Funding information: METAvivor, Grant/Award Number: research grant; National Cancer Institute, Grant/Award Number: R37 CA227984; U.S. Army Materiel Command, Grant/Award Number: W81XWH1910033; U.S. Department of Defense, Grant/Award Number: W81XWH-15-1-0352
Abstract
Background
While sex hormones and their receptors play well-known roles in progression of primary tumors through direct action on sex steroid hormone-responsive cancer cells, emerging evidence suggest that hormones also play important roles in metastatic progression by modulating the tumor microenvironment. Estrogens and androgens synthesized in gonads and within the brain influence memory, behavior, and outcomes of brain pathologies. Yet, their impact on brain metastatic colonization and progression is just beginning to be explored.
Recent findings
Estradiol and testosterone cross the blood-brain barrier and are synthesized de novo in astrocytes and other cells within the adult brain. Circulating and brain-synthesized estrogens have been shown to promote brain metastatic colonization of tumors lacking estrogen receptors (ERs), through mechanisms involving the upregulation of growth factors and neurotrophins in ER+ reactive astrocytes. In this review, we discuss additional mechanisms by which hormones may influence brain metastases, through modulation of brain endothelial cells, astrocytes, and microglia.
Conclusion
A greater understanding of hormone-brain-tumor interactions may shed further light on the mechanisms underlying the adaptation of cancer cells to the brain niche, and provide therapeutic alternatives modulating the brain metastatic niche.
1 INTRODUCTION
In the past decade, major advances have been made in understanding the cellular and molecular mechanisms that regulate brain metastasis (reviewed in 1). Specific interactions within the tumor microenvironment (TME) have emerged as key to the ability of cancer cells to colonize distant sites, including the brain.2-6 While some of the interactions between cancer cells and the microenvironment are common between primary tumors and metastases to various organs, cancer cells that reach the brain encounter a more isolated, highly reactive microenvironment protected by the blood-brain barrier (BBB), which blocks entry of many circulating molecules and cells found in other organs.7 It is now known that brain metastatic colonization is a relatively rare event in which circulating tumor cells are arrested at brain capillaries, extravasate into the brain parenchyma, survive anti-tumorigenic effects of brain immune-surveillance cells, including microglia and astrocytes, and colonize the brain niche by growing around existing vessels and adapting to the unique brain microenvironment.3, 8-11 Interaction of cancer cells with brain endothelial cells, glial cells, and pericytes are critical for the initial steps of metastatic colonization as well as outgrowth to large macrometastases, in a process that spans months to years from primary tumor diagnosis.
It is well recognized that sex hormones influence both normal and pathological brain functions (reviewed in 12-15), but until recently, how the hormonal milieu could influence brain metastases remained unexplored. While both males and females develop brain metastases, age and sex influence the metastatic incidence within specific tumor types. For example, lung brain metastases occur with similar frequency between men and women (18.9% vs 21.8%, respectively), while melanoma brain metastases occur more frequently in men than women (8.7% vs 4.8%, respectively).16 Younger age and female sex were reported as significant risk factors for subsequent brain metastasis in a subset of advanced non-small cell lung cancer cases.17 In predominantly female breast cancer, young age is a predictive factor for the development of brain metastasis independent of tumor subtype, suggesting age-dependent and host-specific factors promote metastases in younger women.18-20
Although the function of sex hormones (estrogens and androgens) in cancer progression has been best defined in terms of their pro-tumorigenic function on sex hormone-receptor expressing cancer cells (see key reviews on this topic21-25), their roles in the brain are more varied and complex. Sex hormones act throughout the entire brain of both males and females modulating many cellular and molecular processes, which in turn alter the structure and function of specific brain compartments. In this review, we will discuss the known roles of estrogens and androgens in brain function, and how they may impact brain metastasis through their action on key components of TME.
1.1 Peripheral synthesis of sex steroid hormones
The predominant circulating sex steroid hormones after puberty are estrogens in females and testosterone in males. In both sexes, the gonads and adrenal glands synthesize estrogens and androgens and release them into circulation. In females, three major forms of physiological estrogens are present: estrone (E1), estradiol (E2), and estriol (E3). Before menopause, E2 is the most potent circulating estrogen, while E1 is more important after menopause. E3 is the least potent estrogen, though it plays a larger role during pregnancy when it is produced in large quantities by the placenta.26
In premenopausal women estrogens are synthetized from cholesterol, mainly in the ovaries and during pregnancy by the placenta, acting as an endocrine factor to maintain ovulation and reproductive capability (reviewed in 27-29). The level of circulating estrogens depends upon the reproductive status of the individual and is highest during the reproductive years30 (Table 1). At menopause, circulating estrogen levels undergo a sustained drop, but androgen production experiences only a small, gradual decline by the ovaries and adrenal glands. After menopause, the ovaries maintain secretion of testosterone and androstenedione, which are converted to E2 and E1 in the breast and other tissues by aromatase enzyme (CYP19).33
TABLE 1.
Circulating levels of sex hormones in adults
Note: To facilitate comparisons, values originally reported as pg/mL, ng/mL, and ng/dL were converted to SI units (pM) using the following molecular weights: E1, 207.366 g/mol; E2, 272.29 g/mol; and Testosterone, 288.42 g/mol. *Values are mean ± SD. All others are median (range).
In males, the much greater levels of circulating testosterone produced by the mature testes generate and maintain the sexual phenotype (Table 1). However, testes also produce about ~20% of circulating estrogens, with the remainder from local production by adipose tissue, brain, skin, and bone, through conversion of testosterone to estrogen by aromatase.34, 36, 37 Estrogens have thus emerged as the active factors in mediating many of testosterone's effects on target tissues in adult males.37, 38
While peripheral sex hormone synthesis is generally well-regulated in healthy individuals, cytotoxic drugs and radiation therapies used to treat systemic and brain metastatic tumors can derail these processes in some patients. When radiotherapy is used to treat brain metastasis, inclusion of the hypothalamic-pituitary axis in the radiation fields can lead to neuroendocrine dysfunction. In a longitudinal trial of patients with brain gliomas, half of premenopausal women at study entry developed premature menopause, and 37% of men aged less than 50 years had low levels of testosterone.39 Similarly, cytotoxic drug treatments in premenopausal women can induce premature ovarian failure, with consequently altered synthesis of ovarian E2.40 Thus, sex hormone function alterations as a result of brain metastasis treatments could also play a role in the progression of brain metastasis.
2 CENTRAL SYNTHESIS OF SEX-STEROID HORMONES IN THE ADULT BRAIN
Because of their lipophilic natures, estradiol and testosterone can cross the BBB. However, the movement of these hormones across the BBB is thought to reflect the combined effects of their lipid solubility and the presence of circulating binding proteins such as albumin or sex-hormone binding globulins.41-45 Therefore, the levels of circulating hormones may not reflect local availability and function of sex hormones in the brain. Korneyev et al demonstrated that pregnenolone, the first steroid formed by mitochondrial oxidative cleavage of cholesterol, is synthesized in the forebrain, cerebellum, and olfactory bulb of adrenalectomized and castrated Sprague-Dawley male rats treated with trilostane (an inhibitor of pregnenolone metabolism from progesterone), demonstrating the functionality of brain cytochrome P450 side-chain cleavage (CYP11A1), as well as the ability of the brain to produce sex hormones de novo.46 In the brain, astrocytes are the most active steroidogenic cells (at least in murine models), expressing CYP11A1, 17alpha-hydroxylase/C17-20-lyase (CYP17), 3beta-hydroxysteroid dehydrogenase (3β-HSD), 17beta-hydroxysteroid dehydrogenase (17β-HSD), and cytochrome P450 aromatase (CYP19A1).47 Astrocytes have been shown to produce pregnenolone, progesterone, dehydroepiandrosterone (DHEA), androstenedione, testosterone, E2, and E1. Oligodendrocytes express only CYP11A1 and 3β-HSD to produce pregnenolone, progesterone, and androstenedione, but lack the enzymes necessary to produce DHEA, testosterone, or estrogens. Neurons express CYP11A1, CYP17, 3β-HSD, and CYP19A1 to produce pregnenolone, DHEA, androstenedione, and estradiol, but do not express 17β-HSD or produce testosterone47, 48 (Figure 1).
Steroid hormone synthesis by the mammalian brain. Steroid hormones are derived from cholesterol, and several cells within the brain niche possess all the enzymes required to synthesize sex-hormones de novo. Astrocytes and neuron express all enzymatic machinery StAR (Steroidogenic acute regulatory protein); CYP11A1 or P450scc (Cholesterol side-chain cleavage enzyme); 3β-HSD (3-beta-hydroxysteroid dehydrogenase); CYP17 (17-alpha-hydroxylase/17,20 lyase) and 17β-HDS (17β-Hydroxysteroid dehydrogenase) and aromatase to produce progesterone, testosterone, and E2 from cholesterol. Microglia express StAR and 17β-HDS and are able to synthetize testosterone and androstenodiol from androgenic C19-steroids. Oligodendrocytes express StAR /CYP11A1 and 3β-HSD to produce progesterone in the brain47, 49-53
Brain E2 biosynthesis from androgenic precursors (testosterone and androstenedione) by the aromatase enzyme has been recognized for several years.54-56 Early studies using 3H-testosterone demonstrated aromatase activity in homogenates of hypothalamus,57 amygdala, and hippocampus58 from human fetuses, and also other mammals.59-64 However, brain aromatase expression is regulated in a species- and region-selective manner. In humans, the regional distribution pattern of aromatase was found to be strikingly different from the distribution reported in non-human primates (baboon and rhesus monkey) and rodents.65, 66 Biegon et al used a radiolabeled aromatase ligand ([11C]vorozole) alone or in combination with the aromatase inhibitor letrozole to trace regional aromatase distribution by positron emission tomography, they reported a highly heterogeneous distribution of aromatase activity, with the highest levels in the thalamic nuclei, followed by moderately high levels in amygdala, preoptic area (POA), and medulla and low levels in cortex, putamen, cerebellum, and cortical white matter.65 Premenopausal women showed higher aromatase activity than postmenopausal women but brain uptake of C-vorozole did not vary across the menstrual cycle in pre-menopausal women,66 suggesting that brain E2 synthesis (or at least aromatase function) is not regulated by circulating levels of E2. Males showed increased brain aromatase activity compared to healthy females, supporting the notion that brain estrogens mediate brain-functions in males and females. Given that testosterone can also be reduced to dihydrotestosterone in the brain (Figure 1), and that dihydrotestosterone is a more potent androgen than testosterone,15 regulation of aromatase and 17β-HSD is likely to play key roles in defining the ultimate action of sex hormones in the male and female brain.
Neurons, astrocytes, and endothelial cells from rodents and humans express aromatase.38, 67-72 Increased brain aromatase expression or activity have been demonstrated following hypoxia and ischemia,73, 74 increased pressure,68 and mechanically or chemically induced brain injuries in male and female rodents.75 Similarly, aromatase upregulation appears to be a response to reactive gliosis initiated by brain metastases, as aromatase can be detected in reactive astrocytes from resected breast cancer brain metastasis (unpublished data). Recent studies showed that ovariectomy combined with the aromatase inhibitor Letrozole was more effective in blocking brain metastatic colonization in an experimental model of estrogen-unresponsive (triple negative) breast cancer brain metastasis, compared to ovariectomy alone.76, 77 This provided the first pre-clinical evidence that both ovarian and peripheral E2 contribute to brain metastatic colonization by acting on the brain microenvironment rather than the tumor itself. However, whether aromatase inhibition could decrease metastatic colonization by other primary tumors, and whether this function is similar in males and females, remains unclear.
3 SEX HORMONE SIGNALING IN BRAIN METASTASIS: DIRECT ACTION ON CANCER CELLS
Direct mechanisms of sex hormone function on cancer cells have been extensively described in the context of hormone receptor-positive (HR+) breast and prostate cancer (reviewed in 24, 78, and 79), and similar mechanisms likely impact HR+ growth in the brain (Figure 2A,B). In this section, we will briefly review mechanisms of direct hormone signaling in cancer cells, followed by evidence linking hormone receptor function to brain metastasis progression of HR+ brain metastasis. In Figure 2, we provide an integrated summary of how sex hormones acting directly on cancer cells and the brain niche may participate in brain metastasis.
Mechanisms by which sex hormones may influence brain metastasis. A, Direct action of local estrogens is likely to influence proliferation of ER+ breast cancer brain metastasis. B, Direct action of androgens on AR+ brain metastasis can have proliferative and anti-proliferative effects in tumor cells. C, Estrogen may influence the ability of metastatic cancer cells to form pseudosynapses with neurons. D, Estrogens can modulate BBB permeability by regulating endothelial tight junctions and pro-angiogenic factors (VEGF, HIF). E, E2 acts through ER+ reactive astrocyte to increase secretion of growth factors that activate canonical oncogenic epidermal growth factor receptor (EGFR) and TRKB on brain metastatic cancer cells. F, Estradiol may promote microglia polarization and recruitment of immunosuppressive cells to the brain niche
Estrogens act via estrogen receptors (ERs, ERα, and ERβ) or through G protein-coupled ER1 (GPER1, also known as GPR30).80, 81 Estrogen binding to these receptors initiates classic nuclear-initiated steroid signaling (NISS, also known as genomic signaling) and membrane-initiated steroid signaling (MISS, previously known as non-genomic signaling).82 During NISS, E2 binding triggers intracellular localization of ERα and ERβ, which dimerize and enter the nucleus, binding to E2 response elements (EREs), or activator protein-1 (Ap1) and specificity protein-1 (Sp1), on the promoter of E2 responsive genes to regulate transcription. In the MISS pathway, E2 binds to membrane-bound ERα and ERβ receptors as well as GPR30 to rapidly activate nuclear transcription factors via the MAPK pathway.56, 83-85 By contrast, androgens bind to one androgen receptor (AR) and signal through NISS.86 The AR is primarily located in the cytoplasm. Following ligand-binding, AR translocates to the nucleus to bind androgen response elements. This enables recruitment of histone acetyltransferases, coactivators, and other proteins crucial for transcriptional machinery.87-89 Some studies suggest that MISS can be induced by testosterone; however, whether this occurs via membrane-bound AR or an unknown receptor remains unclear.83, 90, 91
Estrogens are well known to play a mitogenic role in breast cancer via direct activation of ERα and endocrine therapy (ie, Selective ER Modulators, ER-antagonists, and aromatase inhibitors) are the main form of targeted therapy for these tumors. The direct role for estrogens on brain metastasis of ER+ breast tumors has received less attention, due to the clinical observations that (a) the incidence of brain metastasis is lowest in patients with ER+ breast tumors compared to breast tumors lacking ER (ie, HER2+ or Triple Negative Breast cancer, TNBC)92, 93; (b) receptor conversion, particularly loss of hormone receptors, is a common event in brain metastases from breast cancer, and endocrine therapy may increase its incidence94-96; and (c) metastatic disease in ER+ disease is often a late life event occurring mostly in post-menopausal women with low levels of circulating estrogens. Yet, around 11% of ER + Her2- and 15% of Her2+ ER+ patients with metastatic breast cancer develop brain metastases,97 suggesting that extragonadal hormone production in the brain and other organs might drive the slow but eventual development of hormone-dependent metastases in postmenopausal women. Supporting this hypothesis, a recent study investigating the impact of endocrine therapy after diagnosis of brain metastasis on outcome and clinical course of disease in patients with ER+ metastatic breast cancer, found that continuing endocrine therapy after brain metastasis diagnosis was associated with a significantly prolonged overall survival.98 Unfortunately, models of brain metastatic ER+ breast cancer are limited, and ER+ breast cancer cells commonly used to study primary tumor growth in xenograft models require estrogen supplementation to mimic estrogen levels found in humans. Thus, a model of ER+ brain metastasis that grows in a postmenopausal setting common to ER+ breast cancer brain metastasis remains a challenge.
Androgens also play a role in breast, melanoma, and lung cancer primary tumor growth, and may impact their metastasis through similar mechanisms.24, 25, 99, 100 AR is expressed in several types of lung cancer, including small cell, adenocarcinoma, and squamous cell carcinoma,100 and dihydrotestosterone (DHT) induces a proliferative response in lung cancer cells through cross-talk of AR and EGFR.99 In breast cancer, 60% to 90% of human breast tumors express AR,101, 102 though biological effects of androgens vary in different breast cancer models,103-105 with both anti-proliferative106-109 and proliferative effects reported.110, 111 However, the extent to which androgen/AR function plays a direct role on brain metastases from breast or other tumors remains to be defined.
4 SEX HORMONE SIGNALING IN BRAIN METASTASIS: INDIRECT ACTION ON THE BRAIN NICHE
Androgens regulate a wide array of neural functions, from reproduction to mood and cognitive abilities.91, 112 In the human brain, nuclear and cytoplasmic AR immunoreactivity has been shown in frozen and paraffin-embedded sections of the temporal cortex, specifically in neurons, astrocytes, oligodendrocytes, and microglia cells.113 In rodents, both male and female brains show strong AR activity in specific regions of the brain including the cerebral cortex, thalamus, and pituitary gland.114
Estrogens act via ERα and ERβ and signal through NISS and MISS.115, 116 ERs are expressed widely in different brain regions and cells, mediating E2-signaling in a cell type and region specific manner.117 The expression of ERα is strongly in brain regions such as the POA, bed nucleus stria terminalis (BNST), amygdala, periventricular nucleus, ventrolateral part of the hypothalamic ventromedial nucleus, and the arcuate nucleus. Likewise, ERβ is found in many of the same regions than ERα but is more highly expressed in the BNST, POA, paraventricular nucleus of the hypothalamus, and supraoptic nuclei, with some variation across species. ERα and ERβ are also found in other brain regions including the hippocampus, midbrain, cortex, diagonal band of Broca, and basal nucleus of Meynert (reviewed in 117). High GPR30 expression has been shown in the hypothalamic-pituitary axis, hippocampal formation, and brainstem autonomic nuclei.118 There is evidence that GPR30 receptors are two to four times more abundant than ERα or ERβ in the prefrontal cortex of the rat.119
Expression of specific ERs varies during aging120 and also during brain pathology. For example, brain ischemia models demonstrate a twofold to threefold increase in ERα, whereas ERβ expression in neurons decreases.121, 122 Importantly, ERα, ERβ, and/or GPR30 are expressed in key subcellular compartments within the brain metastatic niche, including neurons, endothelial cells, astrocytes, and microglia.123, 124 In the next section, we explore the mechanisms by which sex hormones can influence these cell types and their potential impact on brain metastases. Given the difficulty in differentiating between the effects of T vs those of brain-produced E2, we will focus on the better-understood roles of estrogen in regulating neuronal function, the BBB, and neuro inflammation that could influence brain metastasis.
4.1 Hormonal regulation of neuronal function
Recent studies have demonstrated that brain metastatic growth of breast cancer cells depends on their ability to form synapses with glutamatergic neurons.125 Breast cancer lines selected for their proficiency in causing brain metastases showed high expression of phospho-GluN2B and other subunits of N-methyl-D-aspartate receptor (NMDAR), as well as NMDAR-mediated currents and calcium transients in response to glutamate or NMDA.126 Similar findings in glioma cells127, 128 suggest that the existence of synaptic structures involving presynaptic neuros and tumor cells post-synapsis may be common to brain metastasis and primary brain tumors. Hormones could influence this newly recognized “synaptic” interaction of cancer cells with neurons.
For example, estrogen plays important roles in cognitive function, mediated by its ability to rapidly enhance excitatory synaptic transmission, especially via NMDAR-mediated synaptic activity and long-term potentiation (LTP).129-133 Estrogen promotes the formation of new dendritic spines and excitatory synapses in the hippocampus and cortex134 via the activation of membrane ERs,135 which subsequently enhance NMDAR transmission and LTP.136, 137 Acute E2 treatment results in the recruitment of postsynaptic density protein 95 (PSD-95) to novel dendritic spines, and the NMDA receptor subunit GluN1 is recruited to nascent synapses in cortical neurons.138 Androgens and the receptor have also been shown to play a role in remodeling of spine synapsis in limbic brain areas,139 suggesting that both estrogens and androgens could influence the ability of cancer cells to form synapses in some brain regions. Future studies should address the extent to which estrogen and potentially androgens modify the ability of cancer cells to form pseudosynapses.
4.2 Hormonal regulation of brain endothelial function
Brain endothelial cells are the first cell-type encountered by disseminated cancer cells at metastatic sites, and studies have shown that cancer cells produce a variety of molecules that destabilize endothelial-tight junctions and allow extravasation. Brain endothelial cells express ERs and E2 is a well-known regulator of endothelial cell function,140 however, a comprehensive characterization of the effects of E2 on brain endothelial cells and its impact on the blood-tumor barrier are largely unknown.
E2 has been shown to promote primary tumor progression and metastasis by increasing intratumoral vessel density and improving vessel stabilization to prevent tumor hypoxia and necrosis,141 thus it could be predicted that similar pro-tumorigenic effects take place at the blood-tumor barrier. Since brain metastases have been shown to grow in close attachment to existing vessels (vessel co-option), it is likely that estrogenic function in brain endothelial cells plays a role regulating cell migration and intravasation of circulating cancer cells through the brain parenchyma and outgrowth of micrometastases. However, E2 regulates endothelial cells and BBB function in manners that would predict pro-metastatic and anti-metastatic effects. For example, E2 induces vasodilatation by increasing nitric oxide synthesis through ERα-dependent MISS, activation of Pi3K/AKT pathway and GPR30 signaling.142-145 Increased vascular permeability and vasodilation at early stages of brain metastases may promote brain metastasis by increasing tumor blood flow. On the other hand, estrogens decrease BBB permeability by directly regulating expression of tight junction proteins such as claudin 5.146 In a mouse model of lipopolysaccharide (LPS)-induced inflammation, ovariectomy alone did not affect the degree of basal Evans blue dye extravasation, but the presence of LPS (3 mg/kg body weight i.p. assessed 4 hours post-injection) significantly enhanced paracellular permeability.147 Thus, increased E2 levels could predict a decrease in paracellular permeability that would oppose cancer cell extravasation and brain colonization. How paracrine effects of E2 on brain endothelial function may impact brain metastases remains to be addressed experimentally.
4.3 Hormonal regulation of neuroinflammation
4.3.1 Direct effects on astrocytes
Interactions of metastatic cells with astrocytes occur at both early and late stages of the colonization process and play pro-tumorigenic as well as anti-tumorigenic role. Induction of astrogliosis (activation of astrocytes and microglia) is an early event during metastatic colonization.148 Valiente et al proposed that early contacts between tumor cells and astrocytes result in tumor cell death of the majority of tumor cells reaching brain, and only a subset of cells can adapt to avoid this pro-apoptotic fate.10 Both testosterone and estrogens decrease astrogliosis elicited by various brain insults in animal models. For example, early and delayed administration of testosterone or E2 resulted in a significant decrease in the number of vimentin-immunoreactive astrocytes as well as reactive microglia in a model of brain injury in rodents.149 Thus, it is tempting to speculate that increased local levels of estrogens (ie, in brains of younger females and males) or testosterone (in male brains) could suppress early cancer-cell elicited astrogliosis and favor the survival of disseminated tumor cells.
Accumulating evidence shows that reactive astrocytes switch from a tumor-suppressive stage to a tumor-promoting role at later steps of the metastatic cascade and that astrogliosis is exploited by the tumor cells to support their growth.5, 150-152 Reactive astrocytes surrounding human brain metastasis as well as experimental brain metastasis models express ERs, and E2-treated astrocytes have been shown to activate key pro-metastatic pathways in cancer cells to promote brain metastatic colonization.76, 77 For example, E2 was shown to upregulate brain-derived neurotrophic factor (BDNF) and epidermal growth factor in ER+ astrocytes, which then activate their cognate receptors tropomyosin receptor kinase B (TrkB) and EGFR in breast cancer cells, promoting tumor-initiating capability, migration and invasion. These studies provided a molecular mechanism by which E2 can influence oncogenic signaling in cancer cells to promote brain colonization. Importantly, testosterone has been shown to increase BDNF expression in hippocampal neurons,153 suggesting that upregulation of BDNF by androgens may impact metastatic colonization in males as well. This can be particularly relevant for brain metastasis derived from primary lung cancer, which is often dependent on tyrosine receptor kinase signaling.154
It has been shown that the formation of gap junctions between astrocytes and tumor cells through connexin 43 (CX43), allows the passage of cyclic guanosine monophosphate-adenosine monophosphate, which activates the stimulator of interferon genes (STING) pathway in astrocytes and promotes expression of IFNα and TNFα to further facilitate brain metastatic growth.155 Interestingly, hypothalamic CX43 expression is regulated by steroid hormones in a brain region-specific and sexually dimorphic manner. Estrogen alone or in combination with progesterone significantly increased CX43 protein levels in some regions of the female rat brain, while both hormones significantly decreased CX43 levels in equivalent regions of male rat brains.156 Therefore, gap junctional communication with astrocytes can differentially influence brain metastatic colonization in males and females under various hormonal stages.
Immunosuppressive mechanisms are vital to the survival and outgrowth of disseminated cancer cells157 and astrocytes play key roles in the modulation of innate and acquired immunity in brain metastases.3, 158 Consistent with pro and anti-tumorigenic roles of astrocytes in brain metastatic progression, estrogens can induce pro-inflammatory (tumor suppressive) and immune-suppressive (tumor promoting) features in astrocytes. Physiological and pharmacological concentrations of E2 exhibit potent anti-inflammatory activity in the central nervous system (CNS) by suppressing production of pro-inflammatory cytokines, such as IL-6, IL-1β, and TNFα in many neurological disorders.159 However, many studies that address the anti-inflammatory properties of E2 in the brain and periphery have yielded different results depending on many factors, including the disease model, species, experimental outcome, whether the study is in vivo or in vitro, estrogenic formulation, and concentration of E2 or other estrogens.160-162 In the brain, activation of ERα by E2, directly repressed NFκβ-dependent transcription and suppressed TNFα-induced NFκB recruitment to the CCL2 enhancer in reactive astrocytes, suppressing astrocytic CCL2 production in a model of experimental allergic encephalomyelitis.163-165 In contrast, E2 did not attenuate CCL2 levels in a model of LPS-induced neuroinflammation.166 Given that CCL2-expressing astrocytes mediate the extravasation of T lymphocytes in the brain, and that CCL2 facilitates the process of both migration and infiltration of several cell systems such as monocytes, natural killer cells, T lymphocytes, and memory cells,167 defining how E2 modulates astrocytic CCL2 expression during different stages of metastatic colonization may shed light into the specific contribution of E2 to the transition of a tumor-suppressive early brain niche to a tumor-promoting immunosuppressive late brain niche.
4.3.2 Direct effects on microglia
Additional to their effects on astrocytes, sex hormones can also directly influence microglia activation and polarization states. Following brain lesions, microglia become active and assume an amoeboid phenotype and a high metabolic rate, synthesizing and secreting several cytokines, such as interleukin IL6, IL1β, and TNFα.168, 169 E2 also inhibited microglia activation via GRP30 in a model of ischemic stroke.170 In ovariectomized rats, low-dose E2 profoundly suppressed microglia activation and quantitatively shifted microglia from their “activated,” amoeboid morphology to a “resting” state. Further studies using previously defined nomenclature to classify macrophage polarization states into pro-inflammatory, anti-tumor M1 macrophages, and immune-suppressive, tumor-promoting M2 macrophages, showed that E2 robustly suppressed the “pro-inflammatory” M1 phenotype, while enhancing the “anti-inflammatory/immune-suppressive” M2 microglia phenotype in the hippocampus after global cerebral ischemia.171In vitro, E2 acts via estrogen receptor β (ERβ) to enhance the phagocytic clearance of apoptotic cells, and stimulation of either ERβ or GPR30 promoted the adoption of an anti-inflammatory/immune suppressive phenotype.172, 173 However, in a model of endotoxin-induced brain inflammation, expression of genes encoding key cytokines involved in the transfer from the innate to adaptive immunity (TLR2, TNF-α, and IL-12) in microglial cells was largely inhibited in the brain of ovariectomized mice at 24 hours post-challenge, and E2 rescues this effect in an manner dependent on ERα.174 Specific studies addressing how hormones alter microglial function in the context of brain metastases are necessary to fully understand their contribution to the pathogenesis of brain metastasis.
4.3.3 Effects on systemic immune infiltration in the brain
Recent studies have shown that E2 plays a pro-tumorigenic role in primary tumors that lack ERs through driving the mobilization of myeloid-derived suppressor cells (MDSCs) and enhancing their intrinsic immunosuppressive activity in vivo.175-177 This effect of E2 appears to involve direct as well as paracrine function of E2 on MDSC. Direct E2 binding to ERα activates the STAT3 pathway in human and mouse bone marrow myeloid precursors by enhancing JAK2 and SRC activity,175 and directly enhances both the expansion and suppressive activity of M-MDSCs.178 Indirectly, estrogen stimulates cancer-associated fibroblasts to secrete SDF-1α, which can recruit tumor-infiltrating MDSCs to the TME.179 Additional to effects on MDSCs, E2 can increase the immune tolerance of tumors by promoting proliferation of the immunosuppressive CD4+ CD25+ Treg phenotype,180 and increasing FOXP3 expression in Treg via ERα.181 However, there are no reports defining whether similar mechanisms play a role in the context of brain metastasis, particularly in early stages of brain metastatic colonization where the BBB is still intact and traffic of MDSCs to the brain niche might be limited.
5 CONCLUDING REMARKS
While the critical role of the brain microenvironment in brain metastases is now well recognized, the complexity of the interactions between cancer cells and specialized cells within the brain niche adds to the challenge of identifying effective and safe therapeutic strategies to target the pro-metastatic events occurring within the brain niche. Preclinical data using ER- breast cancer models in female mice suggest that sex-hormones, particularly E2, plays a pro-metastatic function at least in this subset of tumors. While the mechanisms by which E2 promotes metastasis remain to be fully elucidated, a promising therapeutic strategy may involve the use of estrogen-depletion therapies including aromatase inhibitors, which are already FDA-approved for the treatment of ER+ breast cancers. Given the pleiotropic roles of estrogens in normal brain functioning, it is critically important to balance the antitumoral benefits of such therapies with the possible mood and cognitive impairments associated with brain estrogen-deprivation. Additional studies are needed to define whether aromatase inhibition has therapeutic value in males and in brain metastases from lung cancer and melanoma, and to further elucidate the mechanisms by which sex-hormones alter early and late stages of brain metastatic colonization.
ETHICAL STATEMENT
Not Applicable.
ACKNOWLEDGMENTS
M.J.C. is supported by Department of Defense Breast Cancer Research Program - Breakthrough Fellowship Award No. W81XWH1910033. D.C. is supported by R37 R37 CA227984, DoD BCRP W81XWH-15-1-0352, and the Metavivor Research Foundation.
CONFLICT OF INTEREST
The authors have stated explicitly that there are no conflicts of interest in connection with this article.
AUTHOR CONTRIBUTIONS
All authors had full access to the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Conceptualization, M.J.C. and D.C.; Formal Analysis, M.J.C. and D.C; Writing - Original Draft, M.J.C. and D.C; Writing - Review & Editing, M.J.C. and D.C.; Funding Acquisition, M.J.C. and D.C
3Priego N, Zhu L, Monteiro C, et al. STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis. Nat Med. 2018; 24: 1024-1035. https://doi.org/10.1038/s41591-018-0044-4.
5Schwartz H, Blacher E, Amer M, et al. Incipient melanoma brain metastases instigate astrogliosis and neuroinflammation. Cancer Res. 2016; 76: 4359-4371. https://doi.org/10.1158/0008-5472.CAN-16-0485.
8Bos PD, Zhang XHF, Nadal C, et al. Genes that mediate breast cancer metastasis to the brain. Nature. 2009; 459: 1005-1009. https://doi.org/10.1038/nature08021.
9Lorger M, Felding-Habermann B. Capturing changes in the brain microenvironment during initial steps of breast cancer brain metastasis. Am J Pathol. 2010; 176: 2958-2971. https://doi.org/10.2353/ajpath.2010.090838.
10Valiente M, Obenauf AC, Jin X, et al. Serpins promote cancer cell survival and vascular co-option in brain metastasis. Cell. 2014; 156: 1002-1016. https://doi.org/10.1016/j.cell.2014.01.040.
11Neman J et al. Human breast cancer metastases to the brain display GABAergic properties in the neural niche. Proc Natl Acad Sci U S A. 2014; 111: 984-989. https://doi.org/10.1073/pnas.1322098111.
12Hyer MM, Phillips LL, Neigh GN. Sex differences in synaptic plasticity: hormones and beyond. Front Mol Neurosci. 2018; 11: 266. https://doi.org/10.3389/fnmol.2018.00266.
17Lee JS, Hong JH, Sun DS, et al. The impact of systemic treatment on brain metastasis in patients with non-small-cell lung cancer: a retrospective nationwide population-based cohort study. Sci Rep. 2019; 9:18689. https://doi.org/10.1038/s41598-019-55150-6.
18Hung MH, Liu CY, Shiau CY, et al. Effect of age and biological subtype on the risk and timing of brain metastasis in breast cancer patients. PLoS ONE. 2014; 9:e89389. https://doi.org/10.1371/journal.pone.0089389.
19Xue J, Peng G, Yang J-S, Ding Q, Cheng J. Predictive factors of brain metastasis in patients with breast cancer. Med Oncol. 2013; 30:337. https://doi.org/10.1007/s12032-012-0337-2.
20Brufsky AM, Mayer M, Rugo HS, et al. Central nervous system metastases in patients with HER2-positive metastatic breast cancer: incidence, treatment, and survival in patients from registHER. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2011; 17: 4834-4843. https://doi.org/10.1158/1078-0432.CCR-10-2962.
21Carroll JS, Hickey TE, Tarulli GA, Williams M, Tilley WD. Deciphering the divergent roles of progestogens in breast cancer. Nat Rev Cancer. 2017; 17: 54-64. https://doi.org/10.1038/nrc.2016.116.
22Deblois G, Giguere V. Oestrogen-related receptors in breast cancer: control of cellular metabolism and beyond. Nat Rev Cancer. 2013; 13: 27-36. https://doi.org/10.1038/nrc3396.
23Christopoulos PF, Vlachogiannis NI, Vogkou CT, Koutsilieris M. The role of the androgen receptor signaling in breast malignancies. Anticancer Res. 2017; 37: 6533-6540. https://doi.org/10.21873/anticanres.12109.
24Kono M, Fujii T, Lim B, Karuturi MS, Tripathy D, Ueno NT. Androgen receptor function and androgen receptor-targeted therapies in breast cancer: a review. JAMA Oncol. 2017; 3: 1266-1273. https://doi.org/10.1001/jamaoncol.2016.4975.
25Wang Y et al. Androgen receptor promotes melanoma metastasis via altering the miRNA-539-3p/USP13/MITF/AXL signals. Oncogene. 2017; 36: 1644-1654. https://doi.org/10.1038/onc.2016.330.
26Cui J, Shen Y, Li R. Estrogen synthesis and signaling pathways during aging: from periphery to brain. Trends Mol Med. 2013; 19: 197-209. https://doi.org/10.1016/j.molmed.2012.12.007.
29Barakat R, Oakley O, Kim H, Jin J, Ko CJ. Extra-gonadal sites of estrogen biosynthesis and function. BMB Rep. 2016; 49: 488-496. https://doi.org/10.5483/bmbrep.2016.49.9.141.
30Stricker R et al. Establishment of detailed reference values for luteinizing hormone, follicle stimulating hormone, estradiol, and progesterone during different phases of the menstrual cycle on the Abbott ARCHITECT analyzer. Clin Chem Lab Med. 2006; 44: 883-887. https://doi.org/10.1515/CCLM.2006.160.
31Skiba MA, Bell RJ, Islam RM, Handelsman DJ, Desai R, Davis SR. Androgens during the reproductive years, what's normal for women? J Clin Endocrinol Metab.2019:5382-5392. https://doi.org/10.1210/jc.2019-01357.
32Onizuka Y, Nagai K, Ideno Y, et al. Association between FSH, E1, and E2 levels in urine and serum in premenopausal and postmenopausal women. Clin Biochem.2019;73:105-108. https://doi.org/10.1016/j.clinbiochem.2019.08.009.
33Kotsopoulos J et al. The relationship between bilateral oophorectomy and plasma hormone levels in postmenopausal women. Hormones Cancer. 2015; 6: 54-63. https://doi.org/10.1007/s12672-014-0209-7.
38Yague JG, Muñoz A, de Monasterio-Schrader P, DeFelipe J, Garcia-Segura LM, Azcoitia I. Aromatase expression in the human temporal cortex. Neuroscience. 2006; 138: 389-401. https://doi.org/10.1016/j.neuroscience.2005.11.054.
39Handisurya A, Rumpold T, Caucig-Lütgendorf C, et al. Are hypothyroidism and hypogonadism clinically relevant in patients with malignant gliomas? A longitudinal trial in patients with Glioma. Radiother Oncol J Eur Soc Therapeut Radiol Oncol. 2019; 130: 139-148. https://doi.org/10.1016/j.radonc.2018.10.014.
41Gebhart VM, Caldwell JD, Rodewald A, Kalyvianaki K, Kampa M, Jirikowski GF. Estrogen receptors and sex hormone binding globulin in neuronal cells and tissue. Steroids. 2019; 142: 94-99. https://doi.org/10.1016/j.steroids.2018.06.015.
42Xargay-Torrent S et al. Circulating sex hormone binding globulin: an integrating biomarker for an adverse cardio-metabolic profile in obese pregnant women. PLoS ONE. 2018; 13:e0205592. https://doi.org/10.1371/journal.pone.0205592.
43Watanabe S, Tani T, Watanabe S, Seno M. Transport of steroid hormones facilitated by serum proteins. Biochim Biophys Acta. 1991; 1073: 275-284. https://doi.org/10.1016/0304-4165(91)90132-z.
44Diotel N, Charlier TD, Lefebvre d'Hellencourt C, et al. Steroid transport, local synthesis, and signaling within the brain: roles in neurogenesis, neuroprotection, and sexual behaviors. Front Neurosci. 2018; 12: 84-84. https://doi.org/10.3389/fnins.2018.00084.
45Pardridge WM, Mietus LJ. Transport of steroid hormones through the rat blood-brain barrier. Primary role of albumin-bound hormone. J Clin Invest. 1979; 64: 145-154. https://doi.org/10.1172/JCI109433.
46Korneyev A, Pan BS, Polo A, Romeo E, Guidotti A, Costa E. Stimulation of brain pregnenolone synthesis by mitochondrial diazepam binding inhibitor receptor ligands in vivo. J Neurochem. 1993; 61: 1515-1524. https://doi.org/10.1111/j.1471-4159.1993.tb13647.x.
47Zwain IH, Yen SS. Neurosteroidogenesis in astrocytes, oligodendrocytes, and neurons of cerebral cortex of rat brain. Endocrinology. 1999; 140: 3843-3852. https://doi.org/10.1210/endo.140.8.6907.
48Martini L, Melcangi RC, Maggi R. Androgen and progesterone metabolism in the central and peripheral nervous system. J Steroid Biochem Mol Biol. 1993; 47: 195-205. https://doi.org/10.1016/0960-0760(93)90075-8.
49Gottfried-Blackmore A, Sierra A, Jellinck PH, McEwen BS, Bulloch K. Brain microglia express steroid-converting enzymes in the mouse. J Steroid Biochem Mol Biol.2008;109:96-107.
50Giatti S, Boraso M, Melcangi RC, Viviani B. Neuroactive steroids, their metabolites, and neuroinflammation. J Mol Endocrinol. 2012;49:R125-R134. https://doi.org/10.1530/jme-12-0127.
51González-Orozco JC, Camacho-Arroyo I. Progesterone actions during central nervous system development. Front Neurosci. 2019;13:503-503. https://doi.org/10.3389/fnins.2019.00503.
53Sinchak K, Mills RH, Tao L, LaPolt P, Lu JKH, Micevych P. Estrogen induces de novo progesterone synthesis in astrocytes. Dev Neurosci. 2003;25:343-348.
57Naftolin F, Ryan KJ, Petro Z. Aromatization of androstenedione by the diencephalon. J Clin Endocrinol Metab. 1971; 33: 368-370. https://doi.org/10.1210/jcem-33-2-368.
58Naftolin F, Ryan KJ, Petro Z. Aromatization of androstenedione by limbic system tissue from human foetuses. J Endocrinol. 1971; 51: 795-796. https://doi.org/10.1677/joe.0.0510795.
59MacLusky NJ, Naftolin F, Goldman-Rakic PS. Estrogen formation and binding in the cerebral cortex of the developing rhesus monkey. Proc Natl Acad Sci U S A. 1986; 83: 513-516. https://doi.org/10.1073/pnas.83.2.513.
61Sholl SA, Goy RW, Kim KL. 5 alpha-reductase, aromatase, and androgen receptor levels in the monkey brain during fetal development. Endocrinology. 1989; 124: 627-634. https://doi.org/10.1210/endo-124-2-627.
62Milewich L, George FW, Wilson JD. Estrogen formation by the ovary of the rabbit embryo. Endocrinology. 1977; 100: 187-196. https://doi.org/10.1210/endo-100-1-187.
64Selmanoff MK, Brodkin LD, Weiner RI, Siiteri PK. Aromatization and 5alpha-reduction of androgens in discrete hypothalamic and limbic regions of the male and female rat. Endocrinology. 1977; 101: 841-848. https://doi.org/10.1210/endo-101-3-841.
65Biegon A et al. Unique distribution of aromatase in the human brain: in vivo studies with PET and [N-methyl-11C]vorozole. Synapse (New York, N.Y.). 2010; 64: 801-807. https://doi.org/10.1002/syn.20791.
66Biegon A et al. Aromatase imaging with [N-methyl-11C]vorozole PET in healthy men and women. J Nucl Med. 2015; 56: 580-585. https://doi.org/10.2967/jnumed.114.150383.
67Yague JG, Azcoitia I, DeFelipe J, Garcia-Segura LM, Munoz A. Aromatase expression in the normal and epileptic human hippocampus. Brain Res. 2010; 1315: 41-52. https://doi.org/10.1016/j.brainres.2009.09.111.
68Gatson JW, Simpkins JW, Yi KD, Idris AH, Minei JP, Wigginton JG. Aromatase is increased in astrocytes in the presence of elevated pressure. Endocrinology. 2011; 152: 207-213. https://doi.org/10.1210/en.2010-0724.
69Azcoitia I, Sierra A, Veiga S, Garcia-Segura LM. Aromatase expression by reactive astroglia is neuroprotective. Ann N Y Acad Sci. 2006; 1007: 298-305. https://doi.org/10.1196/annals.1286.028.
71Zuloaga KL, Davis CM, Zhang W, Alkayed NJ. Role of aromatase in sex-specific cerebrovascular endothelial function in mice. Am J Physiol Heart Circ Physiol. 2014; 306: H929-H937. https://doi.org/10.1152/ajpheart.00698.2013.
75Garcia-Segura LM et al. Aromatase expression by astrocytes after brain injury: implications for local estrogen formation in brain repair. Neuroscience. 1999; 89: 567-578. https://doi.org/10.1016/s0306-4522(98)00340-6.
76Sartorius CA, Hanna CT, Gril B, et al. Estrogen promotes the brain metastatic colonization of triple negative breast cancer cells via an astrocyte-mediated paracrine mechanism. Oncogene. 2015; 35: 2881-2892. https://doi.org/10.1038/onc.2015.353.
77Contreras-Zarate MJ et al. Estradiol induces BDNF/TrkB signaling in triple-negative breast cancer to promote brain metastases. Oncogene. 2019; 38: 4685-4699. https://doi.org/10.1038/s41388-019-0756-z.
79Yip CH, Rhodes A. Estrogen and progesterone receptors in breast cancer. Future Oncology (London, England). 2014; 10: 2293-2301. https://doi.org/10.2217/fon.14.110.
81Razandi M, Pedram A, Greene GL, Levin ER. Cell membrane and nuclear estrogen receptors (ERs) originate from a single transcript: studies of ERalpha and ERbeta expressed in Chinese hamster ovary cells. Mol Endocrinol. 1999; 13: 307-319. https://doi.org/10.1210/mend.13.2.0239.
83Boonyaratanakornkit V et al. Extranuclear signaling by sex steroid receptors and clinical implications in breast cancer. Molecular and Cellular Endocrinology. 2018; 466: 51-72. https://doi.org/10.1016/j.mce.2017.11.010.
84Björnström L, Sjöberg M. Mechanisms of estrogen receptor signaling: convergence of genomic and nongenomic actions on target genes. Mol Endocrinol. 2005; 19: 833-842. https://doi.org/10.1210/me.2004-0486.
88Jaiswal B, Gupta A. Modulation of nuclear receptor function by chromatin modifying factor TIP60. Endocrinology. 2018; 159: 2199-2215. https://doi.org/10.1210/en.2017-03190.
89Wang Q, Carroll JS, Brown M. Spatial and temporal recruitment of androgen receptor and its coactivators involves chromosomal looping and polymerase tracking. Molecular Cell. 2005; 19: 631-642. https://doi.org/10.1016/j.molcel.2005.07.018.
93Witzel I, Oliveira-Ferrer L, Pantel K, Müller V, Wikman H. Breast cancer brain metastases: biology and new clinical perspectives. Breast Cancer Res. 2016; 18: 8. https://doi.org/10.1186/s13058-015-0665-1.
94Duchnowska R et al. Conversion of epidermal growth factor receptor 2 and hormone receptor expression in breast cancer metastases to the brain. Breast Cancer Research: BCR. 2012; 14: R119. https://doi.org/10.1186/bcr3244.
95Bachmann C, Grischke EM, Staebler A, Schittenhelm J, Wallwiener D. Receptor change-clinicopathologic analysis of matched pairs of primary and cerebral metastatic breast cancer. J Cancer Res Clin Oncol. 2013; 139: 1909-1916. https://doi.org/10.1007/s00432-013-1511-4.
96Jung J, Lee SH, Park M, et al. Discordances in ER, PR, and HER2 between primary breast cancer and brain metastasis. J Neurooncol. 2018; 137: 295-302. https://doi.org/10.1007/s11060-017-2717-0.
98Bergen ES, Berghoff AS, Medjedovic M, et al. Continued endocrine therapy is associated with improved survival in patients with breast cancer brain metastases. Clin Cancer Res: Off J Am Assoc Cancer Res. 2019; 25: 2737-2744. https://doi.org/10.1158/1078-0432.Ccr-18-1968.
99Recchia AG, Musti AM, Lanzino M, et al. A cross-talk between the androgen receptor and the epidermal growth factor receptor leads to p38MAPK-dependent activation of mTOR and cyclinD1 expression in prostate and lung cancer cells. Int J Biochem Cell Biol. 2009; 41: 603-614. https://doi.org/10.1016/j.biocel.2008.07.004.
101Collins LC et al. Androgen receptor expression in breast cancer in relation to molecular phenotype: results from the nurses' health study. Modern Pathol: Off J United States Can Acad Pathol, Inc. 2011; 24: 924-931. https://doi.org/10.1038/modpathol.2011.54.
102Vera-Badillo FE et al. Androgen receptor expression and outcomes in early breast cancer: a systematic review and meta-analysis. J Natl Cancer Inst. 2014; 106:djt319. https://doi.org/10.1093/jnci/djt319.
104Fioretti FM, Sita-Lumsden A, Bevan CL, Brooke GN. Revising the role of the androgen receptor in breast cancer. J Mol Endocrinol. 2014; 52: R257-R265. https://doi.org/10.1530/jme-14-0030.
106Ortmann J, Prifti S, Bohlmann MK, Rehberger-Schneider S, Strowitzki T, Rabe T. Testosterone and 5 alpha-dihydrotestosterone inhibit in vitro growth of human breast cancer cell lines. Gynecol Endocrinol: Off J Int Soc Gynecol Endocrinol. 2002; 16: 113-120.
107Greeve MA, Allan RK, Harvey JM, Bentel JM. Inhibition of MCF-7 breast cancer cell proliferation by 5alpha-dihydrotestosterone: a role for p21(Cip1/Waf1). J Mol Endocrinol. 2004; 32: 793-810. https://doi.org/10.1677/jme.0.0320793.
108Cops EJ, Bianco-Miotto T, Moore NL, et al. Antiproliferative actions of the synthetic androgen, mibolerone, in breast cancer cells are mediated by both androgen and progesterone receptors. J Steroid Biochem Mol Biol. 2008; 110: 236-243. https://doi.org/10.1016/j.jsbmb.2007.10.014.
109Nahleh Z. Androgen receptor as a target for the treatment of hormone receptor-negative breast cancer: an unchartered territory. Future Oncology (London, England). 2008; 4: 15-21. https://doi.org/10.2217/14796694.4.1.15.
110Maggiolini M, Donze O, Jeannin E, Ando S, Picard D. Adrenal androgens stimulate the proliferation of breast cancer cells as direct activators of estrogen receptor alpha. Cancer Res. 1999; 59: 4864-4869.
111Barton VN, D'Amato NC, Gordon MA, et al. Multiple molecular subtypes of triple-negative breast cancer critically rely on androgen receptor and respond to enzalutamide in vivo. Mol Cancer Ther. 2015; 14: 769-778. https://doi.org/10.1158/1535-7163.Mct-14-0926.
113Puy L et al. Immunocytochemical detection of androgen receptor in human temporal cortex: characterization and application of polyclonal androgen receptor antibodies in frozen and paraffin-embedded tissues. J Steroid Biochem Mol Biol. 1995; 55: 197-209. https://doi.org/10.1016/0960-0760(95)00165-V.
115Vasudevan N, Pfaff DW. Non-genomic actions of estrogens and their interaction with genomic actions in the brain. Front Neuroendocrinol. 2008; 29: 238-257. https://doi.org/10.1016/j.yfrne.2007.08.003.
116Vasudevan N, Kow LM, Pfaff D. Integration of steroid hormone initiated membrane action to genomic function in the brain. Steroids. 2005; 70: 388-396. https://doi.org/10.1016/j.steroids.2005.02.007.
118Brailoiu E, Dun SL, Brailoiu GC, et al. Distribution and characterization of estrogen receptor G protein-coupled receptor 30 in the rat central nervous system. J Endocrinol. 2007; 193: 311-321. https://doi.org/10.1677/joe-07-0017.
120Cai M et al. The loss of estrogen efficacy against cerebral ischemia in aged postmenopausal female mice. Neurosci Lett. 2014; 558: 115-119. https://doi.org/10.1016/j.neulet.2013.11.007.
122Wise PM. Estrogen therapy: does it help or hurt the adult and aging brain? Insights derived from animal models. Neuroscience. 2006; 138: 831-835. https://doi.org/10.1016/j.neuroscience.2005.08.046.
123Lee E, Sidoryk-Wêgrzynowicz M, Wang N, et al. GPR30 regulates glutamate transporter GLT-1 expression in rat primary astrocytes. J Biol Chem. 2012; 287: 26817-26828. https://doi.org/10.1074/jbc.M112.341867.
124Razmara A, Sunday L, Stirone C, et al. Mitochondrial effects of estrogen are mediated by estrogen receptor α in brain endothelial cells. J Pharmacol Exp Ther. 2008; 325: 782-790. https://doi.org/10.1124/jpet.107.134072.
126Yu W et al. cDNA clone, prokaryotic expression and purification of human interleukin-13 receptor [alpha]2 chain. Cancer Immunol Immunother. 2009; 58: 409-413. https://doi.org/10.1007/s00262-008-0566-6.
129Milner TA et al. Ultrastructural evidence that hippocampal alpha estrogen receptors are located at extranuclear sites. J Comp Neurol. 2001; 429: 355-371.
132Smith CC, McMahon LL. Estrogen-induced increase in the magnitude of long-term potentiation occurs only when the ratio of NMDA transmission to AMPA transmission is increased. J Neurosci. 2005; 25: 7780-7791. https://doi.org/10.1523/jneurosci.0762-05.2005.
133Clements L, Harvey J. Activation of estrogen receptor alpha induces a novel form of long-term potentiation at hippocampal temporoammonic-CA1 synapses. Br J Pharmacol. 2019; 177: 642-655. https://doi.org/10.1111/bph.14880.
134Wong M, Moss RL. Electrophysiological evidence for a rapid membrane action of the gonadal steroid, 17 beta-estradiol, on CA1 pyramidal neurons of the rat hippocampus. Brain Res. 1991; 543: 148-152. https://doi.org/10.1016/0006-8993(91)91057-8.
137McEwen B et al. Tracking the estrogen receptor in neurons: implications for estrogen-induced synapse formation. PNAS. 2001; 98: 7093. https://doi.org/10.1073/pnas.121146898.
139Hajszan T, MacLusky NJ, Leranth C. Role of androgens and the androgen receptor in remodeling of spine synapses in limbic brain areas. Horm Behav. 2008; 53: 638-646. https://doi.org/10.1016/j.yhbeh.2007.12.007.
140Iorga A, Cunningham CM, Moazeni S, Ruffenach G, Umar S, Eghbali M. The protective role of estrogen and estrogen receptors in cardiovascular disease and the controversial use of estrogen therapy. Biol Sex Differ. 2017; 8: 33-33. https://doi.org/10.1186/s13293-017-0152-8.
141Pequeux C, Raymond-Letron I, Blacher S, et al. Stromal estrogen receptor-alpha promotes tumor growth by normalizing an increased angiogenesis. Cancer Res. 2012; 72: 3010-3019. https://doi.org/10.1158/0008-5472.Can-11-3768.
142Chambliss KL, Yuhanna IS, Mineo C, et al. Estrogen receptor alpha and endothelial nitric oxide synthase are organized into a functional signaling module in caveolae. Circ Res. 2000; 87: E44-E52. https://doi.org/10.1161/01.res.87.11.e44.
146Burek M, Steinberg K, Forster CY. Mechanisms of transcriptional activation of the mouse claudin-5 promoter by estrogen receptor alpha and beta. Mol Cell Endocrinol. 2014; 392: 144-151. https://doi.org/10.1016/j.mce.2014.05.003.
148Wasilewski D, Priego N, Fustero-Torre C, Valiente M. Reactive astrocytes in brain metastasis. Front Oncol. 2017; 7: 298. https://doi.org/10.3389/fonc.2017.00298.
149Barreto G, Veiga S, Azcoitia I, Garcia-Segura LM, Garcia-Ovejero D. Testosterone decreases reactive astroglia and reactive microglia after brain injury in male rats: role of its metabolites, oestradiol and dihydrotestosterone. Eur J Neurosci. 2007; 25: 3039-3046. https://doi.org/10.1111/j.1460-9568.2007.05563.x.
150Klein A, Schwartz H, Sagi-Assif O, et al. Astrocytes facilitate melanoma brain metastasis via secretion of IL-23. J Pathol. 2015; 236: 116-127. https://doi.org/10.1002/path.4509.
152Xing F et al. Reactive astrocytes promote the metastatic growth of breast cancer stem-like cells by activating notch signalling in brain. EMBO Mol Med. 2013; 5: 384-396. https://doi.org/10.1002/emmm.201201623.
154Drilon A, Siena S, Ou SHI, et al. Safety and antitumor activity of the multitargeted Pan-TRK, ROS1, and ALK inhibitor entrectinib: combined results from two phase I trials (ALKA-372-001 and STARTRK-1). Cancer Discov. 2017; 7: 400-409. https://doi.org/10.1158/2159-8290.CD-16-1237.
155Chen Q, Boire A, Jin X, et al. Carcinoma-astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature. 2016; 533: 493-498. https://doi.org/10.1038/nature18268.
156Gulinello M, Etgen AM. Sexually dimorphic hormonal regulation of the gap junction protein, CX43, in rats and altered female reproductive function in CX43+/− mice. Brain Res. 2005; 1045: 107-115. https://doi.org/10.1016/j.brainres.2005.03.021.
158Priego N, Valiente M. The potential of astrocytes as immune modulators in brain tumors. Frontiers in Immunology. 2019; 10:1314. https://doi.org/10.3389/fimmu.2019.01314.
159Czlonkowska A, Ciesielska A, Gromadzka G, Kurkowska-Jastrzebska I. Gender differences in neurological disease: role of estrogens and cytokines. Endocrine. 2006; 29: 243-256. https://doi.org/10.1385/ENDO:29:2:243.
160Curtis KS et al. Temporal and site-specific changes in central Neuroimmune factors during rapid weight gain after ovariectomy in rats. Neurochem Res. 2018; 43: 1802-1813. https://doi.org/10.1007/s11064-018-2596-6.
162Sarvari M et al. Estrogens regulate neuroinflammatory genes via estrogen receptors alpha and beta in the frontal cortex of middle-aged female rats. J Neuroinflammation. 2011; 8: 82. https://doi.org/10.1186/1742-2094-8-82.
164Spence RD, Wisdom AJ, Cao Y, et al. Estrogen mediates neuroprotection and anti-inflammatory effects during EAE through ERalpha signaling on astrocytes but not through ERbeta signaling on astrocytes or neurons. J Neurosci. 2013; 33: 10924-10933. https://doi.org/10.1523/jneurosci.0886-13.2013.
165Spence RD, Hamby ME, Umeda E, et al. Neuroprotection mediated through estrogen receptor-alpha in astrocytes. Proc Natl Acad Sci U S A. 2011; 108: 8867-8872. https://doi.org/10.1073/pnas.1103833108.
166Brown CM, Mulcahey TA, Filipek NC, Wise PM. Production of proinflammatory cytokines and chemokines during neuroinflammation: novel roles for estrogen receptors alpha and beta. Endocrinology. 2010; 151: 4916-4925. https://doi.org/10.1210/en.2010-0371.
167Carrillo-de Sauvage MA et al. CCL2-expressing astrocytes mediate the extravasation of T lymphocytes in the brain. Evidence from patients with glioma and experimental models in vivo. PLoS ONE. 2012; 7:e30762. https://doi.org/10.1371/journal.pone.0030762.
169von Bernhardi R, Heredia F, Salgado N, Muñoz P. In: R Bernhardi, ed. Glial Cells in Health and Disease of the CNS. Springer International Publishing; 2016: 67-92.
171Thakkar R, Wang R, Wang J, Vadlamudi RK, Brann DW. 17beta-estradiol regulates microglia activation and polarization in the hippocampus following global cerebral ischemia. Oxid Med Cell Longev. 2018; 2018:4248526. https://doi.org/10.1155/2018/4248526.
172Loiola RA, Wickstead ES, Solito E, McArthur S. Estrogen promotes pro-resolving microglial behavior and phagocytic cell clearance through the actions of Annexin A1 microglia. Front Endocrinol (Lausanne). 2019; 10: 420. https://doi.org/10.3389/fendo.2019.00420.
174Soucy G, Boivin G, Labrie F, Rivest S. Estradiol is required for a proper immune response to bacterial and viral pathogens in the female brain. J Immunol. 2005; 174: 6391-6398. https://doi.org/10.4049/jimmunol.174.10.6391.
175Svoronos N, Perales-Puchalt A, Allegrezza MJ, et al. Tumor cell-independent estrogen signaling drives disease progression through mobilization of myeloid-derived supressor cells. Cancer Discovery. 2017;7:72-85. https://doi.org/10.1158/2159-8290.Cd-16-0502.
176Ozerova M, Nefedova Y. Estrogen promotes multiple myeloma through enhancing the immunosuppressive activity of MDSC. Leuk Lymphoma. 2019;60(6):1557-1562. https://doi.org/10.1080/10428194.2018.1538511.
177Kozasa K et al. Estrogen stimulates female cancer progression by inducing myeloid-derived suppressive cells: investigations on pregnant and non-pregnant experimental models. Oncotarget. 2019; 10: 1887-1902. https://doi.org/10.18632/oncotarget.26711.
178Pan T, Zhong L, Wu S, et al. 17beta-Oestradiol enhances the expansion and activation of myeloid-derived suppressor cells via signal transducer and activator of transcription (STAT)-3 signalling in human pregnancy. Clin Exp Immunol. 2016; 185: 86-97. https://doi.org/10.1111/cei.12790.
180Prieto GA, Rosenstein Y. Oestradiol potentiates the suppressive function of human CD4 CD25 regulatory T cells by promoting their proliferation. Immunology. 2006; 118: 58-65. https://doi.org/10.1111/j.1365-2567.2006.02339.x.
181Adurthi S, Kumar MM, Vinodkumar HS, et al. Oestrogen receptor-α binds the FOXP3 promoter and modulates regulatory T-cell function in human cervical cancer. Sci Rep.2017;7:17289. https://doi.org/10.1038/s41598-017-17102-w.
Please check your email for instructions on resetting your password.
If you do not receive an email within 10 minutes, your email address may not be registered,
and you may need to create a new Wiley Online Library account.
Request Username
Can't sign in? Forgot your username?
Enter your email address below and we will send you your username
If the address matches an existing account you will receive an email with instructions to retrieve your username
The full text of this article hosted at iucr.org is unavailable due to technical difficulties.