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 [5,6,8,9,10,11]. 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 develo** 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,

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, 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 [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 [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.