Skip to main content
  • Research article
  • Open access
  • Published:

Tamoxifen suppresses brain metastasis of estrogen receptor-deficient breast cancer by skewing microglia polarization and enhancing their immune functions

Abstract

Background

Brain metastasis of breast cancer exhibits exceedingly poor prognosis, and both triple negative (TN) and Her2+ subtypes have the highest incidence of brain metastasis. Although estrogen blockers are considered to be ineffective for their treatment, recent evidence indicates that estrogen blockade using tamoxifen showed certain efficacy. However, how estrogen affects brain metastasis of triple negative breast cancer (TNBC) remains elusive.

Methods

To examine the effect of estrogen on brain metastasis progression, nude mice were implanted with brain metastatic cells and treated with either estrogen supplement, tamoxifen, or ovariectomy for estrogen depletion. For clinical validation study, brain metastasis specimens from pre- and post-menopause breast cancer patients were examined for microglia polarization by immunohistochemistry. To examine the estrogen-induced M2 microglia polarization, microglia cells were treated with estrogen, and the M1/M2 microglia polarization was detected by qRT-PCR and FACS. The estrogen receptor-deficient brain metastatic cells, SkBrM and 231BrM, were treated with conditioned medium (CM) derived from microglia that were treated with estrogen in the presence or absence of tamoxifen. The effect of microglia-derived CM on tumor cells was examined by colony formation assay and sphere forming ability.

Results

We found that M2 microglia were abundantly infiltrated in brain metastasis of pre-menopausal breast cancer patients. A similar observation was made in vivo, when we treated mice systemically with estrogen. Blocking of estrogen signaling either by tamoxifen treatment or surgical resection of mice ovaries suppressed M2 microglial polarization and decreased the secretion of C-C motif chemokine ligand 5, resulting in suppression of brain metastasis. The estrogen modulation also suppressed stemness in TNBC cells in vitro. Importantly, estrogen enhanced the expression of signal regulatory protein α on microglia and restricted their phagocytic ability.

Conclusions

Our results indicate that estrogen promotes brain metastasis by skewing polarity of M2 microglia and inhibiting their phagocytic ability, while tamoxifen suppresses brain metastasis by blocking the M2 polarization of microglia and increasing their anti-tumor phagocytic ability. Our results also highlight a potential therapeutic utility of tamoxifen for treating brain metastasis of hormone receptor-deficient breast cancer.

Background

Metastatic brain tumors are the most frequently occurring intracranial neoplasms in adults with the annual incidence of over 200,000 cases in the USA [1]. The majority of brain metastases originate from primary tumors of the lungs, melanoma, and breast [2, 3]. Patients with brain metastasis of breast cancer have extremely poor prognosis, high mortality rate, and frequent incidence of tumor recurrence. Even with aggressive treatments involving surgical intervention, irradiation, and chemotherapy, only a fraction of these patients with brain metastasis survives longer than 2 years after diagnosis [4]. Therefore, understanding the pathological mechanism of brain metastasis of breast cancer is urgently needed to identify a novel and effective therapeutic strategy.

One of the major risk factors for brain metastasis of breast cancer is age. A previous study showed that the cumulative 5-years incidence of brain metastasis is higher in younger patients (20.5%) than in older patients (7.5%) [5]. Importantly, premenopausal women have 1.5–2 times higher incidence of brain metastasis than postmenopausal women [6]. This difference is considered to be attributed to the female sex hormones, especially estrogen. Several studies have shown that the incidence of brain metastasis is associated with the expression of estrogen receptor (ER) in luminal subtype breast cancer [7,8,9,10]. Blocking ER by antagonist prolonged the onset of brain metastases from breast cancer [8,9,10]. Although the function of estrogen in promoting primary cancer is well documented, the physiological role of estrogen on brain metastasis and tumor microenvironment is poorly understood.

Among all subtypes of breast cancer, triple-negative breast cancer (TNBC) has a significantly poorer outcome, and approximately 30 to 46% TNBC patients will eventually develop brain metastasis prior to death [11, 12]. Although tamoxifen is used as the first-line treatment for ER+ breast cancer, it is not a standard treatment for ER negative or TNBC patients due to a lack hormone receptor [13,14,15]. Interestingly, however, recent studies showed that tamoxifen exhibited anti-tumor effect [16, 17] in TNBC, suggesting that tamoxifen may modulate estrogen-related tumor microenvironment that suppresses tumor progression. Tumor metastasis in the brain is a complex process that involves communication between the neoplastic cells and the normal brain cells. The non-neoplastic cells such as endothelial cells, macrophages, lymphocytes, astrocytes, and microglial cells present in the brain tumor microenvironment play a critical role in the formation of brain metastasis. Among these cells, microglia accounts for 30–50% of the total brain tumor mass [18, 19]. Previous in vitro and in vivo studies have demonstrated that activated microglia accelerate the growth and invasion of brain tumors [18,19,20]. A lack of microglia/macrophage significantly inhibited the metastatic spread of tumors [21,22,23], suggesting that microglia plays a critical role in the tumor invasion and metastasis in the brain.

Here, we investigated the effect of estrogen and tamoxifen on brain metastasis of hormone receptor-deficient breast cancer and examined the role of estrogen-induced polarization of microglia in tumor progression. We found that estrogen strongly skewed microglia to M2 phenotype, which significantly suppressed anti-tumor immune functions. M2 microglia also promoted tumor stem cell growth by secreting C-C motif chemokine ligand 5 (CCL5) in response to estrogen. Blocking of estrogen signaling by tamoxifen and ovariectomy exhibited anti-tumor effect on hormone receptor-negative tumor growth by modulating M2 microglial polarization and suppressing their anti-tumor immune function.

Methods

Animal

Animals were treated in accordance with the US National Institutes of Health Animal.

Protection Guidelines and approved by the Wake Forest Baptist Health Institutional Animal Care and Use Committee. Five to 6 weeks old of female nude mice were used. The mice were housed (five per cage) with a stable temperature (24 ± 1 °C), a 12-h light/dark cycle, and unrestricted access to food and water. The housing environment and animal health were monitored by the laboratory animal center. To examine the role of estrogen on brain metastasis, nude mice were anesthetized and randomly divided into five groups: (1) tumor transplant only, (2) tumor transplant plus 17β-estradiol (E2), (3) tumor transplant plus ovariectomy (OVX), (4) tumor transplant plus OVX and E2 supplement, (5) tamoxifen (Tamo), and (6) OVX + E2 + Tamo. For OVX, the mouse was anesthetized by inhalation of isofluorane. After wiping the injection site with three times of betadine and 70% alcohol, a midline dorsal incision (approx 1 cm) was made using a sharp scissor. A smaller incision (1 cm) in the muscle layer on either side of midline incision was then made to allow entry into the peritoneal cavity. The ovaries and uterus were identified in a fat pad. The end of bilateral uterus were ligated by 6-0 polyproplene suture and the ovaries were removed. The sham group received the same incision but the ovaries were not removed. For E2 supplement, a 0.5-cm incision was made under the neck skin and implanted with 1.5 mg/pellet E2 (Innovative Research of America, 60 day release) and incision was closed by 6–0 polypropylene suture. Finally, the 6-0 polyproplene suture was used to close the incision site of body wall and skin. One dose of buprenorphine (0.05 mg/kg) was subcutaneously administered and animals were allowed to wake up on the heated pad. The animals were positioned laterally and kept warm for 30 min until they recovered from anesthesia. One week later, mice received luciferase-labeled 231BrM cells by intracardiac injection (i.c.) at a concentration of 2 × 105 cells in 100 μL PBS into the left cardiac ventricle. After 3 days of tumor transplantation, mice in the tamoxifen group received tamoxifen (20 mg/kg) treatment by i.p. injection every 3 days for 30 days. The whole body photon flux of mice was measured immediately after injection to confirm a successful injection using IVIS Xenogen bioimager (Caliper). Tumor growth was monitored by bioluminescence until day 30. For bioluminescent imaging, the mice were injected with D-luciferin intraperitoneally (100 mg/kg), followed by capturing images every week using IVIS Xenogen bioimager. The brain metastasis was monitored and the luminescence was quantified once per week. At the endpoint, whole brain was removed, incubated in PBS with 0.6 mg/mL luciferin for 5 min and ex vivo photon flux was measured by IVIS.

Human subject

Human breast cancer specimens were obtained from surgical pathology archives of the Wake Forest Baptist Comprehensive Cancer Center (WFBCCC), and Cooperative Human Tissue Network, and Pathology Shared Resource at WFBCCC. All tissue sections were obtained by surgical resection and the patient’s information about age, cancer type, and menstrual period and status were record by Wake Forest Baptist hospital. This study was approved by the Wake Forest School of Medicine Institutional Review Board and written informed consent was obtained from all participants.

Cell culture and reagents

Human breast cancer line, MDA-MB231BrM2a (231BrM), was a kind gift from Dr. Massague (Memorial Sloan-Kettering Cancer Center). SKBrM3 cell line was derived from parental SKBr3 cells through three rounds of in vivo selection [24, 25]. SkBrM and 231BrM cell lines were authenticated by using GenePrint® 10 STR System (Promega, # B9510). Human and mouse microglia lines, HMC3 and SIM-A9, were purchased from American Type Culture Collection (ATCC) and were authenticated by ATCC. The SkBrM and 231BrM were cultured in DMEM supplemented with 10% FBS, streptomycin (100 mg/mL) and penicillin (100 U/mL) and HMC3 and SIM-A9 cells were cultured in DMEM/F12 medium supplemented with 5% FBS. All cells were grown at 37 °C under 5% CO2. The HMC3 cells were seeded in a 10-cm dish. After reaching 70% confluence, cells were incubated with DMEM/F12 medium supplemented with 2% FBS or in medium containing 1 nM E2 (Sigma) or in 1 nM E2 plus the STAT3 inhibitor, STATTIC (Selleckchem.com) at 0.5 μM concentration or in 1 nM E2 plus the estrogen receptor antagonist, tamoxifen (Sigma), at 1 μM concentration. We also knocked down the expression of ERα and β in the human microglia cell line using shRNA and examined the effect of estrogen on modulating M2 microglia-mediated brain metastasis. After 24 h, cells were washed twice with PBS and then incubated in the fresh DMEM/F12 medium supplemented with 2% FBS for 24 h. The conditioned medium (CM) harvested from the cell culture was centrifuged at 300×g for 10 min to remove the cells and stored at − 80 °C. All cell lines were ensured to be mycoplasma negative by using a universal mycoplama detection kit (ATCC, #30-1012 k, Lot: 70008746). The cells were collected and qRT-PCR and western blotting were used to quantify protein and mRNA levels. In another round of cell culturing, E2-treated cells were washed again and cultured for an additional 24 h in fresh medium. The CM were collected to identify cytokines using the cytokines array (Raybio). To examine the effect of microglia on T cell proliferation, the E2-treated SIM-A9 cells were cultured with fluorescent-labeled (CFSE Cell Proliferation Kit; Thermo Fisher) primary mouse T cell for 24 h, and the cell proliferation was measured by flow cytometry.

Immunohistochemistry

The human brain sections were stained using goat anti-CD206 (1:200, R&D systems) for M2 microglia and anti-CD47 (1:100, Invitrogen) for tumor cells and anti-SIRP (1:500, Cell signaling). Brain sections were then incubated with appropriate HRP-conjugated secondary antibodies using diaminobenzidine as the substrate. The signals were evaluated based on their intensities after subtracting the signals of the primary antibody-omitted negative controls. In some cases, the primary antibodies were replaced by isotype antibodies to control for non-specific binding of the antibodies. To determine the area of CD206+ cells in brain metastasis, we chose 3 randomly selected fields in each tumor and measured staining intensity by using the Image-Pro software. We also chose and measured staining intensities of 3 non-tumor areas in the consecutive slide, and this background intensity was subtracted to normalize the tumor intensity in the specimens. The average normalized intensity of 3 fields was used as the score of that patient. The scoring range was set from 0 (lowest) to 3 (highest).

Western blot

The cultured microglia cells were homogenized (1:3, in cultured cells) in the RIPA buffer, and then centrifuged at 17,000g for 30 min at 4 °C. The protein concentrations of the supernatants were determined and adjusted to the same concentration. Supernatants (30 g of total protein) were mixed with sample buffer containing 0.5 M of dithiothreitol, heated to 80 °C for 10 min, loaded into each well of 4% polyacrylamide gel and resolved at 120 V for 2 ;h. The separated proteins were transferred to a polyvinylidene fluoride membrane (Bio-Rad Laboratories, Hercules, CA, USA), blocked in 5% milk, and probed with respective primary antibodies: JAK (1:1000, Cell signaling), STAT3 (1:1000, Cell signaling), p-STAT3 (1:3000, Cell signaling), GAPDH (1:10000) (Cell Signaling), and PD-L1 (1:2000) (Cell Signaling). The bound antibodies were detected using an enhanced chemiluminescence detection kit (PerkinElmer, Boston, MA, USA). The band densities were measured using an imaging system (BioChemi; UVP, Upland, CA, USA) and analyzed using ImageJ (1.43u) (http://rsb.info.nih.gov/ij/). For gel loading control, membranes were stained with monoclonal-GAPDH antibody (1:50000, Cell signaling).

Flow cytometry

Nude mice were anesthetized with an overdose of isoflurane and their brains were removed and placed in ice-cold PBS containing 2.5 mg/ml of trypsin and dissociated using mechanical shearing. The cell suspension homogenates were passed through a 40-μm nylon membrane (Becton Dickinson Labware, Franklin Lakes, NJ, USA) and then centrifuged at 500 g for 10 min. The pellets were suspended in PBS, and microglial cells were isolated using a combination of protocols of differential density (stepwise Percoll) gradient centrifugation and immunomagnetic Iba1+ cell separation. Purified microglial cells were fixed in 4% paraformaldehyde at 4 °C for 1 h and then incubated with F4/80 and CD86 antibody (1:250) (eBioscience), Tmem119 (1:250) (eBioscience), CD206 (1:250) (eBioscience), CD44 (1:10000) (BioLegend), and ESA (1:250) (Invitrogen) at 4 °C for 16 h. After they were washed twice with PBS, the stained cells were subjected to flow cytometric analysis (FACScan; Becton-Dickinson, Mountain View, CA, USA). The isotype antibodies were be used as control. The percentage of cells was calculated using Cell-QuestTM software (Becton-Dickinson).

Phagocytosis assay

231BrM cells were labeled with PKH26 dye (Sigma, USA). The labeled cells were then washed three times and cultured overnight to reduce nonspecific leakage of dye during the assay. Labeled 231BrM cells were mixed with HMC3 human microglia cells that were pre-treated with or without E2 (1 nM) or E2 plus tamoxifen at 1 μM concentration. Co-cultured 231BrM and HMC3 cells in the culture slides were harvested after 8 h and fixed with 4% paraformaldehyde. Microglial cells were counterstained with anti-Iba1 antibody overnight. Secondary antibody conjugated with fluorescent dye Alexa-fluor 488 (1:1000, Invitrogen) was used to detect the microglia. The phagocytic activities were measured by immunofluorescence microscope and flow cytometry. For quantification of phagocytosis, the phagocytic percentage was calculated as: 100 × [percent Iba1+/PKH26+ cells/(percent Iba1+/PKH26 cells + percent Iba1+/PKH26+ cells)].

CCL5 and TNF-α ELISA

Microglial cells (HMC3) were treated with or without E2 (1nM) or E2 plus tamoxifen (1 μM) for 24h. Cells were washed with PBS twice and incubated in the fresh DMEM/F12 medium supplemented with 2% FBS for additional 24h. Conditioned medium was collected and analyzed by the human CCL5 and TNF-α ELISA kit (Sigma, USA). The HMC3 cells were homogenized in RIPA lysis buffer and centrifuged at 13,000g for 30min at 4°C. The supernatant was collected, and protein concentrations were measured and adjusted to 1mg/mL. Briefly, 96-well plates were first coated with anti-CCL5 or TNF-α monoclonal antibodies followed by addition of 100μL of the microglia conditional medium or the same volume of the CCL5 and TNF-α standards. The plate was incubated at 4 °C overnight on a rocking platform. After washing the plates, the detection antibody and streptavidin solution were added to each well. After incubation, the TMB and stop solution were added to each well. Finally, the plates were examined at an absorbance wavelength of 450nm. Standard curves were obtained from values generated from known concentrations of mouse and human CCL5 or TNF-α provided by the kit.

Arginase-1 gene promoter reporter assay and natural compound screening

To examine the estrogen-induced M2 microglial polarization and examine the effect of tamoxifen on estrogen-induced microglial polarization, HMC3 cells were first infected with lentivirus containing green fluorescent protein (GFP) gene and GFP+ cells were sorted by FACS. The GFP+ HMC3 cells were seeded in 96-well plates for one day and were transfected with the Arginase-1 promoter reporter plasmid (Addgene) using Lipofectamine 2000 (Invitrogen; Carlsbad, CA, USA). After 24h of incubation, the cells were treated with only E2 or E2 plus tamoxifen in presence of 2% FBS and cultured for another 24h. Cells were washed twice and were treated with 100μL D-luciferin for 5min. The expression of luciferase was detected by using IVIS Xenogen bioimager. For arginase-1 promoter luciferase normalization, the photon flux was divided by GFP signal which was measured by Multi-Mode Reader (Biocompare). Each experiment was conducted a minimum of three times.

Statistical analysis

The total number (n) of observations in each group is indicated in figure captions. Data are represented as mean ± standard deviation (SD). Significance was set at p0.05. All studies with more than two groups were analyzed using one-way ANOVA and then Bonferroni post-hoc tests if the overall effect was significant. Two-way ANOVA was used to analyze the effect of E2, OVX, and tamoxifen on brain metastasis of breast cancer in vivo. Bonferroni post-hoc tests were done if the main effects or interactions were significant. One-way ANOVA was used to analyze the effect of estrogen, OVX, STATTIC or tamoxifen treatment on M1/M2 gene polarization and tumor progression, effect of different concentrations of TNF-α or CCL5 on cancer progression. Significance between groups was represented as *, p0.05; **, p0.01; ***, p0.001.

Results

Estrogen promotes brain metastasis of breast cancer

To test the effect of female hormones on brain metastasis of ER-negative breast cancer and to understand how tamoxifen modulate the metastatic growth in the brain, we first performed bilateral ovariectomy (OVX) in 5–6 week-old female mice. One week after the surgery, mice were intracardially implanted with luciferase-labeled human ER-negative breast cancer cells, 231BrM. The mice were then implanted with or without 17β-estradiol (E2) by subcutaneous implantation of a disc or received tamoxifen treatment (20 mg/kg) by i.p. injection every 3 days for 1 month (Fig. 1a). Tamoxifen is known to be blood brain barrier (BBB) permeable [26]. As shown in Fig. 1a–d, E2 significantly increased the incidence and growth of brain metastasis [brain metastasis: Veh (33%) vs. E2 (89%)]. On the other hand, OVX dramatically suppressed brain metastasis progression [brain metastasis: E2 (89%) vs. OVX (11%)]. However, the OVX-mediated suppression of brain metastasis was rescued by E2 treatment [brain metastasis: OVX (11%) vs. E2 + OVX (33%)]. Interestingly, tamoxifen alone or OVX-treated with E2 plus tamoxifen significantly repressed brain metastasis growth to the similar level observed in the OVX group (Fig. 1b–d). Since MDA-MB-231 cells are resistant to tamoxifen [27], we sought of a possibility that the tumor-suppressive effect is due to the tumor microenvironment. Previous studies showed that activation of microglia, especially pro-tumor M2 microglia, promoted the progression of glioma [28,29,30,31]. Therefore, we examined M1 and M2 microglia in the brain of OVX-, E2-, tamoxifen-, OVX plus tamoxifen-, and OVX plus E2 and tamoxifen-treated mice by flow cytometry. The result of FACS analysis revealed that the population of M1 microglial cell (F4/80+) was significantly decreased (Fig. 1e and Supplemental Figure 1A) and high levels of M2 microglial cells (CD206+) accumulated in the lesions of E2-treated mice (Fig. 1f and Supplemental Figure 1B), suggesting that E2 induced microglia polarization to M2 phenotype in the brain metastatic lesions. In contrast, the effect of E2 in microglia polarization was abrogated by OVX, tamoxifen, or OVX with E2 plus tamoxifen (Fig. 1e, f and Supplemental Figure 1A and B). These results indicate that E2 promotes brain metastasis by skewing microglia polarization to M2 phenotype. To validate this notion in a clinical setting, we examined the status of M2 microglia activation in brain metastatic lesions of pre- and post-menopausal breast cancer patients (Fig. 1g). We observed that M2 microglia (CD206+ cells) abundantly infiltrated in the brain lesions of pre-menopause patients compared to post-menopause patients (Fig. 1g). Microglia and peripheral macrophages are known to be recruited by brain metastasis lesion. Although microglia and macrophage share many common surface markers and similar physiological functions, recent studies have demonstrated that microglia and macrophages are two distinct myeloid populations with different developmental origins [32, 33], and they can be distinguished by CD14 macrophage marker [34,35,36]. We found that the macrophages were recruited in the brain metastasis lesion of breast cancer patients (Fig. 1g).

Fig. 1
figure 1

Estrogen promotes brain metastasis by activating microglia in vivo. a Scheme of the experimental design is shown in the left panel. The OVX, OVX + E2, and OVX + E2 + tamoxifen (Tamo) groups received the OVX treatment 1 week before the tumor injection. 231BrM (6 × 105 cells) were intracardially injected into nude mice (n = 9) followed by administration of E2 pellet. After 3 days of intracardiac transplantation of the tumor cells, mice in tamoxifen group received tamoxifen (20 mg/kg) treatment by i.p. injection every 3 days till day 32. Right upper panel: Representative BLI images of mice from each experimental group (vehicle alone or E2, OVX, OVX + E2, Tamo or OVX + E2 + Tamo treatment). Right lower panel: Representative images of total ex vivo photon flux of brain metastatic lesions measured by BLI at the end point (Day 32). b Percentage of brain metastasis of breast cancer in nude mice with or without estrogen and/or tamoxifen treatment. Chi-square test, *: p 0.05; ***: p0.001. c Quantitative data of in vivo brain metastasis of breast cancer. Two-way ANOVA, **: p < 0.01; ***: p < 0.001 versus the opposite group; ##: p < 0.01. ###: p < 0.001versus related Veh group. (n = 9). d Quantitative data of ex vivo signals in the brain at the endpoint. One-way ANOVA, *: p < 0.05; ***: p < 0.001. (n = 9). e and f Metastatic brain tumors from (a) were isolated and examined by flow cytometry for M1 (F4/80+/Tmem119+) and M2 (CD206+/Tmem119+) microglial polarization. One-way ANOVA, **: p < 0.01; ***: p < 0.001. (n = 4). g Representative images of immunohistochemical analysis for CD206+ (M2 microglia) and CD45+ (macrophage) cells in the brain metastatic lesions of pre-menopause (n = 7) and post-menopause (n = 7) breast cancer patients. Scale bar: 200 μm. Right panels: CD206 and CD14 staining cells were quantified in premenopausal and postmenopausal patients. t test, *: p < 0.05; ***: p < 0.001. The data are presented as the mean ± SD

However, the population of CD45+ macrophage was significantly less than that of CD206+ M2 microglia. (Fig. 1g). We also examined the infiltrated microglia and macrophage in the mouse brain using the macrophage marker CD45 and the microglia marker Tmem119. We found that the microglia (CD45/Tmem119+) are more abundantly infiltrated than macrophages (CD45+/Tmem119) in the brain metastatic lesions (Supplemental Figure 1C). We also observed the increase in CD45+ cells in the metastatic mice brain after E2 treatment (Supplemental Figure 1D), which is consistent with the previous studies [37, 38].

Estrogen skewed M2 microglial polarization

Because M2 microglia are abundantly infiltrated in brain metastatic lesions of pre-menopause patients, we examined whether estrogen promotes brain metastasis by polarizing microglia to M2 microglia. We found that the expression of estrogen receptor (ER) on microglia was upregulated in the mice brain after E2 supplement (Supplemental Figure 2A). We examined the expression of ERs in HMC3 (Human) and SIM-A9 (Mouse) microglia cells and found that the expressions of estrogen receptor α and β are upregulated by E2 treatment (Supplemental Figure 2B). To further investigate the effect of estrogen on microglia polarization, HMC3 and SIM-A9 cells were treated with E2 followed by examining the expression of M1 and M2 markers. Our results showed that E2 significantly increased the mRNA expression of M2-related genes, arginase-1 (Arg1), arginase-2 (Arg2), and CD206, in both human and mouse microglial cells lines (Fig. 2a and b). On the other hand, blocking estrogen receptor (ER) signaling by tamoxifen or knockdown of ERα or ERβ by shRNA significantly suppressed E2-induced M2 microglia polarization (Fig. 2c, d, and Supplemental Figure 2C and D). Phenol red in tissue culture media is a weak estrogen [39], we also examined the microglia’s polarization by incubating human microglia cells with or without Phenol red and with or without 2% of FBS or charcoal-stripped FBS for 1 and 3 days. The cell viability (Supplemental Figure 3A) and M1/M2 phenotype (Supplemental Figure 3B, 3C) had no significant difference between the groups. This result is consistent with other published studies [40]. As the JAK-STAT3 pathway is known to induce M2 microglia polarization [41], we examined the status of phospho-STAT3 and JAK expression in microglia after E2 treatment. Indeed, E2 treatment strongly enhanced the activation of JAK/STAT3 in microglia, while tamoxifen suppressed E2-induced activation of JAK and STAT3 (Fig. 2e). We then treated microglia with E2 in the presence or absence of the STAT3 inhibitor (STATTIC) or tamoxifen and examined their effects on polarization of M1/M2 microglia using the Arg1 promoter reporter [42]. We found that E2 upregulated the Arg1 gene promoter activity, and this effect was significantly suppressed by STATTIC and/or tamoxifen treatment (Fig. 2f). Moreover, inhibition of STAT3 and estrogen receptor reversed the E2-mediated suppression of M1 (CD86+/Iba1+) and promotion of M2 (CD206+/Iba1+) polarization (Fig. 2g, h). These results suggest that the E2 promotes M2 polarization of microglia via activation of the STAT3 pathway.

Fig. 2
figure 2

Estrogen promotes M2 microglial polarization through activation of STAT3. a, b Expressions of M1/M2 microglia markers were examined after the vehicle or E2 (1 nM) or E2 plus tamoxifen (1 μM) 1 treatment of human microglia (HMC3) (a) and mouse microglia (SIM-A9) (b). The value of qRT-PCR in each Figure was normalized using actin as a control. The Y-axis indicates arbitrary unit. c, d Population of M1 and M2 cells was examined by FACS after the treatment of human microglia (HMC3) with vehicle alone, E2 (1 nM) or E2 plus tamoxifen (1 μM) (c) and SIM-A9 (d) cells. e HMC3 cells were treated with or without E2 (1 nM) or tamoxifen (1 μM) for 24 h and the expressions of JAK and STAT3 were examined by western blot. f The HMC3 cells were infected with lentivirus containing green fluorescent protein (GFP) gene and GFP+ cells were sorted by flow cytometry (FACS). The GFP+ HMC3 cells were seeded in 96-well plates for 1 day and they were transfected with the Arginase-1 promoter reporter plasmid using Lipofectamine 2000 (Invitrogen; Carlsbad, CA, USA). After 24 h, the cells were treated with only estrogen (1 nM) or estrogen plus STATTIC (0.5 μM) or tamoxifen (1 μM) and cultured for another 24 h. The expression of luciferase was measured by using IVIS Xenogen bioimager. The Arginase-1 reporter luciferase activity (M2) was normalized with the expression of GFP+. g, h Human microglia (HMC3) were treated with or without E2 (1 nM) and the STAT3 inhibitor (0.5 μM) or tamoxifen (1 μM) for 24 h. Cells were then subjected to flow cytometry for quantifying CD206+/Iba1+ (M2 cell) (g) and CD86+/Iba1+ (M1 cell) (h). The data are presented as the mean ± SD. One-way ANOVA, *: p < 0.05; **: p < 0.01; ***: p < 0.001. (n = 3)

Condition medium (CM) of estrogen-treated microglia promotes tumor cell growth and stem cell self-renewal

To examine how E2 promotes brain metastasis by upregulating M2 microglia polarization, we treated breast cancer brain metastatic cells, SKBrM (ER/Her2+) and 231BrM (ER/PR/Her2), with conditioned medium (CM) that was generated from human HMC3 microglia treated with or without E2. To avoid direct effect of E2 on tumor growth, microglia were treated with E2 (1 nM) for 24 h, washed with PBS to remove E2, and incubated in the fresh DMEM/F12 medium supplemented with 2% FBS for 24 h. The CM was then collected and added to SKBrM and 231BrM cells. We found that the CM derived from E2-treated microglia significantly increased cell viability (Fig. 3a) and colony forming ability (Fig. 3b) of both SKBrM and 231BrM cells. The expression of stemness-inducing genes, SOX2, OCT4, and NANOG, were also significantly upregulated when the brain metastatic cells were treated with the CM derived from E2-treated microglia (Fig. 3c and d). Furthermore, treatment of SKBrM and 231BrM cells with CM generated from E2-treated microglia showed significant increase in their sphere forming ability (Fig. 3e) and CD44+/ESA+ cancer stem cell (CSC) population (Fig. 3f). In contrast, direct treatment of cancer cells with the same dose of E2 (1 nM) did not affect the stem cell renewal or expression of stemness genes (Supplemental Figure 4A-D). Furthermore, E2-mediated promotion of the CSC population (CD44+/ESA+) was also significantly suppressed by STATTIC treatment (Fig. 3g). These results suggest that E2 induces secretory factor(s) from microglia, which in turn promotes tumor cell growth and self-renewal. We also examined the direct cytotoxic effect of tamoxifen on tumor cell viability in vitro (Fig. 3h). Our results showed that tamoxifen suppressed cell viability at the concentration of 100 μΜ, but not at 1 μΜ (the dose used throughout our microglia experiments) (Fig. 3h), indicating that the anti-tumor effect of tamoxifen is not mediated by direct cytotoxic effect on tumor cells, and it is rather mediated by the effect on microglia. To further investigate the effect of estrogen on microglia-derived tumor progression, 231BrM cells were treated with CM from microglia that were pre-treated with E2 or E2 plus tamoxifen or E2 plus shERα/β. We found that CM from E2 plus tamoxifen or shERα/β treated microglia significantly suppressed the tumor promoting effect of E2 treated microglia CM (Fig. 3i, j, Supplemental Figure 4E and F). Similarly, we found that CM derived from microglia that were treated with E2 plus tamoxifen or shERα/β significantly suppressed the population of CSC (Supplemental Figure 4G) and the expression of stemness-related genes SOX2 and NANOG (Fig. 3k-m, Supplemental Figure 4H and I).

Fig. 3
figure 3

Estrogen-mediated M2 microglial polarization promotes tumor growth and stemness. a SKBrM and 231BrM cells were incubated with the CM derived from E2-treated microglia for 24 h. They were then examined for cell viability. Veh CM or E2 CM: microglia cells were treated with PBS or E2 (1 nM) for 24 h, and they were washed with PBS and incubated in the fresh DMEM/F12 medium supplemented with 2% FBS for 24 h. t test, *: p < 0.05; **: p < 0.01. (n = 3). b Colony forming abilities of SKBrM and 231BrM cells were measured in the presence or absence of the CM derived from E2-treated human microglia. t test, **: p < 0.01. (n = 3). c, d SKBrM and 231BrM cells were incubated with the CM derived from E2-treated microglia for 24 h, and the expressions of stemness genes were examined by qRT-PCR., t test, **: p < 0.01; ***: p < 0.001. (n = 3). e Brain metastatic breast cancer cells (SKBrM and 231BrM) were treated with the non-E2 or E2 CM for 24 h and their sphere forming abilities were measured. t test, *: p < 0.05. (n = 3). f Effect of the CM derived from E2-treated microglia on cancer stem cell markers, CD44+/ ESA+ were examined using flow cytometry. t test, *: p < 0.05. (n = 3). g SKBrM (Left) and 231BrM (Right) cells were examined for CD44+/ESA+ cancer stem cell population by FACS. One-way ANOVA, **: p < 0.01. (n = 3). h 231BrM cells were incubated with tamoxifen at different concentrations for 24 h. They were then examined for cell viability. One-way ANOVA, ***: p < 0.001. (n = 3). i 231BrM cells were incubated with the CM derived from microglia that were treated with E2 alone or E2 plus tamoxifen for 24 h. They were then examined for cell viability. E2 or E2 + Tamo CM: microglia cells were treated with E2 (1 nM) or E2 plus tamoxifen (1 μM) for 24 h, and they were washed with PBS and incubated in the fresh DMEM/F12 medium supplemented with 2% FBS for 24 h. One-way ANOVA, *: p < 0.05. (n = 3). j Colony forming abilities of 231BrM cells were measured in the presence or absence of the CM derived from E2− or E2 plus tamoxifen-treated human microglia. One-way ANOVA, **: p < 0.01. (n = 3). k–m 231BrM cells were incubated with the CM derived from E2− or E2 plus tamoxifen-treated microglia for 24 h, and the expressions of stemness genes were examined by qRT-PCR. The data are presented as the mean ± SD. One-way ANOVA, **: p < 0.01; ***: p < 0.001. (n = 3)

Estrogen induces secretion of CCL5 from microglia and promotes tumor growth and stemness

To identify the factor(s) induced by E2 in microglia, we performed cytokine array analysis (consisting of 99 cytokines and growth factors) and found that E2 treatment significantly increased the secretion of C-C motif chemokine ligand 5 (CCL5) whereas it decreased tumor necrosis factor-α (TNF-α) secretion in the microglial CM (Fig. 4a). The effect of E2 in increasing CCL5 and decreasing TNF-α secretion was further verified in human microglia by performing ELISA (Fig. 4b and c). In contrast, blocking estrogen receptor by tamoxifen or shERα/β significantly suppressed E2-mediated secretion of CCL5 and upregulated the production of TNF-α (Fig. 4b, c and Supplemental Figure 5A-D). TNF-α and CCL5 are known to be associated with M1 and M2 phenotypes, respectively [43,44,45]. To examine the functional effects of TNF-α and CCL5, we treated breast cancer cells with recombinant TNF-α or CCL5 and examined their effect on these cells. We found that TNF-α treatment significantly decreased cell viability while CCL5 significantly increased viability of both SKBrM and 231BrM cells (Fig. 4d, e). It should be noted that TNF-α has been shown to kill some tumor cells through activation of apoptotic signaling [46, 47]. We also found that the mRNA expression of CSC markers, SOX2, OCT4, and NANOG, was increased in 231BrM cells when they were treated with recombinant CCL5 (Fig. 4f). Similarly, CCL5 augmented the population of ESA+/CD44+ CSC (Fig. 4g). To examine whether the upregulation of stemness genes by the CM is indeed mediated by CCL5, CM of E2-treated microglia was treated with CCL5 neutralizing antibody followed by measuring the expression of CSC genes. Our result indicates that depletion of CCL5 in the CM significantly suppressed the expression of stemness genes SOX2, OCT4, and NANOG (Fig. 4h). Moreover, the stimulatory effect of CM on stem cell population (CD44+/ESA+ CSC) was also suppressed after the CCL5 depletion (Fig. 4i). In addition, the E2-mediated increase in the population of CSC (CD44+/ESA+) was significantly suppressed by tamoxifen treatment (Fig. 4i). Because cancer cells may also directly affect polarization of microglia [48, 49], we examined the effect of tumor cell-derived conditioned medium. Our results indicated that the direct treatment of microglia with E2 promoted M2 microglia polarization significantly more than the treatment with tumor conditioned medium (Supplemental Figure 5E and F). These results further support our notion that E2 stimulates the secretion of CCL5 from microglia, which in turn promotes self-renewal of tumor cells.

Fig. 4
figure 4

Estrogen induces secretion of CCL5 from microglia and promotes tumor growth and stemness. a Human microglial (HMC3) cells were treated with PBS or E2 (1 nM) for 24 h and they were washed with PBS and incubated in the fresh DMEM/F12 medium supplemented with 2% FBS for 24 h. CM was collected and analyzed by using the cytokine/growth factor array (Raybio). b, c Human microglial (HMC3) cells were treated with or without E2 (1 nM) or E2 plus tamoxifen (1 μM) for 24 h and cells were washed with PBS twice and incubated in the fresh DMEM/F12 medium supplemented with 2% FBS for additional 24 h. Cell lysates (b) and CM (c) were examined for the amount of TNF-α and CCL5 by ELISA (n = 5/group). One-way ANOVA, *: p < 0.05; **: p < 0.01; ***: p < 0.001. d, e SKBrM (d) and 231BrM (e) cells were treated with the indicated concentrations of recombinant TNFα and CCL5 for 24 h. They were then examined for cell viability by the MTS assay. One-way ANOVA, *: p < 0.05; **: p < 0.01; ***: p < 0.001. (n = 4). f 231BrM cells were treated with CCL5 for 24 h followed by measuring the stemness genes by qRT-PCR. t test, ***: p < 0.001. (n = 4). g 231BrM cells were treated with CCL5 for 24 h followed by measuring the population of ESA+/CD44+ stem cell by flow cytometry One-way ANOVA, ***: p < 0.001. (n = 4). h and i Human microglia were treated with or without E2 (1 nM) for 24 h, and their CM was prepared. The CM was then added with anti-CCL5 antibody to deplete CCL5. 231BrM cells were then treated with the CCL5-depleted CM followed by assaying stemness-related genes by qRT-PCR (h) and ESA+/CD44+ stem cell population by FACS (i). The data are presented as the mean ± SD. One-way ANOVA, *: p < 0.05; **: p < 0.01; ***: p < 0.001. (n = 4)

Estrogen suppressed anti-tumor immune function of microglia

It is known that tumor cells express programmed death-ligand 1 (PD-L1) that binds PD-1 on T cell, resulting in dysfunction of cytotoxic T cell activity [50]. Several other studies have also shown that microglia expresses PD-L1, which interacts with T cell PD-1 to suppress adaptive immune function and tumor infiltration of T cells [50,51,52]. We therefore examined whether E2 affects PD-L1 expression in microglia. As shown in Fig. 5a, both mRNA and protein expression of PD-L1 in microglia were significantly upregulated by E2, and inhibiting STAT3 activation by STATTIC or blocking estrogen receptor by tamoxifen treatment significantly suppressed E2-induced upregulation of PD-L1 (Fig. 5a). Moreover, the expression of PD-L1 was elevated on the membrane of microglia after E2 treatment (Fig. 5b). To investigate how upregulation of PD-L1 on microglia by E2 affects T cell function, T cells were incubated with microglia cells that were pre-treated with E2. We found that the proliferation of T cells was significantly suppressed when T cell were incubated with E2-treated microglia (Fig. 5c). Blocking STAT3 activation in microglia by STATTIC treatment restored E2-mediated suppression of T cell proliferation (Fig. 5c).

Fig. 5
figure 5

Estrogen suppresses innate anti-tumor immune function of microglia. a PD-L1 expression was examined in human microglia (HMC3) cells after treating them with or without E2, STAT3 inhibitor (0.5 μM STATTIC), or tamoxifen (1 μΜ) by RT-PCR and western blot. One-way ANOVA, *: p < 0.05; ***: p < 0.001. (n = 3). b Representative images of immunohistochemical analysis of PD-L1 in human microglia (HMC3) cells that were treated with or without E2 for 24 h. Scale bar: 50 μm. c T cells were incubated with E2− or E2 plus STATTIC-treated microglia. Proliferation of T cells was measured by flow cytometry. One-way ANOVA, ***: p < 0.001. (n = 3). d Human (HMC3) microglia cells were treated with or without E2 (1 nM) or E2 plus tamoxifen (1 μM) for 24 h, and the expression of SIRPα was examined by qRT-PCR. One-way ANOVA, **: p < 0.01; ***: p < 0.001. (n = 3). e SKBrM and 231BrM cells were treated with or without E2 for 24 h, and the expression of CD47 was examined by qRT-PCR. t test, ***: p < 0.001. (n = 3). f The brain metastatic lesions from pre- or post-menopause patients in Fig. 1g were sectioned and they were subjected to immunohistochemical analysis for CD47+. Arrows indicate CD47+ cells (brown) in pre-menopause (n = 7) or post-menopause (n = 7) patients. Scale bar: 100 μm. g Phagocytic activities of HMC3 human microglial cells (green labeled) with or without E2 or E2 plus tamoxifen treatment were examined using PKH26-1 labeled 231BrM cells (red labeled). Both cells were incubated together for 24 h, photographed (left panels), and the number of microglia that phagocytosed tumor cells was counted to quantify microglial phagocytic activity as shown in the right panel. h A parallel set of cells in (g) was analyzed by flow cytometry to quantify phagocytic abilities of microglia after E2 or E2 plus tamoxifen treatment. One-way ANOVA, ***: p < 0.001. (n = 3). The data are presented as the mean ± SD

Microglia cells are known to exhibit anti-tumor functions by phagocytosis and by releasing cytotoxic factors [53]. However, tumor cells express CD47 that interacts with signal regulatory protein α (SIRPα) on microglia [54] and triggers the ‘do not eat me’ signal which enables cancer cells to evade microglia’s phagocytic ability. To examine if E2 functionally affects the “do not eat me” signal, we measured the expression of SIRPα on microglia and CD47 on tumor cells. We found that SIRPα was significantly upregulated in human and mouse microglia upon E2 treatment (Fig. 5d). Tamoxifen dramatically decreased E2-mediated upregulation of SIRPα (Fig. 5d). We also found that E2 significantly increased the expression of CD47 in both SKBrM and 231BrM cells (Fig. 5e). Although the SKBrM and 231BrM cells are considered to be “ER-negative cells”, several studies also have found that 30% of TNBCs express ERα and β by an antibody validation analysis [55,56,57,58]. Therefore, we suspected that upregulation of CD47 by E2 was still through the activation of the ER signaling. To test this possibility, we infected 231BrM with lentivirus including shRNA to ERα and β and examined CD47 expression in the presence and absence of E2. We found that the effect of E2 on CD47 expression was indeed nullified by the shRNAs. (Supplemental Figure 6A). The next question is how ER upregulates CD47. E2 is known to upregulate the PI3K/AKT signaling [59, 60], and previous study have shown that CD47 expression is controlled by PI3K/Akt phosphorylation [61, 62]. Therefore, to examine whether E2 upregulates the expression of CD47 by activating PI3K, 231BrM cells were treated with PI3K inhibitor, LY294002. We found that blocking PI3K activation in tumor cells impaired the E2-induced upregulation of CD47 (Supplemental Figure 6B), suggesting that activation of CD47 by E2 is likely to be mediated through the interaction with ER followed by activation of the PI3K signaling. Importantly, the expression of CD47 was shown to be strongly upregulated in brain metastatic lesions of pre-menopause patients (Fig. 5f). We then examined the effect of E2 on phagocytic ability of microglia. 231BrM cells were pre-labeled with the red-fluorescent dye, PKH26 [63], and co-cultured with microglia that were treated with or without E2 or E2 plus tamoxifen. After 24 h of incubation, small pieces of PKH26+ cells were accumulated inside of microglia in the control group, suggesting that tumor cells were rapidly degraded by microglia through phagocytosis (Fig. 5g). However, the microglia suppressed phagocytic activities toward tumor cells after E2 pre-treatment (Fig. 5g and h). In contrast, tamoxifen reversed E2-mediated suppression of phagocytic activities (Fig. 5g and h). These results suggest that E2 suppresses microglia’s immune function by blocking the phagocytic ability and upregulating PD-L1, which in turn promotes brain metastasis progression.

Discussion

The tumor promoting effect of estrogen is well established; however, how estrogen affects tumor microenvironment and support brain metastasis is still poorly understood. Our results indicate the critical roles of estrogen on tumor microenvironment in promoting brain metastasis by skewing microglia to M2 phenotype and inhibiting microglial phagocytic ability. This phenotypic conversion of microglia by estrogen is mediated through activation of STAT3, and that blocking STAT3 activation suppressed the M2 conversion, down-regulated the secretion of pro-tumor M2 cytokines and suppressed their phagocytic activity toward tumor cells. We also found that the population of M2 microglial cells were decreased in OVX and tamoxifen-treated mice, while the estrogen-induced brain metastasis was suppressed in both OVX and tamoxifen-treated mice (Fig. 6). These results suggest that overall homeostasis of sex hormones play a key role in microglial polarization, which affects the progression of brain metastasis in TNBC women.

Fig. 6
figure 6

A proposed model illustrating an estrogen-induced brain metastasis

Skewing polarization of microglia by estrogen was reported by multiple previous studies [64,65,66,67], and several possible mechanisms were suggested. Saijo et al. showed that estrogen could activate CCAAT/enhancer-binding proteins and inhibit the production of M1 pro-inflammatory cytokines in microglial cells [68]. It was also reported that estrogen suppressed the expression of M1 pro-inflammatory genes by blocking the nuclear translocation of NF-κB that suppressed DNA binding activity of NF-κB and upregulated IκB, an inhibitor of NF-κB signaling [69, 70]. In addition, Vejito et al. found that estrogen binding to estrogen receptor downregulated toll-like receptor 4 expression and inhibited M1 pro-inflammatory cytokines production [71]. These results suggest that estrogen modulates microglial polarization through multiple pathways. In the current study, we found a mechanism by which estrogen promotes M2 microglia by upregulating STAT3 expression. It should be noted that STAT3 activation has been associated with promotion of immunosuppressive microenvironment [72]. The activated STAT3 was known to reduce the expression of the genes that are necessary for antigen presentation such as MHC-II, CD80, and CD86 in microglia [73, 74]. Activation of STAT3 also promotes cell proliferation of microglia by enhancing cell survival genes [75]. Treating primary culture of microglia with the STAT3 inhibiter blocked their proliferation [76, 77]. We also observed that the number of M2 microglia was increased in the brain of premenopausal breast cancer patients as well as in the mouse supplemented with E2. On the other hand, the estrogen-associated proliferation of M2 microglia was blocked by the OVX and Tamoxifen treatment in mice, suggesting that estrogen deficiency suppresses microglia proliferation, which may well explain why the number of microglia is lower in brain metastasis of older breast cancer patients and in female mice with an OVX or tamoxifen treatment.

We have shown that estrogen decreased the secretion of M1-associated TNF-α and increased the M2-associated CCL5 in the conditioned medium of microglial cells. TNF-α is known to inhibit cell survival and proliferation by activating apoptosis signaling [46, 47]. The CCL5 and its receptor CCR5 have been detected in hematological malignancies and lymphomas. The interaction of CCL5 and CCR5 has been shown to promote tumor development by multiple mechanisms including acting as growth factors [78], enhancing migration and invasiveness [79], stimulating angiogenesis [80], and recruitment of macrophage [81]. Furthermore, knockdown or knockout of CCL5 in tumor cells was shown to suppress metastatic capability in vivo and in vitro of 4 T1 breast cancer model [78, 82]. In our study, we found that stem cell genes, NANOG/OCT4/SOX2, were significantly upregulated in CCL5-treated breast cancer cells, suggesting that E2-induced CCL5 promotes brain metastasis by enhancing the stemness of breast cancer brain metastatic cells. The depletion of CCL5 from CM of E2-treated microglia significantly suppressed E2-induced stemness genes and decreased the population of stem cells. Consistent with these results, CM from microglia treated with tamoxifen repressed the population of cancer stem cells by downregulating CCL5. These results also suggest that CCL5 in serum may serve as a novel therapeutic target as well as a biomarker for brain metastasis of breast cancer.

The current study focused on the effect of estrogen on tumor microenvironment; however, there are abundant reports showing direct effect of estrogen on tumor cells. Our study here showed that estrogen promotes brain metastasis by suppressing the phagocytosis function of microglia through activation of the “don’t eat me” signal. Our results showed that the estrogen promoted the expression of CD47 on tumor cells and SIRPα on microglia. Interestingly, we found that brain metastasis lesion of pre-menopause breast cancer patient had higher expression of CD47 in tumor and SIRPα in microglia than post-menopausal patients. These results suggest that estrogen-induced upregulation of tumor CD47 and SIRPα compromises phagocytic function of microglia, resulting in further tumor growth. In this context, it is noteworthy that high CD47 expression in breast cancer tissues showed significant association with reduced 5-year disease-free survival [83]. Therefore, CD47 may serve as a biomarker to predict breast cancer progression. We found that tamoxifen suppressed estrogen-induced upregulation of SIRPα expression, resulting in enhanced phagocytic ability. In addition to this suppressive function of estrogen on innate immune function, we found that the expression of PD-L1 was increased in the estrogen-treated microglia. In contrast, a combination of estrogen and tamoxifen suppressed E2-induced elevation of PD-L1 expression. The proliferation of T cells decreased when T cells were co-cultured with microglia cells that were pretreated with E2. Since PD-L1 on microglia interacts with PD-1 on T cells, it also decreases T cell proliferation and their cytotoxic functions as we have shown. These results suggest that estrogen promotes brain metastasis by modulating both innate and adaptive immune functions in the brain.

Estrogens such as 17β-estradiol are known to affect various types of brain microenvironment cells, not only microglia but also neuron, astrocyte, and endothelial cells. Estrogen influences brain function by directly affecting neuronal membranes. E2 was showed to upregulate the expression of glutamate transporter on neuron and increased the production of neurotransmitter glutamate that promoted brain tumor progression [84, 85]. A previous study in preclinical models showed that pre-menopausal levels of E2 promoted TNBC brain metastasis through activation of brain-derived neurotrophic factor and its receptor TrkB in reactive astrocytes [86, 87]. Estrogen also plays a role in increasing BBB permeability. E2 promotes ethanol-induced BBB disruption and changes/or increases in iNOS activity and decreases the tight junction protein expression, occludin and claudin-1, resulting in increased BBB permeability [88, 89]. These results suggest that E2 alters microglia and other brain microenvironment components, which together promotes brain metastasis progression.

Patients with TNBC generally show poor outcomes including early CNS metastasis and a short survival [11, 90]. Hormonal therapy is a standard of care for ER+ tumors, however, it has been considered to be not useful for hormone receptor negative patients. Consequently, TNBC has limited treatment approaches. We found that tamoxifen did not directly affect tumor progression on ER-negative breast cancer or TNBC, however, tamoxifen exhibited strong anti-tumor ability in vivo by skewing microglial polarization to M1 and enhancing their phagocytic functions. This result strongly suggests that the effect of tamoxifen on hormone receptor-deficient brain metastasis is mediated via modulation of brain microenvironment such as reprograming the function of microglia. A previous study demonstrated that treatment of recurrent malignant gliomas with chronic oral high-dose tamoxifen prolonged the patient survival [91], suggesting that tamoxifen can readily cross the BBB [26, 92] and affect brain tumor microenvironment. Moreover, Yang et al. showed that tamoxifen increased the overall survival and disease-free survival rate in ER/PR+ breast cancer patients [93]. In addition, treatment with combination of tamoxifen and radiation increased the overall survival rate and reduced the mortality rate of patients with brain metastasis of triple negative tumors [94]. It is noteworthy that Phase I/II clinical trial for testing efficacy of Tamoxifen and anastrozole on TNBC is currently ongoing (NCT01194908 and NCT01234532). Tamoxifen has been shown to upregulate autophagy and inhibits AMPK/mTOR pathway, resulting in tumor suppression in TNBC [95]. Previous studies also showed that tamoxifen suppressed ER-negative breast cancer cell invasion and metastasis by modulating miR-200c and transcription factor Twist1 [16, 17], suggesting that tamoxifen can suppress the progression of ER negative breast cancer by ER-independent pathways.

Conclusions

Our results showed that estrogen promotes TNBC-brain metastasis by skewing microglia polarization and diminishing their innate and adaptive immune functions. We also found that the M2 microglia and tumor progression were suppressed by OVX and tamoxifen treatment. Considering the inhibitory effect of tamoxifen on estrogen-related M2 microglial activation and their minimum toxicity, tamoxifen may serve as a potential treatment option for the brain metastasis, particularly for patients with ER-negative subtype.

Availability of data and materials

All data generated or analyzed during this study are included in this published article and its supplementary information files.

Abbreviations

TNBC:

Triple negative breast cancer

ER:

Estrogen receptor

CCL5:

C-C motif chemokine ligand 5

OVX:

Ovariectomy

CM:

Conditioned medium

GFP:

Green fluorescent protein

E2:

17β-estradiol

BBB:

Blood brain barrier

Arg1:

Arginase-1

Arg2:

Arginase-2

PD-L1:

Programmed death-ligand 1

SIRPα:

Signal regulatory protein α

FACS:

Flow cytometry

References

  1. Gavrilovic IT, Posner JB. Brain metastases: epidemiology and pathophysiology. J Neuro-Oncol. 2005;75(1):5–14.

    Article  Google Scholar 

  2. Barnholtz-Sloan JS, Sloan AE, Davis FG, Vigneau FD, Lai P, Sawaya RE. Incidence proportions of brain metastases in patients diagnosed (1973 to 2001) in the Metropolitan Detroit Cancer Surveillance System. J Clin Oncol. 2004;22(14):2865–72.

    Article  PubMed  Google Scholar 

  3. Schouten LJ, Rutten J, Huveneers HA, Twijnstra A. Incidence of brain metastases in a cohort of patients with carcinoma of the breast, colon, kidney, and lung and melanoma. Cancer. 2002;94(10):2698–705.

    Article  PubMed  Google Scholar 

  4. Tabouret E, Chinot O, Metellus P, Tallet A, Viens P, Goncalves A. Recent trends in epidemiology of brain metastases: an overview. Anticancer Res. 2012;32(11):4655–62.

    PubMed  Google Scholar 

  5. Polivka J Jr, Kralickova M, Polivka J, Kaiser C, Kuhn W, Golubnitschaja O. Mystery of the brain metastatic disease in breast cancer patients: improved patient stratification, disease prediction and targeted prevention on the horizon? EPMA J. 2017;8(2):119–27.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Gori S, Rimondini S, De Angelis V, Colozza M, Bisagni G, Moretti G, Sidoni A, Basurto C, Aristei C, Anastasi P, et al. Central nervous system metastases in HER-2 positive metastatic breast cancer patients treated with trastuzumab: incidence, survival, and risk factors. Oncologist. 2007;12(7):766–73.

    Article  PubMed  Google Scholar 

  7. Arvold ND, Oh KS, Niemierko A, Taghian AG, Lin NU, Abi-Raad RF, Sreedhara M, Harris JR, Alexander BM. Brain metastases after breast-conserving therapy and systemic therapy: incidence and characteristics by biologic subtype. Breast Cancer Res Treat. 2012;136(1):153–60.

    Article  PubMed  Google Scholar 

  8. Pors H, von Eyben FE, Sorensen OS, Larsen M. Longterm remission of multiple brain metastases with tamoxifen. J Neuro-Oncol. 1991;10(2):173–7.

    Article  CAS  Google Scholar 

  9. Salvati M, Cervoni L, Innocenzi G, Bardella L. Prolonged stabilization of multiple and single brain metastases from breast cancer with tamoxifen. Report of three cases. Tumori. 1993;79(5):359–62.

    Article  CAS  PubMed  Google Scholar 

  10. Bergen ES, Berghoff AS, Medjedovic M, Rudas M, Fitzal F, Bago-Horvath Z, Dieckmann K, Mader RM, Exner R, Gnant M, et al. Continued endocrine therapy is associated with improved survival in patients with breast cancer brain metastases. Clin Cancer Res. 2019;25(9):2737–44.

    Article  CAS  PubMed  Google Scholar 

  11. Hines SL, Vallow LA, Tan WW, McNeil RB, Perez EA, Jain A. Clinical outcomes after a diagnosis of brain metastases in patients with estrogen- and/or human epidermal growth factor receptor 2-positive versus triple-negative breast cancer. Ann Oncol. 2008;19(9):1561–5.

    Article  CAS  PubMed  Google Scholar 

  12. Jin J, Gao Y, Zhang J, Wang L, Wang B, Cao J, Shao Z, Wang Z. Incidence, pattern and prognosis of brain metastases in patients with metastatic triple negative breast cancer. BMC Cancer. 2018;18(1):446.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Lobbezoo DJ, van Kampen RJ, Voogd AC, Dercksen MW, van den Berkmortel F, Smilde TJ, van de Wouw AJ, Peters FP, van Riel JM, Peters NA, et al. Prognosis of metastatic breast cancer: are there differences between patients with de novo and recurrent metastatic breast cancer? Br J Cancer. 2015;112(9):1445–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Mahmood H, Faheem M, Mahmood S, Sadiq M, Irfan J. Impact of age, tumor size, lymph node metastasis, stage, receptor status and menopausal status on overall survival of breast cancer patients in Pakistan. Asian Pac J Cancer Prev. 2015;16(3):1019–24.

    Article  PubMed  Google Scholar 

  15. Ren Z, Li Y, Shen T, Hameed O, Siegal GP, Wei S. Prognostic factors in advanced breast cancer: race and receptor status are significant after development of metastasis. Pathol Res Pract. 2016;212(1):24–30.

    Article  CAS  PubMed  Google Scholar 

  16. Ma G, He J, Yu Y, Xu Y, Yu X, Martinez J, Lonard DM, Xu J. Tamoxifen inhibits ER-negative breast cancer cell invasion and metastasis by accelerating Twist1 degradation. Int J Biol Sci. 2015;11(5):618–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang Q, Cheng Y, Wang Y, Fan Y, Li C, Zhang Y, Wang Y, Dong Q, Ma Y, Teng YE, et al. Tamoxifen reverses epithelial-mesenchymal transition by demethylating miR-200c in triple-negative breast cancer cells. BMC Cancer. 2017;17(1):492.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Graeber MB, Scheithauer BW, Kreutzberg GW. Microglia in brain tumors. Glia. 2002;40(2):252–9.

    Article  PubMed  Google Scholar 

  19. Watters JJ, Schartner JM, Badie B. Microglia function in brain tumors. J Neurosci Res. 2005;81(3):447–55.

    Article  CAS  PubMed  Google Scholar 

  20. Wagner S, Czub S, Greif M, Vince GH, Suss N, Kerkau S, Rieckmann P, Roggendorf W, Roosen K, Tonn JC. Microglial/macrophage expression of interleukin 10 in human glioblastomas. Int J Cancer. 1999;82(1):12–6.

    Article  CAS  PubMed  Google Scholar 

  21. Giraudo E, Inoue M, Hanahan D. An amino-bisphosphonate targets MMP-9-expressing macrophages and angiogenesis to impair cervical carcinogenesis. J Clin Invest. 2004;114(5):623–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pukrop T, Dehghani F, Chuang HN, Lohaus R, Bayanga K, Heermann S, Regen T, Van Rossum D, Klemm F, Schulz M, et al. Microglia promote colonization of brain tissue by breast cancer cells in a Wnt-dependent way. Glia. 2010;58(12):1477–89.

    Article  PubMed  Google Scholar 

  23. Robinson-Smith TM, Isaacsohn I, Mercer CA, Zhou M, Van Rooijen N, Husseinzadeh N, McFarland-Mancini MM, Drew AF. Macrophages mediate inflammation-enhanced metastasis of ovarian tumors in mice. Cancer Res. 2007;67(12):5708–16.

    Article  CAS  PubMed  Google Scholar 

  24. Xing F, Liu Y, Sharma S, Wu K, Chan MD, Lo HW, Carpenter RL, Metheny-Barlow LJ, Zhou X, Qasem SA, et al. Activation of the c-met pathway mobilizes an inflammatory network in the brain microenvironment to promote brain metastasis of breast Cancer. Cancer Res. 2016;76(17):4970–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sharma S, Wu SY, Jimenez H, Xing F, Zhu D, Liu Y, Wu K, Tyagi A, Zhao D, Lo HW, et al. Ca(2+) and CACNA1H mediate targeted suppression of breast cancer brain metastasis by AM RF EMF. EBioMedicine. 2019;44:194–208.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Lien EA, Solheim E, Ueland PM. Distribution of tamoxifen and its metabolites in rat and human tissues during steady-state treatment. Cancer Res. 1991;51(18):4837–44.

    CAS  PubMed  Google Scholar 

  27. Strobl JS, Peterson VA. Tamoxifen-resistant human breast cancer cell growth: inhibition by thioridazine, pimozide and the calmodulin antagonist, W-13. J Pharmacol Exp Ther. 1992;263(1):186–93.

    CAS  PubMed  Google Scholar 

  28. da Fonseca AC, Badie B. Microglia and macrophages in malignant gliomas: recent discoveries and implications for promising therapies. Clin Dev Immunol. 2013;2013:264124.

    PubMed  Google Scholar 

  29. He BP, Wang JJ, Zhang X, Wu Y, Wang M, Bay BH, Chang AY. Differential reactions of microglia to brain metastasis of lung cancer. Mol Med. 2006;12(7–8):161–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Roesch S, Rapp C, Dettling S, Herold-Mende C. When immune cells turn bad-tumor-associated microglia/macrophages in glioma. Int J Mol Sci. 2018;19(2):436.

  31. Wei J, Gabrusiewicz K, Heimberger A. The controversial role of microglia in malignant gliomas. Clin Dev Immunol. 2013;2013:285246.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F, et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature. 2015;518(7540):547–51.

    Article  PubMed  CAS  Google Scholar 

  33. Prinz M, Priller J. Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease. Nat Rev Neurosci. 2014;15(5):300–12.

    Article  CAS  PubMed  Google Scholar 

  34. Feng X, Szulzewsky F, Yerevanian A, Chen Z, Heinzmann D, Rasmussen RD, Alvarez-Garcia V, Kim Y, Wang B, Tamagno I, et al. Loss of CX3CR1 increases accumulation of inflammatory monocytes and promotes gliomagenesis. Oncotarget. 2015;6(17):15077–94.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Rangaraju S, Raza SA, Li NX, Betarbet R, Dammer EB, Duong D, Lah JJ, Seyfried NT, Levey AI. Differential phagocytic properties of CD45(low) microglia and CD45(high) brain mononuclear phagocytes-activation and age-related effects. Front Immunol. 2018;9:405.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Greter M, Lelios I, Croxford AL. Microglia versus myeloid cell nomenclature during brain inflammation. Front Immunol. 2015;6:249.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Zhang L, Yao J, Wei Y, Zhou Z, Li P, Qu J, Badu-Nkansah A, Yuan X, Huang YW, Fukumura K, et al. Blocking immunosuppressive neutrophils deters pY696-EZH2-driven brain metastases. Sci Transl Med. 2020;12(545):eaaz5387.

  38. Tyagi A, Sharma S, Wu K, Wu SY, Xing F, Liu Y, Zhao D, Deshpande RP, D'Agostino RB Jr, Watabe K. Nicotine promotes breast cancer metastasis by stimulating N2 neutrophils and generating pre-metastatic niche in lung. Nat Commun. 2021;12(1):474.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Berthois Y, Katzenellenbogen JA, Katzenellenbogen BS. Phenol red in tissue culture media is a weak estrogen: implications concerning the study of estrogen-responsive cells in culture. Proc Natl Acad Sci U S A. 1986;83(8):2496–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. de Faria AN, Zancanela DC, Ramos AP, Torqueti MR, Ciancaglini P. Estrogen and phenol red free medium for osteoblast culture: study of the mineralization ability. Cytotechnology. 2016;68(4):1623–32.

    Article  PubMed  CAS  Google Scholar 

  41. Qin C, Fan WH, Liu Q, Shang K, Murugan M, Wu LJ, Wang W, Tian DS. Fingolimod protects against ischemic white matter damage by modulating microglia toward M2 polarization via STAT3 pathway. Stroke. 2017;48(12):3336–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yi WJ, Kim TS. Melatonin protects mice against stress-induced inflammation through enhancement of M2 macrophage polarization. Int Immunopharmacol. 2017;48:146–58.

    Article  CAS  PubMed  Google Scholar 

  43. Ruytinx P, Proost P, Van Damme J, Struyf S. Chemokine-induced macrophage polarization in inflammatory conditions. Front Immunol. 2018;9:1930.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Zhou Y, Guo W, Zhu Z, Hu Y, Wang Y, Zhang X, Wang W, Du N, Song T, Yang K, et al. Macrophage migration inhibitory factor facilitates production of CCL5 in astrocytes following rat spinal cord injury. J Neuroinflammation. 2018;15(1):253.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Lu HL, Huang XY, Luo YF, Tan WP, Chen PF, Guo YB. Activation of M1 macrophages plays a critical role in the initiation of acute lung injury. Biosci Rep. 2018;38(2):BSR20171555.

  46. Robaye B, Mosselmans R, Fiers W, Dumont JE, Galand P. Tumor necrosis factor induces apoptosis (programmed cell death) in normal endothelial cells in vitro. Am J Pathol. 1991;138(2):447–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Tartaglia LA, Ayres TM, Wong GH, Goeddel DV. A novel domain within the 55 kd TNF receptor signals cell death. Cell. 1993;74(5):845–53.

    Article  CAS  PubMed  Google Scholar 

  48. Wu SY, Xing F, Sharma S, Wu K, Tyagi A, Liu Y, Zhao D, Deshpande RP, Shiozawa Y, Ahmed T, et al. Nicotine promotes brain metastasis by polarizing microglia and suppressing innate immune function. J Exp Med. 2020;217(8):e20191131.

  49. Xing F, Liu Y, Wu SY, Wu K, Sharma S, Mo YY, Feng J, Sanders S, Jin G, Singh R, et al. Loss of XIST in breast cancer activates MSN-c-met and reprograms microglia via exosomal miRNA to promote brain metastasis. Cancer Res. 2018;78(15):4316–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Juneja VR, McGuire KA, Manguso RT, LaFleur MW, Collins N, Haining WN, Freeman GJ, Sharpe AH. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med. 2017;214(4):895–904.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kim H, Khanna V, Kucaba TA, Zhang W, Ferguson DM, Griffith TS, Panyam J. Combination of sunitinib and PD-L1 blockade enhances anticancer efficacy of TLR7/8 agonist-based nanovaccine. Mol Pharm. 2019;16(3):1200–10.

    Article  CAS  PubMed  Google Scholar 

  52. Yuan C, Liu Z, Yu Q, Wang X, Bian M, Yu Z, Yu J. Expression of PD-1/PD-L1 in primary breast tumours and metastatic axillary lymph nodes and its correlation with clinicopathological parameters. Sci Rep. 2019;9(1):14356.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Hutter G, Theruvath J, Graef CM, Zhang M, Schoen MK, Manz EM, Bennett ML, Olson A, Azad TD, Sinha R, et al. Microglia are effector cells of CD47-SIRPalpha antiphagocytic axis disruption against glioblastoma. Proc Natl Acad Sci U S A. 2019;116(3):997–1006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Folkes AS, Feng M, Zain JM, Abdulla F, Rosen ST, Querfeld C. Targeting CD47 as a cancer therapeutic strategy: the cutaneous T-cell lymphoma experience. Curr Opin Oncol. 2018;30(5):332–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Reese JM, Suman VJ, Subramaniam M, Wu X, Negron V, Gingery A, Pitel KS, Shah SS, Cunliffe HE, McCullough AE, et al. ERbeta1: characterization, prognosis, and evaluation of treatment strategies in ERalpha-positive and -negative breast cancer. BMC Cancer. 2014;14:749.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Wu X, Subramaniam M, Negron V, Cicek M, Reynolds C, Lingle WL, Goetz MP, Ingle JN, Spelsberg TC, Hawse JR. Development, characterization, and applications of a novel estrogen receptor beta monoclonal antibody. J Cell Biochem. 2012;113(2):711–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Weitsman GE, Skliris G, Ung K, Peng B, Younes M, Watson PH, Murphy LC. Assessment of multiple different estrogen receptor-beta antibodies for their ability to immunoprecipitate under chromatin immunoprecipitation conditions. Breast Cancer Res Treat. 2006;100(1):23–31.

    Article  CAS  PubMed  Google Scholar 

  58. Nelson AW, Groen AJ, Miller JL, Warren AY, Holmes KA, Tarulli GA, Tilley WD, Katzenellenbogen BS, Hawse JR, Gnanapragasam VJ, et al. Comprehensive assessment of estrogen receptor beta antibodies in cancer cell line models and tissue reveals critical limitations in reagent specificity. Mol Cell Endocrinol. 2017;440:138–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ciruelos Gil EM. Targeting the PI3K/AKT/mTOR pathway in estrogen receptor-positive breast cancer. Cancer Treat Rev. 2014;40(7):862–71.

    Article  CAS  PubMed  Google Scholar 

  60. Meng Q, Li J, Chao Y, Bi Y, Zhang W, Zhang Y, Ji T, Fu Y, Chen Q, Zhang Q, et al. beta-estradiol adjusts intestinal function via ERbeta and GPR30 mediated PI3K/AKT signaling activation to alleviate postmenopausal dyslipidemia. Biochem Pharmacol. 2020;180:114134.

    Article  CAS  PubMed  Google Scholar 

  61. Liu Y, Chang Y, He X, Cai Y, Jiang H, Jia R, Leng J. CD47 enhances cell viability and migration ability but inhibits apoptosis in endometrial carcinoma cells via the PI3K/Akt/mTOR signaling pathway. Front Oncol. 2020;10:1525.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Sick E, Boukhari A, Deramaudt T, Ronde P, Bucher B, Andre P, Gies JP, Takeda K. Activation of CD47 receptors causes proliferation of human astrocytoma but not normal astrocytes via an Akt-dependent pathway. Glia. 2011;59(2):308–19.

    Article  PubMed  Google Scholar 

  63. Munn DH, Cheung NK. Phagocytosis of tumor cells by human monocytes cultured in recombinant macrophage colony-stimulating factor. J Exp Med. 1990;172(1):231–7.

    Article  CAS  PubMed  Google Scholar 

  64. Thakkar R, Wang R, Wang J, Vadlamudi RK, Brann DW. 17beta-estradiol regulates microglia activation and polarization in the Hippocampus following global cerebral ischemia. Oxidative Med Cell Longev. 2018;2018:4248526.

    Article  CAS  Google Scholar 

  65. Siani F, Greco R, Levandis G, Ghezzi C, Daviddi F, Demartini C, Vegeto E, Fuzzati-Armentero MT, Blandini F. Influence of estrogen modulation on glia activation in a murine model of Parkinson’s disease. Front Neurosci. 2017;11:306.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Chen Y, Tian Y, Tian H, Huang Q, Fang Y, Wang W, Wan Y, Pan D, Xie M. Tamoxifen promotes white matter recovery and cognitive functions in male mice after chronic hypoperfusion. Neurochem Int. 2019;131:104566.

    Article  CAS  PubMed  Google Scholar 

  67. Ishihara Y, Itoh K, Ishida A, Yamazaki T. Selective estrogen-receptor modulators suppress microglial activation and neuronal cell death via an estrogen receptor-dependent pathway. J Steroid Biochem Mol Biol. 2015;145:85–93.

    Article  CAS  PubMed  Google Scholar 

  68. Saijo K, Collier JG, Li AC, Katzenellenbogen JA, Glass CK. An ADIOL-ERbeta-CtBP transrepression pathway negatively regulates microglia-mediated inflammation. Cell. 2011;145(4):584–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Murphy AJ, Guyre PM, Pioli PA. Estradiol suppresses NF-kappa B activation through coordinated regulation of let-7a and miR-125b in primary human macrophages. J Immunol. 2010;184(9):5029–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ghisletti S, Meda C, Maggi A, Vegeto E. 17beta-estradiol inhibits inflammatory gene expression by controlling NF-kappaB intracellular localization. Mol Cell Biol. 2005;25(8):2957–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Vegeto E, Ghisletti S, Meda C, Etteri S, Belcredito S, Maggi A. Regulation of the lipopolysaccharide signal transduction pathway by 17beta-estradiol in macrophage cells. J Steroid Biochem Mol Biol. 2004;91(1–2):59–66.

    Article  CAS  PubMed  Google Scholar 

  72. Jones LM, Broz ML, Ranger JJ, Ozcelik J, Ahn R, Zuo D, Ursini-Siegel J, Hallett MT, Krummel M, Muller WJ. STAT3 establishes an immunosuppressive microenvironment during the early stages of breast carcinogenesis to promote tumor growth and metastasis. Cancer Res. 2016;76(6):1416–28.

    Article  CAS  PubMed  Google Scholar 

  73. Kowalczyk A, D'Souza CA, Zhang L. Cell-extrinsic CTLA4-mediated regulation of dendritic cell maturation depends on STAT3. Eur J Immunol. 2014;44(4):1143–55.

    Article  CAS  PubMed  Google Scholar 

  74. Chan LL, Cheung BK, Li JC, Lau AS. A role for STAT3 and cathepsin S in IL-10 down-regulation of IFN-gamma-induced MHC class II molecule on primary human blood macrophages. J Leukoc Biol. 2010;88(2):303–11.

    Article  CAS  PubMed  Google Scholar 

  75. Shen Y, Devgan G, Darnell JE Jr, Bromberg JF. Constitutively activated Stat3 protects fibroblasts from serum withdrawal and UV-induced apoptosis and antagonizes the proapoptotic effects of activated Stat1. Proc Natl Acad Sci U S A. 2001;98(4):1543–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Zhang L, Handel MV, Schartner JM, Hagar A, Allen G, Curet M, Badie B. Regulation of IL-10 expression by upstream stimulating factor (USF-1) in glioma-associated microglia. J Neuroimmunol. 2007;184(1–2):188–97.

    Article  CAS  PubMed  Google Scholar 

  77. Komohara Y, Horlad H, Ohnishi K, Fujiwara Y, Bai B, Nakagawa T, Suzu S, Nakamura H, Kuratsu J, Takeya M. Importance of direct macrophage-tumor cell interaction on progression of human glioma. Cancer Sci. 2012;103(12):2165–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Stormes KA, Lemken CA, Lepre JV, Marinucci MN, Kurt RA. Inhibition of metastasis by inhibition of tumor-derived CCL5. Breast Cancer Res Treat. 2005;89(2):209–12.

    Article  CAS  PubMed  Google Scholar 

  79. Singh SK, Mishra MK, Eltoum IA, Bae S, Lillard JW Jr, Singh R. CCR5/CCL5 axis interaction promotes migratory and invasiveness of pancreatic cancer cells. Sci Rep. 2018;8(1):1323.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Suffee N, Richard B, Hlawaty H, Oudar O, Charnaux N, Sutton A. Angiogenic properties of the chemokine RANTES/CCL5. Biochem Soc Trans. 2011;39(6):1649–53.

    Article  CAS  PubMed  Google Scholar 

  81. Walens A, DiMarco AV, Lupo R, Kroger BR, Damrauer JS, Alvarez JV. CCL5 promotes breast cancer recurrence through macrophage recruitment in residual tumors. Elife. 2019;8:e43653.

  82. Zhang Y, Lv D, Kim HJ, Kurt RA, Bu W, Li Y, Ma X. A novel role of hematopoietic CCL5 in promoting triple-negative mammary tumor progression by regulating generation of myeloid-derived suppressor cells. Cell Res. 2013;23(3):394–408.

    Article  CAS  PubMed  Google Scholar 

  83. Yuan J, He H, Chen C, Wu J, Rao J, Yan H. Combined high expression of CD47 and CD68 is a novel prognostic factor for breast cancer patients. Cancer Cell Int. 2019;19:238.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Zeng Q, Michael IP, Zhang P, Saghafinia S, Knott G, Jiao W, McCabe BD, Galvan JA, Robinson HPC, Zlobec I, et al. Synaptic proximity enables NMDAR signalling to promote brain metastasis. Nature. 2019;573(7775):526–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Lee E, Sidoryk-Wegrzynowicz M, Yin Z, Webb A, Son DS, Aschner M. Transforming growth factor-alpha mediates estrogen-induced upregulation of glutamate transporter GLT-1 in rat primary astrocytes. Glia. 2012;60(7):1024–36.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Contreras-Zarate MJ, Day NL, Ormond DR, Borges VF, Tobet S, Gril B, Steeg PS, Cittelly DM. Estradiol induces BDNF/TrkB signaling in triple-negative breast cancer to promote brain metastases. Oncogene. 2019;38(24):4685–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Sartorius CA, Hanna CT, Gril B, Cruz H, Serkova NJ, Huber KM, Kabos P, Schedin TB, Borges VF, Steeg PS, et al. Estrogen promotes the brain metastatic colonization of triple negative breast cancer cells via an astrocyte-mediated paracrine mechanism. Oncogene. 2016;35(22):2881–92.

    Article  CAS  PubMed  Google Scholar 

  88. Kuruca SE, Karadenizli S, Akgun-Dar K, Kapucu A, Kaptan Z, Uzum G. The effects of 17beta-estradiol on blood brain barrier integrity in the absence of the estrogen receptor alpha; an in-vitro model. Acta Histochem. 2017;119(6):638–47.

    Article  CAS  PubMed  Google Scholar 

  89. Sandoval KE, Witt KA. Age and 17beta-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 

  90. Smid M, Wang Y, Zhang Y, Sieuwerts AM, Yu J, Klijn JG, Foekens JA, Martens JW. Subtypes of breast cancer show preferential site of relapse. Cancer Res. 2008;68(9):3108–14.

    Article  CAS  PubMed  Google Scholar 

  91. Couldwell WT, Hinton DR, Surnock AA, DeGiorgio CM, Weiner LP, Apuzzo ML, Masri L, Law RE, Weiss MH. Treatment of recurrent malignant gliomas with chronic oral high-dose tamoxifen. Clin Cancer Res. 1996;2(4):619–22.

    CAS  PubMed  Google Scholar 

  92. Toney TW, Katzenellenbogen BS. An evaluation of the interactions of antiestrogens with pituitary and striatal dopamine receptors. J Recept Res. 1987;7(5):695–712.

    Article  CAS  PubMed  Google Scholar 

  93. Yang LH, Tseng HS, Lin C, Chen LS, Chen ST, Kuo SJ, Chen DR. Survival benefit of tamoxifen in estrogen receptor-negative and progesterone receptor-positive low grade breast cancer patients. J Breast Cancer. 2012;15(3):288–95.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Payandeh M, Sadeghi M, Sadeghi E, Aeinfar M. Clinicopathology figures and long-term effects of Tamoxifen plus radiation on survival of women with invasive ductal carcinoma and triple negative breast cancer. Asian Pac J Cancer Prev. 2015;16(12):4863–7.

    Article  PubMed  Google Scholar 

  95. Wu ST, Sun GH, Cha TL, Kao CC, Chang SY, Kuo SC, Way TD. CSC-3436 switched tamoxifen-induced autophagy to apoptosis through the inhibition of AMPK/mTOR pathway. J Biomed Sci. 2016;23(1):60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Dr. Joan Massagué for providing the brain metastasis cell lines.

Funding

This work was supported by NIH grant RO1CA173499, R01CA185650 and R01CA205067, and by DOD (W81XWH2110075) (to KW). We also used Cancer Center Shared Resources, The Tumor Tissue and Pathology, Cell Engineering, Flow Cytometry, Biostatistics and Bioinformatics that are supported by the Comprehensive Cancer Center of Wake Forest University NCI, National Institutes of Health Grant (P30CA012197). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute. SW is a trainee of the T32 training grant (T32CA247819).

Author information

Authors and Affiliations

Authors

Contributions

SW and KW designed the study and wrote the manuscript. SW, SS and KWu conducted experiments and acquired, analyzed and interpreted the data. SS, AT and KWu performed animal experiment. SW and KWu performed intracardiac injections. DZ and RPD performed clinical sample analysis, SW, SS, KWu, AT, DZ, RPD, and KW reviewed and edited manuscript and interpreted the data. KW supervised the study. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Kounosuke Watabe.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the institutional review board of the Wake Forest University.

Consent for publication

Not applicable.

Competing interests

No other authors have any particular competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Supplemental Figure 1.

Expression of M1/M2 microglia polarization after E2, OVX and tamoxifen and the infiltration of microglia/macrophage by FACS. Supplemental Figure 2. Blocking estrogen receptors (ERs) by shRNA suppressed E2-related M2 microglia polarization. Supplemental Figure 3. Effect of phenol red free culture medium and charcoal-stripped fetal bovine serum on cell viability and microglia polarization. Supplemental Figure 4. Blocking estrogen receptors (ERs) on microglia suppressed E2-derived microglia condition medium (CM)-mediated stemness and growth. Supplemental Figure 5. Effect of estrogen on cytokines production and microglia polarization in microglia cells after shERα/β or tumor CM treatment. Supplemental Figure 6. Blocking ERs by shRNA on tumor cells down-regulated the expression of CD47.

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

Wu, SY., Sharma, S., Wu, K. et al. Tamoxifen suppresses brain metastasis of estrogen receptor-deficient breast cancer by skewing microglia polarization and enhancing their immune functions. Breast Cancer Res 23, 35 (2021). https://doi.org/10.1186/s13058-021-01412-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13058-021-01412-z

Keywords