Skip to main content

Estrogen-immuno-neuromodulation disorders in menopausal depression

Abstract

A significant decrease in estrogen levels puts menopausal women at high risk for major depression, which remains difficult to cure despite its relatively clear etiology. With the discovery of abnormally elevated inflammation in menopausal depressed women, immune imbalance has become a novel focus in the study of menopausal depression. In this paper, we examined the characteristics and possible mechanisms of immune imbalance caused by decreased estrogen levels during menopause and found that estrogen deficiency disrupted immune homeostasis, especially the levels of inflammatory cytokines through the ERα/ERβ/GPER-associated NLRP3/NF-κB signaling pathways. We also analyzed the destruction of the blood-brain barrier, dysfunction of neurotransmitters, blockade of BDNF synthesis, and attenuation of neuroplasticity caused by inflammatory cytokine activity, and investigated estrogen-immuno-neuromodulation disorders in menopausal depression. Current research suggests that drugs targeting inflammatory cytokines and NLRP3/NF-κB signaling molecules are promising for restoring homeostasis of the estrogen-immuno-neuromodulation system and may play a positive role in the intervention and treatment of menopausal depression.

Introduction

Depression is a major cause of suicide and mortality worldwide [1, 2]. The vast majority of patients with depression show insufficient secretion of monoamine neurotransmitters, making selective serotonin reuptake inhibitors (SSRIs) the main antidepressant drugs. SSRIs maintain a high level of 5-hydroxytryptamine (5-HT) in the synaptic cleft, which can effectively alleviate depressive episodes in the short term [2, 3] but cannot repair the autonomous regulation of neurotransmitter synthesis, secretion, release and absorption by neurons [4, 5]. Long-term use of SSRIs induces neuronal dysfunction and gradually leads to drug dependence in depressed patients [5,6,7]; therefore, tracking the cause of neuronal dysfunction and developing corresponding treatments may be the best way to cure depression. With the deterioration of ovarian function, the secretion of estrogen in menopausal women decreases sharply, and the serious imbalance of endocrine hormone levels results in a high incidence of major depression in menopausal women [8,9,10,11]. The relatively clear etiology of menopausal depression provides promising prospects for the investigation of neuronal dysfunction.

Some early breakthroughs in menopausal depression focused on the link between estrogen and depression [12,13,14]. Initially, an epidemiological survey of 16,080 women aged 35–60 years led to a preliminary understanding of menopausal depression and revealed that menopausal women had significantly more severe depressive symptoms, suggesting that changes in estrogen homeostasis are related to the development of depression [12]. Further analysis confirmed that estrogen can exert antidepressant effects by regulating the level of 5-HT and the expression of 5-HT receptors (5-HTR) on presynaptic or postsynaptic membranes [13]. Subsequently, Chhibber et al. reported that decreased estrogen receptor (ER) expression reduces brain-derived neurotrophic factor (BDNF) levels in the hippocampus, thereby inhibiting the tropomyosin receptor kinase B (TrkB) signaling pathway, leading to 5-HT2A dysfunction, attenuated synaptic plasticity, and increased susceptibility to depression in menopausal women [14]. To date, abnormalities in signaling pathways caused by estrogen deficiency have attracted much attention.

Recently, significantly abnormal proportions of T-cell subsets, activation of microglia, and elevated levels of inflammatory cytokines in the central nervous system and peripheral blood were detected during the onset of menopausal depression [15,16,17], and these immune disorders increase the activity of the estrogen-immuno-neuromodulation system, which may be the key to resolving neuronal dysfunction in menopausal depression. However, due to the involvement of multiple ERs [18,19,20,21], immune responses and regulatory factors [19,20,21,22,23,24,25,26,27] and nerve signaling molecules [26,27,28,29,30], the disturbance of signaling pathways in this complex system remains unclear and has not been comprehensively reviewed in menopausal depression.

To this end, focusing on the decline of estrogen levels in menopausal women, we summarize the immune system imbalance and neurological impairments caused by estrogen deficiency and analyze the detailed process and possible mechanism of estrogen-immuno-neuromodulation disorders in menopausal depression, with the aim of providing scientific directions for further elucidating the pathogenesis of menopausal depression and developing novel targeted therapeutic drugs.

Immune imbalance in menopause

Characteristic changes in the immune system during menopause

Characteristic changes in the immune system in menopausal women are the first clue to understanding the disorder of the estrogen-immuno-neuromodulation system in menopausal depression in order to trace the relevant signaling pathways from estrogen to specific indicators of immune imbalance.

An analysis of peripheral blood lymphocyte subsets in menopausal women and reproductive women revealed that the total number of lymphocytes in the menopausal women was lower; specifically, the number of B lymphocytes and CD4+ T cells were significantly lower, and the ratio of CD4+ to CD8+ T cells was also significantly lower [31]. Further diagnostic findings of serological biochemical factors revealed that the serum level of interleukin (IL)-4 (a Th2 cytokine) was increased and the level of interferon (IFN)-γ (a Th1 cytokine) was decreased in menopausal women, suggesting that the cellular immune activity of the body tended to be attenuated after the decrease in estrogen levels [32].

Another important characteristic change in the immune system of menopausal women is an increased susceptibility to inflammation. Malutan et al. compared the levels of inflammatory factors in women who were fertile, perimenopausal, postmenopausal, ovariectomized, or chronically inflammatory and observed that the levels of the inflammatory factors IL-1β, IL-8 and tumor necrosis factor (TNF)-α were significantly greater in menopausal women, while the levels of the anti-inflammatory factor IL-20 were lower [33]. Patients with perimenopausal depression have increased levels of inflammation [34]. Animal models of perimenopausal depression exhibit activation of microglia and astrocytes, as well as increased neuroinflammation and nerve damage [25, 35, 36].

Effect of estrogen on immune imbalance during menopause

Changes in the immune system in menopausal women indicate that a decrease in estrogen levels had an important impact on immune homeostasis, leading to the development of estrogen replacement therapy (ERT). Estrogens are cholesterol-derived steroid hormones with wide ranges of biological activities that mainly include estrone (E1), estradiol (17β-estradiol, E2) and estriol (E3). Among them, E2 is the main form of estrogen in the human body, and its physiological activity is considered to be the strongest [37]. Kumru et al. demonstrated that E2 could reduce the number of CD8+ T cells to a certain extent, restore the ratio of CD4+ to CD8+ T cells in peripheral blood to a normal level, and significantly increase the proportion of CD19+ B cells and the level of IFN-γ [38]. E2 also had a restorative effect on the proportion of different lymphocyte subsets in menopausal women, but the number of CD4+ T cells and CD20+ B cells was still lower than that in reproductive women, which indicates that estrogen plays an important role in regulating the immune status of menopausal women but cannot reverse the decline in immune function caused by aging [39]. Estrogen has also been found to have a positive regulatory effect on the preservation of naïve B cells, which is conducive to the occurrence of a humoral immune response. In addition, estrogen can decrease the level of the proinflammatory cytokine IL-6 in peripheral blood to a certain extent, inhibit the secretion of inflammatory cytokines by CD4+ T cells, and reduce the inflammatory response in menopausal women [39]. All of this evidence suggests that estrogen can repair immune imbalances and maintain immune homeostasis during menopause.

Possible mechanisms of immune imbalance caused by estrogen deficiency

The mechanism of immune imbalance caused by estrogen deficiency has not yet been well investigated in menopausal women [32, 40], but the understanding of this process is gradually being explored in menopausal animal models and related cell lines. The effect of estrogen is known to be exerted mainly through its receptors. ERs consists of classical nuclear receptors (ERα, ERβ and their subtypes), G protein-coupled estrogen receptor (GPER), ER-X and Gaq-ER, which are specifically expressed in the cell membrane, cytoplasm and nucleus of monocytes, macrophages, dendritic cells, neutrophils, NK cells, CD4+ and CD8+ T cells, regulatory T (Treg) cells, B cells, microglia, astrocytes, and neurons, among others [18, 20, 22, 41, 42]. When activated by estrogen, different ERs can regulate gene expression directly or in conjunction with transcription factors or can also regulate cell signal transduction pathways through second messengers such as cAMP [18, 20, 22, 41]. Recent studies on the mechanism of immune imbalance caused by estrogen deficiency during menopause have focused mainly on the regulation of inflammatory signaling pathways by ERα, ERβ and GPER. Understanding these pathways is crucial for elucidating the mechanisms of immune imbalance caused by estrogen deficiency and their potential contribution to menopausal depression.

NLRP3 signaling pathways mediated by the classical ER

Nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), the triggering component of inflammasome formation, is involved in the occurrence and treatment of menopausal depression. Xu et al. reported that estrogen deficiency increased the expression of purinergic ligand-gated ion channel 7 receptor (P2X7R), toll-like receptor (TLR) 2 and TLR4 in the hippocampus of ovariectomized mice, promoted the inflammatory cascade reaction and the formation of the NLRP3 inflammasome, stimulated nuclear factor-κB (NF-κB), increased the expression of pro-IL-1β and pro-IL-18, and finally induced depression and anxiety-like behavior in mice. Administration of the inflammasome inhibitors VX-765, E2 and ERβ agonists to ovariectomized mice blocked these signaling pathways by inhibiting P2X7R, TLR2 and TLR4 expression, thereby reversing depression and anxiety-like behaviors caused by estrogen deficiency (Fig. 1) [23]. Resveratrol (RSV), a potential replacement for E2, can inhibit the activation of NLRP3 and NF-κB in the hippocampus by increasing the levels of silent information regulator factor 2-related enzyme 1 (sirtuin 1, SIRT1), thereby restraining the increase in caspase-1 and NLRP3 inflammasome effectors caused by the activation of NLRP3, the conversion of pro-IL-1β to mature IL-1β and the strong release of IL-1β and IL-18, effectively combating depression and anxiety-like behaviors caused by estrogen deficiency (Fig. 1) [43]. Menze et al. analyzed the mechanism of simvastatin (SIM) in neuroprotection and depression-like behavior resistance and revealed that SIM can also inhibit the expression of P2X7R, TLR2 and TLR4 in the hippocampus of ovariectomized rats and block the activation of the NLRP3 inflammasome, thus decreasing the levels of the proinflammatory cytokines IL-1β and IL-18 and reducing the expression of ionized calcium-binding adapter molecule 1 (IBA1) and the activation of microglia. SIM also significantly increased the expression of ERα and ERβ in the hippocampus and the expression of ERβ in the uterus of ovariectomized mice but did not increase uterine weight, suggesting that SIM may be a safer alternative to hormone replacement therapy for the management of postmenopausal depression (Fig. 1) [44]. These studies suggest that the NLRP3 inflammasome may become a potential therapeutic target for estrogen deficiency-related affective disorders such as depression.

Fig. 1
figure 1

NLRP3 signaling pathways regulated by E2. Estrogen deficiency in ovariectomized mice or rats increases the expression of P2X7R, TLR2 and TLR4 in the hippocampus and further leads to the activation of NLRP3, which promotes the inflammatory cascade and increases the expression of pro-IL-1β and pro-IL-18. Moreover, NLRP3 can lead to caspase-1 activation, which in turn promotes the transformation of pro-IL-1β and pro-IL-18 into mature IL-1β and IL-18, and then, IL-1β facilitates the increase in IBA1 expression and the activation of microglia. Administration of the inflammasome inhibitors VX-765, E2 and ERβ agonists to ovariectomized mice can inhibit the expression of P2X7R, TLR2 and TLR4, block these signaling pathways, and reverse the depression and anxiety-like behavior caused by estrogen deficiency. RSV inhibits depression-like behavior by increasing the level of SIRT1 and inhibiting the activation of the NLRP3 inflammasome. SIM can also inhibit the expression of P2X7R, TLR2 and TLR4 and the activation of the NLRP3 inflammasome and its downstream signaling pathways in the hippocampus of ovariectomized rats, as well as reduce the activation of microglia induced by IL-1β, and alleviate depression in ovariectomized rats. (➝, positive regulation; , negative regulation. The meaning of these two indicators is the same across all the figures in this article.)

NF-κB signaling pathways mediated by the classical ER

NF-κB, an important nuclear transcription factor, can be regulated by estrogen through multiple signaling pathways. Ovariectomy experiments in animals have demonstrated that the decreased levels of E2 activate NF-κB signaling pathway in microglia, converting microglia from the M2 subtype to the M1 subtype and resulting in the production and secretion of large amounts of inflammatory cytokines such as TNF-α, IL-1β and IL-6, which leads to cognitive impairment and depressive behavior [45, 46]. Another comparative study showed that E2 supplementation improved depression-like behavior in aging female mice by reducing NF-κB activation and the expression of the inflammatory cytokines TNF-α, IL-1β, and IL-6. Both ERα and SIRT1 antagonists can reverse the effects of E2, suggesting that E2 may protect aging female mice from depression through the E2/ERα/SIRT1/NF-κB signaling pathway [19] (Fig. 2). By implanting E2 release pellets into ovariectomized middle-aged female rats, Pratap et al. reported that E2 could decrease the level of p-NF-κB and increase the expression of the molecular markers phosphorylated extracellular signal regulated protein kinase (p-ERK), phosphorylated cAMP-responsive element binding protein (p-CREB), phosphorylated protein kinase B (p-PKB or AKT), and nitric oxide in the frontal cortex, striatum, and other brain areas [24] (Fig. 2). In addition, E2 can inhibit the activation of microglia and astrocytes and reduce the expression of proinflammatory cytokines such as TNF-α, IL-1β and IL-6 through TLR4/NF-κB signaling pathway [22], and our previous studies indicated that the occurrence and exacerbation of depression are closely related to the NF-κB-related signaling pathway in microglia [27] (Fig. 2). E2 can also activate marrow stromal kinesin family member 18 A (MS-KIF18A) in the cytoplasm after binding to ERα, and MS-KIF18A further activates the rapid phosphorylation of ERK, thereby inhibiting the activation of NF-κB [41] (Fig. 2).

The above results [19, 22, 24, 41, 45,46,47] support the idea that estrogen inhibits the activation of the NF-κB signaling pathway and the secretion of inflammatory cytokines in reproductive women, maintaining immune homeostasis in the body (especially in the central nervous system), while during menopause, a decrease in estrogen levels triggers the activation of NF-κB signaling pathways and the expression of inflammatory cytokines, resulting in an immune imbalance in the body, which further promotes neuronal damage and menopausal depression.

Fig. 2
figure 2

NF-κB signaling pathways mediated by classical ER. In general, upon binding to ERα in the membrane, cytoplasm or nucleus, E2 can inhibit the activation of NF-κB by regulating TLR4, MS-KIF18A, ERK, AKT, CREB, SIRT1 and other signal transduction molecules, thereby decreases the expression of the proinflammatory cytokines TNF-α, IL-1β and IL-6. However, supraphysiologic doses of E2 (sE2) abnormally activate NF-κB in microglia, resulting in increased expression of IBA1, CD86 and the proinflammatory cytokines TNF-α, IL-1β, and IL-6 and decreased expression of the anti-inflammatory cytokines TGF-β, IL-10, and IL-4. Moreover, E2 can promote the proliferation, migration and invasion of ER+ breast cancer cells by activating the PI3K/AKT/NF-κB/TWIST1/CCL2 signaling pathway

Unexpectedly, Li et al. reported that supraphysiologic doses of E2 can induce neuronal damage and aggravate the depressive behavior of ovariectomized mice by abnormally activating the expression of IBA1 and CD86 in microglia and disrupting brain glycerophospholipid metabolism through ERα/NF-κB, increasing the expression of the proinflammatory cytokines TNF-α, IL-1β, and IL-6 and decreasing the expression of the anti-inflammatory cytokines TGF-β, IL-10, and IL-4 [48] (Fig. 2). In addition, studies on the relationship between E2 and breast cancer showed that E2 can upregulate the transcription factor TWIST1 (a member of the basic helix-loop-helix transcription factor family) and NF-κB by activating the ERα/PI3K (phosphatidylinositol 3-hydroxy kinase)/AKT/NF-κB signal transduction pathway, thus positively regulating C-C motif chemokine ligand 2 (CCL2) synthesis and autocrine signaling to promote the proliferation, migration and invasion of ER+ breast cancer cells [49] (Fig. 2). These results suggest that the E2/ERα/NF-κB signaling pathway regulates cellular or body inflammation levels with diverse outcomes [19, 22, 24, 41, 47,48,49]. Whether the reason for this phenomenon is related to the dose of E2 or the different ways in which E2 regulates the central nervous system and peripheral immune system requires further precise investigation.

Inflammatory signaling pathways mediated by GPER

G protein-coupled receptor (GPR) 30 is a new type of ER, also known as GPER, which is widely distributed in the central nervous system (hippocampus, cortex, hypothalamus, striatum and brain stem) and is associated with emotional regulation [50,51,52,53,54]. Estrogen can not only regulates the NLRP3 and NF-κB signaling pathways through ERα and ERβ but also regulates inflammation levels, microglial polarization and astrocyte activity in the central nervous system through GPER-related signaling pathways to maintain immune homeostasis in the brain and rescue brain injury, hypomnesia and cognitive impairment [20, 21, 52, 54,55,56] (Fig. 3).

Zhao et al. demonstrated that E2 can inhibit the expression of inflammatory cytokines in microglia through GPER, and the GPER agonist G1 can promote the anti-inflammatory effect of E2, but GPER antagonist G15 or GPER knockdown can eliminate the anti-inflammatory effect of E2 [21]. In addition to inhibiting the production of TNF-α, IL-1β and IL-6 by microglia, the GPER agonist G1 can also reduce the expression of CD86, CD11b, macrophage cationic peptide 1 (MCP-1, also known as CCL2), IBA1, nitric oxide synthase 2 (NOS2), macrophage inflammatory protein 2 (MIP-2, also known as CXCL2), and NF-κB and increase the expression of the anti-inflammatory factors IL-4, IL-10, arginase 1 (Arg1) and CD206 in microglia, polarize microglia to the M2 phenotype, and rescue the damage of inflammatory factors to neurons [52, 55, 56]. The mechanism of the GPER agonist G1 is closely related to the phosphorylation of AKT and the inhibition of TLR4 [24, 52, 55, 57]. GPER activation can also reduce microglial activation, NLRP3 inflammasome formation, caspase-1 activation mediated by the NLRP3 inflammasome and downstream IL-1β production and signal transduction, as well as downstream NF-κB signaling, and strongly upregulate the expression of the anti-inflammatory factor IL-1RA (an endogenous IL-1 receptor antagonist) mediated by p-CREB (a transcription factor known to enhance the expression of IL-1RA) in hippocampal CA1 neurons. The GPER agonist G1 enhances this anti-inflammatory effect of GPER, while the GPER antagonist G36 reverses this effect [54]. The loss of GPER in astrocytes can lead to the transformation of astrocytes to the A1 phenotype, increase the expression of TNF-α, IL-6 and IL-1β, increase the accumulation of Serpina3n in the cells, damage the function of neurons, and disrupt learning and memory in female mice. Serpina3n is a molecular marker of neuroinflammation in astrocytes. Praja-1 (PJA1) mediated the effects of astrocytic GPER on learning and memory by binding to Serpina3n. GPER can positively regulate PJA1 expression by stimulating the G protein (Gs)/cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)/CREB signaling pathway in cultured rat and human astrocytes [20]. In exosomes isolated from the plasma of postmenopausal women, the levels of GPER and PJA1 mRNA were decreased, while the level of Serpina3n in plasma was increased [20]. These results indicate that the astrocyte GPER can regulate learning and memory through the GPER/PJA1/Serpina3n signaling pathway in female mice. Although E2 beneficially regulates inflammation in the central nervous system and exerts neuronal protection by binding to its membrane receptor GPER [20, 21, 52, 54,55,56], too high of a level of E2 can also lead to the overexpression of GPER, sarcoma protein (Src) and matrix metalloproteinase 9 (MMP-9) in neurons and subsequently activate IL-1β to result in harmful effects on the nervous system. The GPER antagonist G15 can reverse the damage caused by E2-induced GPER overactivation [58] (Fig. 3). Considering the positive regulatory effects of GPER on neuroinflammation, microglial and astrocyte activation, neuronal damage, and cognitive and memory impairment, GPER may be a potential therapeutic target for menopausal depression.

Fig. 3
figure 3

Inflammatory signaling pathways mediated by GPER. E2 can inhibit the expression of TNF-α and IL-1β through GPER, and the GPER agonist G1 can promote the anti-inflammatory effect of E2. G1 can also reduce the expression of IL-6, CD86, CD11b, MCP-1, IBA1, NOS2, and MIP-2, as well as the activation of NF-κB, and increase the expression of the anti-inflammatory factors IL-4, IL-10, Arg1 and CD206. The mechanism of the GPER agonist G1 is closely related to the phosphorylation of AKT and the inhibition of TLR4. GPER activation can also reduce NLRP3 inflammasome formation, caspase-1 activation mediated by the NLRP3 inflammasome and downstream pro-IL-1β transformation into IL-1β, as well as downstream NF-κB signaling, and upregulate the expression of IL-1RA mediated by p-CREB (IL-1RA enhances the defense of neurons against IL-1β). The GPER agonist G1 enhances this anti-inflammatory effect of GPER, while the GPER antagonist G36 reverses this effect. Additionally, GPER can decrease the expression of TNF-α, IL-6 and IL-1β, positively regulate PJA1 expression, and decrease the accumulation of Serpina3n through the Gs/cAMP/PKA/CREB signaling pathway. A too high level of E2 can also lead to the overexpression of GPER, Src and MMP-9 in neurons and subsequently activate IL-1β to result in harmful effects on the nervous system. The GPER antagonist G15 can reverse the damage caused by E2-induced GPER overactivation

As a cell membrane receptor, GPER is more responsive to exogenous estrogen agonist therapeutic drugs, making its therapeutic mechanism is relatively easier to address and elucidate [59]. Additionally, GPER expression is notably significant in brain tissues such as the hippocampus [60, 61]. These factors suggest that GPER-based antidepressants may be more acceptable to clinical patients. However, this does not imply that ERα and ERβ are poor targets. Although the majority of classical estrogen receptors (ERs) are expressed in the nucleus, a small number of these classical ER subtypes are expressed on the cell membrane. Moreover, the activation of classical ERs can regulate gene expression more briefly and rapidly. Therefore, further analysis of the differences in the expression of various ERs in key brain regions associated with depression and the affinity preferences of estrogen alternative therapeutic drugs for these ERs will help determine the most appropriate ER target for developing antidepressant therapies. In any case, therapeutic drugs targeting key molecules such as inflammatory cytokines within the estrogen-immune modulatory signaling pathway should play an effective role in treating menopausal depression.

Neurological impairments caused by immune imbalance

As discussed above, due to decreased estrogen levels, menopausal depression is characterized by an immune imbalance that includes abnormal activity of immune factors such as TNF-α, IL-1β, IL-6, IL-10, IL-17, IL-33, and IFN-γ. In this section, we will further focus on neurological impairments caused by immune imbalance and the detailed pathogenesis of these impairments, providing a comprehensive overview of this complex phenomenon based on the findings of menopausal depression and its related fields.

BBB destruction

The blood-brain barrier (BBB) refers to the isolating material between brain cells or cerebrospinal fluid (CSF) and plasma and is mainly composed of capillary endothelial cells with tight junctions, the endothelial basement membrane and the astrocyte foot plate. Under normal conditions, the BBB controls the selective permeability between the components of the plasma and brain tissue and plays a protective role in brain tissue. Under aging or pathological conditions, the integrity of the BBB structure changes, and peripheral substances enter brain tissue, leading to dysfunction of brain cells or CSF.

Studies on changes in BBB structure after a decrease in estrogen levels have shown that the expression of the tight junction protein claudin-5 decreases during menopause and that the permeability of paracellular junctions to sucrose increases, thereby increasing the probability of brain edema and stroke [62, 63]. In the brain tissue of menopausal women or animal models, a decrease in estrogen levels caused an increase in the number of M1 microglia and activated astrocytes, which can secrete a large number of inflammatory cytokines. Therefore, inflammatory cytokines may play an important role in changes in BBB permeability [25, 64,65,66,67,68,69,70,71,72,73,74]. Several subsequent studies have demonstrated that TNF-α, IL-6 and IL-17 can reduce the expression of claudin-5, zona occludens-1 (ZO-1) and other tight junction proteins by activating NF-κB signaling pathways in endothelial cells, destroying the structure of the BBB and promoting depressive behavior [69, 70]. A large amount of TNF-α can also induce necrotic apoptosis of endothelial cells, directly leading to an increase in BBB permeability [71]. Stress-induced depression in humans is accompanied by a decrease in claudin-5 expression and an increase in IL-6 expression. Stress triggers BBB disruption, allowing IL-6 to penetrate the brain and increases the risk of depression in humans [68]. IL-1β can degrade the tight junction proteins occludin and ZO-1 by enhancing the expression of MMP-9 in endothelial cells, leading to the destruction of BBB integrity [72]. Moreover, IL-1β can inhibit the production of hedgehog proteins by astrocytes, thereby eliminating the protective effect of astrocytes on BBB integrity by downregulating the expression of tight junction proteins. IL-1β can also increase the levels of the proinflammatory chemokines CCL2, CCL20 and CXC-chemokine ligand 2 (CXCL2) in astrocytes, which further exacerbates BBB destruction and neuroinflammation after immune cell recruitment [73]. In addition, during persistent inflammation, active microglia engulf astrocyte terminals, which directly results in a loss of BBB integrity [74] (Fig. 4). Our previous studies have shown that the expression of C-reactive protein (CRP) is increased in patients with poststroke depression [75]. Cossette et al. reported that CRP can induce endothelial cell activation and that activated endothelial cells can produce more inflammatory factors, such as CRP, IL-6, IL-8 and vascular cell adhesion molecule-1 (VCAM-1), which further aggravate vascular endothelial cell injury. E2 pretreatment can reduce the production of the above inflammatory factors and promote vascular repair [76].

Based on the above findings, we hypothesize that some peripheral inflammatory cytokines first attack vascular epithelial cells and decrease the expression of tight junction proteins, leading to an increase in BBB permeability. Then, these inflammatory cytokines penetrate the BBB to activate microglia, which in turn secrete more inflammatory cytokines and chemokines and, by inducing necrotic apoptosis of endothelial cells and triggering phagocytosis of the astrocyte footplates, severely exacerbate the destruction of the BBB. Ultimately, this process leads to neuronal damage and menopausal depression.

Fig. 4
figure 4

BBB destruction was triggered by immune imbalance during menopause. During menopause, a decrease in estrogen levels causes an increase in inflammatory cytokines (TNF-α/IL-6/IL-17) in the peripheral blood, which gradually decreases expression of claudin-5, ZO-1 and occludin by activating the NF-κB signaling pathway and downregulating MMP-9 expression in endothelial cells, thereby increasing the permeability of the BBB. Subsequently, inflammatory cytokines penetrate the BBB to activate microglia, which in turn secrete more inflammatory cytokines and chemokines (CCL2/CCL20/CXCL2) and recruit more aggressive immune cells, further inducing necrotic apoptosis of endothelial cells, inhibiting hedgehog protein secretion (which can improve the expression of tight junction proteins in endothelial cells) in astrocytes and triggering the phagocytosis of astrocyte footplates, thereby severely exacerbating BBB destruction. CRP can induce endothelial cell activation, and activated endothelial cells can produce more CRP, IL-6, IL-8, and VCAM-1, which further aggravate vascular endothelial cell injury. E2 pretreatment can reduce the production of the above inflammatory factors and promote vascular repair

Neurotransmitter dysfunction

Neurotransmitters in the central nervous system of patients with major depression, including 5-HT [77, 78], dopamine (DA) [78], norepinephrine (NE) [78], γ-aminobutyric acid (GABA) [79] and glutamate (Glu) [79] have received increasing amounts of attention. We next outline the effects of inflammatory cytokines on neurotransmitter dysfunction and the associated pathogenesis in order to enhance the understanding of neurological impairments caused by immune imbalance.

5-HT

5-HT, also called serotonin, is an indoleamine neurotransmitter in brain tissue that can regulate pleasant emotions. TNF-α, IL-1β and IL-6 promote depression and inhibit the functional activity of serotonergic neurons.

On the one hand, TNF-α, IL-1β and IL-6 can activate indoleamine 2,3-dioxygenase (IDO), tryptophan 2,3-dioxygenase (TDO) and monoamine oxidase (MAO), which degrade tryptophan (Trp). IDO and TDO catalyze the oxidation of Trp to initiate the metabolic cascade, leading to Trp entering the kynurenine (Kyn) metabolic pathway and no longer synthesizing 5-HT [80,81,82]. MAO catalyzes the degradation of 5-HT to 5-hydroxyindoleacetic acid (5-HIAA). 5-HIAA levels are significantly associated with depression and anxiety scores in patients with somatic symptom disorder, and elevated IL-6 levels exhibit mediating effects [83].

On the other hand, TNF-α, IL-1β and IL-6 can increase the expression of 5-HT transporters (5-HTT or SERT) in the presynaptic membrane of neurons and promote 5-HT transport from the synaptic cleft to the inner cell body of neurons, leading to a reduction in 5-HT in the synaptic cleft and subsequent weakening of the excitatory potential at the receptor [82, 84]. In addition, inflammatory cytokines also inhibit the expression of 5-HTR, thereby blocking or attenuating the functional activity of serotonergic neurons [85] (Fig. 5).

Fig. 5
figure 5

Effects of inflammatory cytokines on 5-HT synthesis, degradation, reuptake and 5-HTR expression. TNF-α/IL-1β/IL-6 activate IDO and TDO to catalyze Trp to enter the Kyn metabolic pathway, reduce 5-HT synthesis, stimulate MAO to promote 5-HT degradation into 5-HIAA, increase the expression of 5-HTT in the presynaptic membrane, aggravate 5-HT reuptake in the synaptic cleft, inhibit 5-HTR expression in the postsynaptic membrane and attenuate 5-HT efficacy

DA

DA is a catecholamine neurotransmitter in the brain that induces pleasure. Studies have shown that increased levels of TNF-α and IL-1β in CSF are positively correlated with decreased levels of DA and the severity of dopaminergic neuron damage [86, 87]. The influence of inflammatory cytokines on DA involves synthesis, packaging, metabolism, release and reuptake.

DA synthesis depends on tyrosine hydroxylase (TH), the rate-limiting enzyme that converts tyrosine (Tyr) to levodopa (L-DOPA). Tyr is derived from phenylalanine (Phe) via phenylalanine hydroxylase (PAH), and TH and PAH require the common cofactor tetrahydrobiopterin (BH4). However, high levels of TNF-α and IL-1β can cause an increase in intracellular reactive oxide species (ROS), which promotes the oxidation of BH4 to dihydroxanthopterin (XPH2) and dihydrobiopterin (BH2), thereby blocking TH and PAH activity and inhibiting DA synthesis [88] (Fig. 6). E2 treatment can not only decrease the expression of TNF-α, IL-1β and IL-6 but also improve the production of BH4 [89].

TNF-α and IL-1β are also thought to reduce the expression of vesicle monoamine transporter (VMAT), which are not conducive to DA packaging and transport [90, 91] (Fig. 6). Unpackaged DA can be degraded to 3,4-dihydroxyphenzene acetaldehyde (DOPAL) by MAO on the mitochondrial membrane; the latter is further oxidized by aldehyde dehydrogenase (ALDH) to 3,4-dihydroxyphenylacetic acid (DOPAC), resulting in the massive metabolism of the synthesized DA, which cannot be released from the presynaptic membrane [92].

In addition, TNF-α can increase the degradation of DA by extracellular MAO and produce a large amount of the metabolite homovanillic acid (HVA), which is not conducive to the activation of receptors on the postsynaptic membrane by high concentrations of DA [93] (Fig. 6).

Other studies have shown that TNF-α and IL-1β can activate 5-HTT and DA transporter (DAT) through the receptor-coupled mitogen-activated protein kinase (MAPK) signaling pathway, thereby increasing the reuptake of 5-HT and DA by presynaptic membranes, reducing the transmitter effect, and triggering depressive behaviors [94,95,96,97] (Fig. 6).

Fig. 6
figure 6

Effects of inflammatory cytokines on DA synthesis, packaging, transport, degradation and reuptake. TNF-α and IL-1β can increase ROS levels and subsequently promote the oxidation of BH4 to XPH2 and BH2, which further leads to the inactivation of TH and PAH and a reduction in DA synthesis. TNF-α and IL-1β are also thought to interfere with DA packaging and transport by reducing VMAT expression, and unpackaged DA is easily degraded to DOPAL and HVA by MAO. Furthermore, DOPAL is oxidized by ALDH to DOPAC, resulting in the massive metabolism of the synthesized DA. In addition, TNF-α and IL-1β can activate DAT through the MAPK signaling pathway, increasing the reuptake of DA

NE

NE is a catecholamine neurotransmitter synthesized and secreted by sympathetic postganglionic neurons and norepinephrine neurons in the brain and is involved in the dual regulation of the hypothalamic pituitary adrenal (HPA) axis and hypothalamic pituitary gonadal (HPG) axis. NE is closely related to anxiety and sex hormone secretion. Studies on the emotional centers and areas of the brain that regulate sex hormone production have shown that IL-1β can increase NE levels in the central amygdala (CeA), bed nucleus of the stria terminalis (BNST) and paraventricular nucleus (PVN) of the HPA axis but decrease NE levels in the diagonal band of Broca (DBB), organum vasculosum lamina terminalis (OVLT), medial preoptic area (MPA), suprachiasmatic nucleus (SCN) and arcuate nucleus (Arc) of the HPG axis [98] (Fig. 7). IL-1β differentially regulates NE in different brain regions to promote the body’s adaptation to systemic immune challenges.

The mechanism by which IL-1β affects NE has been analyzed mainly through its synthesis and release. L-DOPA is a precursor substance for NE synthesis, and the supply of L-DOPA can reverse the effect of IL-1β on NE in several brain regions of rats. These results suggest that TH, the rate-limiting enzyme of L-DOPA synthesis, may be the key target of IL-1β in regulating NE synthesis [99, 100]. Similar findings have been observed for IL-6. IL-6 inhibits TH expression by activating Janus kinase-signal transducer and activator of transcription (JAK-STAT) 3 and reduces NE synthesis [101]. The anti-inflammatory cytokine IL-4 can upregulate the expression of TH and tryptophan hydroxylase (TPH), thereby reversing the negative regulatory effect of inflammatory cytokines on neurotransmitters such as NE and 5-HT and alleviating depressive behavior in rats [102]. The release of NE is regulated by a variety of transmitters. The decrease in NE in the MPA region caused by IL-1β is mainly the result of its activation of GABAergic neurons, which further exerts inhibitory effects on adrenergic neurons [103] (Fig. 7).

The effects of proinflammatory cytokines on the function of NE also include the regulation of its receptors. The binding of NE to β2-adrenoceptors on the surface of astrocytes induces the production of a large number of nerve growth factors and promotes the growth of neuronal axons, while IFN-γ can activate the MAPK signaling pathway, inhibit the expression of β2-adrenoceptors in astrocytes and attenuate the regeneration and repair of axonal branches induced by NE [104, 105].

Fig. 7
figure 7

Effects of immune cytokines on NE synthesis and release. IL-1β can reduce NE synthesis by inhibiting TH activity and regulating the HPG axis, increase NE synthesis by regulating the HPA axis, and reduce NE release by activating GABAergic neurons. IL-6 can also reduce NE synthesis by inhibiting TH activity via the JAK-STAT3 signaling pathway, but the anti-inflammatory cytokine IL-4 can activate TH activity and increase NE synthesis

GABA

GABA is an inhibitory neurotransmitter widely distributed in the brain and spinal cord, and it mainly exists in interneurons that send out negative feedback signals, thereby inhibiting 5-HT, DA and other neurotransmitters, and regulating hormone secretion, muscle contractions, blood pressure, heart rate, feeding, sleep, mood and other physiological activities. Normally, the inhibitory effect of GABAergic neurons helps coordinate the body’s homeostasis, but abnormal GABA activity can also lead to a variety of physiological dysfunctions [106].

GABA is primarily synthesized through the decarboxylation of glutamate, a process catalyzed by glutamic acid decarboxylase (GAD), which is the main rate-limiting enzyme for GABA synthesis. After synthesized GABA is released into the synaptic cleft, it first interacts with receptors on the postsynaptic membrane before being removed from the synaptic cleft by diffusion effect and actively taken up by glial cells. The ingested GABA is transformed into Glu through transamination, and the latter is further transformed into glutamine (Gln, an amide of glutamate) under the catalysis of glutamine synthetase (GS). As Gln regurgitates to neurons, it is transformed again into Glu by glutaminase (Glut), which is subsequently transformed into new GABA under the action of GAD. Studies have shown that IL-33 has a negative regulatory effect on GABA synthesis. LPS treatment of mice can lead to the overexpression of IL-33 in amygdala astrocytes. After being captured by IL-1Rs on neurons in the CeA, IL-33 can activate NF-κB and downregulate the expression of BDNF directly or indirectly through the MAPK signaling pathway, which further decreases the GAD expression region and GABA synthesis in the medial prefrontal cortex (mPFC), resulting in an attenuated inhibitory effect on GABAergic neuronal circuitry between the mPFC and CeA, and ultimately leading to anxiety-like behaviors related to excitatory neurotoxicity in the mice [29, 107] (Fig. 8A).

IL-1β, another ligand of IL-1R, can regulate GABA release in the CeA. The frequency and amplitude of miniature inhibitory postsynaptic currents (mIPSCs) in GABAergic neurons change regularly with increasing or decreasing IL-1β levels, indicating that IL-1β has a bidirectional regulatory effect on GABA release from the presynaptic membrane. This forward or reverse regulatory effect is different in various clusters of GABAergic neurons in the CeA region [108, 109]. An endogenous antagonist of IL-1β can competitively bind to IL-1R but generally does not cause downstream signals. However, its regulatory effect on spontaneous inhibitory postsynaptic currents (sIPSCs) of GABAergic neurons in the CeA region was opposite to that of IL-1β. These results indicate that IL-1β can also indirectly affect the action potential of presynaptic neurons to induce GABA release [109, 110] (Fig. 8B, C).

IL-6 has also been reported to have a regulatory effect on GABAergic neurons, mainly by reducing the density of functional GABA receptor A (GABAAR), increasing the probability of GABAAR leaving the plasma membrane or decreasing the probability of GABAAR insertion into the plasma membrane. Therefore, the effect of GABA is attenuated, and the inhibitory or excitatory balance of postsynaptic neurons is disrupted [111]. The regulatory effect of TNF-α on GABA is also related to its receptor. Studies have shown that TNF-α binds to TNF-R1, activates the MAPK, PI3K, protein phosphatase 1 and dynein GTPase 1 signaling pathways, downregulates the levels of the α1, α2, β2, β3 and γ2 subunits of GABAAR on the cell membrane surface, and rapidly and sustainably decreases inhibitory synaptic strength in hippocampal GABAergic neurons of adult rat and mouse [112]. The mixed administration of IL-1β, TNF-α and GM-CSF to mice also showed that proinflammatory cytokines can increase the dysfunction of GABAAR by increasing the conversion of the GABAAR α1-subunit, accelerating the attenuation of GABA IPSC, and weakening its neuroinhibitory effect [113] (Fig. 8B).

Recent studies have shown that IL-10 can regulate the release of GABA from presynaptic neurons in the amygdala through the MAPK and PI3K pathways or indirectly affect the release of GABA from presynaptic neurons induced by action potentials, thereby alleviating anxiety-like behaviors. One study also revealed that the regulatory effect of IL-10 on GABA release in the amygdala of mice with alcohol addiction was comprehensively weakened, and the level of GABA release in the amygdala of mice with alcohol addiction increased after the remission of alcohol addiction, suggesting that the key mechanism involved in the effect of alcohol addiction is the dysfunction of IL-10 on GABA regulation. Therefore, IL-10 can be used as a diagnostic indicator for alcohol addiction and can be applied in the treatment of alcohol addiction through the regulation of GABA release [30] (Fig. 8C).

Fig. 8
figure 8

Effects of inflammatory cytokines on GABA synthesis (A) and release (B, C). (A) GABA is primarily synthesized by Glu, and GAD is the main rate-limiting enzyme for GABA synthesis. The binding of IL-33 to IL-1R first activates MAPK and further activates the NF-κB signaling pathway to inhibit BDNF synthesis, leading to a decrease in GAD expression and GABA synthesis. (B, C) IL-1β can regulate different GABAergic neurons, resulting in decreased or increased GABA release and corresponding decreases or increases in the number of IPSCs. TNF-α/IL-1β/IL-6 can also decrease the expression of postsynaptic GABAAR and attenuate GABA-induced IPSC. However, IL-10 promotes GABA release from GABAergic neurons and enhances IPSC through the MAPK/PI3K signaling pathway

Glu

Glu, also known as alpha-amino glutaric acid, is the most abundant amino acid neurotransmitter in the central nervous system. As the precursor of GABA production in the brain, Glu release causes the excitation of postsynaptic neurons, and together with GABA, Glu regulates the balance between excitation and inhibition in the brain [114]. Glu in neurons can be produced by glucose bypass metabolism through tricarboxylic acid (TCA) cycle. After release, Glu is ingested by excitatory amino acid transporters (EAATs) of astrocytes and synthesized into Gln for storage. Gln from astrocytes is then transported to neurons, where it is metabolized back into Glu in response to Glut (Figs. 8A and 9). Insufficient Glu leads to decreased excitability of neurons, and excessive release of Glu leads to hyperexcitability poisoning of postsynaptic neurons [114]. Abnormal Glu synthesis and release are believed to be related to memory disorders, cognitive impairment, schizophrenia, depression, epilepsy and other neurological and psychiatric diseases [79].

Fig. 9
figure 9

Effects of inflammatory cytokines on Glu synthesis, release and uptake. TNF-α and IL-1β can promote the synthesis and release of Glu, activate mGluR and AMPAR on the postsynaptic membrane, and cause mEPSC enhancement and neuronal excitotoxicity in postsynaptic neurons. Moreover, microglia-derived IL-1β can inhibit the expression of EAAT, reduce the transfer of Glu from the synaptic cleft to astrocytes, and aggravate the hyperexcited stimulation of postsynaptic neurons by Glu. In addition, astrocytes absorb less Glu, transform and generate less Gln, and transport less Gln to glutamatergic neurons, a process that would balance Glu release from glutamatergic neurons to a certain extent

Studies on the regulation and mechanism of inflammatory cytokines in glutamatergic neurons have focused mainly on the effect of inflammatory cytokines on Glu uptake by glial cells. For example, activated microglia release large amounts of IL-1β, which can lead to a decrease in EAAT activity and Glu uptake in astrocytes. The excessive presence of glutamate in the synaptic cleft leads to sustained activation of Ca2+-permeable α-amino-3-hydroxy-5-methyl-4-isoxazole-propionicacid receptor (AMPAR) in postsynaptic neurons, resulting in hyperexcitability [115, 116] (Fig. 9). Similar results have been demonstrated in mouse models of multiple sclerosis (MS), where IL-1β decreases EAAT expression in cerebellar astrocytes and enhances the effect of Glu in Purkinje cells. The duration of the miniature excitatory postsynaptic currents (mEPSCs) is increased, resulting in hyperexcitability poisoning of postsynaptic neurons [117] (Fig. 9). However, in the treatment of glaucomatous retinal detachment, IL-1 plays a positive regulatory role in Glu uptake by glial cells. The binding of IL-1 to IL-1R in Muller cells can activate the P38 MAPK pathway and increase the expression of caspase 11, which can promote actin fiber depolymerization mediated by cofilin. As a result, actin-loaded Na+/K+-ATPase is more localized to the cell membrane, and the Na+ in Muller cells is transported to the outside of the cell. However, the glutamate transporter (GLAST) on the cell membrane needs to transfer three Na+ ions in coordination with each Glu ingested, so Na+/K+-ATPase increases the efficiency of Glu ingestion through GLAST [118].

In addition, Shim et al. treated normal and TNFR1-shRNA-transfected Bergmann glial cells with TNF-α and found that TNF-α promoted the release of Glu from Bergmann cells to the synaptic cleft of Purkinje cells. It also enhanced the excitability of postsynaptic neurons by activating metabotropic glutamate receptor 1 (mGluR1) [119] (Fig. 9). It has also been shown that inflammatory cytokines can directly regulate the synthesis and release of Glu by neurons. When exploring the neurotoxicity of TNF-α and IL-1β, Ye et al. reported that both could lead to simultaneous increases in intracellular and extracellular Glu content in human cortical neuron cells supplemented with Gln in culture medium, and Glut was no longer localized only in the mitochondrial membrane, but also in other intracellular or extracellular regions. These results indicate that TNF-α and IL-1β can cause neuronal excitatory intoxication by promoting the synthesis and release of Glu [120] (Fig. 9). In contrast, Zhou et al. reported that IL-1β can upregulate the degradation of the synaptophysin protein by the E3 ligase siah1 via the ERK signaling pathway and subsequently inhibit the release of Glu, which impaired the learning and memory ability of rats [121].

BDNF deficiency and dysfunction

BDNF typically exists as a dimer protein, with each monomer being a secreted, mature polypeptide composed of 119 amino acid residues. BDNF is predominantly expressed in the central nervous system and exerts significant neurotrophic effects. It is the most abundant neurotrophic factor in the adult brain, particularly in the hippocampus and cerebral cortex, where it plays crucial roles in axon growth, myelination, synaptogenesis, and synaptic plasticity [122,123,124]. Compared to other neurotrophic factors, BDNF has garnered considerable attention in depression research due to its involvement in neuronal repair during depression remission [124,125,126,127]. BDNF’s significance in this context has led to a focus on its mechanisms of action and interactions with inflammatory factors, which can impact neural activities through various neurotrophic pathways. Given its importance, BDNF will be used as a representative neurotrophic factor to discuss the effects and mechanisms of inflammatory factors on neural activities mediated by neurotrophic factors. Blood samples and depression scale scores from elderly patients with depression show that the levels of TNF-α and BDNF are negatively correlated, and the ratio of TNF-α to BDNF is consistent with the Montgomery-Asberg Depression Rating Scale (MADRS) [128]. Other studies have confirmed that TNF-α, IL-1β and IL-6 reduce the expression of BDNF in the brain [129, 130], which further attenuates the activation of PI3K/Akt and MAPK P38, downstream signaling molecules of TrkB (transmembrane receptor of BDNF on neurons), leading to structural damage and dysfunction of neurons [26, 131, 132]. These results suggest that inflammatory cytokines can induce BDNF deficiency and dysfunction.

Furthermore, BDNF/TrkB signaling often occurs at the end of axons, and BDNF and TrkB can first form endosomes through endocytosis and then undergo retrograde transport to the cell body, leading to ERK5 phosphorylation and subsequent downstream effects. Carlos et al. cultured the cell body and axon compartments of primary neurons to explore the mechanism by which IL-1β affects BDNF and found that IL-1β attenuated the density of BDNF endosomes in various regions of neurons and that BDNF endosomes accumulated only in the terminal region of axons. Subsequent investigations confirmed that IL-1β induces the ubiquitination of BDNF endosomes, and these ubiquitinated endosomes are discarded after sorting by the cell transport system, and the failure of retrograde transport blocked the downstream signal activation of BDNF [133, 134]. In addition, in a study showing that IL-1β affects long-term potentiation (LTP) in the dendritic spines of hippocampal neurons, IL-1β inhibited the phosphorylation of insulin receptor substrate 1, a protein that links the activation of the BDNF receptor TrkB to downstream signaling pathways regulating CREB, Arc and cofilin, thereby blocking the effect of BDNF, attenuating P38 MAPK activation and actin fiber polymerization, and interfering with memory consolidation [135].

Neuroplasticity attenuation

Neuroplasticity mainly refers to the regulation and rearrangement of neuronal genesis, differentiation and apoptosis, among which the plasticity of neuronal synapses is the most commonly studied target in disease development and repair. Synaptic plasticity mainly includes short-term synaptic plasticity and long-term synaptic plasticity. Short-term synaptic plasticity includes facilitation, inhibition and enhancement. Long-term synaptic plasticity is primarily manifested as LTP and long-term depression (LTD). In menopause model mice subjected to bilateral ovariectomy, neuronal plasticity in the hippocampus, hypothalamus and amygdala was altered, synaptic transmission and LTP of neurons were significantly weakened, and mice exhibited obvious spatial memory deficits and depressive behaviors [136].

Inflammatory cytokines levels during menopause play important roles in regulating synaptic plasticity. It has been reported that a high concentration of TNF (1 µg/mL) can impair the plasticity of hippocampal CA1 pyramidal neurons in mice without affecting baseline synaptic transmission or previously established LTP. In contrast, a low concentration of TNF (1 ng/mL) promotes LTP in neurons, and the release of intracellular Ca2+ can prevent the negative effect of a high concentration of TNF on synaptic plasticity, but when Ca2+ stores are depleted, the ability of a low concentration of TNF to promote synaptic plasticity is lost [137]. Recent studies have shown that the MAPK signaling pathway may mediate the inhibitory effect of TNF-α on hippocampal LTP [138].

IL-6 also interferes with synaptic plasticity. The paired pulse current (presynaptic and postsynaptic) in mPFC neurons was measured by the patch clamp technique in IL-6 signal-blocking and wild-type social failure chronic stress model mice. The ratio of current generated by N-methyl-D-aspartate receptor (NMDAR) and AMPAR in stress-tolerant wild-type mice was significantly lower than that in IL-6 signal-blocking mice, and the ratio of current generated by AMPA and GABAAR in stress-sensitive wild-type mice was also significantly lower than that in IL-6 signal-blocking mice, indicating that IL-6 drives the negative rearrangement of synaptic plasticity in mPFC neurons during the formation of social failure behaviors [139]. IL-6 has also been shown to play a key role in synaptic plasticity dysfunction in amygdala neurons in mice with peripheral nerve injury-induced depressive behavior. IL-6 activates the palmitoylated transferase DHHC3 through the PI3K signaling pathway and promotes the palmitoylation of postsynaptic density protein-95 (PSD-95), which binds to the postsynaptic membrane Glu receptor 1 and aspartate receptor subunit NR2B, impinging on amygdala neurons from the rest of the brain, resulting in depression-like behavior in mice [140]. Tocilizumab, an IL-6 receptor antagonist, can inhibit the activation of hippocampal microglia and astrocytes and the subsequent “inflammatory storm” through the Wnt/β-catenin signaling pathway and restore hippocampal synaptic plasticity [141].

Additionally, INF-γ is involved in the regulation of neuroplasticity, which can lead to synaptic structural instability by reshaping the morphology of neurons in the nucleus accumbens, thus affecting opioid-induced addictive withdrawal behavior [142]. INF-γ can induce an acute increase in the amplitude of IPSCs in GABAergic neurons in a postsynaptic protein kinase C (PKC)-dependent manner and an acute increase in the frequency of IPSCs in GABAergic neurons in an inducible nitric oxide synthase- and soluble guanylate cyclase-dependent manner. IFN-γ was observed to shift short-term synaptic plasticity toward facilitation [143].

The immune factors IL-1β, IL-10 and IL-33 also play roles in the regulation of neuroplasticity. IL-1β inhibits the expression of the synaptic formation-related molecules synaptophysin (SYP and SYN1), the postsynaptic protein α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor GluA1 subunit, the N-methyl-d-aspartate receptor NR2B subunit and PSD-95 through histone deacetylase 4 (HDAC4), thus regulating the transcriptional inhibitor MeCP2 SUMO, resulting in LTP deficits and decreased synaptic plasticity [144].

IL-10 has been found to significantly enhance synaptic transmission and synaptic plasticity in hippocampal glutamatergic neurons, increasing the frequency of mEPSCs in a dose-dependent manner. In addition, IL-10 can induce postsynaptic compensatory changes such as synaptic expansion, increased mEPSC amplitude, and enhanced Ca2+ responsiveness to the AMPA agonist 5-fluorouracil alanine in primary cultured hippocampal glial synapses [145].

The regulation of neuroplasticity by IL-33 was discovered in the study of the mechanism of memory consolidation. Both memory accuracy and the expression of IL-33 decreased simultaneously in aged mice, and IL-33 administration significantly alleviated age-related dendritic spine regression. IL-33 can also guide microglia to phagocytose the extracellular matrix, which is conducive to the remodeling of synaptic connections between neurons and the functional integration of new neurons and increases the accuracy of fear memory [146]. As a new member of the IL-1 family, IL-33 is considered to play a role similar to that of a warning activator in the positive regulation of synaptic plasticity; however, the detailed mechanism is still unclear.

Therapeutic drugs for menopausal depression that target estrogen-immuno-neuromodulation

Currently, the primary therapeutic drugs used to treat menopausal depression include fluoxetine, duloxetine, escitalopram, mirtazapine, desvenlafaxine, quetiapine, modafinil and other neuromodulators [147,148,149,150,151,152,153]. However, these drugs do not address the decline in ovarian function and estrogen levels in menopausal women, so their therapeutic effect on menopausal depression is not ideal. Consequently, ERT has gained attention as a potential treatment. E2, ERβ selective ligand C-1, Vivelle-Dot, G03C, G03F, E2 + progesterone and tibolone have been used to treat model animals or menopausal depression patients [154,155,156,157,158,159]. Additionally, immune imbalances in menopausal women are also of interest to researchers, and immunomodulatory drugs and agents, such as RSV, SIM, astragalin, Erxian decoction, omega-3 (ω-3, also called N-3) polyunsaturated fatty acid, Fagopyrum tataricum seed extract, Acer tegmentosum, Saikosaponin A and Chaihu-Guizhi-Ganjiang tang, have been used to treat menopausal depression model animals or perimenopausal and postmenopausal women with mood disorders and shown good curative efficacy. However, the targets of most of these drugs and agents remain unclear [25, 43,44,45, 67, 160,161,162,163,164]. Here, we summarize the names, known targets, subjects and efficacy of these therapeutic drugs and agents to provide a reference for further evaluation, improvement or clinical trial research (Table 1).

Table 1 Therapeutic drugs/agents for menopausal depression targeting the estrogen-immune-neuromodulation signaling pathway

Conclusion and future directions

In conclusion, a significant decrease in estrogen levels disrupts immune homeostasis, particularly the levels of the immune factors TNF-α, IL-1β, IL-4, IL-6, IL-10 and IL-18 through the ERα/ERβ/GPER-associated NLRP3/NF-κB signaling pathway, which involves many signaling molecules, such as TLR2, TLR4, P2X7R, SIRT1, CREB, MS-KIF18A, PI3K, AKT, Gs, cAMP, PKA, PJA1, Serpina3n, Src, and MMP-9 [19,20,21,22,23,24, 41, 43, 44, 47,48,49, 52, 54,55,56, 58]. Subsequently, the increase of inflammatory cytokine levels causes BBB destruction [25, 62,63,64,65,66,67,68,69,70,71,72,73,74, 165], the dysfunction of neurotransmitters such as 5-HT [80,81,82,83,84,85], DA [86,87,88,89,90,91,92,93,94,95,96,97], NE [98,99,100,101,102,103,104,105], GABA [29, 30, 106,107,108,109,110,111,112,113] and Glu [79, 114,115,116,117,118,119,120,121], BDNF deficiency and dysfunction [26, 128,129,130,131,132,133,134,135], and attenuation of neuroplasticity [136,137,138,139,140,141,142,143,144,145,146], which are key factors in the onset and progression of depression (Fig. 10). All of these studies have shown that there is a complex regulatory system from estrogen to immune signaling molecules to neurons, which we call the estrogen-immune-neuromodulation system, and estrogen-immune-neuromodulation disorders cause susceptibility to depression in menopausal women (Fig. 10). Therefore, drugs targeting inflammatory cytokines and NLRP3/NF-κB signaling pathway-associated molecules are promising for restoring homeostasis of the estrogen-immuno-neuromodulation system and may play a positive role in the intervention and treatment of menopausal depression.

Fig. 10
figure 10

Estrogen-immune-neuromodulation disorders promote menopausal depression. A significant decrease in estrogen levels in menopausal women disrupts the homeostasis of the estrogen-immune-neuromodulation system, which first leads to immune imbalance, then induces nervous disorders, and finally gradually causes depression in menopausal women. Current research on immune imbalance has focused mainly on microglia, astrocytes, proinflammatory factors such as TNF-α, IL-1β, and IL-6, and anti-inflammatory factors such as IL-4 and IL-10. The investigation of neurological impairments involves BBB permeability, neurotransmitter activity, BDNF synthesis and neuronal plasticity. All these advances provide clues for the targeted treatment of menopausal depression

Although the main framework of the estrogen-immune-neuromodulation system is understood, many details involving this regulatory system remain unclear. For example, studies on the downstream signaling molecules of ERs and the stimulating factors of the NLRP3/NF-κB signaling pathway are very extensive, and few studies on the activation of NLRP3 and NF-κB caused by estrogen deficiency have clarified the interactions between various molecules step by step, which has limited the development of targeted antidepressants. In addition, there are some opposing and unified equilibrium points in the estrogen-immune-neuromodulation system. Multiple studies focusing on IL-1β have shown that IL-1β can not only decrease NE synthesis by disrupting the HPG system, GABA neurons and the expression of TH but also promote an increase in NE synthesis through the HPA system [98, 99, 103] (Fig. 7). IL-1β acts on different GABA neuron clusters in the amygdala and shows positive and negative regulatory effects [108,109,110] (Fig. 8B). Coincidentally, low levels of IL-33 and other new members of the IL-1 family show a positive regulatory effect on neuroplasticity, while high levels of IL-33 lead to a decrease in BDNF synthesis [29, 107] (Fig. 8A), disrupting the construction of neural synapses. Our understanding is that under normal circumstances, different elements of the body control a physiological activity in an appropriate state through mutual restriction and coordination, so different doses of stimulus factors affected by time and space are critical to maintaining or upsetting the balance. The dramatic decrease in estrogen levels in menopausal women triggers an imbalance in the estrogen-immune-neuromodulation system, which is the underlying reason why menopausal women are more susceptible to depression. Therefore, accurately understanding the appropriate balance in the nervous system and the regulatory mechanism is key for improving the clinical treatment of menopausal depression.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Wang Y, Cai X, Ma Y, Yang Y, Pan CW, Zhu X, Ke C. Metabolomics on depression: a comparison of clinical and animal research. J Affect Disord. 2024;349:559–68.

    Article  CAS  PubMed  Google Scholar 

  2. Kim JW, Suzuki K, Kavalali ET, Monteggia LM. Ketamine: mechanisms and relevance to treatment of depression. Annu Rev Med. 2024;75:129–43.

    Article  CAS  PubMed  Google Scholar 

  3. Wong S, Kwan ATH, Teopiz KM, Le GH, Meshkat S, Ho R, d’Andrea G, Cao B, Di Vincenzo JD, Rosenblat JD, McIntyre RS. A comparison between psilocybin and esketamine in treatment-resistant depression using number needed to treat (NNT): a systematic review. J Affect Disord. 2024;350:698–705.

    Article  CAS  PubMed  Google Scholar 

  4. Duman RS, Aghajanian GK, Sanacora G, Krystal JH. Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants. Nat Med. 2016;22(3):238–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Demireva EY, Suri D, Morelli E, Mahadevia D, Chuhma N, Teixeira CM, Ziolkowski A, Hersh M, Fifer J, Bagchi S, Chemiakine A, Moore H, Gingrich JA, Balsam P, Rayport S, Ansorge MS. 5-HT2C receptor blockade reverses SSRI-associated basal ganglia dysfunction and potentiates therapeutic efficacy. Mol Psychiatry. 2020;25(12):3304–21.

    Article  PubMed  Google Scholar 

  6. Bravo K, González-Ortiz M, Beltrán-Castillo S, Cáceres D, Eugenín J. Development of the placenta and brain are affected by selective serotonin reuptake inhibitor exposure during critical periods. Adv Exp Med Biol. 2023;1428:179–98.

    Article  CAS  PubMed  Google Scholar 

  7. Hong X, Chen R, Zhang L, Yan L, Xin J, Li J, Zha J. Long-term exposure to SSRI citalopram induces neurotoxic effects in zebrafish. Environ Sci Technol. 2022;56(17):12380–90.

    Article  CAS  PubMed  Google Scholar 

  8. Campbell KE, Dennerstein L, Finch S, Szoeke CE. Impact of menopausal status on negative mood and depressive symptoms in a longitudinal sample spanning 20 years. Menopause. 2017;24(5):490–6.

    Article  PubMed  Google Scholar 

  9. Gordon JL, Girdler SS, Meltzer-Brody SE, Stika CS, Thurston RC, Clark CT, Prairie BA, Moses-Kolko E, Joffe H, Wisner KL. Ovarian hormone fluctuation, neurosteroids, and HPA axis dysregulation in perimenopausal depression: a novel heuristic model. Am J Psychiatry. 2015;172(3):227–36.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Brown L, Hunter MS, Chen R, Crandall CJ, Gordon JL, Mishra GD, Rother V, Joffe H, Hickey M. Promoting good mental health over the menopause transition. Lancet. 2024;403(10430):969–83.

    Article  PubMed  Google Scholar 

  11. Herson M, Kulkarni J. Hormonal agents for the treatment of depression associated with the menopause. Drugs Aging. 2022;39(8):607–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Tangen T, Mykletun A. Depression and anxiety through the climacteric period: an epidemiological study (HUNT-II). J Psychosom Obstet Gynaecol. 2008;29(2):125–31.

    Article  CAS  PubMed  Google Scholar 

  13. Osterlund MK. Underlying mechanisms mediating the antidepressant effects of estrogens. Biochim Biophys Acta. 2010;1800(10):1136–44.

    Article  PubMed  Google Scholar 

  14. Chhibber A, Woody SK, Karim Rumi MA, Soares MJ, Zhao L. Estrogen receptor β deficiency impairs BDNF-5-HT(2A) signaling in the hippocampus of female brain: a possible mechanism for menopausal depression. Psychoneuroendocrinology. 2017;82:107–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Wang H, He Y, Sun Z, Ren S, Liu M, Wang G, Yang J. Microglia in depression: an overview of microglia in the pathogenesis and treatment of depression. J Neuroinflammation. 2022;19(1):132.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Dolotov OV, Inozemtseva LS, Myasoedov NF, Grivennikov IA. Stress-induced depression and alzheimer’s disease: focus on astrocytes. Int J Mol Sci. 2022;23(9):4999.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wu A, Zhang J. Neuroinflammation, memory, and depression: new approaches to hippocampal neurogenesis. J Neuroinflammation. 2023;20(1):283.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chakraborty B, Byemerwa J, Krebs T, Lim F, Chang CY, McDonnell DP. Estrogen receptor signaling in the immune system. Endocr Rev. 2023;44(1):117–41.

    Article  PubMed  Google Scholar 

  19. Jiang X, Chen Z, Yu X, Chen J, Sun C, Jing C, Xu L, Liu F, Ni W, Chen L. Lipopolysaccharide-induced depression is associated with estrogen receptor-α/SIRT1/NF-κB signaling pathway in old female mice. Neurochem Int. 2021;148:105097.

    Article  CAS  PubMed  Google Scholar 

  20. Wang X, Jiang Y, Feng B, Ma X, Zhang K, Yang F, Liu Z, Yang L, Yue J, Lu L, Song D, Guo Q, Qi J, Li X, Wang M, Zhang H, Huang J, Zhao M, Liu S. PJA1 mediates the effects of astrocytic GPR30 on learning and memory in female mice. J Clin Invest. 2023;133(18):e165812.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhao TZ, Ding Q, Hu J, He SM, Shi F, Ma LT. GPER expressed on microglia mediates the anti-inflammatory effect of estradiol in ischemic stroke. Brain Behav. 2016;6(4):e00449.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Wang J, Hou Y, Zhang L, Liu M, Zhao J, Zhang Z, Ma Y, Hou W. Estrogen attenuates traumatic brain injury by inhibiting the activation of microglia and astrocyte-mediated neuroinflammatory responses. Mol Neurobiol. 2021;58(3):1052–61.

    Article  CAS  PubMed  Google Scholar 

  23. Xu Y, Sheng H, Bao Q, Wang Y, Lu J, Ni X. NLRP3 inflammasome activation mediates estrogen deficiency-induced depression- and anxiety-like behavior and hippocampal inflammation in mice. Brain Behav Immun. 2016;56:175–86.

    Article  CAS  PubMed  Google Scholar 

  24. Pratap UP, Patil A, Sharma HR, Hima L, Chockalingam R, Hariharan MM, Shitoot S, Priyanka HP, ThyagaRajan S. Estrogen-induced neuroprotective and anti-inflammatory effects are dependent on the brain areas of middle-aged female rats. Brain Res Bull. 2016;124:238–53.

    Article  CAS  PubMed  Google Scholar 

  25. Yao G, Bai Z, Niu J, Zhang R, Lu Y, Gao T, Wang H. Astragalin attenuates depression-like behaviors and memory deficits and promotes M2 microglia polarization by regulating IL-4R/JAK1/STAT6 signaling pathway in a murine model of perimenopausal depression. Psychopharmacology. 2022;239(8):2421–43.

    Article  CAS  PubMed  Google Scholar 

  26. Liu B, Zhang Y, Yang Z, Liu M, Zhang C, Zhao Y, Song C. ω-3 DPA protected neurons from neuroinflammation by balancing microglia M1/M2 polarizations through inhibiting NF-κB/MAPK p38 signaling and activating neuron-BDNF-PI3K/AKT pathways. Mar Drugs. 2021;19(11):587.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Tang CZ, Zhang DF, Yang JT, Liu QH, Wang YR, Wang WS. Overexpression of microRNA-301b accelerates hippocampal microglia activation and cognitive impairment in mice with depressive-like behavior through the NF-κB signaling pathway. Cell Death Dis. 2019;10(4):316.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Xu Y, Sheng H, Tang Z, Lu J, Ni X. Inflammation and increased IDO in hippocampus contribute to depression-like behavior induced by estrogen deficiency. Behav Brain Res. 2015;288:71–8.

    Article  CAS  PubMed  Google Scholar 

  29. Zhuang X, Zhan B, Jia Y, Li C, Wu N, Zhao M, Chen N, Guo Y, Du Y, Zhang Y, Cao B, Li Y, Zhu F, Guo C, Wang Q, Li Y, Zhang L. IL-33 in the basolateral amygdala integrates neuroinflammation into anxiogenic circuits via modulating BDNF expression. Brain Behav Immun. 2022;102:98–109.

    Article  CAS  PubMed  Google Scholar 

  30. Patel RR, Wolfe SA, Bajo M, Abeynaike S, Pahng A, Borgonetti V, D’Ambrosio S, Nikzad R, Edwards S, Paust S, Roberts AJ, Roberto M. IL-10 normalizes aberrant amygdala GABA transmission and reverses anxiety-like behavior and dependence-induced escalation of alcohol intake. Prog Neurobiol. 2021;199:101952.

    Article  CAS  PubMed  Google Scholar 

  31. Giglio T, Imro MA, Filaci G, Scudeletti M, Puppo F, De Cecco L, Indiveri F, Costantini S. Immune cell circulating subsets are affected by gonadal function. Life Sci. 1994;54(18):1305–12.

    Article  CAS  PubMed  Google Scholar 

  32. Priyanka HP, Sharma U, Gopinath S, Sharma V, Hima L, ThyagaRajan S. Menstrual cycle and reproductive aging alters immune reactivity, NGF expression, antioxidant enzyme activities, and intracellular signaling pathways in the peripheral blood mononuclear cells of healthy women. Brain Behav Immun. 2013;32:131–43.

    Article  CAS  PubMed  Google Scholar 

  33. Malutan AM, Dan M, Nicolae C, Carmen M. Proinflammatory and anti-inflammatory cytokine changes related to menopause. Prz Menopauzalny. 2014;13(3):162–8.

    PubMed  PubMed Central  Google Scholar 

  34. Wan Z, Qin X, Tian Y, Ouyang F, Wang G, Wan Q. Long-term consumption of green tea can reduce the degree of depression in postmenopausal women by increasing estradiol. Nutrients. 2023;15(21):4514.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zare Z, Tehrani M, Zarbakhsh S, Mohammadi M. Protective effects of treadmill exercise on apoptotic neuronal damage and astrocyte activation in ovariectomized and/or diabetic rat prefrontal cortex: molecular and histological aspects. Int J Neurosci. 2022:1–9.

  36. Rana AK, Sharma S, Patial V, Singh D. Lithium therapy subdues neuroinflammation to maintain pyramidal cells arborization and rescues neurobehavioural impairments in ovariectomized rats. Mol Neurobiol. 2022;59(3):1706–23.

    Article  CAS  PubMed  Google Scholar 

  37. Caiazza F, Ryan EJ, Doherty G, Winter DC, Sheahan K. Estrogen receptors and their implications in colorectal carcinogenesis. Front Oncol. 2015;5:19.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Kumru S, Godekmerdan A, Yilmaz B. Immune effects of surgical menopause and estrogen replacement therapy in peri-menopausal women. J Reprod Immunol. 2004;63(1):31–8.

    Article  CAS  PubMed  Google Scholar 

  39. Engelmann F, Rivera A, Park B, Messerle-Forbes M, Jensen JT, Messaoudi I. Impact of estrogen therapy on lymphocyte homeostasis and the response to seasonal influenza vaccine in post-menopausal women. PLoS ONE. 2016;11(2):e0149045.

    Article  PubMed  PubMed Central  Google Scholar 

  40. McCarthy M, Raval AP. The peri-menopause in a woman’s life: a systemic inflammatory phase that enables later neurodegenerative disease. J Neuroinflammation. 2020;17(1):317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zusev M, Benayahu D. The regulation of MS-KIF18A expression and cross talk with estrogen receptor. PLoS ONE. 2009;4(7):e6407.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Toran-Allerand CD. Estrogen and the brain: beyond ER-alpha, ER-beta, and 17beta-estradiol. Ann N Y Acad Sci. 2005;1052:136–44.

    Article  CAS  PubMed  Google Scholar 

  43. Liu T, Ma Y, Zhang R, Zhong H, Wang L, Zhao J, Yang L, Fan X. Resveratrol ameliorates estrogen deficiency-induced depression- and anxiety-like behaviors and hippocampal inflammation in mice. Psychopharmacology. 2019;236(4):1385–99.

    Article  CAS  PubMed  Google Scholar 

  44. Menze ET, Ezzat H, Shawky S, Sami M, Selim EH, Ahmed S, Maged N, Nadeem N, Eldash S, Michel HE. Simvastatin mitigates depressive-like behavior in ovariectomized rats: possible role of NLRP3 inflammasome and estrogen receptors’ modulation. Int Immunopharmacol. 2021;95:107582.

    Article  CAS  PubMed  Google Scholar 

  45. Wu B, Song Q, Zhang Y, Wang C, Yang M, Zhang J, Han W, Jiang P. Antidepressant activity of ω-3 polyunsaturated fatty acids in ovariectomized rats: role of neuroinflammation and microglial polarization. Lipids Health Dis. 2020;19(1):4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zheng X, Wang J, Bi F, Li Y, Xiao J, Chai Z, Li Y, Miao Z, Wang Y. Protective effects of Lycium barbarum polysaccharide on ovariectomy–induced cognition reduction in aging mice. Int J Mol Med. 2021;48(1):121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Mishra P, Davies DA, Albensi BC. The interaction between NF-κB and estrogen in alzheimer’s disease. Mol Neurobiol. 2023;60(3):1515–26.

    Article  CAS  PubMed  Google Scholar 

  48. Li M, Zhang J, Chen W, Liu S, Liu X, Ning Y, Cao Y, Zhao Y. Supraphysiologic doses of 17β-estradiol aggravate depression-like behaviors in ovariectomized mice possibly via regulating microglial responses and brain glycerophospholipid metabolism. J Neuroinflammation. 2023;20(1):204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Han R, Gu S, Zhang Y, Luo A, Jing X, Zhao L, Zhao X, Zhang L. Estrogen promotes progression of hormone-dependent breast cancer through CCL2-CCR2 axis by upregulation of twist via PI3K/AKT/NF-κB signaling. Sci Rep. 2018;8(1):9575.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Alexander A, Irving AJ, Harvey J. Emerging roles for the novel estrogen-sensing receptor GPER1 in the CNS. Neuropharmacology. 2017;113(Pt B):652–60.

    Article  CAS  PubMed  Google Scholar 

  51. Tang H, Zhang Q, Yang L, Dong Y, Khan M, Yang F, Brann DW, Wang R. GPR30 mediates estrogen rapid signaling and neuroprotection. Mol Cell Endocrinol. 2014;387(1–2):52–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhang Z, Qin P, Deng Y, Ma Z, Guo H, Guo H, Hou Y, Wang S, Zou W, Sun Y, Ma Y, Hou W. The novel estrogenic receptor GPR30 alleviates ischemic injury by inhibiting TLR4-mediated microglial inflammation. J Neuroinflammation. 2018;15(1):206.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Brailoiu E, Dun SL, Brailoiu GC, Mizuo K, Sklar LA, Oprea TI, Prossnitz ER, Dun NJ. Distribution and characterization of estrogen receptor G protein-coupled receptor 30 in the rat central nervous system. J Endocrinol. 2007;193(2):311–21.

    Article  CAS  PubMed  Google Scholar 

  54. Bai N, Zhang Q, Zhang W, Liu B, Yang F, Brann D, Wang R. G-protein-coupled estrogen receptor activation upregulates interleukin-1 receptor antagonist in the hippocampus after global cerebral ischemia: implications for neuronal self-defense. J Neuroinflammation. 2020;17(1):45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Pan MX, Li J, Ma C, Fu K, Li ZQ, Wang ZF. Sex-dependent effects of GPER activation on neuroinflammation in a rat model of traumatic brain injury. Brain Behav Immun. 2020;88:421–31.

    Article  CAS  PubMed  Google Scholar 

  56. Xue Y, Zhang Y, Wu Y, Zhao T. Activation of GPER-1 attenuates traumatic brain injury-induced neurological impairments in mice. Mol Neurobiol. 2024.

  57. Notas G, Kampa M, Castanas E. G protein-coupled estrogen receptor in immune cells and its role in immune-related diseases. Front Endocrinol (Lausanne). 2020;11:579420.

    Article  PubMed  Google Scholar 

  58. Jiang M, Liu Y, Wu H, Ma Z, Gu X. High estrogen level modifies postoperative hyperalgesia via GPR30 and MMP-9 in dorsal root ganglia neurons. Neurochem Res. 2020;45(7):1661–73.

    Article  CAS  PubMed  Google Scholar 

  59. Prossnitz ER, Barton M. The G protein-coupled oestrogen receptor GPER in health and disease: an update. Nat Rev Endocrinol. 2023;19(7):407–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Pemberton K, Rosato M, Dedert C, DeLeon C, Arnatt C, Xu F. Differential effects of the G-protein-coupled estrogen receptor (GPER) on rat embryonic (E18) hippocampal and cortical neurons. eNeuro 2022; 9(4).

  61. DeLeon C, Pemberton K, Green M, Kalajdzic V, Rosato M, Xu F, Arnatt C. Novel GPER agonist, CITFA, increases neurite growth in rat embryonic (E18) hippocampal neurons. ACS Chem Neurosci. 2022;13(8):1119–28.

    Article  CAS  PubMed  Google Scholar 

  62. Sandoval KE, Witt KA. Age and 17β-estradiol effects on blood-brain barrier tight junction and estrogen receptor proteins in ovariectomized rats. Microvasc Res. 2011;81(2):198–205.

    Article  CAS  PubMed  Google Scholar 

  63. Shin JA, Oh S, Ahn JH, Park EM. Estrogen receptor-mediated resveratrol actions on blood-brain barrier of ovariectomized mice. Neurobiol Aging. 2015;36(2):993–1006.

    Article  CAS  PubMed  Google Scholar 

  64. Butler T, Glodzik L, Wang XH, Xi K, Li Y, Pan H, Zhou L, Chiang GC, Morim S, Wickramasuriya N, Tanzi E, Maloney T, Harvey P, Mao X, Razlighi QR, Rusinek H, Shungu DC, de Leon M, Atwood CS, Mozley PD. Positron Emission Tomography reveals age-associated hypothalamic microglial activation in women. Sci Rep. 2022;12(1):13351.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Itoh N, Itoh Y, Meyer CE, Suen TT, Cortez-Delgado D, Rivera Lomeli M, Wendin S, Somepalli SS, Golden LC, MacKenzie-Graham A, Voskuhl RR. Estrogen receptor beta in astrocytes modulates cognitive function in mid-age female mice. Nat Commun. 2023;14(1):6044.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Wei X, Li R, Li X, Wang B, Huang J, Mu H, Zhang Q, Zhang Z, Ru Y, Wu X, Qiu Y, Ye Y, Feng Y, Wang S, Chen H, Yi C, Wang J. iPSCs-derived mesenchymal stromal cells mitigate anxiety and neuroinflammation in aging female mice. Int J Biochem Cell Biol. 2023;155:106347.

    Article  CAS  PubMed  Google Scholar 

  67. Rana AK, Sharma S, Kumar R, Singh D. Buckwheat tartary regulates the Gsk-3β/β-catenin pathway to prevent neurobehavioral impairments in a rat model of surgical menopause. Metab Brain Dis. 2023;38(6):1859–75.

    Article  CAS  PubMed  Google Scholar 

  68. Gal Z, Torok D, Gonda X, Eszlari N, Anderson IM, Deakin B, Juhasz G, Bagdy G, Petschner P. Inflammation and blood-brain barrier in depression: interaction of CLDN5 and IL6 gene variants in stress-induced depression. Int J Neuropsychopharmacol. 2023;26(3):189–97.

    Article  CAS  PubMed  Google Scholar 

  69. Voirin AC, Perek N, Roche F. Inflammatory stress induced by a combination of cytokines (IL-6, IL-17, TNF-α) leads to a loss of integrity on bEnd.3 endothelial cells in vitro BBB model. Brain Res. 2020;1730:146647.

    Article  CAS  PubMed  Google Scholar 

  70. Dudek KA, Dion-Albert L, Lebel M, LeClair K, Labrecque S, Tuck E, Ferrer Perez C, Golden SA, Tamminga C, Turecki G, Mechawar N, Russo SJ, Menard C. Molecular adaptations of the blood-brain barrier promote stress resilience vs. depression. Proc Natl Acad Sci U S A. 2020;117(6):3326–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Chen AQ, Fang Z, Chen XL, Yang S, Zhou YF, Mao L, Xia YP, Jin HJ, Li YN, You MF, Wang XX, Lei H, He QW, Hu B. Microglia-derived TNF-α mediates endothelial necroptosis aggravating blood brain-barrier disruption after ischemic stroke. Cell Death Dis. 2019;10(7):487.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Yang F, Zhao K, Zhang X, Zhang J, Xu B. ATP induces disruption of tight junction proteins via IL-1 beta-dependent MMP-9 activation of human blood-brain barrier in vitro. Neural Plast. 2016;2016:8928530.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Wang Y, Jin S, Sonobe Y, Cheng Y, Horiuchi H, Parajuli B, Kawanokuchi J, Mizuno T, Takeuchi H, Suzumura A. Interleukin-1β induces blood-brain barrier disruption by downregulating sonic hedgehog in astrocytes. PLoS ONE. 2014;9(10):e110024.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Haruwaka K, Ikegami A, Tachibana Y, Ohno N, Konishi H, Hashimoto A, Matsumoto M, Kato D, Ono R, Kiyama H, Moorhouse AJ, Nabekura J, Wake H. Dual microglia effects on blood brain barrier permeability induced by systemic inflammation. Nat Commun. 2019;10(1):5816.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Tang CZ, Zhang YL, Wang WS, Li WG, Shi JP. Serum levels of high-sensitivity C-reactive protein at admission are more strongly associated with poststroke depression in acute ischemic stroke than homocysteine levels. Mol Neurobiol. 2016;53(4):2152–60.

    Article  CAS  PubMed  Google Scholar 

  76. Cossette É, Cloutier I, Tardif K, DonPierre G, Tanguay JF. Estradiol inhibits vascular endothelial cells pro-inflammatory activation induced by C-reactive protein. Mol Cell Biochem. 2013;373(1–2):137–47.

    Article  CAS  PubMed  Google Scholar 

  77. Lan XY, Yu H, Chen QJ, Zhai S, Zhang CF, Li F, Wang CZ, Yuan CS. Effect of liquiritin on neuroendocrine-immune network in menopausal rat model. Phytother Res. 2020;34(10):2665–74.

    Article  CAS  PubMed  Google Scholar 

  78. Cao LH, Qiao JY, Huang HY, Fang XY, Zhang R, Miao MS, Li XM. PI3K-AKT signaling activation and icariin: the potential effects on the perimenopausal depression-like rat model. Molecules. 2019;24(20):3700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Zhang K, Liu R, Gao Y, Ma W, Shen W. Electroacupuncture relieves LPS-induced depression-like behaviour in rats through IDO-mediated tryptophan-degrading pathway. Neuropsychiatr Dis Treat. 2020;16:2257–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Joaquim HPG, Costa AC, Gattaz WF, Talib LL. Kynurenine is correlated with IL-1β in plasma of schizophrenia patients. J Neural Transm (Vienna). 2018;125(5):869–73.

    Article  CAS  PubMed  Google Scholar 

  82. Correia AS, Vale N. Tryptophan metabolism in depression: a narrative review with a focus on serotonin and kynurenine pathways. Int J Mol Sci. 2022;23(15):8493.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Park B, Lee S, Jang Y, Park HY. Affective dysfunction mediates the link between neuroimmune markers and the default mode network functional connectivity, and the somatic symptoms in somatic symptom disorder. Brain Behav Immun. 2024;118:90–100.

    Article  CAS  PubMed  Google Scholar 

  84. Qiu X, Lu P, Zeng X, Jin S, Chen X. Study on the mechanism for SIRT1 during the process of exercise improving depression. Brain Sci. 2023;13(5):719.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Atmaca HT. Expression of serotonin 2A, 2 C, 6 and 7 receptor and IL-6 mRNA in experimental toxoplasmic encephalitis in mice. Heliyon. 2019;5(11):e02890.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Lian TH, Guo P, Zhang YN, Li JH, Li LX, Ding DY, Li DN, Zhang WJ, Guan HY, Wang XM, Zhang W. Parkinson’s disease with depression: the correlations between neuroinflammatory factors and neurotransmitters in cerebrospinal fluid. Front Aging Neurosci. 2020;12:574776.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Dong L, Zheng YM, Luo XG, He ZY. High inflammatory tendency induced by malignant stimulation through imbalance of CD28 and CTLA-4/PD-1 contributes to dopamine neuron injury. J Inflamm Res. 2021;14:2471–82.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Vancassel S, Fanet H, Castanon N, De Monchaux C, Cussotto S, Capuron L. Tetrahydrobiopterin modulates the behavioral neuroinflammatory response to an LPS challenge in mice. Brain Behav Immun. 2022;105:139–48.

    Article  CAS  PubMed  Google Scholar 

  89. Sprouse J, Sampath C, Gangula P. 17β-Estradiol suppresses gastric inflammatory and apoptotic stress responses and restores nNOS-mediated gastric emptying in streptozotocin (STZ)-induced diabetic female mice. Antioxid (Basel). 2023;12(3):758.

    Article  CAS  Google Scholar 

  90. Singh G, Mittra N, Singh C. Tempol and silymarin rescue from zinc-induced degeneration of dopaminergic neurons through modulation of oxidative stress and inflammation. Mol Cell Biochem. 2023;478(8):1705–18.

    Article  CAS  PubMed  Google Scholar 

  91. Lee HJ, Choe K, Park JS, Khan A, Kim MW, Park TJ, Kim MO. O-cyclic phytosphingosine-1-phosphate protects against motor dysfunctions and glial cell mediated neuroinflammation in the Parkinson’s disease mouse models. Antioxid (Basel). 2022;11(11):2107.

    Article  CAS  Google Scholar 

  92. Graves SM, Xie Z, Stout KA, Zampese E, Burbulla LF, Shih JC, Kondapalli J, Patriarchi T, Tian L, Brichta L, Greengard P, Krainc D, Schumacker PT, Surmeier DJ. Dopamine metabolism by a monoamine oxidase mitochondrial shuttle activates the electron transport chain. Nat Neurosci. 2020;23(1):15–20.

    Article  CAS  PubMed  Google Scholar 

  93. Erbaş O, Taşkıran D. Sepsis-induced changes in behavioral stereotypy in rats; involvement of tumor necrosis factor-alpha, oxidative stress, and dopamine turnover. J Surg Res. 2014;186(1):262–68.

    Article  PubMed  Google Scholar 

  94. Wu Y, Na X, Zang Y, Cui Y, Xin W, Pang R, Zhou L, Wei X, Li Y, Liu X. Upregulation of tumor necrosis factor-alpha in nucleus accumbens attenuates morphine-induced rewarding in a neuropathic pain model. Biochem Biophys Res Commun. 2014;449(4):502–7.

    Article  CAS  PubMed  Google Scholar 

  95. Zhu CB, Lindler KM, Owens AW, Daws LC, Blakely RD, Hewlett WA. Interleukin-1 receptor activation by systemic lipopolysaccharide induces behavioral despair linked to MAPK regulation of CNS serotonin transporters. Neuropsychopharmacology. 2010;35(13):2510–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Morón JA, Zakharova I, Ferrer JV, Merrill GA, Hope B, Lafer EM, Lin ZC, Wang JB, Javitch JA, Galli A, Shippenberg TS. Mitogen-activated protein kinase regulates dopamine transporter surface expression and dopamine transport capacity. J Neurosci. 2003;23(24):8480–8.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Felger JC, Mun J, Kimmel HL, Nye JA, Drake DF, Hernandez CR, Freeman AA, Rye DB, Goodman MM, Howell LL, Miller AH. Chronic interferon-α decreases dopamine 2 receptor binding and striatal dopamine release in association with anhedonia-like behavior in nonhuman primates. Neuropsychopharmacology. 2013;38(11):2179–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Sirivelu MP, MohanKumar PS, MohanKumar SM. Interleukin-1 beta simultaneously affects the stress and reproductive axes by modulating norepinephrine levels in different brain areas. Life Sci. 2012;91(17–18):878–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Sirivelu MP, Shin AC, Perez GI, MohanKumar PS, MohanKumar SM. Effect of L-dopa on interleukin-1 beta-induced suppression of luteinizing hormone secretion in intact female rats. Hum Reprod. 2009;24(3):718–25.

    Article  CAS  PubMed  Google Scholar 

  100. Nagatsu T, Nakashima A, Watanabe H, Ito S, Wakamatsu K, Zucca FA, Zecca L, Youdim M, Wulf M, Riederer P, Dijkstra JM. The role of tyrosine hydroxylase as a key player in neuromelanin synthesis and the association of neuromelanin with Parkinson’s disease. J Neural Transm (Vienna). 2023;130(5):611–25.

    Article  CAS  PubMed  Google Scholar 

  101. Li R, Hou J, Xu Q, Liu QJ, Shen YJ, Rodin G, Li M. High level interleukin-6 in the medium of human pancreatic cancer cell culture suppresses production of neurotransmitters by PC12 cell line. Metab Brain Dis. 2012;27(1):91–100.

    Article  PubMed  Google Scholar 

  102. Park HJ, Shim HS, An K, Starkweather A, Kim KS, Shim I. IL-4 inhibits IL-1β-induced depressive-like behavior and central neurotransmitter alterations. Mediators Inflamm. 2015; 2015:941413.

  103. Sirivelu MP, Burnett R, Shin AC, Kim C, MohanKumar PS, MohanKumar SM. Interaction between GABA and norepinephrine in interleukin-1beta-induced suppression of the luteinizing hormone surge. Brain Res. 2009;1248:107–14.

    Article  CAS  PubMed  Google Scholar 

  104. Day JS, O’Neill E, Cawley C, Aretz NK, Kilroy D, Gibney SM, Harkin A, Connor TJ. Noradrenaline acting on astrocytic β₂-adrenoceptors induces neurite outgrowth in primary cortical neurons. Neuropharmacology. 2014;77:234–48.

    Article  CAS  PubMed  Google Scholar 

  105. Ryan KM, Boyle NT, Harkin A, Connor TJ. Dexamethasone attenuates inflammatory-mediated suppression of β(2)-adrenoceptor expression in rat primary mixed glia. J Neuroimmunol. 2020;338:577082.

    Article  CAS  PubMed  Google Scholar 

  106. Milanez MIO, Silva AM, Perry JC, Faber J, Nishi EE, Bergamaschi CT, Campos RR. Pattern of sympathetic vasomotor activity induced by GABAergic inhibition in the brain and spinal cord. Pharmacol Rep. 2020;72(1):67–79.

    Article  CAS  PubMed  Google Scholar 

  107. Vainchtein ID, Chin G, Cho FS, Kelley KW, Miller JG, Chien EC, Liddelow SA, Nguyen PT, Nakao-Inoue H, Dorman LC, Akil O, Joshita S, Barres BA, Paz JT, Molofsky AB, Molofsky AV. Astrocyte-derived interleukin-33 promotes microglial synapse engulfment and neural circuit development. Science. 2018;359(6381):1269–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Bajo M, Varodayan FP, Madamba SG, Robert AJ, Casal LM, Oleata CS, Siggins GR, Roberto M. IL-1 interacts with ethanol effects on GABAergic transmission in the mouse central amygdala. Front Pharmacol. 2015;6:49.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Patel RR, Khom S, Steinman MQ, Varodayan FP, Kiosses WB, Hedges DM, Vlkolinsky R, Nadav T, Polis I, Bajo M, Roberts AJ, Roberto M. IL-1β expression is increased and regulates GABA transmission following chronic ethanol in mouse central amygdala. Brain Behav Immun. 2019;75:208–19.

    Article  CAS  PubMed  Google Scholar 

  110. Bajo M, Herman MA, Varodayan FP, Oleata CS, Madamba SG, Harris RA, Blednov YA, Roberto M. Role of the IL-1 receptor antagonist in ethanol-induced regulation of GABAergic transmission in the central amygdala. Brain Behav Immun. 2015;45:189–97.

    Article  CAS  PubMed  Google Scholar 

  111. Pribiag H, Stellwagen D. TNF-α downregulates inhibitory neurotransmission through protein phosphatase 1-dependent trafficking of GABA(A) receptors. J Neurosci. 2013;33(40):15879–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Garcia-Oscos F, Salgado H, Hall S, Thomas F, Farmer GE, Bermeo J, Galindo LC, Ramirez RD, D’Mello S, Rose-John S, Atzori M. The stress-induced cytokine interleukin-6 decreases the inhibition/excitation ratio in the rat temporal cortex via trans-signaling. Biol Psychiatry. 2012;71(7):574–82.

    Article  CAS  PubMed  Google Scholar 

  113. Giacco V, Panattoni G, Medelin M, Bonechi E, Aldinucci A, Ballerini C, Ballerini L. Cytokine inflammatory threat, but not LPS one, shortens GABAergic synaptic currents in the mouse spinal cord organotypic cultures. J Neuroinflammation. 2019;16(1):127.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Andersen JV, Schousboe A, Verkhratsky A. Astrocyte energy and neurotransmitter metabolism in Alzheimer’s disease: integration of the glutamate/GABA-glutamine cycle. Prog Neurobiol. 2022;217:102331.

    Article  CAS  PubMed  Google Scholar 

  115. Charles-Messance H, Blot G, Couturier A, Vignaud L, Touhami S, Beguier F, Siqueiros L, Forster V, Barmo N, Augustin S, Picaud S, Sahel JA, Rendon A, Grosche A, Tadayoni R, Sennlaub F, Guillonneau X. IL-1β induces rod degeneration through the disruption of retinal glutamate homeostasis. J Neuroinflammation. 2020;17(1):1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Chen Y, Li X, Xiong Q, Du Y, Luo M, Yi L, Pang Y, Shi X, Wang YT, Dong Z. Inhibiting NLRP3 inflammasome signaling pathway promotes neurological recovery following hypoxic-ischemic brain damage by increasing p97-mediated surface GluA1-containing AMPA receptors. J Transl Med. 2023;21(1):567.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Mandolesi G, Musella A, Gentile A, Grasselli G, Haji N, Sepman H, Fresegna D, Bullitta S, De Vito F, Musumeci G, Di Sanza C, Strata P, Centonze D. Interleukin-1β alters glutamate transmission at purkinje cell synapses in a mouse model of multiple sclerosis. J Neurosci. 2013;33(29):12105–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Namekata K, Harada C, Kohyama K, Matsumoto Y, Harada T. Interleukin-1 stimulates glutamate uptake in glial cells by accelerating membrane trafficking of Na+/K+-ATPase via actin depolymerization. Mol Cell Biol. 2008;28(10):3273–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Shim HG, Jang SS, Kim SH, Hwang EM, Min JO, Kim HY, Kim YS, Ryu C, Chung G, Kim Y, Yoon BE, Kim SJ. TNF-α increases the intrinsic excitability of cerebellar Purkinje cells through elevating glutamate release in Bergmann Glia. Sci Rep. 2018;8(1):11589.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Ye L, Huang Y, Zhao L, Li Y, Sun L, Zhou Y, Qian G, Zheng JC. IL-1β and TNF-α induce neurotoxicity through glutamate production: a potential role for neuronal glutaminase. J Neurochem. 2013;125(6):897–908.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Zhou Q, Lin L, Li H, Li Y, Liu N, Wang H, Jiang S, Li Q, Chen Z, Lin Y, Jin H, Deng Y. Intrahippocampal injection of IL-1β upregulates Siah1-mediated degradation of synaptophysin by activation of the ERK signaling in male rat. J Neurosci Res. 2023;101(6):930–51.

    Article  CAS  PubMed  Google Scholar 

  122. Albini M, Krawczun-Rygmaczewska A, Cesca F. Astrocytes and brain-derived neurotrophic factor (BDNF). Neurosci Res. 2023;197:42–51.

    Article  CAS  PubMed  Google Scholar 

  123. Li Y, Li F, Qin D, Chen H, Wang J, Wang J, Song S, Wang C, Wang Y, Liu S, Gao D, Wang ZH. The role of brain derived neurotrophic factor in central nervous system. Front Aging Neurosci. 2022;14:986443.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Wang CS, Kavalali ET, Monteggia LM. BDNF signaling in context: from synaptic regulation to psychiatric disorders. Cell. 2022;185(1):62–76.

    Article  CAS  PubMed  Google Scholar 

  125. Zhang JC, Yao W, Hashimoto K. Brain-derived neurotrophic factor (BDNF)-TrkB signaling in inflammation-related depression and potential therapeutic targets. Curr Neuropharmacol. 2016;14(7):721–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Correia AS, Cardoso A, Vale N. BDNF unveiled: exploring its role in major depression disorder serotonergic imbalance and associated stress conditions. Pharmaceutics. 2023;15(8):2081.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Baek DC, Kang JY, Lee JS, Lee EJ, Son CG. Linking alterations in estrogen receptor expression to memory deficits and depressive behavior in an ovariectomy mouse model. Sci Rep. 2024;14(1):6854.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Loef D, Vansteelandt K, Oudega ML, van Eijndhoven P, Carlier A, van Exel E, Rhebergen D, Sienaert P, Vandenbulcke M, Bouckaert F, Dols A. The ratio and interaction between neurotrophin and immune signaling during electroconvulsive therapy in late-life depression. Brain Behav Immun Health. 2021;18:100389.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Feng X, Ma X, Li J, Zhou Q, Liu Y, Song J, Liu J, Situ Q, Wang L, Zhang J, Lin F. Inflammatory pathogenesis of post-stroke depression. Aging and disease.; 2024.

  130. Zhang YM, Wei RM, Feng YZ, Zhang KX, Ge YJ, Kong XY, Li XY, Chen GH. Sleep deprivation aggravates lipopolysaccharide-induced anxiety, depression and cognitive impairment: the role of pro-inflammatory cytokines and synaptic plasticity-associated proteins. J Neuroimmunol. 2024;386:578252.

    Article  CAS  PubMed  Google Scholar 

  131. Wu Y, Zhu Z, Lan T, Li S, Li Y, Wang C, Feng Y, Mao X, Yu S. Levomilnacipran improves lipopolysaccharide-induced dysregulation of synaptic plasticity and depression-like behaviors via activating BDNF/TrkB mediated PI3K/Akt/mTOR signaling pathway. Mol Neurobiol. 2023.

  132. Gao R, Ali T, Liu Z, Li A, Hao L, He L, Yu X, Li S. Ceftriaxone averts neuroinflammation and relieves depressive-like behaviors via GLT-1/TrkB signaling. Biochem Biophys Res Commun. 2024;701:149550.

    Article  CAS  PubMed  Google Scholar 

  133. Carlos AJ, Tong L, Prieto GA, Cotman CW. IL-1β impairs retrograde flow of BDNF signaling by attenuating endosome trafficking. J Neuroinflammation. 2017;14(1):29.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Bronfman FC, Escudero CA, Weis J, Kruttgen A. Endosomal transport of neurotrophins: roles in signaling and neurodegenerative diseases. Dev Neurobiol. 2007;67(9):1183–203.

    Article  CAS  PubMed  Google Scholar 

  135. Tong L, Prieto GA, Kramár EA, Smith ED, Cribbs DH, Lynch G, Cotman CW. Brain-derived neurotrophic factor-dependent synaptic plasticity is suppressed by interleukin-1β via p38 mitogen-activated protein kinase. J Neurosci. 2012;32(49):17714–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Yan YD, Chen YQ, Wang CY, Ye CB, Hu ZZ, Behnisch T, Huang ZL, Yang SR. Chronic modafinil therapy ameliorates depressive-like behavior, spatial memory and hippocampal plasticity impairments, and sleep-wake changes in a surgical mouse model of menopause. Transl Psychiatry. 2021;11(1):116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Maggio N, Vlachos A. Tumor necrosis factor (TNF) modulates synaptic plasticity in a concentration-dependent manner through intracellular calcium stores. J Mol Med (Berl). 2018;96(10):1039–47.

    Article  CAS  PubMed  Google Scholar 

  138. Yang Q, Zhang Y, Zhang L, Li X, Dong R, Song C, Cheng L, Shi M, Zhao H. Combination of tea polyphenols and proanthocyanidins prevents menopause-related memory decline in rats via increased hippocampal synaptic plasticity by inhibiting p38 MAPK and TNF-α pathway. Nutr Neurosci. 2022;25(9):1909–27.

    Article  CAS  PubMed  Google Scholar 

  139. Esquivel-Rendón E, Vargas-Mireles J, Cuevas-Olguín R, Miranda-Morales M, Acosta-Mares P, García-Oscos F, Pineda JC, Salgado H, Rose-John S, Atzori M. Interleukin 6 dependent synaptic plasticity in a social defeat-susceptible prefrontal cortex circuit. Neuroscience. 2019;414:280–96.

    Article  PubMed  Google Scholar 

  140. Liu L, Dai L, Xu D, Wang Y, Bai L, Chen X, Li M, Yang S, Tang Y. Astrocyte secretes IL-6 to modulate PSD-95 palmitoylation in basolateral amygdala and depression-like behaviors induced by peripheral nerve injury. Brain Behav Immun. 2022;104:139–54.

    Article  CAS  PubMed  Google Scholar 

  141. Xu D, Xu Y, Gao X, Yan M, Zhang C, Wu X, Xia Q, Ge J. Potential value of Interleukin-6 as a diagnostic biomarker in human MDD and the antidepressant effect of its receptor antagonist tocilizumab in lipopolysaccharide-challenged rats. Int Immunopharmacol. 2023;124Pt B:110903.

    Article  Google Scholar 

  142. Zhu Y, Yan P, Wang R, Lai J, Tang H, Xiao X, Yu R, Bao X, Zhu F, Wang K, Lu Y, Dang J, Zhu C, Zhang R, Dang W, Zhang B, Fu Q, Zhang Q, Kang C, Chen Y, Chen X, Liang Q, Wang K. Opioid-induced fragile-like regulatory T cells contribute to withdrawal. Cell. 2023;186(3):591–e606523.

    Article  CAS  PubMed  Google Scholar 

  143. Döhne N, Falck A, Janach GMS, Byvaltcev E, Strauss U. Interferon-γ augments GABA release in the developing neocortex via nitric oxide synthase/soluble guanylate cyclase and constrains network activity. Front Cell Neurosci. 2022;16:913299.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Li Puma DD, Colussi C, Bandiera B, Puliatti G, Rinaudo M, Cocco S, Paciello F, Re A, Ripoli C, De Chiara G, Bertozzi A, Palamara AT, Piacentini R, Grassi C. Interleukin 1β triggers synaptic and memory deficits in herpes simplex virus type-1-infected mice by downregulating the expression of synaptic plasticity-related genes via the epigenetic MeCP2/HDAC4 complex. Cell Mol Life Sci. 2023;80(6):172.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Nenov MN, Konakov MV, Teplov IY, Levin SG. Interleukin-10 facilitates glutamatergic synaptic transmission and homeostatic plasticity in cultured hippocampal neurons. Int J Mol Sci. 2019;20(13):3375.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Nguyen PT, Dorman LC, Pan S, Vainchtein ID, Han RT, Nakao-Inoue H, Taloma SE, Barron JJ, Molofsky AB, Kheirbek MA, Molofsky AV. Microglial remodeling of the extracellular matrix promotes synapse plasticity. Cell. 2020;182(2):388–e403315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Zhou J, Wang X, Feng L, Xiao L, Yang R, Zhu X, Shi H, Hu Y, Chen R, Boyce P, Wang G. Venlafaxine vs. fluoxetine in postmenopausal women with major depressive disorder: an 8-week, randomized, single-blind, active-controlled study. BMC Psychiatry. 2021;21(1):260.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Joffe H, Soares CN, Petrillo LF, Viguera AC, Somley BL, Koch JK, Cohen LS. Treatment of depression and menopause-related symptoms with the serotonin-norepinephrine reuptake inhibitor duloxetine. J Clin Psychiatry. 2007;68(6):943–50.

    Article  CAS  PubMed  Google Scholar 

  149. Li S, Li ZF, Wu Q, Guo XC, Xu ZH, Li XB, Chen R, Zhou DY, Wang C, Duan Q, Sun J, Luo D, Li MY, Wang JL, Xie H, Xuan LH, Su SY, Huang DM, Liu ZS, Fu WB. A Multicenter, Randomized, Controlled trial of electroacupuncture for perimenopause women with mild-moderate depression. Biomed Res Int. 2018; 2018:5351210.

  150. Joffe H, Groninger H, Soares CN, Nonacs R, Cohen LS. An open trial of mirtazapine in menopausal women with depression unresponsive to estrogen replacement therapy. J Womens Health Gend Based Med. 2001;10(10):999–1004.

    Article  CAS  PubMed  Google Scholar 

  151. Kornstein SG, Clayton AH, Bao W, Guico-Pabia CJ. A pooled analysis of the efficacy of desvenlafaxine for the treatment of major depressive disorder in perimenopausal and postmenopausal women. J Womens Health (Larchmt). 2015;24(4):281–90.

    Article  PubMed  Google Scholar 

  152. Soares CN, Thase ME, Clayton A, Guico-Pabia CJ, Focht K, Jiang Q, Kornstein SG, Ninan PT, Kane CP. Open-label treatment with desvenlafaxine in postmenopausal women with major depressive disorder not responding to acute treatment with desvenlafaxine or escitalopram. CNS Drugs. 2011;25(3):227–38.

    Article  CAS  PubMed  Google Scholar 

  153. Soares CN, Frey BN, Haber E, Steiner M. A pilot, 8-week, placebo lead-in trial of quetiapine extended release for depression in midlife women: impact on mood and menopause-related symptoms. J Clin Psychopharmacol. 2010;30(5):612–15.

    Article  PubMed  Google Scholar 

  154. Ibrahim WW, Safar MM, Khattab MM, Agha AM. 17β-Estradiol augments antidepressant efficacy of escitalopram in ovariectomized rats: neuroprotective and serotonin reuptake transporter modulatory effects. Psychoneuroendocrinology. 2016;74:240–50.

    Article  CAS  PubMed  Google Scholar 

  155. Sasayama D, Sugiyama N, Yonekubo S, Pawlak A, Murasawa H, Nakamura M, Hayashi M, Ogawa T, Moro M, Washizuka S, Amano N, Hongo K, Ohnota H. Novel oestrogen receptor β-selective ligand reduces obesity and depressive-like behaviour in ovariectomized mice. Sci Rep. 2017;7(1):4663.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Schmidt PJ, Ben Dor R, Martinez PE, Guerrieri GM, Harsh VL, Thompson K, Koziol DE, Nieman LK, Rubinow DR. Effects of estradiol withdrawal on mood in women with past perimenopausal depression: a randomized clinical trial. JAMA Psychiatry. 2015;72(7):714–26.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Wium-Andersen MK, Jørgensen TSH, Halvorsen AH, Hartsteen BH, Jørgensen MB, Osler M. Association of hormone therapy with depression during menopause in a cohort of Danish women. JAMA Netw Open. 2022;5(11):e2239491.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Gordon JL, Rubinow DR, Eisenlohr-Moul TA, Xia K, Schmidt PJ, Girdler SS. Efficacy of transdermal estradiol and micronized progesterone in the prevention of depressive symptoms in the menopause transition: a randomized clinical trial. JAMA Psychiatry. 2018;75(2):149–57.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Kulkarni J, Gavrilidis E, Thomas N, Hudaib AR, Worsley R, Thew C, Bleeker C, Gurvich C. Tibolone improves depression in women through the menopause transition: a double-blind randomized controlled trial of adjunctive tibolone. J Affect Disord. 2018;236:88–92.

    Article  CAS  PubMed  Google Scholar 

  160. Zhang L, Li J, Chen Q, Di L, Li N. Erxian decoction, a famous Chinese medicine formula, ameliorate depression- like behavior in perimenopausal mice. Endocr Metab Immune Disord Drug Targets. 2021;21(12):2203–12.

    Article  CAS  PubMed  Google Scholar 

  161. Choi JE, Borkowski K, Newman JW, Park Y. N-3 PUFA improved post-menopausal depression induced by maternal separation and chronic mild stress through serotonergic pathway in rats-effect associated with lipid mediators. J Nutr Biochem. 2021;91:108599.

    Article  CAS  PubMed  Google Scholar 

  162. Park HJ, Shim HS, Park S, Shim I. Antidepressant effect and neural mechanism of Acer tegmentosum in repeated stress-induced ovariectomized female rats. Anim Cells Syst (Seoul). 2020;24(4):205–13.

    Article  CAS  PubMed  Google Scholar 

  163. Chen XQ, Chen SJ, Liang WN, Wang M, Li CF, Wang SS, Dong SQ, Yi LT, Li CD. Saikosaponin A attenuates perimenopausal depression-like symptoms by chronic unpredictable mild stress. Neurosci Lett. 2018;662:283–9.

    Article  CAS  PubMed  Google Scholar 

  164. Ushiroyama T, Ikeda A, Sakuma K, Ueki M. Chai-Hu-Gui-Zhi-Gan-Jiang-Tang regulates plasma interleukin-6 and soluble interleukin-6 receptor concentrations and improves depressed mood in climacteric women with insomnia. Am J Chin Med. 2005;33(5):703–11.

    Article  PubMed  Google Scholar 

  165. Nusslock R, Alloy LB, Brody GH, Miller GE. Annual research review: neuroimmune network model of depression: a developmental perspective. J Child Psychol Psychiatry. 2024.

Download references

Acknowledgements

Not applicable.

Funding

This research was supported by the Henan Provincial Science and Technology Research Projects (No. 232102210110 and 242102310555) to CT.

Author information

Authors and Affiliations

Authors

Contributions

Y.Z. and X.T. wrote the original draft. C.T supervised, reviewed and edited the manuscript. C.T also participated in funding acquisition. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Chaozhi Tang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

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.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

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, Y., Tan, X. & Tang, C. Estrogen-immuno-neuromodulation disorders in menopausal depression. J Neuroinflammation 21, 159 (2024). https://doi.org/10.1186/s12974-024-03152-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-024-03152-1

Keywords