Hsa-miRNA-765 as a key mediator for inhibiting growth, migration and invasion in fulvestrant-treated prostate cancer

Yuet-Kin Leung, Queeny Kwan-Yi Chan, Chi-Fai Ng, Fanny Man-Ting Ma, Ho-Man Tse, Ka-Fai To, Jodi Maranchie, Shuk-Mei Ho, Kin-Mang Lau, Yuet-Kin Leung, Queeny Kwan-Yi Chan, Chi-Fai Ng, Fanny Man-Ting Ma, Ho-Man Tse, Ka-Fai To, Jodi Maranchie, Shuk-Mei Ho, Kin-Mang Lau

Abstract

Fulvestrant (ICI-182,780) has recently been shown to effectively suppress prostate cancer cell growth in vitro and in vivo. But it is unclear whether microRNAs play a role in regulating oncogene expression in fulvestrant-treated prostate cancer. Here, this study reports hsa-miR-765 as the first fulvestrant-driven, ERβ-regulated miRNA exhibiting significant tumor suppressor activities like fulvestrant, against prostate cancer cell growth via blockage of cell-cycle progression at the G2/M transition, and cell migration and invasion possibly via reduction of filopodia/intense stress-fiber formation. Fulvestrant was shown to upregulate hsa-miR-765 expression through recruitment of ERβ to the 5'-regulatory-region of hsa-miR-765. HMGA1, an oncogenic protein in prostate cancer, was identified as a downstream target of hsa-miR-765 and fulvestrant in cell-based experiments and a clinical study. Both the antiestrogen and the hsa-miR-765 mimic suppressed HMGA1 protein expression. In a neo-adjuvant study, levels of hsa-miR-765 were increased and HMGA1 expression was almost completely lost in prostate cancer specimens from patients treated with a single dose (250 mg) of fulvestrant 28 days before prostatectomy. These findings reveal a novel fulvestrant signaling cascade involving ERβ-mediated transcriptional upregulation of hsa-miR-765 that suppresses HMGA1 protein expression as part of the mechanism underlying the tumor suppressor action of fulvestrant in prostate cancer.

Conflict of interest statement

Competing Interests: Dr. Jodi Maranchie received the Investigator-Sponsored Study Program of AstraZeneca. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Fulvestrant inhibits DU145 cell growth,…
Figure 1. Fulvestrant inhibits DU145 cell growth, migration, and invasion.
(A) Fulvestrant induces growth inhibition of DU145 cells via an ERβ-dependent mechanism. Growth of the fulvestrant-treated DU145 cells with or without ERβ siRNA knockdown for 4 days relative to the ethanol-treated control cells with negative-control siRNA are presented and compared (n = 8). ERβ expression was also knocked down by another siRNA (siRNA#2) and the similar results were obtained (Figure S5). (B) Fulvestrant induces DU145 cell-cycle arrest at G2/M phase. Representative DNA histograms of 48 hrs fulvestrant -or ethanol- (control) treated cells and percentage distributions of the cells at G0/G1 and G2/M phases (n = 3) are presented and compared. (C) Fulvestrant induces expression of G2/M markers. DU145 cells were treated with fulvestrant or ethanol for 2 days (control) and cell cycle markers were determined by Western blot analysis. Two independent experiments were performed and one representative set of data was presented. (D) Fulvestrant suppresses cell migration. A wound-healing assay was performed on the fulvestrant- and ethanol (EtOH)-treated DU145 cells (n = 3). Representative micrographs of the fulvestrant- and ethanol-treated cell cultures with scratches at 0 h and after 16 h are shown. The wound is marked by dotted lines. (E) Fulvestrant inhibits transwell migration (left panel) and invasion (right panel) in DU145 cells (n = 3) after 5 hrs of fulvestrant treatment. (F) Reductions of filopodial cells and cells with intense stress fibers by fulvestrant (treated with 48 hrs) via an ERβ-dependent mechanism. Representative micrographs and the percentages of the cells with intense stress fibers and the filopodial cells (n = 3) are presented. Student t-test was performed to determine significance with a cutoff p value of 0.05. ** p

Figure 2. Fulvestrant upregulates hsa-miR-765 expression in…

Figure 2. Fulvestrant upregulates hsa-miR-765 expression in DU145 cells.

(A) Hsa-miR-765 is highly expressed in…

Figure 2. Fulvestrant upregulates hsa-miR-765 expression in DU145 cells.
(A) Hsa-miR-765 is highly expressed in fulvestrant-treated DU145 cells. Total RNAs of treated cells were labeled directly and arrayed on the NCode Human miRNA Microarray. The median- and linear regression-normalized data are presented in a scatterplot. (B) Hsa-miR-765 is induced by fulvestrant in two prostate cancer cell lines. The hsa-miR-765 in the fulvestrant- and ethanol-treated control DU145 and PC-3 cells was quantified by miRNA qRT-PCR analysis. Relative fold changes between the expression of hsa-miR-765 in the fulvestrant-treated and control cells are presented. Student t-test was performed to determine their significance using a cutoff p value of 0.05 (n = 3). **p<0.01; bars = S.D.

Figure 3. Hsa-miR-765 suppresses DU145 cell growth,…

Figure 3. Hsa-miR-765 suppresses DU145 cell growth, migration, and invasion.

(A) Hsa-miR-765 mimic effectively recognizes…

Figure 3. Hsa-miR-765 suppresses DU145 cell growth, migration, and invasion.
(A) Hsa-miR-765 mimic effectively recognizes reporter with complementary sequence of hsa-miR-765 in DU145 cells. Fold changes of luciferase activities of the hsa-miR-765 mimic treated cells relative to the cells treated with the negative-control mimic are presented (n = 3). Transfection reagents were used as control. (B) Hsa-miR-765 mimic reduces DU145 cell growth. MTS assay was performed on the cells treated with hsa-miR-765 mimic or negative-control mimic or transfection control for 4 days (n = 8). (C) Hsa-miR-765 mimic significant reduces G0/G1 to G2/M ratio in DU145. Representative DNA histograms (n = 3) are presented. (D) Hsa-miR-765 mimic treatment causes up-regulation of cyclin A, cyclin B, and phosphorylated-cdc2 expression in DU145 cells. Protein expression levels of cell cycle regulator proteins were determined by Western blot analyses. Two independent experiments were performed and one representative set of data was presented. (E) Hsa-miR-765 mimic suppresses DU145 cell migration and invasion as shown in transwell migration assay (top left) and invasion assay (top right), respectively. Representative micrographs of the cells after transwell migration (top left) or invasion assay (top right) are presented. Fold changes of migration (bottom left) and invasion (bottom right) of DU145 cells with either hsa-miR-765 mimic or negative-control mimic relative to the control cells with negative-control mimic are presented (n = 3). (F) Hsa-miR-765 mimic significantly reduces stress fibers and filopodia formations in DU145 cells. Representative micrographs and the percentages of the cells with intense stress fibers and the filopodial cells (n = 3) are presented. Student's t-test was used for comparisons with a cutoff p value of 0.05. ** p<0.01; bar = S.D.

Figure 4. ERβ is involved in fulvestrant-induced…

Figure 4. ERβ is involved in fulvestrant-induced upregulation of hsa-miR-765 expression.

(A) ERβ siRNA knockdown…

Figure 4. ERβ is involved in fulvestrant-induced upregulation of hsa-miR-765 expression.
(A) ERβ siRNA knockdown blocks fulvestrant-induced upregulation of hsa-miR-765 expression in DU145 cells. Expression levels of hsa-miR-765 determined by qRT-PCR analysis of the fulvestrant-treated cells with ERβ-siRNA (siERβ) or scramble negative-control (siNeg) were compared (n = 3). (B) SiRNA knockdown of ERβ blocks fulvestrant-induced transactivation of the 5′ upstream regulatory region of hsa-miR-765 in DU145 cells. 5′ upstream regulatory region of hsa-miR-765 was cloned into a luciferase vector. The reporter activities with ERβ-knockdown (siERβ) or scramble negative-control (siNeg) in the presence of fulvestrant were compared (n = 3). (C) Deletion mapping analysis defines a fulvestrant-responsive segment in hsa-miR-765 regulatory region in DU145 cells. The 5′ upstream DNA sequence of hsa-miR-765 from nt. −3208 to +100 was analyzed using luciferase reporter system. Serial deletions from the 5′ end of the cloned sequence in the vector were conducted. Reporter activities were compared between the fulvestrant-treated (Fulvestrant) and control (ETOH) cells for each reporter vector (n = 3). (D) Fulvestrant-induces recruitment of ERβ onto the putative hsa-miR-765 regulatory region. Chromatin-immunoprecipitation revealed the recruitment of ERβ to a sequence in the 5′-regulatory region of hsa-miR765. Mouse IgG and RNA polymerase II serve as negative and positive control, respectively. Fulvestrant induced 17 fold increase in ERβ recruitment when compared with non-ERβ binding region (the 0N promoter of ERβ [33], [41]). Student's t-test was performed to determine significance of between groups using a cutoff p value of 0.05. ** p<0.01; bar = S.D.

Figure 5. HMGA1 is a direct target…

Figure 5. HMGA1 is a direct target of hsa-miR-765 .

(A) The 3′UTR of HMGA1 from…

Figure 5. HMGA1 is a direct target of hsa-miR-765.
(A) The 3′UTR of HMGA1 from +8910 to +8929 is predicted to be hsa-miR-765 binding site. (B) Hsa-miR-765 interacts with 3′UTR of HMGA1 in a targeting reporter assay. DU145 cells were transfected with either pMIR-empty or pMIR-HMGA1-3UTR in which 3′ UTR of HMGA1 (+8026–+9332) was cloned into the 3′ end of luciferase. Reporter activities of the pMIR-HMGA1-3UTR transfected cells treated with hsa-miR-765 mimic or negative-control mimic are compared (n = 3). (C) Hsa-miR-765 mimic reduced HMGA1 protein expression in DU145 cells. Protein and mRNA levels of HMGA1 in the hsa-miR-765 mimic- and negative-control mimic-treated cells were determined by Western blot analysis (upper) and real-time RT-PCR analysis (lower), respectively. Results from miR-765 mimic vs negative control mimic are compared (n = 3). (D) Fulvestrant reduces HMGA1 protein expression in DU145 cells. Protein level of HMGA1 and β-actin in the fulvestrant-treated and ethanol-treated control (CTL) cells were determined by Western blot analysis. (E) Ectopic expression of HMGA1 blocks fulvestrant-induced DU145 cell growth inhibition. The relative cell growth was determined after 4 days of treatment with fulvestrant or ethanol after stable transfection of HMGA1 (or empty vector for control) for a week. Protein levels of HMGA1 were shown in Figure S6. The cell growth of fulvestrant-treated cells with HMGA1 overexpression vs empty vector are compared (n = 8). Student's t-test was performed to determine significance between groups using a cutoff p value of 0.05. **p<0.01; bar = S.D.

Figure 6. Significant reduction of HMGA1 protein…

Figure 6. Significant reduction of HMGA1 protein correlates with enhanced expression of hsa-miR-765 in fulvestrant-treated…

Figure 6. Significant reduction of HMGA1 protein correlates with enhanced expression of hsa-miR-765 in fulvestrant-treated clinical PCa specimens.
(A) Higher level of hsa-miR-765 is detected in fulvestrant-treated clinical PCa specimens. Relative fold changes between expression of hsa-miR-765 in the fulvestrant-treated (n = 7) and untreated (n = 7) clinical specimen are presented. Student's t-test was performed to determine significance between two groups. *p<0.05; bar = S.E.M. (B) Nuclear expression of HMGA1 and AR is reduced in fulvestrant-treated clinical PCa specimens. HMGA1 immunostaining was performed in the clinical PCa specimens from the fulvestrant-treated (n = 5) and untreated (n = 5) patients. Representative micrographs (100×) are shown. In upper panel, a magnified view (400×) of a selected region (dashed rectangle) in each micrograph is shown as a small insert to show the immunostaining of HGMA1 in the nuclei of Gleason grade 3/4 cancer foci. Imunnopositivity of nuclear AR is reduced in fulvestrant-treated Gleason grade 3/4 foci as shown in lower panel (400×). (C) Expression of both HMGA1 and AR is significantly reduced in fulvestrant-treated clinical PCa specimens when compared with their respective untreated samples (*p<0.05; **p<0.01; n = 9 (from 5 patients) for untreated samples; n = 10 (from 5 patients) for fulvestrant-treated samples, bar = S.E.M).
Figure 2. Fulvestrant upregulates hsa-miR-765 expression in…
Figure 2. Fulvestrant upregulates hsa-miR-765 expression in DU145 cells.
(A) Hsa-miR-765 is highly expressed in fulvestrant-treated DU145 cells. Total RNAs of treated cells were labeled directly and arrayed on the NCode Human miRNA Microarray. The median- and linear regression-normalized data are presented in a scatterplot. (B) Hsa-miR-765 is induced by fulvestrant in two prostate cancer cell lines. The hsa-miR-765 in the fulvestrant- and ethanol-treated control DU145 and PC-3 cells was quantified by miRNA qRT-PCR analysis. Relative fold changes between the expression of hsa-miR-765 in the fulvestrant-treated and control cells are presented. Student t-test was performed to determine their significance using a cutoff p value of 0.05 (n = 3). **p<0.01; bars = S.D.
Figure 3. Hsa-miR-765 suppresses DU145 cell growth,…
Figure 3. Hsa-miR-765 suppresses DU145 cell growth, migration, and invasion.
(A) Hsa-miR-765 mimic effectively recognizes reporter with complementary sequence of hsa-miR-765 in DU145 cells. Fold changes of luciferase activities of the hsa-miR-765 mimic treated cells relative to the cells treated with the negative-control mimic are presented (n = 3). Transfection reagents were used as control. (B) Hsa-miR-765 mimic reduces DU145 cell growth. MTS assay was performed on the cells treated with hsa-miR-765 mimic or negative-control mimic or transfection control for 4 days (n = 8). (C) Hsa-miR-765 mimic significant reduces G0/G1 to G2/M ratio in DU145. Representative DNA histograms (n = 3) are presented. (D) Hsa-miR-765 mimic treatment causes up-regulation of cyclin A, cyclin B, and phosphorylated-cdc2 expression in DU145 cells. Protein expression levels of cell cycle regulator proteins were determined by Western blot analyses. Two independent experiments were performed and one representative set of data was presented. (E) Hsa-miR-765 mimic suppresses DU145 cell migration and invasion as shown in transwell migration assay (top left) and invasion assay (top right), respectively. Representative micrographs of the cells after transwell migration (top left) or invasion assay (top right) are presented. Fold changes of migration (bottom left) and invasion (bottom right) of DU145 cells with either hsa-miR-765 mimic or negative-control mimic relative to the control cells with negative-control mimic are presented (n = 3). (F) Hsa-miR-765 mimic significantly reduces stress fibers and filopodia formations in DU145 cells. Representative micrographs and the percentages of the cells with intense stress fibers and the filopodial cells (n = 3) are presented. Student's t-test was used for comparisons with a cutoff p value of 0.05. ** p<0.01; bar = S.D.
Figure 4. ERβ is involved in fulvestrant-induced…
Figure 4. ERβ is involved in fulvestrant-induced upregulation of hsa-miR-765 expression.
(A) ERβ siRNA knockdown blocks fulvestrant-induced upregulation of hsa-miR-765 expression in DU145 cells. Expression levels of hsa-miR-765 determined by qRT-PCR analysis of the fulvestrant-treated cells with ERβ-siRNA (siERβ) or scramble negative-control (siNeg) were compared (n = 3). (B) SiRNA knockdown of ERβ blocks fulvestrant-induced transactivation of the 5′ upstream regulatory region of hsa-miR-765 in DU145 cells. 5′ upstream regulatory region of hsa-miR-765 was cloned into a luciferase vector. The reporter activities with ERβ-knockdown (siERβ) or scramble negative-control (siNeg) in the presence of fulvestrant were compared (n = 3). (C) Deletion mapping analysis defines a fulvestrant-responsive segment in hsa-miR-765 regulatory region in DU145 cells. The 5′ upstream DNA sequence of hsa-miR-765 from nt. −3208 to +100 was analyzed using luciferase reporter system. Serial deletions from the 5′ end of the cloned sequence in the vector were conducted. Reporter activities were compared between the fulvestrant-treated (Fulvestrant) and control (ETOH) cells for each reporter vector (n = 3). (D) Fulvestrant-induces recruitment of ERβ onto the putative hsa-miR-765 regulatory region. Chromatin-immunoprecipitation revealed the recruitment of ERβ to a sequence in the 5′-regulatory region of hsa-miR765. Mouse IgG and RNA polymerase II serve as negative and positive control, respectively. Fulvestrant induced 17 fold increase in ERβ recruitment when compared with non-ERβ binding region (the 0N promoter of ERβ [33], [41]). Student's t-test was performed to determine significance of between groups using a cutoff p value of 0.05. ** p<0.01; bar = S.D.
Figure 5. HMGA1 is a direct target…
Figure 5. HMGA1 is a direct target of hsa-miR-765.
(A) The 3′UTR of HMGA1 from +8910 to +8929 is predicted to be hsa-miR-765 binding site. (B) Hsa-miR-765 interacts with 3′UTR of HMGA1 in a targeting reporter assay. DU145 cells were transfected with either pMIR-empty or pMIR-HMGA1-3UTR in which 3′ UTR of HMGA1 (+8026–+9332) was cloned into the 3′ end of luciferase. Reporter activities of the pMIR-HMGA1-3UTR transfected cells treated with hsa-miR-765 mimic or negative-control mimic are compared (n = 3). (C) Hsa-miR-765 mimic reduced HMGA1 protein expression in DU145 cells. Protein and mRNA levels of HMGA1 in the hsa-miR-765 mimic- and negative-control mimic-treated cells were determined by Western blot analysis (upper) and real-time RT-PCR analysis (lower), respectively. Results from miR-765 mimic vs negative control mimic are compared (n = 3). (D) Fulvestrant reduces HMGA1 protein expression in DU145 cells. Protein level of HMGA1 and β-actin in the fulvestrant-treated and ethanol-treated control (CTL) cells were determined by Western blot analysis. (E) Ectopic expression of HMGA1 blocks fulvestrant-induced DU145 cell growth inhibition. The relative cell growth was determined after 4 days of treatment with fulvestrant or ethanol after stable transfection of HMGA1 (or empty vector for control) for a week. Protein levels of HMGA1 were shown in Figure S6. The cell growth of fulvestrant-treated cells with HMGA1 overexpression vs empty vector are compared (n = 8). Student's t-test was performed to determine significance between groups using a cutoff p value of 0.05. **p<0.01; bar = S.D.
Figure 6. Significant reduction of HMGA1 protein…
Figure 6. Significant reduction of HMGA1 protein correlates with enhanced expression of hsa-miR-765 in fulvestrant-treated clinical PCa specimens.
(A) Higher level of hsa-miR-765 is detected in fulvestrant-treated clinical PCa specimens. Relative fold changes between expression of hsa-miR-765 in the fulvestrant-treated (n = 7) and untreated (n = 7) clinical specimen are presented. Student's t-test was performed to determine significance between two groups. *p<0.05; bar = S.E.M. (B) Nuclear expression of HMGA1 and AR is reduced in fulvestrant-treated clinical PCa specimens. HMGA1 immunostaining was performed in the clinical PCa specimens from the fulvestrant-treated (n = 5) and untreated (n = 5) patients. Representative micrographs (100×) are shown. In upper panel, a magnified view (400×) of a selected region (dashed rectangle) in each micrograph is shown as a small insert to show the immunostaining of HGMA1 in the nuclei of Gleason grade 3/4 cancer foci. Imunnopositivity of nuclear AR is reduced in fulvestrant-treated Gleason grade 3/4 foci as shown in lower panel (400×). (C) Expression of both HMGA1 and AR is significantly reduced in fulvestrant-treated clinical PCa specimens when compared with their respective untreated samples (*p<0.05; **p<0.01; n = 9 (from 5 patients) for untreated samples; n = 10 (from 5 patients) for fulvestrant-treated samples, bar = S.E.M).

References

    1. Ho SM, Lee MT, Lam HM, Leung YK (2011) Estrogens and prostate cancer: etiology, mediators, prevention, and management. Endocrinol Metab Clin North Am 40: 591–ix, 591-614, ix S0889-8529(11)00051-X [pii];10.1016/j.ecl.2011.05.002 [doi]
    1. Lai JS, Brown LG, True LD, Hawley SJ, Etzioni RB, et al. (2004) Metastases of prostate cancer express estrogen receptor-beta. Urology 64: 814–820.
    1. Leav I, Lau KM, Adams JY, McNeal JE, Taplin ME, et al. (2001) Comparative studies of the estrogen receptors beta and alpha and the androgen receptor in normal human prostate glands, dysplasia, and in primary and metastatic carcinoma. Am J Pathol 159: 79–92.
    1. Oh WK (2002) The evolving role of estrogen therapy in prostate cancer. Clin Prostate Cancer 1: 81–89.
    1. Byar DP (1973) Proceedings: The Veterans Administration Cooperative Urological Research Group's studies of cancer of the prostate. Cancer 32: 1126–1130.
    1. von SB, Carlstrom K, Collste L, Eriksson A, Henriksson P, et al. (1989) Estrogen therapy and liver function–metabolic effects of oral and parenteral administration. Prostate 14: 389–395.
    1. Langley RE, Godsland IF, Kynaston H, Clarke NW, Rosen SD, et al. (2008) Early hormonal data from a multicentre phase II trial using transdermal oestrogen patches as first-line hormonal therapy in patients with locally advanced or metastatic prostate cancer. BJU Int 102: 442–445 BJU7583 [pii];10.1111/j.1464-410X.2008.07583.x [doi]
    1. Stein M, Goodin S, Doyle-Lindrud S, Silberberg J, Kane M, et al. (2012) Transdermal estradiol in castrate and chemotherapy resistant prostate cancer. Med Sci Monit 18: CR260–CR264 882626 [pii].
    1. Ockrim JL, Lalani EN, Banks LM, Svensson WE, Blomley MJ, et al. (2004) Transdermal estradiol improves bone density when used as single agent therapy for prostate cancer. J Urol 172: 2203–2207 00005392-200412010-00021 [pii].
    1. Bergan RC, Reed E, Myers CE, Headlee D, Brawley O, et al. (1999) A Phase II study of high-dose tamoxifen in patients with hormone-refractory prostate cancer. Clin Cancer Res 5: 2366–2373.
    1. Hamilton M, Dahut W, Brawley O, Davis P, Wells-Jones T, et al. (2003) A phase I/II study of high-dose tamoxifen in combination with vinblastine in patients with androgen-independent prostate cancer. Acta Oncol 42: 195–201.
    1. Shazer RL, Jain A, Galkin AV, Cinman N, Nguyen KN, et al. (2006) Raloxifene, an oestrogen-receptor-beta-targeted therapy, inhibits androgen-independent prostate cancer growth: results from preclinical studies and a pilot phase II clinical trial. BJU Int 97: 691–697 BJU5974 [pii];10.1111/j.1464-410X.2006.05974.x [doi]
    1. Stein S, Zoltick B, Peacock T, Holroyde C, Haller D, et al. (2001) Phase II trial of toremifene in androgen-independent prostate cancer: a Penn cancer clinical trials group trial. Am J Clin Oncol 24: 283–285.
    1. Lane KE, Leav I, Ziar J, Bridges RS, Rand WM, et al. (1997) Suppression of testosterone and estradiol-17beta-induced dysplasia in the dorsolateral prostate of Noble rats by bromocriptine. Carcinogenesis 18: 1505–1510.
    1. Tam NN, Szeto CY, Sartor MA, Medvedovic M, Ho SM (2008) Gene expression profiling identifies lobe-specific and common disruptions of multiple gene networks in testosterone-supported, 17beta-estradiol- or diethylstilbestrol-induced prostate dysplasia in Noble rats. Neoplasia 10: 20–40.
    1. Tam NN, Szeto CY, Freudenberg JM, Fullenkamp AN, Medvedovic M, et al. (2010) Research resource: estrogen-driven prolactin-mediated gene-expression networks in hormone-induced prostatic intraepithelial neoplasia. Mol Endocrinol 24: 2207–2217 me.2010-0179 [pii];10.1210/me.2010-0179 [doi]
    1. Thompson CJ, Tam NN, Joyce JM, Leav I, Ho SM (2002) Gene expression profiling of testosterone and estradiol-17 beta-induced prostatic dysplasia in Noble rats and response to the antiestrogen ICI 182,780. Endocrinology 143: 2093–2105.
    1. Lau KM, LaSpina M, Long J, Ho SM (2000) Expression of estrogen receptor (ER)-alpha and ER-beta in normal and malignant prostatic epithelial cells: regulation by methylation and involvement in growth regulation. Cancer Res 60: 3175–3182.
    1. Leung YK, Gao Y, Lau KM, Zhang X, Ho SM (2006) ICI 182,780-regulated gene expression in DU145 prostate cancer cells is mediated by estrogen receptor-beta/NFkappaB crosstalk. Neoplasia 8: 242–249 10.1593/neo.05853.
    1. Nakajima Y, Akaogi K, Suzuki T, Osakabe A, Yamaguchi C, et al. (2011) Estrogen regulates tumor growth through a nonclassical pathway that includes the transcription factors ERbeta and KLF5. Sci Signal 4: ra22 4/168/ra22 [pii];10.1126/scisignal.2001551 [doi]
    1. Bhattacharyya RS, Krishnan AV, Swami S, Feldman D (2006) Fulvestrant (ICI 182,780) down-regulates androgen receptor expression and diminishes androgenic responses in LNCaP human prostate cancer cells. Mol Cancer Ther 5: 1539–1549 5/6/1539 [pii];10.1158/1535-7163.MCT-06-0065 [doi]
    1. Chadha MK, Ashraf U, Lawrence D, Tian L, Levine E, et al. (2008) Phase II study of fulvestrant (Faslodex) in castration resistant prostate cancer. Prostate 68: 1461–1466 10.1002/pros.20813 [doi]
    1. Gasent Blesa JM, Alberola CV, Giner MV, Giner-Bosch V, Provencio PM, et al. (2010) Experience with fulvestrant acetate in castration-resistant prostate cancer patients. Ann Oncol 21: 1131–1132 mdq010 [pii];10.1093/annonc/mdq010 [doi]
    1. He L, Hannon GJ (2004) MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet 5: 522–531 10.1038/nrg1379 [doi];nrg1379 [pii]
    1. Brennecke J, Stark A, Russell RB, Cohen SM (2005) Principles of microRNA-target recognition. PLoS Biol 3: e85 10.1371/journal.pbio.0030085 [doi]
    1. Doench JG, Sharp PA (2004) Specificity of microRNA target selection in translational repression. Genes Dev 18: 504–511 10.1101/gad.1184404 [doi];1184404 [pii]
    1. Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB (2003) Prediction of mammalian microRNA targets. Cell 115: 787–798 S0092867403010183 [pii].
    1. Coppola V, De MR, Bonci D (2010) MicroRNAs and prostate cancer. Endocr Relat Cancer 17: F1–17 ERC-09-0172 [pii];10.1677/ERC-09-0172 [doi]
    1. Sevli S, Uzumcu A, Solak M, Ittmann M, Ozen M (2010) The function of microRNAs, small but potent molecules, in human prostate cancer. Prostate Cancer Prostatic Dis 13: 208–217 pcan201021 [pii];10.1038/pcan.2010.21 [doi]
    1. Shi XB, Tepper CG, White RW (2008) MicroRNAs and prostate cancer. J Cell Mol Med 12: 1456–1465 JCMM420 [pii];10.1111/j.1582-4934.2008.00420.x [doi]
    1. Schaefer A, Jung M, Kristiansen G, Lein M, Schrader M, et al. (2010) MicroRNAs and cancer: current state and future perspectives in urologic oncology. Urol Oncol 28: 4–13 S1078-1439(08)00283-4 [pii];10.1016/j.urolonc.2008.10.021 [doi]
    1. Lau KM, Chan QK, Pang JC, Ma FM, Li KK, et al. (2012) Overexpression of HMGA1 deregulates tumor growth via cdc25A and alters migration/invasion through a cdc25A-independent pathway in medulloblastoma. Acta Neuropathol 123: 553–571 10.1007/s00401-011-0934-8 [doi]
    1. Zhu X, Leav I, Leung YK, Wu M, Liu Q, et al. (2004) Dynamic regulation of estrogen receptor-beta expression by DNA methylation during prostate cancer development and metastasis. Am J Pathol 164: 2003–2012 S0002-9440(10)63760-1.
    1. Lau KM, Chan QK, Pang JC, Li KK, Yeung WW, et al. (2010) Minichromosome maintenance proteins 2, 3 and 7 in medulloblastoma: overexpression and involvement in regulation of cell migration and invasion. Oncogene 29: 5475–5489 onc2010287 [pii];10.1038/onc.2010.287 [doi]
    1. John B, Enright AJ, Aravin A, Tuschl T, Sander C, et al. (2004) Human MicroRNA targets. PLoS Biol 2: e363 10.1371/journal.pbio.0020363 [doi]
    1. Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP, et al. (2007) MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell 27: 91–105 S1097-2765(07)00407-8 [pii];10.1016/j.molcel.2007.06.017 [doi]
    1. Kruger J, Rehmsmeier M (2006) RNAhybrid: microRNA target prediction easy, fast and flexible. Nucleic Acids Res 34: W451–W454 34/suppl_2/W451 [pii];10.1093/nar/gkl243 [doi]
    1. Gaidatzis D, van NE, Hausser J, Zavolan M (2007) Inference of miRNA targets using evolutionary conservation and pathway analysis. BMC Bioinformatics 8: 69 1471-2105-8-69 [pii];10.1186/1471-2105-8-69 [doi]
    1. Leung YK, Lam HM, Wu S, Song D, Levin L, et al. (2010) Estrogen receptor beta2 and beta5 are associated with poor prognosis in prostate cancer, and promote cancer cell migration and invasion. Endocr Relat Cancer 17: 675–689 ERC-09-0294;10.1677/ERC-09-0294.
    1. Wei JJ, Wu X, Peng Y, Shi G, Basturk O, et al. (2011) Regulation of HMGA1 expression by microRNA-296 affects prostate cancer growth and invasion. Clin Cancer Res 17: 1297–1305 1078-0432.CCR-10-0993 [pii];10.1158/1078-0432.CCR-10-0993 [doi]
    1. Fernandes SA, Gomes GR, Siu ER, Damas-Souza DM, Bruni-Cardoso A, et al. (2011) The anti-oestrogen fulvestrant (ICI 182,780) reduces the androgen receptor expression, ERK1/2 phosphorylation and cell proliferation in the rat ventral prostate. Int J Androl 34: 486–500 10.1111/j.1365-2605.2010.01109.x [doi]
    1. Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, et al. (1997) Differential ligand activation of estrogen receptors ERalpha and ERbeta at AP1 sites. Science 277: 1508–1510.
    1. Zou A, Marschke KB, Arnold KE, Berger EM, Fitzgerald P, et al. (1999) Estrogen receptor beta activates the human retinoic acid receptor alpha-1 promoter in response to tamoxifen and other estrogen receptor antagonists, but not in response to estrogen. Mol Endocrinol 13: 418–430.
    1. Schultz JR, Petz LN, Nardulli AM (2003) Estrogen receptor alpha and Sp1 regulate progesterone receptor gene expression. Mol Cell Endocrinol 201: 165–175 S030372070200415X [pii].
    1. Saini S, Majid S, Dahiya R (2010) Diet, microRNAs and prostate cancer. Pharm Res 27: 1014–1026 10.1007/s11095-010-0086-x [doi]
    1. Takaha N, Resar LM, Vindivich D, Coffey DS (2004) High mobility group protein HMGI(Y) enhances tumor cell growth, invasion, and matrix metalloproteinase-2 expression in prostate cancer cells. Prostate 60: 160–167 10.1002/pros.20049 [doi]
    1. Tamimi Y, van der Poel HG, Denyn MM, Umbas R, Karthaus HF, et al. (1993) Increased expression of high mobility group protein I(Y) in high grade prostatic cancer determined by in situ hybridization. Cancer Res 53: 5512–5516.
    1. Tamimi Y, van der Poel HG, Karthaus HF, Debruyne FM, Schalken JA (1996) A retrospective study of high mobility group protein I(Y) as progression marker for prostate cancer determined by in situ hybridization. Br J Cancer 74: 573–578.
    1. Hassan O, Ahmad A, Sethi S, Sarkar FH (2012) Recent updates on the role of microRNAs in prostate cancer. J Hematol Oncol 5: 9 1756-8722-5-9 [pii];10.1186/1756-8722-5-9 [doi]
    1. Murata T, Takayama K, Katayama S, Urano T, Horie-Inoue K, et al. (2010) miR-148a is an androgen-responsive microRNA that promotes LNCaP prostate cell growth by repressing its target CAND1 expression. Prostate Cancer Prostatic Dis 13: 356–361 pcan201032 [pii];10.1038/pcan.2010.32 [doi]
    1. Shi XB, Xue L, Yang J, Ma AH, Zhao J, et al. (2007) An androgen-regulated miRNA suppresses Bak1 expression and induces androgen-independent growth of prostate cancer cells. Proc Natl Acad Sci U S A 104: 19983–19988 0706641104 [pii];10.1073/pnas.0706641104 [doi]
    1. Waltering KK, Porkka KP, Jalava SE, Urbanucci A, Kohonen PJ, et al. (2011) Androgen regulation of micro-RNAs in prostate cancer. Prostate 71: 604–614 10.1002/pros.21276 [doi]
    1. Josson S, Sung SY, Lao K, Chung LW, Johnstone PA (2008) Radiation modulation of microRNA in prostate cancer cell lines. Prostate 68: 1599–1606 10.1002/pros.20827 [doi]
    1. Li B, Shi XB, Nori D, Chao CK, Chen AM, et al. (2011) Down-regulation of microRNA 106b is involved in p21-mediated cell cycle arrest in response to radiation in prostate cancer cells. Prostate 71: 567–574 10.1002/pros.21272 [doi]
    1. Mak P, Leav I, Pursell B, Bae D, Yang X, et al. (2010) ERbeta impedes prostate cancer EMT by destabilizing HIF-1alpha and inhibiting VEGF-mediated snail nuclear localization: implications for Gleason grading. Cancer Cell 17: 319–332 S1535-6108(10)00082-6 [pii];10.1016/j.ccr.2010.02.030 [doi]
    1. Setlur SR, Mertz KD, Hoshida Y, Demichelis F, Lupien M, et al. (2008) Estrogen-dependent signaling in a molecularly distinct subclass of aggressive prostate cancer. J Natl Cancer Inst 100: 815–825 djn150 [pii];10.1093/jnci/djn150 [doi]

Source: PubMed

Подписаться