 

  113. Borsotto M, Veyssiere J, Moha Ou Maati H, Devader C, Mazella J, Heurteaux C. Targeting two-pore domain K(+) channels TREK-1 and TASK-3 for the treatment of depression: a new therapeutic concept. Br J Pharmacol. 2015;172(3):771–84.

    Article  CAS  PubMed  Google Scholar 

  114. Meshkat S, Kwan ATH, Le GH, Wong S, Rhee TG, Ho R, et al. The role of KCNQ channel activators in management of major depressive disorder. J Affect Disord. 2024;15(359):364–72.

    Article  Google Scholar 

  115. 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.

    Article  CAS  PubMed  Google Scholar 

  116. Galeotti N, Ghelardini C, Caldari B, Bartolini A. Effect of potassium channel modulators in mouse forced swimming test. Br J Pharmacol. 1999;126(7):1653–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. 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. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41589-024-01555-y.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Esmaeili MH, Bahari B, Salari AA. ATP-sensitive potassium-channel inhibitor glibenclamide attenuates HPA axis hyperactivity, depression- and anxiety-related symptoms in a rat model of Alzheimer’s disease. Brain Res Bull. 2018;137:265–76.

    Article  CAS  PubMed  Google Scholar 

  119. Guo W, Tang ZY, Cai ZY, Zhao WE, Yang J, Wang XP, et al. Iptakalim alleviates synaptic damages via targeting mitochondrial ATP-sensitive potassium channel in depression. FASEB J Off Publ Fed Am Soc Exp Biol. 2021;35(5):e21581.

    CAS  Google Scholar 

  120. Tsai SJ. Sipatrigine could have therapeutic potential for major depression and bipolar depression through antagonism of the two-pore-domain K+ channel TREK-1. Med Hypotheses. 2008;70(3):548–50.

    Article  CAS  PubMed  Google Scholar 

  121. Yeung SY, Millar JA, Mathie A. Inhibition of neuronal KV potassium currents by the antidepressant drug, fluoxetine. Br J Pharmacol. 1999;128(7):1609–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Jeong I, Choi JS, Hahn SJ. Effects of fluoxetine on cloned Kv4.3 potassium channels. Brain Res. 2013;1500:10–8.

    Article  CAS  PubMed  Google Scholar 

  123. Sung MJ, Ahn HS, Hahn SJ, Choi BH. Open channel block of Kv3.1 currents by fluoxetine. J Pharmacol Sci. 2008;106(1):38–45.

    Article  CAS  PubMed  Google Scholar 

  124. Tytgat J, Maertens C, Daenens P. Effect of fluoxetine on a neuronal, voltage-dependent potassium channel (Kv1.1). Br J Pharmacol. 1997;122(7):1417–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Lin DH, Zhang XR, Ye DQ, Xi GJ, Hui JJ, Liu SS, et al. The role of the two-pore domain potassium channel TREK-1 in the therapeutic effects of escitalopram in a rat model of poststroke depression. CNS Neurosci Ther. 2015;21(6):504–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Božić M, Pirnat S, Fink K, Potokar M, Kreft M, Zorec R, et al. ketamine reduces the surface density of the astroglial Kir4.1 channel and inhibits voltage-activated currents in a manner similar to the action of Ba2+ on K+ Currents. Cells. 2023;12(10):1360.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Stenovec M, Božić M, Pirnat S, Zorec R. Astroglial mechanisms of ketamine action include reduced mobility of Kir4.1-carrying vesicles. Neurochem Res. 2020;45(1):109–21.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported, in part, The National Science Foundation of China (82374586, 82274128), Shandong Province Chinese Medicine High-level Talents Cultivation Program Special Funds, China Postdoctoral Science Foundation (2021M702045, 2024M751906), Natural Science Foundation of Shandong Province(ZR2022QH083), the Postdoctoral Fellowship Program of CPSF (GZC20240959), Shanghai Municipal Health Commission Traditional Chinese Medicine Research Project  (2022CX004).

Author information

Authors and Affiliations

Authors

Contributions

YC, JZ MZ conceived and designed project. JZ, JY, YuanZ prepared the figures. JZ, YaoZ, ZL, ZS prepared the reference. YC, JZ, YaoZ wrote the manuscript. JY, MZ, LY, YuanZ helped revise the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Yongjun Chen.

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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, J., Zhu, Y., Zhang, M. et al. Potassium channels in depression: emerging roles and potential targets. Cell Biosci 14, 136 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13578-024-01319-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13578-024-01319-0

Keywords