Skip to main content
Endocrinology logoLink to Endocrinology
. 2012 Nov 1;153(12):5949–5960. doi: 10.1210/en.2012-1316

The Androgen Metabolite, 5α-androstane-3β,17β-diol, Decreases Cytokine-Induced Cyclooxygenase-2, Vascular Cell Adhesion Molecule-1 Expression, and P-Glycoprotein Expression in Male Human Brain Microvascular Endothelial Cells

Kristen L Zuloaga 1, Sibyl N Swift 1, Rayna J Gonzales 1, T John Wu 1, Robert J Handa 1,
PMCID: PMC3512076  PMID: 23117931

Abstract

P-glycoprotein (Pgp), a multiple drug resistance transporter expressed by vascular endothelial cells, is a key component of the blood-brain barrier and has been shown to increase after inflammation. The nonaromatizable androgen, dihydrotestosterone (DHT), decreases inflammatory markers in vascular smooth muscle cells, independent of androgen receptor (AR) stimulation. The principal metabolite of DHT, 5α-androstane-3β,17β-diol (3β-diol), activates estrogen receptor (ER)β and similarly decreases inflammatory markers in vascular cells. Therefore, we tested the hypothesis that either DHT or 3β-diol decrease cytokine-induced proinflammatory mediators, vascular cell adhesion molecule-1 (VCAM-1) and cyclooxygenase-2 (COX-2), to regulate Pgp expression in male primary human brain microvascular endothelial cells (HBMECs). Using RT-qPCR, the mRNAs for AR, ERα, and ERβ and steroid metabolizing enzymes necessary for DHT conversion to 3β-diol were detected in male HBMECs demonstrating that the enzymes and receptors for production of and responsiveness to 3β-diol are present. Western analysis showed that 3β-diol reduced COX-2 and Pgp expression; the effect on Pgp was inhibited by the ER antagonist, ICI-182,780. IL-1β-caused an increase in COX-2 and VCAM-1 that was reduced by either DHT or 3β-diol. 3β-diol also decreased cytokine-induced Pgp expression. ICI-182,780 blocked the effect of 3β-diol on COX-2 and VCAM-1, but not Pgp expression. Therefore, in cytokine-stimulated male HBMECs, the effect of 3β-diol on proinflammatory mediator expression is ER dependent, whereas its effect on Pgp expression is ER independent. These studies suggest a novel role of 3β-diol in regulating blood-brain barrier function and support the concept that 3β-diol can be protective against proinflammatory mediator stimulation.


A key group of drug efflux transporters at the blood brain barrier (BBB) are the superfamily of ATP-binding cassette transporters (ABC-transporters) (1), which are classified into seven families (2, 3). These drug efflux pumps are located in the luminal membrane of endothelial cells that form the BBB and play an important role in pharmacotherapeutic efficacy by acting as a selective barrier to drugs or other small molecules by transporting them out of the brain tissue. The most highly expressed ABC transporter of the BBB is P-glycoprotein (Pgp; ABCB1), which actively prevents the uptake of some anticancer drugs, antiepileptic drugs, antidepressants, antipsychotics, and corticosteroids (47). In rodent studies, inhibition of Pgp increases the movement of chemotherapy drugs across the BBB and thereby increases the therapeutic efficacy of these drugs (810).

The BBB is defined as the monolayer of vascular endothelial cells connected by high-resistance tight cell-cell junctions devoid of fenestrations and serves as a selective barrier for the movement of molecules from the bloodstream into the brain parenchyma (11). The integrity of the BBB can be compromised by inflammation (12), which is of interest because inflammation occurs in a wide range of brain disorders (7, 1315). Further, BBB breakdown is correlated with advancing disease states such as multiple sclerosis, epilepsy, and Alzheimer's disease (11). Inflammatory responses elicited by neural trauma, such as after ischemic stroke and traumatic brain injury, have also been shown to compromise BBB integrity (11, 16, 17).

Aside from endothelial cell and tight junction damage, BBB integrity may also be altered by changes in the expression of transporter proteins such as Pgp. Studies have shown that factors that promote inflammation increase Pgp expression/activity in both human and rodent cells/tissue (6, 1823). For example, the cytokine, TNFα, has been shown to increase Pgp protein and mRNA expression in human and rat brain endothelial cell lines (6, 22). Both TNFα and the cytokine, IL-1β, also increased Pgp protein expression in primary cultures of rat astrocytes (23). In rat brain endothelial cells, hypoxia followed by reoxygenation increased Pgp expression and nitric oxide, a product of the proinflammatory enzyme, inducible nitric oxide synthase (19). In some cases, increased expression and activity of Pgp could be blocked by TNFα receptor inhibition or did not occur in TNFα receptor null mice (20).

The proinflammatory mediator cyclooxygenase-2 (COX-2) has been shown to play a role in the increases in Pgp protein expression and activity observed after seizures (15, 24). This may be the result of exposure of capillaries to the excitatory neurotransmitter glutamate, which is in excess during seizure activity. Exposure to glutamate, both in vivo and in vitro, increased Pgp expression and transport activity within several hours. This effect could be blocked with a selective COX-2 inhibitor and was not observed in COX-2 null mice. Similar studies in rats also found that seizures induced Pgp expression and this effect was blocked by COX-2 inhibition (15, 24). Of importance, we have recently reported that in both brain and coronary human vascular smooth muscle cells, dihydrotestosterone (DHT), a potent nonaromatizable androgen, decreases COX-2 protein expression that was induced with cytokines or by hypoxia with glucose deprivation (25, 26).

Both androgens and estrogens have been shown to have antiinflammatory effects (2529); however, it is important to note that androgens also have proinflammatory effects as well (3032). In regards to its antiinflammatory properties, DHT can suppress levels of COX-2 after hypoxia with glucose deprivation or cytokine stimulation in human vascular smooth muscle cells, independent of androgen receptor (AR) stimulation (25, 26, 33). Interestingly, and perhaps not commonly appreciated, DHT can have properties beyond that of an AR agonist because it can be further metabolized to 5α-androstane-3β,17β-diol (3β-diol), an estrogen receptor β (ERβ) agonist (3436). 3β-diol has recently been shown to reduce expression of inflammatory markers in human brain vascular smooth muscle cells (33) and human umbilical vein endothelial cells (37) and in a similar fashion, the dehydroepiandrosterone (DHEA) metabolite/ERβ agonist 5α-androstene-3β,17β diol (Adiol), has also been shown to have antiinflammatory effects (38). Therefore, in these studies we tested the hypothesis that DHT and 3β-diol, act through estrogen receptors to decrease cytokine-induced expression of the inflammatory mediator COX-2, leading to decreased inflammation-induced expression of Pgp in male human brain microvascular endothelial cells (HBMECs). In addition to COX-2, we examined expression of vascular cell adhesion molecule-1 (VCAM-1), another common maker for vascular inflammation. Like COX-2, VCAM-1 is up-regulated by cytokine stimulation in endothelial cells. Both DHT and 3β-diol have been shown to decrease cytokine-induced VCAM-1 expression in human umbilical vein endothelial cells (27, 37); however, their effects in human brain endothelial cells are unknown. The results of these studies indicate that 3β-diol can inhibit Pgp, COX-2, and VCAM-1 expression but that its actions on Pgp are not coupled with its antiinflammatory properties.

Materials and Methods

Cell culture and hormone/drug treatment

Primary HBMECs isolated from a 30-yr-old male donor (Lot no. ACBRI 419) were purchased from Cell Systems Corp. (Kirkland, WA). HBMECs were grown in 5% CO2, room air atmosphere at 37 C, in CSC media supplemented with culture boost (Cell Systems Corp.) and 10% fetal bovine serum.

Hormone/drug treatments were performed on cells at 80 to 90% confluency and at passage 7 in which HBMECs expressed the endothelial-specific protein von Willebrand factor (data not shown). Cell treatments were carried out in hormone-free media supplemented with charcoal-stripped fetal bovine serum (Invitrogen Life Technologies, Gaithersburg, MD). A 6-h time point for COX-2 induction, 10 nm DHT dose, and a 5 ng/ml IL-1β dose were selected for these studies based on our previous studies (25, 26). A 6-h time point for Pgp induction was chosen based on a published report (21). Cells were treated with DHT (10 nm), 3β-diol (1 nm or 10 nm), 17β-estradiol (E2; 10 nm), DHEA (10 nm), Adiol (10 nm), or vehicle (VEH; 0.001% ethanol) for 18 h followed by IL-1β (5 ng/ml) for an additional 6 h. In a separate set of experiments, cells were pre-treated for 1 h with the nonselective estrogen receptor (ER) antagonist ICI 182,780 (ICI, 1 μm; Tocris Bioscience; dissolved in ethanol) followed by 18 h of cotreatment with either VEH (0.001% ethanol) or 3β-diol (10 nm), and then 6 h exposure to IL-1β (5 ng/ml). ICI doses of 1 μm were chosen based on results from previous studies (25, 26). In some studies, cells were treated with VEH or hormone alone (Adiol or 3β-diol) for 6 h to determine their effects in the absence of inflammation.

Quantitative real-time RT-PCR (RT-qPCR)

RT-qPCR was used to measure mRNA expression of gonadal steroid hormone receptors and steroid-metabolizing enzymes in adult male cells treated with hormone-free media for 6 h as previously described (33) Negative controls, where water was used in place of template, were used in all experiments. Levels of mRNA expression for each sample were determined by comparison to the standard curve and reported as the absolute concentration of cDNA. Specificity was confirmed via thermal melting curve analysis which showed a single peak at the predicted melting temperature for each primer set. The list of primers used is shown in Supplemental Table 1 published on The endocrine Society's Journals Online web site at http://endo.endojournals.org.

Western blot

Levels of COX-2, VCAM-1, or Pgp protein were examined using standard immunoblotting methods, as previously described (26). Briefly, cells were rinsed with ice-cold PBS containing 100 μm sodium orthovanadate, scraped from flasks over ice, and centrifuged at 200 × g for 10 min. The pellet was resuspended in ice-cold lysis buffer (50 mm Tris, 150 mm NaCl, 0.1% sodium dodecyl sulfate, 1% Nonidet P-40, 1 mm EGTA, 1 mm EDTA, 1 mm dithiothreitol, 20 μm Pepstatin, 20 μm Leupeptin, 0.1 U/ml Aprotinin, 0.1 mm phenylmethylsulfonyl fluoride). Cell/lysis buffer mixture was divided in half to collect the whole-cell lysate for COX-2 and VCAM-1 analysis and membrane fraction lysate for Pgp analysis separately. Whole-cell lysate was prepared as previously described (25, 33). For membrane fraction isolation, the cell/lysis buffer mixture was centrifuged at 17,000 × g for 30 min at 4 C. Pellet (crude membrane fraction) and supernatant (crude cytosolic fraction) were collected. The crude membrane fraction was used for Pgp protein analysis. After total protein content determination, samples were loaded into 7.5% Smart gels (LI-CORE), and proteins were separated via SDS-PAGE and transferred to nitrocellulose membranes. Nonspecific binding was blocked by incubation at room temperature for 30 min in PBS containing 1% Tween (TPBS) and 3% dried milk. Membranes were incubated with COX-2 (1:1000) mouse monoclonal antibody (Cayman Chemical, Ann Arbor, MI), VCAM-1 (H-276; 1:1000) rabbit antibody (Santa Cruz Biotechnology, Inc., Santa Cruz, CA), Pgp (1:500) mouse antibody (Calbiochem, Gibbstown, NJ), and/or β-actin (1:5000) mouse monoclonal antibody (Sigma Aldrich Corp., St. Louis, MO) overnight at 4 C in TPBS. After TPBS washes, the membranes were incubated in goat antimouse IR 800 Dye (1:15,000), goat antirabbit IR 680 Dye (1:15,000), secondary antibody (LI-COR), or goat antimouse IgG-horseradish peroxidase (1:5000) secondary antibody (Bio-Rad Laboratories, Hercules, CA) for 1 h at room temperature. COX-2 antibody specificity was verified with lipopolysaccharide (LPS) stimulated Raw-264.7 (mouse macrophage) cell lysate (Santa Cruz Biotechnology), which is a positive control for COX-2 protein. Endothelial cell phenotype was confirmed (data not shown) in adult male HBMECs at passage 7 with Von Willebrand factor rabbit antibody (1:500, Santa Cruz Biotechnology). After additional TPBS washes, proteins were visualized and quantitated using an Odyssey Infrared Imager, and data were analyzed using Odyssey V3.0 software (LI-COR) or using a Fujifilm Imager, and data were analyzed using the Multi gauge V3.0 Software (Fujifilm Medical Systems USA, Inc., Stamford, CT).

Reagents

All reagents were purchased from Sigma Aldrich Corp. unless otherwise noted.

Statistical analysis

Samples from each treatment group were run on the same Western blot to allow for direct comparison. For each treatment, individual measures were repeated to achieve an appropriate number for statistical analysis (n = 3-17). Data from Western blots are expressed as an optical density ratio relative to VEH and normalized to the optical density values for β-actin bands. Multiple vehicles and treatment groups were included on the same gel and normalized to the first vehicle on the gel to account for variance. Normalizing to both a vehicle and a loading control limits variance between blots and allows pooling of multiple blots from each experiment. For VCAM-1 Western blot data, the combined optical density of the doublet was used for quantification. Data from RT-qPCR studies are expressed as femtograms of cDNA, and all measures were repeated a sufficient number of times for statistical analysis (n = 3-4). All values are reported as means ± sem. Unless otherwise noted, data were compared using one-way ANOVA across treatment groups using Prism Software (Irvine, CA), and when indicated, differences were compared post hoc using Student Newman-Keuls test. A level of P < 0.05 was considered significant.

Results

Gonadal steroid receptor and steroid-metabolizing enzyme mRNAs are expressed in adult male HBMECs

To confirm the expression of necessary gonadal steroid receptors and steroid-metabolizing enzymes for DHT and 3β-diol metabolism/receptor activation, adult male HBMECs were grown in hormone-free media, and mRNA levels were measured by RT-qPCR. The size of the amplified cDNA was confirmed by 2% agarose gel electrophoresis for each primer set (data not shown). Expression of mRNA for AR, ERα, ERβ, 3α-hydroxysteroid dehydrogenase (HSD), 3β-HSD, 17β-HSD, and CYP7B1 was detected in HBMECs (Fig. 1). AR mRNA expression was greatest compared with ERα or ERβ expression (Fig. 1A). 3α-HSD mRNA expression was greatest compared with other steroid-metabolizing enzyme genes tested (Fig. 1B).

Fig. 1.

Fig. 1.

Gonadal steroid receptors and steroid-metabolizing enzyme mRNAs are expressed in male HBMECs. A, Relative levels of AR, ERα, and ERβ mRNA expression assessed by RT-qPCR in adult male HBMECs treated for 6 h with hormone-free media (n = 3-4 per group). B, Relative levels of steroid metabolism enzymes 3α-HSD, 3β-HSD, 17β-HSD, and CYP7B1 mRNA expression assessed by RT-qPCR in adult male HBMECs treated for 6 h with hormone-free media (n = 3-4 per group).

3β-diol decreased COX-2 and Pgp protein expression

To determine the effect of 3β-diol on COX-2 and Pgp expression, male HBMECs were treated for 6 h with VEH or 3β-diol (1 nm and 10 nm). Additionally, to test whether 3β-diol's effects were ER mediated, some cells were pretreated with the non-subtype-selective ER antagonist ICI (1 μm) followed by 6 h of 10 nm 3β-diol. One-way ANOVA showed that a 10 nm dose of 3β-diol significantly decreased COX-2 and Pgp protein expression (P < 0.05 vs. VEH; Fig. 2). 3β-diol's effects on Pgp, but not COX-2, were blocked by ICI.

Fig. 2.

Fig. 2.

3β-diol decreased COX-2 and Pgp expression. COX-2 and Pgp protein expression were assessed using Western analysis in adult male HBMECs treated with VEH, 1 nm 3β-diol, 10 nm 3β-diol, or 10 nm 3β-diol + ICI (1 μm; 1 h pretreatment) for 6 h. A, Representative Western blot for COX-2 and β-actin. Molecular mass is indicated in parentheses. A molecular weight standard (STD) is shown in the far left lane. B, Relative levels of COX-2 after treatment with VEH, 1 nm 3β-diol, 10 nm 3β-diol, or 10 nm 3β-diol + 1 μm ICI. C, Representative Western blot for Pgp and β-actin. Molecular weight is indicated in parentheses. D, Relative levels of Pgp after treatment with VEH, 1 nm 3β-diol, 10 nm 3β-diol, or 10 nm 3β-diol + 1 μm ICI. For panels B and D, COX-2 and Pgp expression were each normalized to β-actin and calculated as the ratio compared with VEH. For panels B and D, each bar represents the mean ± sem of three to six determinations. *, P < 0.05 vs. VEH.

DHT and 3β-diol decreased IL-1β-induced COX-2 and VCAM-1 expression

To determine whether 3β-diol could inhibit inflammatory markers induced by IL-1β, male HBMECs were treated for 18 h with VEH, DHT (10 nm), or 3β-diol (10 nm) followed by VEH or IL-1β (5 ng/ml; 6 h) in the continued presence of hormone. One-way ANOVA showed that IL-1β treatment significantly increased levels of COX-2 (P < 0.05, Fig. 3) and VCAM-1 (P < 0.05; Fig. 4). Furthermore, both DHT and 3β-diol reduced the IL-1β-induced increases in COX-2 (P < 0.05 vs. VEH + IL-1β; Fig. 3) and VCAM-1 protein (P < 0.05 vs. VEH+IL1β; Fig. 4).

Fig. 3.

Fig. 3.

DHT and 3β-diol inhibited cytokine-induced increases in COX-2. COX-2 protein expression was assessed in adult male HBMECs treated with VEH, DHT (10 nm), or 3β-diol (10 nm) for 18 h, and then exposed to IL-1β (5 ng/ml; 6 h) in the continued presence of hormone. Relative changes in COX-2 protein expression are shown after treatment with VEH, 10 nm DHT, or 10 nm 3β-diol in combination with IL-1β. COX-2 expression was normalized to β actin and reported as an intensity ratio compared with VEH-treated cells. Each bar represents the mean ± sem of six to 17 determinations. *, P < 0.05 vs. VEH; #, P < 0.05 vs. IL-1β VEH.

Fig. 4.

Fig. 4.

DHT and 3β-diol inhibited cytokine-induced increases in VCAM-1. VCAM-1 protein expression was assessed in adult male HBMECs treated with VEH, DHT (10 nm), or 3β-diol (10 nm) for 18 h, and then exposed to IL-1β (5 ng/ml; 6 h) in the continued presence of hormone. A, Representative Western blot for VCAM-1 and β actin. Numbers in parentheses indicate the calculated molecular mass of the protein shown. A molecular weight standard (STD) is shown in the far left lane. B, Relative changes in VCAM-1 protein expression after treatment with VEH, 10 nm DHT, or 10 nm 3β-diol in combination with IL-1β. VCAM-1 expression was normalized to β-actin and reported as an intensity ratio compared with VEH-treated cells. For panel B each bar represents the mean ± sem of six to 17 determinations. *, P < 0.05 vs. VEH; #, P < 0.05 vs. IL-1β VEH.

3β-diol, but not DHT or E2, inhibited cytokine-induced increases in Pgp

We sought to determine whether sex steroids would affect cytokine-induced Pgp expression in male HBMECs in a fashion similar to the inflammatory markers COX-2 and VCAM-1. HBMECs were treated for 18 h with VEH, DHT (10 nm), E2 (10 nm), or 3β-diol (10 nm), followed by VEH or IL-1β (5 ng/ml; 6 h) in the continued presence of hormone. One-way ANOVA revealed that IL-1β significantly increased Pgp protein expression (P < 0.05 vs. VEH; Fig. 5). 3β-diol, but not DHT or E2, attenuated the IL-1β-induced increases in Pgp protein (P < 0.05 vs. VEH + IL-1β; Fig. 5).

Fig. 5.

Fig. 5.

3β-diol inhibited cytokine-induced increases in Pgp. Pgp protein expression was assessed in HBMECs treated with VEH, DHT (10 nm), E2 (10 nm), or 3β-diol (10 nm) for 18 h, and then exposed to IL-1β (5 ng/ml; 6 h) in the continued presence of hormone. Relative levels of Pgp are shown after treatment with VEH, DHT, E2, or 3β-diol in combination with IL-1β. Pgp expression was normalized to β-actin and reported as an intensity ratio compared with VEH-treated cells. Each bar represents the mean ± sem of four to 11 determinations. *, P < 0.05 vs. VEH; #, P < 0.05 vs. IL-1β VEH.

3β-diol inhibits cytokine-induced increases in COX-2 and VCAM-1 via ER activation

To test ER dependence of 3β-diol's effects, male HBMECs were pretreated for 1 h with VEH or the nonselective ER antagonist ICI (1 μm), and then treated (18 h) with VEH or 3β-diol (10 nm) followed by VEH or IL-1β (5 ng/ml; 6 h) in the continued presence of hormone/antagonist. One-way ANOVA revealed that 3β-diol significantly reduced the IL-1β-induced increases in COX-2 protein (P < 0.05 vs. IL-1β/VEH), and this effect was prevented by the ER antagonist, ICI (Fig. 6A). 3β-diol also reduced the IL-1β-induced increases in VCAM-1 protein expression (P < 0.05 vs. IL-1β/VEH), an effect that was also prevented by the ER antagonist, ICI (Fig. 6B). ICI alone did not have an effect on COX-2 or VCAM-1 levels.

Fig. 6.

Fig. 6.

3β-diol inhibited cytokine-induced increases in COX-2 and VCAM-1 via ER. HBMECs were pretreated for 1 h with VEH or the nonselective ER antagonist ICI (1 μm), followed by VEH or treated with VEH or 3β-diol (10 nm) for 18 h, and then exposed to IL-1β (5 ng/ml; 6 h) in the continued presence of hormone/antagonist. A, Relative levels of COX-2 after treatment with VEH, 3β-diol, or 3β-diol + ICI in combination with IL-1β. COX-2 expression was normalized to β-actin and reported as an intensity ratio compared with VEH-treated cells. B, Relative levels of VCAM-1 after treatment with VEH, 3β-diol, or 3β-diol + ICI in combination with IL-1β. VCAM-1 expression was normalized to β-actin and reported as an intensity ratio compared with VEH-treated cells. Each bar represents the mean ± sem of five to 17 determinations (panel A) or two to 12 determinations (panel B).*, P < 0.05 vs. VEH; #, P < 0.05 vs. IL-1β VEH.

3β-diol inhibited cytokine-induced increases in Pgp independent of ER activation

To test ER dependence of 3β-diol's effects on Pgp, male HBMECs were pretreated for 1 h with VEH or the nonselective ER antagonist ICI (1 μm), and then treated (18 h) with VEH or 3β-diol (10 nm) followed by VEH or IL-1β (5 ng/ml; 6 h) in the continued presence of hormone/antagonist. 3β-diol prevented the IL-1β-induced increases in Pgp protein expression (P < 0.05 vs. IL-1β/VEH); however, this effect was not inhibited by the ER antagonist ICI (Fig. 7).

Fig. 7.

Fig. 7.

3β-diol inhibited cytokine-induced increases in Pgp independent of ER activation. Pgp protein expression was assessed in HBMECs pretreated for 1 h with VEH or the nonselective ER antagonist ICI (1 μm), followed by VEH or 3β-diol (10 nm or 100 nm) for 18 h, and then exposed to IL-1β (5 ng/ml; 6 h) in the continued presence of hormone/antagonist. Relative changes in Pgp expression are shown after treatment with VEH, 10 nm 3β-diol, 100 nm 3β-diol, or 10 nm 3β-diol + ICI in combination with IL-1β. Pgp expression was normalized to β-actin and reported as an intensity ratio compared with VEH-treated cells. Each bar represents the mean ± sem of four to 11 determinations. *, P < 0.05 vs. VEH; #, P < 0.05 vs. IL-1β VEH.

Effects of DHEA and its metabolite/ERβ agonist Adiol on cytokine-induced COX-2, VCAM-1, and Pgp

Similar to our recent findings with 3β-diol (33), it was recently reported that the DHEA metabolite, Adiol, has antiinflammatory properties in microglia and activates ERβ (38). To determine whether Adiol and DHEA have similar effects as 3β-diol on brain endothelial cells, male HBMEC were treated for 18 h with VEH, DHEA (10 nm), or Adiol (10 nm) followed by VEH or IL-1β (5 ng/ml; 6 h) in the continued presence of hormone. Neither DHEA nor Adiol altered IL-1β-induced COX-2 (Fig. 8A) or Pgp expression (Supplemental Fig. 1). Although DHEA itself did not alter IL-1β-induced VCAM-1 expression, its metabolite, Adiol, modestly decreased IL-1β-induced VCAM-1 expression (P < 0.05 vs. VEH + IL-1β; Fig. 8B).

Fig. 8.

Fig. 8.

Effects of DHEA and the DHEA metabolite/ERβ agonist Adiol on cytokine-induced increases in COX-2 and VCAM-1. Adult male HBMECs were treated with VEH, DHEA (10 nm), or Adiol (10 nm) for 18 h, and then exposed to IL-1β (5 ng/ml; 6 h) in the continued presence of hormone. A, Relative changes in COX-2 expression after treatment with VEH, DHEA, or Adiol in combination with IL-1β. COX-2 expression was normalized to β-actin and reported as an intensity ratio compared with VEH-treated cells. B, Relative changes in VCAM-1 expression after treatment with VEH, DHEA, or Adiol in combination with IL-1β. VCAM-1 expression was normalized to β-actin and reported as an intensity ratio compared with VEH-treated cells. Each bar represents the mean ± sem of three to seven determinations.*, P < 0.05 vs. VEH; #, P < 0.05 vs. IL-1β VEH.

Discussion

This study addressed the effects of the androgen DHT and its metabolite 3β-diol on vascular inflammatory markers and Pgp expression in male HBMECs and determined whether they were mechanistically linked. Using COX-2 and VCAM-1 protein expression as markers for inflammation, we examined the effects of DHT and DHEA and their metabolites, 3β-diol and Adiol, on cytokine-induced inflammation and Pgp expression. Results show that DHT decreases cytokine-induced COX-2 and VCAM-1 and that this may possibly occur via metabolism to 3β-diol and subsequent activation of ER. Interestingly, 3β-diol's effect to reduce cytokine-induced Pgp expression appears to be ER independent and not associated with the up-regulation of inflammatory markers.

In the current studies, we demonstrate that the potent androgen, DHT, reduces cytokine-induced expression of the proinflammatory mediators, COX-2 and VCAM-1, in male HBMECs. These data are supported by numerous studies showing antiinflammatory effects of androgens in a variety of cells/tissues including human monocytes (39), human macrophages (40), mouse macrophages (4143), human prostate cancer cells (44), human endothelial cells (27, 4547), human coronary artery vascular smooth muscle cells (26), and human brain vascular smooth muscle cells (25, 33). Androgen treatment in vivo has also been shown to decrease inflammatory markers (42, 48, 49).

Our results point to ERs as a potential alternate receptor pathway used by DHT during cytokine-induced inflammation in endothelial cells, because the effects of the DHT metabolite 3β-diol on COX-2 and VCAM-1 were blocked by ER antagonism. Because DHT can be converted to 3β-diol by the enzymes 3α-HSD, 3β-HSD, and 17β-HSD (35, 5154), we asked whether these enzymes are found in HBMECs. Our data show that primary HBMECs express the steroid receptors AR, ERα, ERβ, as well as enzymes involved in 3β-diol metabolism, 3α-HSD, 3β-HSD, 17β-HSD, and CYP7B1. This is consistent with previous studies showing that blood vessels contain AR, ERα, ERβ, and 3β-HSD (5558). Although we did not measure expression of these enzymes or receptors after hormone treatment or cytokine treatment, we have previously measured mRNA expression after IL-1β treatment in human brain vascular smooth muscle cells and found that ERβ expression is unchanged (33). In many cases treatment with hormones causes an up-regulation of their receptor. For example, with DHT treatment we have found that AR protein expression increases (26). 3β-diol has also been shown to increase ERβ expression (59). As for effects that inflammation may have on hormone metabolism, it has been shown that cytokine stimulation (IL-1α, IL-4) increases 3β-HSD expression and activity (60). However, it should be pointed out that changes in any one of these enzymes may not necessarily alter levels of 3β-diol because metabolism of DHT can be through the actions of several different enzymes (35, 5154).

These results also show that both DHT and 3β-diol decrease cytokine-induced COX-2 and VCAM-1 expression in HBMECs and that 3β-diol's effects are mediated via ER because a non-subtype-selective ER antagonist can block the actions of 3β-diol. These data are consistent with the recent study by Norata et al. (37) showing that 3β-diol has antiinflammatory actions in human umbilical vein endothelial cells and mouse aorta, and that these effects are mediated by ERβ. Furthermore, we have recently found that DHT reduces cytokine-induced COX-2 expression in human brain vascular smooth muscle cells via ERβ (33). Interestingly, in the absence of cytokine stimulation, 3β-diol decreases COX-2 expression and this effect is not ER mediated, whereas, during cytokine stimulation the inhibition of COX-2 expression was ER mediated. These results suggest that targeting the 3β-diol/ER pathway may be beneficial for reducing vascular inflammation. This could be accomplished therapeutically with 3β-diol treatment, or perhaps treatment with another ERβ-selective agonist or inhibition of CYP7B1, the enzyme that converts 3β-diol to inactive metabolites (35, 61).

HBMECs also express 17β-HSD. This enzyme not only converts DHT to 3β-diol, but also is the enzyme primarily responsible for the conversion of the adrenal androgen, DHEA to Adiol (62), another ERβ agonist (38). Consequently, the possibility exists that other products of androgen metabolism can have similar effects as those in the DHT/3β-diol pathway. However, our results show that at a similar dose, DHEA itself did not alter COX-2 or VCAM-1 expression in HBMECs, whereas its metabolite, Adiol, only modestly decreased cytokine-induced VCAM-1 expression via ER activation. Such data indicate that DHEA conversion to Adiol is likely not an active pathway in HBMEC cells although HBMEC cells can respond to Adiol through its ER activity.

Recent studies show that ERβ activation has a number of antiinflammatory effects in a variety of cells/tissues such as rodent microglial (63, 64), mouse brain (65), a rodent motor neuron cell line (66), astrocytes (67), rat vascular smooth muscle cells (29), and human endothelial cells (37). Saijo et al. (38) identified Adiol as an ERβ agonist that can act on microglia to reduce inflammatory markers. Further, they found that Adiol suppresses inflammatory responses by binding to ERβ and recruiting C-terminal binding protein (CtBP), a corepressor that binds to activator protein-1-dependent promoters, thereby repressing expression of proinflammatory genes. Interesting, E2 does not recruit CtBP and was unable to elicit this antiinflammatory response. CtBP recruitment is likely also required for ERβ-mediated repression of IL-1β induced proinflammatory mediators because the endotoxin LPS, their inflammatory stimulant of choice, and IL-1β act via a converging transcriptional pathway. Furthermore, they found that IL-1β treatment, like LPS treatment, activated protein kinase A, which they showed was one of the key steps in ERβ-mediated repression of proinflammatory mediators (38). Although it is possible that 3β-diol may, at a molecular level, work similarly to Adiol and recruit CtBP to inhibit inflammation, in the current studies 3β-diol was much more effective than Adiol, suggesting that its binding to ER may induce a more effective conformation change to attract CtBP or an alternate set of coregulatory proteins to the transcriptional machinery in HBMECs. Nonetheless, these studies show that ERβ activation has similar protective effects as do androgens and support the theory that some of the beneficial effects of androgens, particularly those associated with antiinflammatory responses, may occur through activation of ERβ. Whether the ER-dependent effects of 3β-diol we observed in the current studies are mediated by ERβ is yet to be determined. Based upon previous studies in other human vascular cell types, including brain vascular smooth muscle (33) and umbilical vein endothelial cells (37), suggesting ERβ receptor subtype involvement leads us to suspect that ERβ is involved. However, future studies using ERβ-selective antagonists or ERβ knockdown would need to be performed to be certain that 3β-diol is working through ERβ.

Our current data show that the cytokine IL-1β increases protein expression of Pgp in a fashion similar to that reported by others, in epithelial cells (18), rat astrocytes (23), a human brain endothelial cell line (6), rat brain endothelial cells (19), and isolated rat brain capillaries (20, 21). This is consistent with the effects elicited by other cytokines such as TNFα, which has been shown to increase Pgp protein and mRNA expression in a human brain endothelial cell line (6) and to increase Pgp protein expression and activity in rat brain capillaries (21).

To our knowledge, this is the first report showing effects of 3β-diol on any multiple drug-resistance transporter in any cell type. We have identified the DHT metabolite 3β-diol, but not DHT itself, as an inhibitor of Pgp expression in HBMECs. Thus, although all enzymes necessary for the conversion of DHT to 3β-diol are present in HBMECs, there may be insufficient levels to convert DHT at a rate sufficiently high to affect Pgp expression. Moreover, the ability of 3β-diol to inhibit Pgp expression occurred in HBMECs even in the absence of an inflammatory signal such as IL-1β. Thus, although it appears that 3β-diol is capable of preventing the inflammatory mediated increase in Pgp, it is more likely that 3β-diol's effect on Pgp expression occurs independent of its effect on COX2 inhibition. Support for this comes from our results after treatment with 3β-diol alone, showing that the effect on Pgp expression is blocked by ICI 182,780, but the effect on COX-2 is not. Furthermore, treatments with 3β-diol inhibited the IL-1β-induced up-regulation of Pgp, and cotreatment with ICI 182,780 decreased the effects of 3β-diol on IL-1β-induced COX-2 expression but did not alter IL-1β induced Pgp expression. In both cases, there was a disassociation of inflammatory marker regulation and of Pgp indicating that their expressions are similar but independently controlled. Interestingly, although 3β-diol was able to inhibit Pgp expression, E2 was not. Another recent paper showed that protective effects of 3β-diol to reduce cell proliferation, cell adhesion, and tumor invasion, which were mediated via ERβ, also could not be mimicked by E2 (68). In addition, we recently showed that recruitment of steroid receptor coactivator 1 and cAMP response element-binding protein-binding protein to the composite response element in the oxytocin promoter occurs after 3β-diol, but not after E2 treatment (69). Further, when examining the regulation of the vasopressin promoter by 3β-diol, occupancy of ERβ by 3β-diol is more effective in inducing transcriptional activity than E2 (70).

In previous studies, we found that DHT decreases COX-2 expression via an AR-independent mechanism in human brain vascular smooth muscle cells in the presence of an inflammatory stimulus (25, 26, 33). Further, we have recently demonstrated that the effect of DHT on COX-2 expression in human vascular smooth muscle cells is mediated by ERβ (33). Our current data support and extend those data in that we show that DHT's antiinflammatory effects in vascular endothelial cells are likely also occurring via metabolism to 3β-diol and activation of ERs. Because COX-2 has been shown to mediate the increases in Pgp expression that are seen after epileptic seizures (15, 24), it would be congruent to suggest that 3β-diol's effect to decrease Pgp in HBMECs could also be COX-2 mediated. However, several observations suggest that this is not the case. After IL-1β administration, 3β-diol inhibited Pgp expression to a much greater degree than it had on COX-2 and VCAM-1 expression. Further, ICI did not block the actions of 3β-diol on Pgp expression, whereas it prevented its actions on COX2 and VCAM-1. Because 3β-diol's effect on Pgp was not blocked by ICI, it is possible that in this case 3β-diol may be acting via a nongenomic mechanism that is not blocked by ER antagonists. This might involve GPR30 or other membrane-bound estrogen receptors, even potentially membrane-bound ERβ (7173). Metabolism to other steroids is also possible (74). Taken together, these data suggest that 3β-diol's ability to block Pgp expression is independent of its actions as an antiinflammatory agent.

Previous studies have shown that Pgp expression can be regulated by steroid hormones. Kim and Benet (75), showed that ethinyl estradiol can increase Pgp and MDR1 expression levels by up to 3-fold (75). Progesterone and E2 have also been shown to up-regulate Pgp expression via transcriptional regulation (76). E2, working via ERβ, has been shown to down-regulate Pgp protein expression via a posttranslational mechanism in human breast cancer cells (77). However, another study showed that E2 treatment can increase Pgp expression in breast cancer cells via ERα (78). Further, hormones such as glucocorticoids have been described as targets for Pgp-mediated transport (79). Thus the possibility exists that changes in gonadal steroid hormones or their metabolites may alter sensitivity to glucocorticoids in the brain through changes in Pgp expression.

In the present studies, the inability of ICI to block 3β-diol's effect on Pgp expression during IL-1β stimulation suggests that, in the presence of an inflammatory stimulus such as IL-1β, 3β-diol is acting through an ER-independent mechanism. A similar mechanism has been reported for E2's effect on another gene encoding a multiple drug resistance transporter, breast cancer-related protein. It was found that E2 rapidly decreases breast cancer-related protein activity via nongenomic signaling in mouse brain capillaries, and that this effect could not be blocked with the nonselective ER antagonist ICI (80). However, Hartz et al., (80) examined only short-term (90 min or less) E2 effects, so alternative genomic effects after longer treatment time may have been missed (80). Similarly, our studies did not examine Pgp regulation after acute exposure to hormones. Because the current studies show that E2 did not alter cytokine-induced Pgp expression, it is likely that the effect of E2 on Pgp expression varies based on experimental conditions/cell types and would not be expected to correspond to changes seen after 3β-diol.

The functional significance for the up-regulation of Pgp expression by cytokines, and the down-regulation after 3β-diol treatment is not currently known. Up-regulation of Pgp would indicate a decrease in BBB permeability, although this has not been tested under the conditions used in the current study. However, because Pgp does not work alone in controlling efflux across the BBB, it is possible that it represents a compensatory change in response to alterations in other ABC cassette transporters. Up-regulation of Pgp may also be working to prevent permeability of substances that can be neurotoxic or neuroendangering. An example of these are the glucocorticoid hormones. Synthetic glucocorticoids such as prednisone (81), dexamethasone (82), and endogenous hormones such as progesterone and cortisol have been suggested to be targets for Pgp (79, 83) and may alter the negative feedback regulation of the hypothalamo-pituitary-adrenal axis (50), particularly because Pgp restricts accumulation of radiolabeled cortisol in hypothalamus, but not other brain areas (82). However, recent studies suggest that this is not always the case (85, 86) and ultimately, transport mechanisms may be tissue specific.

This study used primary cultures of HBMECs to investigate the effects of sex steroids on vascular inflammation and Pgp expression. This model may be more translationally relevant compared with studies using rodent models; however, there are certain limitations with the use of human primary cell cultures. Although these cells originated from a single donor, our previous studies in human coronary artery vascular smooth muscle cells, originating from a different donor, also showed that DHT attenuated IL-1β-induced COX-2 via an AR-independent mechanism (26). Further, we have found that DHT decreases COX-2 expression in response to hypoxia with glucose deprivation in human brain vascular smooth muscle cells, rat pial arteries, and in the human brain vascular smooth muscle cells. The antiinflammatory effect of DHT was also AR independent (25). Our findings correspond with a previous study showing that 3β-diol decreased cytokine-induced vascular inflammation via an ER-dependent mechanism in human umbilical vein endothelial cells (37). Together, these studies minimize the concern of using cells isolated from a single donor. A second limitation of using cultured primary cells that should be noted is that their properties can change over time. Therefore, to minimize the risk of culture-induced changes, all experiments were performed after a small number of passages (7) when the HBMEC still expressed the endothelial cell marker, von Willebrand factor (data not shown). The third limitation of this study is that responses were examined in male cells. There have been reports of sex differences in inflammatory responses (87, 88) and in future studies we hope to determine whether the DHT/3β-diol/ER pathway also plays a role in the female response to vascular inflammation.

In summary, we have shown that DHT, 3β-diol, and Adiol decrease cytokine-induced vascular inflammatory markers in male HBMECs and that 3β-diol decreases Pgp expression in the presence and absence of IL1-β. Although 3β-diol's antiinflammatory effects during cytokine stimulation occur via an ER-dependent mechanism, its ability to decrease Pgp expression appears to be ER independent. Thus, the results of these studies have identified an ER-dependent antiinflammatory effect for DHT in vascular endothelial cells by the DHT metabolite 3β-diol, and the DHEA metabolite Adiol. Further, we have identified 3β-diol, a naturally occurring metabolite of DHT, as a steroid that can down-regulate Pgp expression and potentially influence the function of the BBB. Because Pgp functions to limit the uptake of many therapeutic drugs (46) and hormones (79, 83), the ability to down-regulate its expression could have a major impact on the efficacy of therapeutic approaches to manage brain diseases or traumatic insults as well as inhibit any accompanying inflammatory responses.

Supplementary Material

Supplemental Data

Acknowledgments

We thank Dr. Seibal Dey (Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD) for technical assistance with Pgp assays.

This work was supported by American Heart Association Predoctoral Fellowship (to K.L.Z.), Doris Griswold Research Award for Novel Projects in the Area of Cardiovascular Disease and Medicine from the University of Arizona Sarver Heart Center (to K.L.Z.; R.J.G., SHC member), Department of Defense DMRDP no. D61_I_10_J6_125_ (to T.J.W.) and National Institutes of Health Grants MH082679 and NS039951 (to R.J.H.).

Disclosure Summary: There is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

Footnotes

Abbreviations:
ABC
ATP-binding cassette
Adiol
5α-androstene-3β,17β diol
AR
androgen receptor
BBB
blood-brain barrier
COX-2
cyclooxygenase-2
CtBP
C-terminal binding protein
DHEA
dehydroepiandrosterone
DHT
dihydrotestosterone
3β-diol
5α-androstane-3β,17β-diol
E2
17β-estradiol
ER
estrogen receptor
HBMECs
human brain microvascular endothelial cells
HSD
hydroxysteroid dehydrogenase
ICI
ICI-182,780
LPS
lipopolysaccharide
Pgp
P-glycoprotein
RT-qPCR
quantitative real-time RT-PCR
TPBS
PBS containing 1% Tween
VCAM-1
vascular cell adhesion molecule-1
VEH
vehicle.

References

  • 1. Miller DS. 2010. Regulation of P-glycoprotein and other ABC drug transporters at the blood-brain barrier. Trends Pharmacol Sci 31:246-254 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Schinkel AH, Jonker JW. 2003. Mammalian drug efflux transporters of the ATP binding cassette (ABC) family: an overview. Adv Drug Deliv Rev 55:3-29 [DOI] [PubMed] [Google Scholar]
  • 3. Cordon-Cardo C, O'Brien JP, Casals D, Rittman-Grauer L, Biedler JL, Melamed MR, Bertino JR. 1989. Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc Natl Acad Sci USA 86:695-698 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Schinkel AH, Wagenaar E, van Deemter L, Mol CA, Borst P. 1995. Absence of the mdr1a P-Glycoprotein in mice affects tissue distribution and pharmacokinetics of dexamethasone, digoxin, and cyclosporin A. J Clin Invest 96:1698-1705 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Löscher W, Potschka H. 2005. Blood-brain barrier active efflux transporters: ATP-binding cassette gene family. NeuroRx 2:86-98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Poller B, Drewe J, Krahenbühl S, Huwyler J, Gutmann H. 2010. Regulation of BCRP (ABCG2) and P-glycoprotein (ABCB1) by cytokines in a model of the human blood-brain barrier. Cell Mol Neurobiol 30:63-70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Roberts DJ, Goralski KB. 2008. A critical overview of the influence of inflammation and infection on P-glycoprotein expression and activity in the brain. Expert Opin Drug Metab Toxicol 4:1245-1264 [DOI] [PubMed] [Google Scholar]
  • 8. Fellner S, Bauer B, Miller DS, Schaffrik M, Fankhänel M, Spruss T, Bernhardt G, Graeff C, Färber L, Gschaidmeier H, Buschauer A, Fricker G. 2002. Transport of paclitaxel (Taxol) across the blood-brain barrier in vitro and in vivo. J Clin Invest 110:1309-1318 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Kemper EM, van Zandbergen AE, Cleypool C, Mos HA, Boogerd W, Beijnen JH, van Tellingen O. 2003. Increased penetration of paclitaxel into the brain by inhibition of P-glycoprotein. Clin Cancer Res 9:2849-2855 [PubMed] [Google Scholar]
  • 10. Kemper EM, Verheij M, Boogerd W, Beijnen JH, van Tellingen O. 2004. Improved penetration of docetaxel into the brain by co-administration of inhibitors of P-glycoprotein. Eur J Cancer 40:1269-1274 [DOI] [PubMed] [Google Scholar]
  • 11. Hawkins BT, Davis TP. 2005. The blood-brain barrier/neurovascular unit in health and disease. Pharmacol Rev 57:173-185 [DOI] [PubMed] [Google Scholar]
  • 12. Abbott NJ. 2000. Inflammatory mediators and modulation of blood brain barrier permeability. Cell Mol Neurobiol 20:131-147 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. del Zoppo GJ, Mabuchi T. 2003. Cerebral microvessel responses to focal ischemia. J Cereb Blood Flow Metab 23:879-894 [DOI] [PubMed] [Google Scholar]
  • 14. Emsley HC, Tyrrell PJ. 2002. Inflammation and infection in clinical stroke. J Cereb Blood Flow Metab 22:1399-1419 [DOI] [PubMed] [Google Scholar]
  • 15. Bauer B, Hartz AM, Pekcec A, Toellner K, Miller DS, Potschka H. 2008. Seizure-induced up-regulation of P-glycoprotein at the blood-brain barrier through glutamate and cyclooxygenase-2 signaling. Mol Pharmacol 73:1444-1453 [DOI] [PubMed] [Google Scholar]
  • 16. Morganti-Kossmann MC, Rancan M, Stahel PF, Kossmann T. 2002. Inflammatory response in acute traumatic brain injury: a double-edged sword. Curr Opin Crit Care 8:101-105 [DOI] [PubMed] [Google Scholar]
  • 17. Petty MA, Wettstein JG. 2001. Elements of cerebral microvascular ischaemia. Brain Res Rev 36:23-34 [DOI] [PubMed] [Google Scholar]
  • 18. Al-Bataineh MM, van der Merwe D, Schultz BD, Gehring R. 2010. Tumor necrosis factor α increases P-glycoprotein expression in a BME-UV in vitro model of mammary epithelial cells. Biopharm Drug Dispos 31:506-515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Robertson S, Mokgokong R, Kania K, Guedj AS, Hladky S, Barrand M. 2011. Nitric oxide contributes to hypoxia-reoxygenation-induced P-glycoprotein expression in rat brain endothelial cells. Cell Mol Neurobiol 31:1103-1111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Hartz AM, Bauer B, Block ML, Hong JS, Miller DS. 2008. Diesel exhaust particles induce oxidative stress, proinflammatory signaling, and P-glycoprotein up-regulation at the blood-brain barrier. FASEB J 22:2723-2733 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Bauer B, Hartz AM, Miller DS. 2007. Tumor necrosis factor α and endothelin-1 increase P-glycoprotein expression and transport activity at the blood-brain barrier. Mol Pharmacol 71:667-675 [DOI] [PubMed] [Google Scholar]
  • 22. Yu C, Kastin AJ, Tu H, Waters S, Pan W. 2007. TNF activates P-glycoprotein in cerebral microvascular endothelial cells. Cell Physiol Biochem 20:853-858 [DOI] [PubMed] [Google Scholar]
  • 23. Ronaldson PT, Bendayan R. 2006. HIV-1 viral envelope glycoprotein gp120 triggers an inflammatory response in cultured rat astrocytes and regulates the functional expression of P-glycoprotein. Mol Pharmacol 70:1087-1098 [DOI] [PubMed] [Google Scholar]
  • 24. Zibell G, Unkrüer B, Pekcec A, Hartz AM, Bauer B, Miller DS, Potschka H. 2009. Prevention of seizure-induced up-regulation of endothelial P-glycoprotein by COX-2 inhibition. Neuropharmacology 56:849-855 [DOI] [PubMed] [Google Scholar]
  • 25. Zuloaga KL, Gonzales RJ. 2011. Dihydrotestosterone attenuates hypoxia inducible factor-1α and cyclooxygenase-2 in cerebral arteries during hypoxia or hypoxia with glucose deprivation. Am J Physiol Heart Circ Physiol 301:H1882–H1890 [DOI] [PubMed] [Google Scholar]
  • 26. Osterlund KL, Handa RJ, Gonzales RJ. 2010. Dihydrotestosterone alters cyclooxygenase-2 levels in human coronary artery smooth muscle cells. Am J Physiol Endocrinol Metab 298:E838–E845 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Norata GD, Tibolla G, Seccomandi PM, Poletti A, Catapano AL. 2006. Dihydrotestosterone decreases tumor necrosis factor-α and lipopolysaccharide-induced inflammatory response in human endothelial cells. J Clin Endocrinol Metab 91:546-554 [DOI] [PubMed] [Google Scholar]
  • 28. Malkin CJ, Pugh PJ, Jones RD, Jones TH, Channer KS. 2003. Testosterone as a protective factor against atherosclerosis-immunomodulation and influence upon plaque development and stability. J Endocrinol 178:373-380 [DOI] [PubMed] [Google Scholar]
  • 29. Xing D, Feng W, Miller AP, Weathington NM, Chen YF, Novak L, Blalock JE, Oparil S. 2007. Estrogen modulates TNF-α-induced inflammatory responses in rat aortic smooth muscle cells through estrogen receptor-β activation. Am J Physiol Heart Circ Physiol 292:H2607–H2612 [DOI] [PubMed] [Google Scholar]
  • 30. Death AK, McGrath KC, Sader MA, Nakhla S, Jessup W, Handelsman DJ, Celermajer DS. 2004. Dihydrotestosterone promotes vascular cell adhesion molecule-1 expression in male human endothelial cells via a nuclear factor-κB-dependent pathway. Endocrinology 145:1889-1897 [DOI] [PubMed] [Google Scholar]
  • 31. McCrohon JA, Jessup W, Handelsman DJ, Celermajer DS. 1999. Androgen exposure increases human monocyte adhesion to vascular endothelium and endothelial cell expression of vascular cell adhesion molecule-1. Circulation 99:2317-2322 [DOI] [PubMed] [Google Scholar]
  • 32. Gonzales RJ, Duckles SP, Krause DN. 2009. Dihydrotestosterone stimulates cerebrovascular inflammation through NFκB, modulating contractile function. J Cereb Blood Flow Metab 29:244-253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Zuloaga KL, O'Connor DT, Handa RJ, Gonzales RJ. 2012. Estrogen receptor β dependent attenuation of cytokine-induced cyclooxygenase-2 by androgens in human brain vascular smooth muscle cells and rat mesenteric arteries. Steroids 77:835-844 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA. 1996. Cloning of a novel receptor expressed in rat prostate and ovary. Proc Natl Acad Sci USA 93:5925-5930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Weihua Z, Lathe R, Warner M, Gustafsson JA. 2002. An endocrine pathway in the prostate, ERβ, AR, 5α-androstane-3β,17β-diol, and CYP7B1, regulates prostate growth. Proc Natl Acad Sci USA 99:13589-13594 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Kuiper GG, Carlsson B, Grandien K, Enmark E, Häggblad J, Nilsson S, Gustafsson JA. 1997. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors α and β. Endocrinology 138:863-870 [DOI] [PubMed] [Google Scholar]
  • 37. Norata GD, Cattaneo P, Poletti A, Catapano AL. 2010. The androgen derivative 5α-androstane-3β,17β-diol inhibits tumor necrosis factor α and lipopolysaccharide induced inflammatory response in human endothelial cells and in mice aorta. Atherosclerosis 212:100-106 [DOI] [PubMed] [Google Scholar]
  • 38. Saijo K, Collier JG, Li AC, Katzenellenbogen JA, Glass CK. 2011. An ADIOL-ER[β]-CtBP transrepression pathway negatively regulates microglia-mediated inflammation. Cell 145:584-595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Li ZG, Danis VA, Brooks PM. 1993. Effect of gonadal steroids on the production of IL-1 and IL-6 by blood mononuclear cells in vitro. Clin Exp Rheumatol 11:157-162 [PubMed] [Google Scholar]
  • 40. Corcoran MP, Meydani M, Lichtenstein AH, Schaefer EJ, Dillard A, Lamon-Fava S. 2010. Sex hormone modulation of proinflammatory cytokine and C-reactive protein expression in macrophages from older men and postmenopausal women. J Endocrinol 206:217-224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Padgett DA, Loria RM. 1998. Endocrine regulation of murine macrophage function: effects of dehydroepiandrosterone, androstenediol, and androstenetriol. J Neuroimmunol 84:61-68 [DOI] [PubMed] [Google Scholar]
  • 42. Qiu Y, Yanase T, Hu H, Tanaka T, Nishi Y, Liu M, Sueishi K, Sawamura T, Nawata H. 2010. Dihydrotestosterone suppresses foam cell formation and attenuates atherosclerosis development. Endocrinology 151:3307-3316 [DOI] [PubMed] [Google Scholar]
  • 43. Rettew JA, Huet-Hudson YM, Marriott I. 2008. Testosterone reduces macrophage expression in the mouse of toll-like receptor 4, a trigger for inflammation and innate immunity. Biol Reprod 78:432-437 [DOI] [PubMed] [Google Scholar]
  • 44. Keller ET, Chang C, Ershler WB. 1996. Inhibition of NFκB activity through maintenance of IκBα levels contributes to dihydrotestosterone-mediated repression of the interleukin-6 promoter. J Biol Chem 271:26267-26275 [DOI] [PubMed] [Google Scholar]
  • 45. Hatakeyama H, Nishizawa M, Nakagawa A, Nakano S, Kigoshi T, Uchida K. 2002. Testosterone inhibits tumor necrosis factor-α-induced vascular cell adhesion molecule-1 expression in human aortic endothelial cells. FEBS Lett 530:129-132 [DOI] [PubMed] [Google Scholar]
  • 46. Jin H, Qiu WB, Mei YF, Wang DM, Li YG, Tan XR. 2009. Testosterone alleviates tumor necrosis factor-α-mediated tissue factor pathway inhibitor downregulation via suppression of nuclear factor-κ B in endothelial cells. Asian J Androl 11:266-271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Mukherjee TK, Dinh H, Chaudhuri G, Nathan L. 2002. Testosterone attenuates expression of vascular cell adhesion molecule-1 by conversion to estradiol by aromatase in endothelial cells: implications in atherosclerosis. Proc Natl Acad Sci USA 99:4055-4060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Malkin CJ, Pugh PJ, Jones RD, Kapoor D, Channer KS, Jones TH. 2004. The effect of testosterone replacement on endogenous inflammatory cytokines and lipid profiles in hypogonadal men. J Clin Endocrinol Metab 89:3313-3318 [DOI] [PubMed] [Google Scholar]
  • 49. Kalinchenko SY, Tishova YA, Mskhalaya GJ, Gooren LJ, Giltay EJ, Saad F. 2010. Effects of testosterone supplementation on markers of the metabolic syndrome and inflammation in hypogonadal men with the metabolic syndrome: the double-blinded placebo-controlled Moscow study. Clin Endocrinol (Oxf) 73:602-612 [DOI] [PubMed] [Google Scholar]
  • 50. Müller MB, Keck ME, Binder EB, Kresse AE, Hagemeyer TP, Landgraf R, Holsboer F, Uhr M. 2003. ABCB1 (MDR1)-type P-glycoproteins at the blood-brain barrier modulate the activity of the hypothalamic-pituitary-adrenocortical system: implications for affective disorder. Neuropsychopharmacology 28:1991-1999 [DOI] [PubMed] [Google Scholar]
  • 51. Gangloff A, Shi R, Nahoum V, Lin SX. 2003. Pseudo-symmetry of C19 steroids, alternative binding orientations, and multispecificity in human estrogenic 17β-hydroxysteroid dehydrogenase. FASEB J 17:274-276 [DOI] [PubMed] [Google Scholar]
  • 52. Törn S, Nokelainen P, Kurkela R, Pulkka A, Menjivar M, Ghosh S, Coca-Prados M, Peltoketo H, Isomaa V, Vihko P. 2003. Production, purification, and functional analysis of recombinant human and mouse 17β-hydroxysteroid dehydrogenase type 7. Biochem Biophys Res Commun 305:37-45 [DOI] [PubMed] [Google Scholar]
  • 53. Jin Y, Penning TM. 2001. Steroid 5α-reductases and 3α-hydroxysteroid dehydrogenases: key enzymes in androgen metabolism. Best Pract Res Clin Endocrinol Metab 15:79-94 [DOI] [PubMed] [Google Scholar]
  • 54. Steckelbroeck S, Jin Y, Gopishetty S, Oyesanmi B, Penning TM. 2004. Human cytosolic 3α-hydroxysteroid dehydrogenases of the aldo-keto reductase superfamily display significant 3β-hydroxysteroid dehydrogenase activity: implications for steroid hormone metabolism and action. J Biol Chem 279:10784-10795 [DOI] [PubMed] [Google Scholar]
  • 55. Gonzales RJ, Ansar S, Duckles SP, Krause DN. 2007. Androgenic/estrogenic balance in the male rat cerebral circulation: metabolic enzymes and sex steroid receptors. J Cereb Blood Flow Metab 27:1841-1852 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Snyder GD, Krishna UM, Falck JR, Spector AA. 2002. Evidence for a membrane site of action for 14,15-EET on expression of aromatase in vascular smooth muscle. Am J Physiol Heart Circ Physiol 283:H1936–H1942 [DOI] [PubMed] [Google Scholar]
  • 57. Shih HC, Lin CL, Wu SC, Kwan AL, Hong YR, Howng SL. 2008. Upregulation of estrogen receptor α and mediation of 17β-estradiol vasoprotective effects via estrogen receptor α in basilar arteries in rats after experimental subarachnoid hemorrhage. J Neurosurg 109:92-99 [DOI] [PubMed] [Google Scholar]
  • 58. Nakamura Y, Suzuki T, Inoue T, Tazawa C, Moriya T, Saito H, Ishibashi T, Takahashi S, Yamada S, Sasano H. 2005. 3β-Hydroxysteroid dehydrogenase in human aorta. Endocr J 52:111-115 [DOI] [PubMed] [Google Scholar]
  • 59. Oliveira AG, Coelho PH, Guedes FD, Mahecha GA, Hess RA, Oliveira CA. 2007. 5α-Androstane-3β,17β-diol (3β-diol), an estrogenic metabolite of 5α-dihydrotestosterone, is a potent modulator of estrogen receptor ERβ expression in the ventral prostrate of adult rats. Steroids 72:914-922 [DOI] [PubMed] [Google Scholar]
  • 60. Papacleovoulou G, Hogg K, Fegan KS, Critchley HO, Hillier SG, Mason JI. 2009. Regulation of 3β-hydroxysteroid dehydrogenase type 1 and type 2 gene expression and function in the human ovarian surface epithelium by cytokines. Mol Hum Reprod 15:379-392 [DOI] [PubMed] [Google Scholar]
  • 61. Sundin M, Warner M, Haaparanta T, Gustafsson JA. 1987. Isolation and catalytic activity of cytochrome P-450 from ventral prostate of control rats. J Biol Chem 262:12293-12297 [PubMed] [Google Scholar]
  • 62. Lin SX, Shi R, Qiu W, Azzi A, Zhu DW, Dabbagh HA, Zhou M. 2006. Structural basis of the multispecificity demonstrated by 17β-hydroxysteroid dehydrogenase types 1 and 5. Mol Cell Endocrinol 248:38-46 [DOI] [PubMed] [Google Scholar]
  • 63. Baker AE, Brautigam VM, Watters JJ. 2004. Estrogen modulates microglial inflammatory mediator production via interactions with estrogen receptor β. Endocrinology 145:5021-5032 [DOI] [PubMed] [Google Scholar]
  • 64. Smith JA, Das A, Butler JT, Ray SK, Banik NL. 2011. Estrogen or estrogen receptor agonist inhibits lipopolysaccharide induced microglial activation and death. Neurochem Res 36:1587-1593 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Brown CM, Mulcahey TA, Filipek NC, Wise PM. 2010. Production of proinflammatory cytokines and chemokines during neuroinflammation: novel roles for estrogen receptors α and β. Endocrinology 151:4916-4925 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Das A, Smith JA, Gibson C, Varma AK, Ray SK, Banik NL. 2011. Estrogen receptor agonists and estrogen attenuate TNF-α-induced apoptosis in VSC4.1 motoneurons. J Endocrinol 208:171-182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Lewis DK, Johnson AB, Stohlgren S, Harms A, Sohrabji F. 2008. Effects of estrogen receptor agonists on regulation of the inflammatory response in astrocytes from young adult and middle-aged female rats. J Neuroimmunol 195:47-59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Dondi D, Piccolella M, Biserni A, Della Torre S, Ramachandran B, Locatelli A, Rusmini P, Sau D, Caruso D, Maggi A, Ciana P, Poletti A. 2010. Estrogen receptor β and the progression of prostate cancer: role of 5α-androstane-3β,17β-diol. Endocr Relat Cancer 17:731-742 [DOI] [PubMed] [Google Scholar]
  • 69. Sharma D, Handa RJ, Uht RM. 2012. The ERβ ligand 5α-androstane, 3β,17β-diol (3β-diol) regulates hypothalamic oxytocin (Oxt) gene expression. Endocrinology 153:2353-2361 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Pak TR, Chung WC, Hinds LR, Handa RJ. 2007. Estrogen receptor-β mediates dihydrotestosterone-induced stimulation of the arginine vasopressin promoter in neuronal cells. Endocrinology 148:3371-3382 [DOI] [PubMed] [Google Scholar]
  • 71. Nilsson BO, Olde B, Leeb-Lundberg LM. 2011. G protein-coupled oestrogen receptor 1 (GPER1)/GPR30: a new player in cardiovascular and metabolic oestrogenic signalling. Br J Pharmacol 163:1131-1139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Meyer MR, Haas E, Prossnitz ER, Barton M. 2009. Non-genomic regulation of vascular cell function and growth by estrogen. Mol Cell Endocrinol 308:9-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Razandi M, Pedram A, Merchenthaler I, Greene GL, Levin ER. 2004. Plasma membrane estrogen receptors exist and functions as dimers. Mol Endocrinol 18:2854-2865 [DOI] [PubMed] [Google Scholar]
  • 74. Perusquía M, Stallone JN. 2010. Do androgens play a beneficial role in the regulation of vascular tone? Nongenomic vascular effects of testosterone metabolites. Am J Physiol Heart Circ Physiol 298:H1301–H1307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Kim WY, Benet LZ. 2004. P-glycoprotein (P-gp/MDR1)-mediated efflux of sex-steroid hormones and modulation of P-gp expression in vitro. Pharm Res 21:1284-1293 [DOI] [PubMed] [Google Scholar]
  • 76. Coles LD, Lee IJ, Voulalas PJ, Eddington ND. 2009. Estradiol and progesterone-mediated regulation of P-gp in P-gp overexpressing cells (NCI-ADR-RES) and placental cells (JAR). Mol Pharm 6:1816-1825 [DOI] [PubMed] [Google Scholar]
  • 77. Mutoh K, Tsukahara S, Mitsuhashi J, Katayama K, Sugimoto Y. 2006. Estrogen-mediated post transcriptional down-regulation of P-glycoprotein in MDR1-transduced human breast cancer cells. Cancer Sci 97:1198-1204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Zampieri L, Bianchi P, Ruff P, Arbuthnot P. 2002. Differential modulation by estradiol of P-glycoprotein drug resistance protein expression in cultured MCF7 and T47D breast cancer cells. Anticancer Res 22:2253-2259 [PubMed] [Google Scholar]
  • 79. Uhr M, Holsboer F, Müller MB. 2002. Penetration of endogenous steroid hormones corticosterone, cortisol, aldosterone and progesterone into the brain is enhanced in mice deficient for both mdr1a and mdr1b P-glycoproteins. J Neuroendocrinol 14:753-759 [DOI] [PubMed] [Google Scholar]
  • 80. Hartz AM, Mahringer A, Miller DS, Bauer B. 2010. 17-β-Estradiol: a powerful modulator of blood-brain barrier BCRP activity. J Cereb Blood Flow Metab 30:1742-1755 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Karssen AM, Meijer OC, van der Sandt IC, De Boer AG, De Lange EC, De Kloet ER. 2002. The role of the efflux transporter P-glycoprotein in brain penetration of prednisolone. J Endocrinol 175:251-260 [DOI] [PubMed] [Google Scholar]
  • 82. Mason BL, Pariante CM, Thomas SA. 2008. A revised role for P-glycoprotein in the brain distribution of dexamethasone, cortisol, and corticosterone in wild-type and ABCB1A/B-deficient mice. Endocrinology 149:5244-5253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Karssen AM, Meijer OC, van der Sandt IC, Lucassen PJ, de Lange EC, de Boer AG, de Kloet ER. 2001. Multidrug resistance P-glycoprotein hampers the access of cortisol but not of corticosterone to mouse and human brain. Endocrinology 142:2686-2694 [DOI] [PubMed] [Google Scholar]
  • 84. Mason BL, Pariante CM, Jamel S, Thomas SA. 2010. Central nervous system (CNS) delivery of glucocorticoids is fine-tuned by saturable transporters at the blood-CNS barriers and nonbarrier regions. Endocrinology 151:5294-5305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Heinzmann JM, Thoeringer CK, Knapman A, Palme R, Holsboer F, Uhr M, Landgraf R, Touma C. 2010. Intrahippocampal corticosterone response in mice selectively bred for extremes in stress reactivity: a microdialysis study. J Neuroendocrinol 22:1187-1197 [DOI] [PubMed] [Google Scholar]
  • 86. Card JW, Carey MA, Bradbury JA, DeGraff LM, Morgan DL, Moorman MP, Flake GP, Zeldin DC. 2006. Gender differences in murine airway responsiveness and lipopolysaccharide-induced inflammation. J Immunol 177:621-630 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Siegel C, Turtzo C, McCullough LD. 2010. Sex differences in cerebral ischemia: possible molecular mechanisms. J Neurosci Res 88:2765-2774 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Data

Articles from Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES