BRCA1 and BRCA2 as molecular targets for phytochemicals indole-3-carbinol and genistein in breast and prostate cancer cells

S Fan, Q Meng, K Auborn, T Carter, E M Rosen, S Fan, Q Meng, K Auborn, T Carter, E M Rosen

Abstract

Indole-3-carbinol (I3C) and genistein are naturally occurring chemicals derived from cruciferous vegetables and soy, respectively, with potential cancer prevention activity for hormone-responsive tumours (e.g., breast and prostate cancers). Previously, we showed that I3C induces BRCA1 expression and that both I3C and BRCA1 inhibit oestrogen (E2)-stimulated oestrogen receptor (ER-alpha) activity in human breast cancer cells. We now report that both I3C and genistein induce the expression of both breast cancer susceptibility genes (BRCA1 and BRCA2) in breast (MCF-7 and T47D) and prostate (DU-145 and LNCaP) cancer cell types, in a time- and dose-dependent fashion. Induction of the BRCA genes occurred at low doses of I3C (20 microM) and genistein (0.5-1.0 microM), suggesting potential relevance to cancer prevention. A combination of I3C and genistein gave greater than expected induction of BRCA expression. Studies using small interfering RNAs (siRNAs) and BRCA expression vectors suggest that the phytochemical induction of BRCA2 is due, in part, to BRCA1. Functional studies suggest that I3C-mediated cytoxicity is, in part, dependent upon BRCA1 and BRCA2. Inhibition of E2-stimulated ER-alpha activity by I3C and genistein was dependent upon BRCA1; and inhibition of ligand-inducible androgen receptor (AR) activity by I3C and genistein was partially reversed by BRCA1-siRNA. Finally, we provide evidence suggesting that the phytochemical induction of BRCA1 expression is due, in part, to endoplasmic reticulum stress response signalling. These findings suggest that the BRCA genes are molecular targets for some of the activities of I3C and genistein.

Figures

Figure 1
Figure 1
Indole-3-carbinol (I3C) upregulates BRCA1 and BRCA2 mRNA expression in human breast cancer cells in a time- and dose-dependent manner. For time course studies, subconfluent proliferating MCF-7 (A) or T47D (B) cells were treated with I3C (60 μM) for different times and harvested for mRNA analysis by semiquantitative RT–PCR. For dose–response studies, MCF-7 (C) or T47D (D) cells were treated with different doses of I3C for 24 h and harvested for mRNA analysis. The PCR bands were quantified by densitometry and expressed relative to the control gene, β-actin. The densitometry values are means±s.e.m.'s of at least two independent experiments.
Figure 2
Figure 2
Indole-3-carbinol upregulates BRCA1 and BRCA2 protein levels in human breast and prostate cancer cells in a time- and dose-dependent manner. For time course studies, subconfluent proliferating MCF-7 (A), T47D (B), DU-145 (E), and LNCaP (F) cells were treated with I3C (60 μM) for various times and harvested for Western blot analysis to detect the BRCA1, BRCA2, or actin (control for loading and transfer) proteins. For dose–response studies, MCF-7 (C), T47D (D), DU-145 (G), or LNCaP (H) cells were treated with the indicated dose of I3C for 24 h and then assayed for BRCA1 and BRCA2 protein expression. The protein bands were quantitated by densitometry and expressed relative to actin. The densitometry values are means±s.e.m.'s of at least two independent experiments.
Figure 2
Figure 2
Indole-3-carbinol upregulates BRCA1 and BRCA2 protein levels in human breast and prostate cancer cells in a time- and dose-dependent manner. For time course studies, subconfluent proliferating MCF-7 (A), T47D (B), DU-145 (E), and LNCaP (F) cells were treated with I3C (60 μM) for various times and harvested for Western blot analysis to detect the BRCA1, BRCA2, or actin (control for loading and transfer) proteins. For dose–response studies, MCF-7 (C), T47D (D), DU-145 (G), or LNCaP (H) cells were treated with the indicated dose of I3C for 24 h and then assayed for BRCA1 and BRCA2 protein expression. The protein bands were quantitated by densitometry and expressed relative to actin. The densitometry values are means±s.e.m.'s of at least two independent experiments.
Figure 3
Figure 3
Genistein upregulates BRCA1 and BRCA2 protein levels in breast cancer cells. For time course studies, subconfluent proliferating MCF-7 (A), T47D (B), DU-145 (E), and LNCaP (F) cells were treated with genistein (5 μM) for the indicated times and harvested for Western blotting to detect the BRCA1, BRCA2, or actin proteins. For dose–response studies, MCF-7 (C), T47D (D), DU-145 (G), or LNCaP (H) cells were treated with the indicated dose of genistein for 24 h and assayed for BRCA1 and BRCA2 protein expression, as described above. The densitometry values represent means±s.e.m.'s of at least two independent experiments.
Figure 3
Figure 3
Genistein upregulates BRCA1 and BRCA2 protein levels in breast cancer cells. For time course studies, subconfluent proliferating MCF-7 (A), T47D (B), DU-145 (E), and LNCaP (F) cells were treated with genistein (5 μM) for the indicated times and harvested for Western blotting to detect the BRCA1, BRCA2, or actin proteins. For dose–response studies, MCF-7 (C), T47D (D), DU-145 (G), or LNCaP (H) cells were treated with the indicated dose of genistein for 24 h and assayed for BRCA1 and BRCA2 protein expression, as described above. The densitometry values represent means±s.e.m.'s of at least two independent experiments.
Figure 4
Figure 4
Dependence of phytochemical-induced expression of BRCA2 on BRCA1. (A) BRCA1-siRNA causes time-dependent loss of BRCA1 and BRCA2 in MCF-7 cells. Subconfluent proliferating cells were treated with BRCA1-siRNA (left) or control-siRNA (right) (50 nM) for different times, harvested, and Western blotted for BRCA1, BRCA2, and actin. (B) Effect of BRCA1-siRNA on BRCA2 protein levels and vice versa in DU-145 cells. Cells were treated with BRCA1, BRCA2, or control-siRNA (50 nM) for 72 h and Western blotted for BRCA1, BRCA2, and actin. Results are shown for two separate cell treatments and protein isolations on the same blot. (C) Effect of wtBRCA1 on BRCA2 protein levels and vice versa in DU-145 cells. Cells were transfected overnight with wtBRCA1, wtBRCA2, or empty pcDNA3 vector, washed, postincubated for 24 h to allow gene expression, harvested, and Western blotted for BRCA1, BRCA2, and actin. Results are shown for two separate cell treatments and protein isolations on the same blot. The densitometry values are means±ranges of two experiments. (D) Effect of BRCA1 and BRCA2 siRNAs on BRCA induction by I3C. DU-145 cells were preincubated with the indicated siRNA (50 nM × 72 h) or no siRNA (transfection reagent only), then treated with I3C (40 μM) for 24 h, and then harvested for Western blotting. (E) Effect of BRCA1 and BRCA2 siRNA on induction of BRCA1 and BRCA2 by genistein. DU-145 cells were preincubated with the indicated siRNA (50 nM × 72 h), treated with genistein (5 μM) for 24 h, and harvested for Western blotting as above. (F) Induction of BRCA1 and BRCA2 by a combination of I3C plus genistein. MCF-7 or DU-145 cells were treated with low doses of I3C (25 μM) and/or genistein (1 μM) for 24 h and harvested for Western blotting. The densitometry values are means±ranges of two independent experiments. (G) Effect of ICI182,780 on phytochemical induction of BRCA1 in MCF-7 cells. Proliferating cells were incubated with the indicated agents for 24 h and then harvested for Western blotting for BRCA1, ER-α, or actin. (H) Effect of BRCA1 knockdown and phytochemicals on ER-α protein levels. MCF-7 cells were pretreated with BRCA1 or control siRNA as described above, exposed to the indicated doses of I3C or genistein for 24 h, and then Western blotted for ER-α, BRCA1, or actin.
Figure 4
Figure 4
Dependence of phytochemical-induced expression of BRCA2 on BRCA1. (A) BRCA1-siRNA causes time-dependent loss of BRCA1 and BRCA2 in MCF-7 cells. Subconfluent proliferating cells were treated with BRCA1-siRNA (left) or control-siRNA (right) (50 nM) for different times, harvested, and Western blotted for BRCA1, BRCA2, and actin. (B) Effect of BRCA1-siRNA on BRCA2 protein levels and vice versa in DU-145 cells. Cells were treated with BRCA1, BRCA2, or control-siRNA (50 nM) for 72 h and Western blotted for BRCA1, BRCA2, and actin. Results are shown for two separate cell treatments and protein isolations on the same blot. (C) Effect of wtBRCA1 on BRCA2 protein levels and vice versa in DU-145 cells. Cells were transfected overnight with wtBRCA1, wtBRCA2, or empty pcDNA3 vector, washed, postincubated for 24 h to allow gene expression, harvested, and Western blotted for BRCA1, BRCA2, and actin. Results are shown for two separate cell treatments and protein isolations on the same blot. The densitometry values are means±ranges of two experiments. (D) Effect of BRCA1 and BRCA2 siRNAs on BRCA induction by I3C. DU-145 cells were preincubated with the indicated siRNA (50 nM × 72 h) or no siRNA (transfection reagent only), then treated with I3C (40 μM) for 24 h, and then harvested for Western blotting. (E) Effect of BRCA1 and BRCA2 siRNA on induction of BRCA1 and BRCA2 by genistein. DU-145 cells were preincubated with the indicated siRNA (50 nM × 72 h), treated with genistein (5 μM) for 24 h, and harvested for Western blotting as above. (F) Induction of BRCA1 and BRCA2 by a combination of I3C plus genistein. MCF-7 or DU-145 cells were treated with low doses of I3C (25 μM) and/or genistein (1 μM) for 24 h and harvested for Western blotting. The densitometry values are means±ranges of two independent experiments. (G) Effect of ICI182,780 on phytochemical induction of BRCA1 in MCF-7 cells. Proliferating cells were incubated with the indicated agents for 24 h and then harvested for Western blotting for BRCA1, ER-α, or actin. (H) Effect of BRCA1 knockdown and phytochemicals on ER-α protein levels. MCF-7 cells were pretreated with BRCA1 or control siRNA as described above, exposed to the indicated doses of I3C or genistein for 24 h, and then Western blotted for ER-α, BRCA1, or actin.
Figure 5
Figure 5
Contribution of BRCA1 and BRCA2 to I3C-mediated cytotoxicity. (A) The effects of BRCA1-siRNA (48 and 72 h) and BRCA2-siRNA (72 h) (50 nM) on BRCA1 and BRCA2 protein levels, respectively, in MCF-7 cells. MCF-7 (B and C), T47D (D and E), and DU-145 (F and G) cells were experimentally manipulated to increase (wtBRCA1) or decrease (BRCA1-siRNA) BRCA1 levels, treated with different doses of I3C, and tested for cell viability using MTT assays. In (H) and (I), DU-145 cells were manipulated to increase (wtBRCA2) or decrease (BRCA2-siRNA) BRCA2 levels, exposed to different doses of I3C, and tested for cell viability using MTT assays. Methodology (BI). To increase BRCA1 levels, subconfluent cells in 96-well dishes were transfected with wtBRCA1 overnight (see Materials and Methods), washed, postincubated for 24 h, exposed to different doses of I3C for 24 h, and assayed for MTT dye reduction. To decrease BRCA1 levels, cells were pretreated with BRCA1- or control-siRNA (50 nM × 72 h) or mock-transfected (control) and assayed for sensitivity to I3C as above. For BRCA2 experiments, DU-145 cells were transfected with wtBRCA2 or treated with BRCA2- or control-siRNA (as above) and assayed as described above for sensitivity to I3C. Cell viability values are expressed relative to the 0 I3C control and are means±s.e.m.'s for 10 replicate wells. Statistical comparisons. Cell viability comparisons were made using two-tailed t-tests. Significant differences were as follows: MCF-7 wtBRCA1 vs control, P<0.001 at 100–400 μM I3C; MCF-7 BRCA1-siRNA vs control, P<0.001 at 200–500 μM I3C; T47D wtBRCA1 vs control, P<0.001 at 100–500 μM I3C; T47D BRCA1-siRNA vs control, P<0.001 at 200–500 μM I3C; DU-145 wtBRCA1 vs control, P<0.001 at 100–400 μM I3C; DU-145 BRCA1-siRNA vs control, P<0.001 at 100–500 μM I3C; DU-145 wtBRCA1 vs control, P<0.001, 100–400 μM I3C; and DU-145 BRCA2-siRNA vs control, P<0.001 at 200–500 μM I3C.
Figure 5
Figure 5
Contribution of BRCA1 and BRCA2 to I3C-mediated cytotoxicity. (A) The effects of BRCA1-siRNA (48 and 72 h) and BRCA2-siRNA (72 h) (50 nM) on BRCA1 and BRCA2 protein levels, respectively, in MCF-7 cells. MCF-7 (B and C), T47D (D and E), and DU-145 (F and G) cells were experimentally manipulated to increase (wtBRCA1) or decrease (BRCA1-siRNA) BRCA1 levels, treated with different doses of I3C, and tested for cell viability using MTT assays. In (H) and (I), DU-145 cells were manipulated to increase (wtBRCA2) or decrease (BRCA2-siRNA) BRCA2 levels, exposed to different doses of I3C, and tested for cell viability using MTT assays. Methodology (BI). To increase BRCA1 levels, subconfluent cells in 96-well dishes were transfected with wtBRCA1 overnight (see Materials and Methods), washed, postincubated for 24 h, exposed to different doses of I3C for 24 h, and assayed for MTT dye reduction. To decrease BRCA1 levels, cells were pretreated with BRCA1- or control-siRNA (50 nM × 72 h) or mock-transfected (control) and assayed for sensitivity to I3C as above. For BRCA2 experiments, DU-145 cells were transfected with wtBRCA2 or treated with BRCA2- or control-siRNA (as above) and assayed as described above for sensitivity to I3C. Cell viability values are expressed relative to the 0 I3C control and are means±s.e.m.'s for 10 replicate wells. Statistical comparisons. Cell viability comparisons were made using two-tailed t-tests. Significant differences were as follows: MCF-7 wtBRCA1 vs control, P<0.001 at 100–400 μM I3C; MCF-7 BRCA1-siRNA vs control, P<0.001 at 200–500 μM I3C; T47D wtBRCA1 vs control, P<0.001 at 100–500 μM I3C; T47D BRCA1-siRNA vs control, P<0.001 at 200–500 μM I3C; DU-145 wtBRCA1 vs control, P<0.001 at 100–400 μM I3C; DU-145 BRCA1-siRNA vs control, P<0.001 at 100–500 μM I3C; DU-145 wtBRCA1 vs control, P<0.001, 100–400 μM I3C; and DU-145 BRCA2-siRNA vs control, P<0.001 at 200–500 μM I3C.
Figure 6
Figure 6
Contribution of BRCA genes to regulation of ER-α and AR activity by I3C and genistein. (A) Rescue of I3C inhibition of E2-stimulated ER-α activity by BRCA1-siRNA. MCF-7 cells were pretreated with BRCA1-siRNA, BRCA2-siRNA, control-siRNA (50 nM × 72 h), or no siRNA (vehicle only). After the first 48 h of siRNA treatment, they were transfected with the ERE-TK-Luc reporter overnight, washed, postincubated±17β-estradiol (E2, 1 μM) and ±I3C (100 μM) for 24 h, and tested for luciferase activity. Values are expressed relative to the +E2 positive control (no siRNA, no I3C) and are means±s.e.m.'s of three independent experiments. In each experiment, each assay condition was tested in four replicate wells, and the values were averaged. BRCA1 (but not BRCA2 or control) siRNA reversed the inhibition of E2-stimulated ER-α activity by I3C (P<0.001). (B) Rescue of genistein inhibition of E2-stimulated ER-α activity by BRCA1-siRNA. The experiment was performed as described above, except that the cells were treated ±genistein (5 μM) instead of I3C. Luciferase values are expressed relative to the +E2 positive control (no siRNA, no genistein) and are means±s.e.m.'s of three independent experiments, with each assay condition tested in four replicate wells per experiment. BRCA1 (but not BRCA2 or control) siRNA reversed the inhibition of E2-stimulated ER-α activity by genistein (P<0.001). (C, D) Contribution of BRCA1 to inhibition of DHT-stimulated AR activity by I3C and genistein. LNCaP (C) or PC-3 (D) cells were pretreated with BRCA1-siRNA, control-siRNA (50 nM × 72 h), or no siRNA. LNCaP cells, which are AR-positive, were transfected with an androgen-responsive reporter (ARE-TK-Luc); while PC-3 cells, which are AR-negative, were cotransfected with an AR expression vector plus ARE-TK-Luc. The cells were treated with dihydrotestosterone (DHT, 10 nM), I3C (25 μM), and/or genistein (1.0 μM) for 24 h and assayed for luciferase activity. BRCA1-siRNA enhanced DHT-stimulated AR activity and partially rescued the inhibition of AR activity by I3C and genistein (left panels). The asterisks indicate a significant comparison (P<0.01). (E) Effect of BRCA1-siRNA and phytochemicals on AR protein levels in LNCaP cells. Cells were pretreated with BRCA1 or control siRNA and then treated with I3C, genistein, or DHT as described in (C). The cells were then harvested and Western blotted for AR or actin. (F) Effect of BRCA1-siRNA on AR mRNA levels. LNCaP cells were pretreated with BRCA1- or control-siRNA, treated with DHT for 24 h, and harvested for semiquantitative RT–PCR to detect BRCA1, AR, or β-actin.
Figure 7
Figure 7
Thapsigargin and tunicamycin upregulate BRCA gene expression. (A, B) Dose–response for induction of BRCA mRNAs by thapsigargin in T47D (A) and MCF-7 (B) cells. Subconfluent proliferating cells were incubated with the indicated doses of thapsigargin for 24-h and then harvested for semiquantitative RT–PCR analysis of BRCA1, BRCA2, and β-actin (control gene). (C) Time course for induction of BRCA1 mRNAs by thapsigargin in MCF-7 cells. MCF-7 cells were incubated with thapsigargin (300 nM) for different time intervals up to 48-h and then harvested for semiquantitative RT–PCR analysis of BRCA1, BRCA2, and β-actin. (D, E) Dose–response for induction of BRCA1 protein by thapsigargin in T47D (D) and MCF-7 (E) cells. Cells were incubated with the indicated doses of thapsigargin for 24-h and then harvested for Western blotting to detect BRCA1 and actin (control for loading and transfer). (F, G) Dose–response for induction of BRCA proteins by tunicamycin in T47D (F) and MCF-7 (G) cells. Cells were incubated with the indicated doses of thapsigargin for 24-h and then harvested for Western blotting to detect BRCA1, BRCA2, and actin.
Figure 8
Figure 8
Induction of BRCA expression by I3C requires the kinase PERK (EIFAK3). (A) Dominant negative (DN) PERK blocks I3C induction of BRCA protein. Subconfluent proliferating MCF-7 cells were transfected overnight with control (vehicle only), empty pcDNA3 vector, or DN-PERK (15-μg plasmid DNA per 100-mm dish) using Lipofectamine™. The transfected cells were washed, allowed to recover for several hours, treated±I3C for 24-h, and harvested for Western blotting to detect BRCA1, BRCA2, or actin (control for loading and transfer). (B) Dominant negative IRE1 fails to block I3C induction of BRCA protein. Assays were performed as described in (A), except using DN-IRE1 instead of DN-PERK. (C) Dominant negative ATF4 fails to block I3C induction of BRCA protein. Assays were performed as described in (A), except using DN-ATF4 instead of DN-PERK.
Figure 9
Figure 9
Stimulation of endoplasmic reticulum stress signalling by I3C, genistein, and BRCA1. (A) Stimulation of ERSE-driven reporter activity by I3C and tunicamycin. MCF-7 or T47D cells were transfected overnight with the indicated luciferase reporter, washed, postincubated with tunicamycin (1 μg/ml) and/or I3C (100 μM) for 24-h, and harvested for luciferase assays. Luciferase activity was expressed relative to that observed using the ERSEwt-Luc reporter in the absence of tunicamycin or I3C. Values are means±s.e.m.'s of four replicate wells. The reporters tested were driven by the wt endoplasmic reticulum stress-response element (ERSEwt-Luc), a mutant ERSE (ERSEmut-Luc), and three copies of the ERSE-II element (ERSEII3x-Luc). (B) Stimulation of ERSE reporter activity by genistein. Assays were performed as above, except that after transfection of reporters, the cells were treated with genistein (0.5 or 1.0 μM) for 24 h. (C) Stimulation of CHOP promoter–reporter activity by I3C and gensitein. Cells were transfected overnight with the a reporter composed of the CHOP promoter upstream of a luciferase gene (pCHOP-Luc), washed, and postincubated with different doses of I3C or genistein for 24 h. Luciferase activity was expressed relative to that in the absence of I3C or genistein. The values are means±s.e.m.'s of four replicate wells. (D) Dependence of basal ERSE reporter activity on endogenous BRCA1. Proliferating cells were pretreated with BRCA1-siRNA, control-siRNA (50 nM × 72-h), or no siRNA (vehicle only). After 48 h of siRNA treatment, they were transfected with the indicated ERSE reporter. After siRNA treatment and transfection, the cells were washed, postincubated for 24 h, and harvested for luciferase assays. Values are expressed relative to the ERSEwt-Luc control (no siRNA) and are means±s.e.m.'s of four replicate wells. (E) Stimulation of ERSE reporter activity by wtBRCA1. Cells were cotransfected overnight with wtBRCA1, empty pcDNA3 vector, or no vector (vehicle only) and the indicated reporter. The cells were washed and postincubated for 24 h. Luciferase values are expressed relative to the ERSEwt-Luc reporter in the absence of wtBRCA1 or pcDNA3 and are means±s.e.m.' of quadruplicate wells. (F) Dependence of I3C-stimulated ERSE activity on endogenous BRCA1. Assays were performed as described in (D), except that after siRNA treatment and transfection, the cells were postincubated for 24 h in the absence or presence of I3C (100 μM). (G) Dependence of genistein-stimulated ERSE activity on endogenous BRCA1. Assays were performed as described in (F), except using genistein (1 μM) instead of I3C.
Figure 9
Figure 9
Stimulation of endoplasmic reticulum stress signalling by I3C, genistein, and BRCA1. (A) Stimulation of ERSE-driven reporter activity by I3C and tunicamycin. MCF-7 or T47D cells were transfected overnight with the indicated luciferase reporter, washed, postincubated with tunicamycin (1 μg/ml) and/or I3C (100 μM) for 24-h, and harvested for luciferase assays. Luciferase activity was expressed relative to that observed using the ERSEwt-Luc reporter in the absence of tunicamycin or I3C. Values are means±s.e.m.'s of four replicate wells. The reporters tested were driven by the wt endoplasmic reticulum stress-response element (ERSEwt-Luc), a mutant ERSE (ERSEmut-Luc), and three copies of the ERSE-II element (ERSEII3x-Luc). (B) Stimulation of ERSE reporter activity by genistein. Assays were performed as above, except that after transfection of reporters, the cells were treated with genistein (0.5 or 1.0 μM) for 24 h. (C) Stimulation of CHOP promoter–reporter activity by I3C and gensitein. Cells were transfected overnight with the a reporter composed of the CHOP promoter upstream of a luciferase gene (pCHOP-Luc), washed, and postincubated with different doses of I3C or genistein for 24 h. Luciferase activity was expressed relative to that in the absence of I3C or genistein. The values are means±s.e.m.'s of four replicate wells. (D) Dependence of basal ERSE reporter activity on endogenous BRCA1. Proliferating cells were pretreated with BRCA1-siRNA, control-siRNA (50 nM × 72-h), or no siRNA (vehicle only). After 48 h of siRNA treatment, they were transfected with the indicated ERSE reporter. After siRNA treatment and transfection, the cells were washed, postincubated for 24 h, and harvested for luciferase assays. Values are expressed relative to the ERSEwt-Luc control (no siRNA) and are means±s.e.m.'s of four replicate wells. (E) Stimulation of ERSE reporter activity by wtBRCA1. Cells were cotransfected overnight with wtBRCA1, empty pcDNA3 vector, or no vector (vehicle only) and the indicated reporter. The cells were washed and postincubated for 24 h. Luciferase values are expressed relative to the ERSEwt-Luc reporter in the absence of wtBRCA1 or pcDNA3 and are means±s.e.m.' of quadruplicate wells. (F) Dependence of I3C-stimulated ERSE activity on endogenous BRCA1. Assays were performed as described in (D), except that after siRNA treatment and transfection, the cells were postincubated for 24 h in the absence or presence of I3C (100 μM). (G) Dependence of genistein-stimulated ERSE activity on endogenous BRCA1. Assays were performed as described in (F), except using genistein (1 μM) instead of I3C.

References

    1. Andres JL, Fan S, Turkel GJ, Wang JA, Twu NF, Yuan RQ, Lamszus K, Goldberg ID, Rosen EM (1998) Regulation of BRCA1 and BRCA2 expression in human breast cancer cells by DNA-damaging agents. Oncogene 16: 2229–2241
    1. Aretini P, D'Andrea E, Pasini B, Viel A, Costantini RM, Cortesi L, Ricevuto E, Agata S, Bisegna R, Boiocchi M, Caligo MA, Chieco-Bianchi L, Cipollini G, Crucianelli R, D'Amico C, Federico M, Ghimenti C, De Giacomi C, De Nicolo A, Della Puppa L, Ferrari S, Ficorella C, Iandolo D, Manoukian S, Marchetti P, Marroni F, Menin C, Montagna M, Ottini L, Pensotti V, Pierotti M, Radice P, Santarosa M, Silingardi V, Turchetti D, Bevilacqua G, Presciuttini S (2003) Different expressivity of BRCA1 and BRCA2: analysis of 179 Italian pedigrees with identified mutation. Breast Cancer Res Treat 81: 71–79
    1. Bell MC, Crowley-Nowick P, Bradlow HL, Sepkovic DW, Schmidt-Grimminger D, Howell P, Mayeaux EJ, Tucker A, Turbat-Herrera EA, Mathis JM (2000) Placebo-controlled trial of indole-3-carbinol in the treatment of CIN. Gynecol Oncol 78: 123–129
    1. Bernard-Gallon DJ, De Latour MP, Sylvain V, Vissac C, Aunoble B, Chassagne J, Bignon YJ (2001) Brca1 and Brca2 protein expression patterns in different tissues of murine origin. Int J Oncol 18: 271–280
    1. Bonnesen C, Eggleston IM, Hayes JD (2001) Dietary indoles and isothiocyanates that are generated from cruciferous vegetables can both stimulate apoptosis and confer protection against DNA damage in human colon cell lines. Cancer Res 61: 6120–6130
    1. Bradlow HL, Michnovicz J, Telang NT, Osborne MP (1991) Effects of dietary indole-3-carbinol on estradiol metabolism and spontaneous mammary tumors in mice. Carcinogenesis 12: 1571–1574
    1. Bruhat A, Jousse C, Carraro V, Reimold AM, Ferrara M, Fafournoux P (2000) Amino acids control mammalian gene transcription: activating transcription factor 2 is essential for the amino acid responsiveness of the CHOP promoter. Mol Cell Biol 20: 7192–7204
    1. Cabanes A, Wang M, Olivo S, DeAssis S, Gustafsson JA, Khan G, Hilakivi-Clarke L (2004) Prepubertal estradiol and genistein exposures up-regulate BRCA1 mRNA and reduce mammary tumorigenesis. Carcinogenesis 25: 741–748
    1. Carter TH, Liu K, Ralph Jr W, Chen D, Qi M, Fan S, Yuan F, Rosen EM, Auborn KJ (2002) Diindolylmethane alters gene expression in human keratinocytes. J Nutr 132: 3314–3324
    1. Castle EP, Thrasher JB (2002) The role of soy phytoestrogens in prostate cancer. Urol Clin North Am 29: 71–81
    1. Chen D-Z, Mei Q, Auborn KJ, Carter TH (2001) Indole-3-carbinol and diindolylmethane induce apoptosis of human cervical cancer cells and in murine HPV16-transgenic preneoplastic cervical epithelium. J Nutr 131: 3294–3302
    1. Chen I, McDougal A, Wang F, Safe S (1998) Aryl hydrocarbon-mediated antiestrogenic and antitumorigenic activity of diindolymethane. Carcinogenesis 19: 1631–1639
    1. Chinni SR, Li Y, Upadhyay S, Koppolu PK, Sarkar FH (2001) Indole-3-carbinol (I3C) induced cell growth inhibition, G1 cell cycle arrest and apoptosis in prostate cancer cells. Oncogene 20: 2927–2936
    1. Cover CM, Hsieh SJ, Cram EJ, Hong C, Riby JE, Bjeldanes LF, Firestone GL (1999) Indole-3-carbinol and tamoxifen cooperate to arrest the cell cycle of MCF-7 human breast cancer cells. Cancer Res 59: 1244–1251
    1. Cover CM, Hsieh SJ, Tran SH, Hallden G, Kim GS, Bjeldanes LF, Firestone GL (1998) Indole-3-carbinol inhibits the expression of cyclin-dependent kinase-6 and induces a G1 cell cycle arrest of human breast cancer cells independent of estrogen receptor signaling. J Biol Chem 273: 3838–3847
    1. Edwards SM, Kote-Jarai Z, Meitz J, Hamoudi R, Hope Q, Osin P, Jackson R, Southgate C, Singh R, Falconer A, Dearnaley DP, Ardern-Jones A, Murkin A, Dowe A, Kelly J, Williams S, Oram R, Stevens M, Teare DM, Ponder BA, Gayther SA, Easton DF, Eeles RA, Cancer Research UK/Bristish Prostate Group UK Familial Prostate Cancer Study Collaborators; British Association of Urological Surgeons Section of Oncology (2003) Two percent of men with early-onset prostate cancer harbor germline mutations in the BRCA2 gene. Am J Hum Genet 1: 1–12
    1. Fan S, Ma YX, Wang C, Yuan RQ, Meng Q, Wang JA, Erdos M, Goldberg ID, Webb P, Kushner PJ, Pestell RG, Rosen EM (2001a) Role of direct interaction in BRCA1 inhibition of estrogen receptor activity. Oncogene 20: 77–87
    1. Fan S, Meng Q, Gao B, Grossman J, Yadegari M, Goldberg ID, Rosen EM (2000) Alcohol stimulates estrogen receptor signaling in human breast cancer cell lines. Cancer Res 60: 5635–5639
    1. Fan S, Wang JA, Yuan RQ, Goldberg ID, Rosen EM (1999a) Co-ordinate down-regulation of BRCA1, BRCA2, Rad51, and p300 during the response to damage in human prostate cancer cell lines. Prostate 40: 37–49
    1. Fan S, Wang JA, Yuan RQ, Ma YX, Meng Q, Erdos MR, Brody LC, Goldberg ID, Rosen EM (1998) BRCA1 as a human prostate tumor suppressor: Modulation of proliferation, damage responses, and expression of cell regulatory proteins. Oncogene 16: 3069–3083
    1. Fan S, Wang J, Yuan R, Ma Y, Meng Q, Erdos MR, Pestell RG, Yuan F, Auborn KJ, Goldberg ID, Rosen EM (1999b) BRCA1 inhibits estrogen receptor signaling in transfected cells. Science 284: 1354–1356
    1. Fan S, Yuan RQ, Ma YX, Meng Q, Goldberg ID, Rosen EM (2001b) Mutant BRCA1 genes antagonize phenotype of wild-type BRCA1. Oncogene 20: 8215–8235
    1. Graham S, Marshall J, Mettlin C, Rzepka T, Nemoto T, Byers T (1982) Diet in the epidemiology of breast cancer. Am J Epidemiol 116: 68–75
    1. Hayes JD, McMahon M (2001) Molecular basis for the contribution of the antioxidant responsive element to cancer chemoprevention. Review. Cancer Lett 174: 103–113
    1. Jeffy BD, Chirnomas RB, Chen EJ, Gudas JM, Romagnolo DF (2002) Activation of the aromatic hydrocarbon receptor pathway is not sufficient for transcriptional repression of BRCA-1: requirements for metabolism of benzo[a]pyrene to 7r,8t-dihydroxy-9t,10-epoxy-7,8,9,10-tetrahydrobenzo[a]pyrene. Cancer Res 62: 113–121
    1. Jin L, Qi M, Chen DZ, Anderson A, Yang GY, Arbeit JM, Auborn KJ (1999) Indole-3-carbinol prevents cervical cancer in human papilloma virus type 16 (HPV16) transgenic mice. Cancer Res 59: 3991–3997
    1. Jones LP, Li M, Halama ED, Ma Y, Lubet R, Grubbs CJ, Deng CX, Rosen EM, Furth PA (2005) Promotion of mammary cancer development by tamoxifen in a mouse model of Brca1-mutation-related breast cancer. Oncogene 24: 3554–3562
    1. Kojima T, Tanaka T, Mori H (1994) Chemoprevention of spontaneous endometrial cancer in female donryu rats by indole-3-carbinol. Cancer Res 54: 1446–1449
    1. Kuiper GG, Lemmen JG, Carlsson B, Corton JC, Safe SH, van der Saag PT, van der Burg B, Gustafsson JA (1998) Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology 139: 4252–4263
    1. Kwak MK, Itoh K, Yamamoto M, Kensler TW (2002) Enhanced expression of the transcription factor Nrf2 by cancer chemopreventive agents: role of antioxidant response element-like sequences in the nrf2 promoter. Mol Cell Biol 22: 2883–2892
    1. Lamartiniere CA, Cotroneo MS, Fritz WA, Wang J, Mentor-Marcel R, Elgavish A (2002) Genistein chemoprevention: timing and mechanisms of action in murine mammary and prostate tumors. J Nutr 132: 552S–558S
    1. Le HT, Schaldach CM, Firestone GL, Bjeldanes JF (2003) Plant derived 3,3′-diindolylmethane is a strong androgen antagonist in human prostate cancer cells. J Biol Chem 278: 21136–21145
    1. Maggiolini M, Vivacqua A, Carpino A, Bonofiglio D, Fasanella G, Salerno M, Picard D, Ando S (2002) The mutant androgen receptor T877A mediates the proliferative but not the cytotoxic dose-dependent effects of genistein and quercetin on human LNCaP prostate cancer cells. Mol Pharmacol 62: 1027–1035
    1. Meng Q, Goldberg ID, Rosen EM, Fan S (2001) Inhibitory effects of indole-3-carbinol on invasion and migration in human breast cancer cells. Breast Cancer Res Treat 63: 147–152
    1. Meng Q, Qi M, Chen DZ, Yuan R, Goldberg ID, Rosen EM, Auborn K, Fan S (2000a) Suppression of breast cancer invasion and migration mediated by indole-3-carbinol: associated with up-regulation of BRCA1 and E-cadherin/catenin complexes. J Mol Med 78: 155–165
    1. Meng Q, Yuan F, Goldberg ID, Rosen EM, Auborn K, Fan S (2000b) Indole-3-carbinol is a negative regulator of estrogen receptor-α signaling in human tumor cells. J Nutr 130: 2927–2931
    1. Mentor-Marcel R, Lamartiniere CA, Greenberg NM, Elagavish A (2001) Genistein in the diet reduces the incidence of prostate tumors in a transgenic mouse (TRAMP). Cancer Res 61: 6777–6782
    1. Michnovicz JJ, Bradlow HL (1990) Induction of estradiol metabolism by dietary indole-3-carbinol in humans. J Natl Cancer Inst 82: 947–949
    1. Nacheson-Kedmi M, Yannai S, Haj A, Fares FA (2003) Indole-3-carbinol and 3,3'-diindolylmethane induce apoptosis in human prostate cancer cells. Food Chem Toxicol 41: 745–752
    1. Oyadomari S, Mori M (2004) Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell Death Differ 11: 381–389, Review
    1. Page KM, Heblich F, Davies A, Butcher AJ, Leroy J, Bertaso F, Pratt WS, Dolphin AC (2004) Dominant-negative calcium channel suppression by truncated constructs involves a kinase implicated in the unfolded protein response. J Neurosci 24: 5400–5409
    1. Rahman KM, Aranha O, Sarkar FH (2003) Indole-3-carbinol (I3C) induces apoptosis in tumorigenic but not in nontumorigenic breast epithelial cells. Nutr Cancer 45: 101–112
    1. Rajan JV, Marquis ST, Gardner HP, Chodosh LA (1997) Developmental expression of Brca2 colocalizes with Brca1 and is associated with proliferation and differentiation in multiple tissues. Dev Biol 184: 385–401
    1. Rajan JV, Wang M, Marquis ST, Chodosh LA (1996) Brca2 is coordinately regulated with Brca1 during proliferation and differentiation in mammary epithelial cells. Proc Natl Acad Sci USA 93: 13078–13083
    1. Rice JC, Ozcelik H, Maxeiner P, Andrulis I, Futscher BW (2000) Methylation of the BRCA1 promoter is associated with decreased BRCA1 mRNA levels in clinical breast cancer specimens. Carcinogenesis 21: 1761–1765
    1. Rosen CA, Woodson GE, Thompson JW, Hengesteg AP, Bradlow HL (1998) Preliminary results of the use of indole-3-carbinol for recurrent laryngeal papillomatosis. Otolaryngol Head Neck Surg 118: 810–815
    1. Roybal CN, Yang S, Sun CW, Hurtado D, Vander Jagt DL, Townes TM, Abcouwer SF (2004) Homocysteine increases the expression of vascular endothelial growth factor by a mechanism involving endoplasmic reticulum stress and transcription factor ATF4. J Biol Chem 279: 14844–14852
    1. Rutkowski DT, Kaufman RJ (2003) All roads lead to ATF4. Dev Cell 4: 442–444, Review
    1. Sarkar FH, Li Y (2002) Mechanisms of cancer chemoprevention by soy isoflavone genistein. Cancer Metast Rev 21: 265–280
    1. Sarkar FH, Rahman KM, Li Y (2003) Bax translocation to mitochondria is an important event in inducing apoptotic cell death by indole-3-carbinol (I3C) treatment of breast cancer cells. J Nutr 133: 2434S–2439S
    1. Shen X, Ellis RE, Lee K, Liu CY, Yang K, Solomon A, Yoshida H, Morimoto R, Kurnit DM, Mori K, Kaufman RJ (2001) Complementary signaling pathways regulate the unfolded protein response and are required for C. elegans development. Cell 107: 893–903
    1. Shin S, Verma IM (2003) BRCA2 cooperates with histone acetyltransferases in androgen receptor-mediated transcription. Proc Natl Acad Sci USA 100: 7201–7206
    1. Staff S, Isola J, Tanner M (2003) Haplo-insufficiency of BRCA1 in sporadic breast cancer. Cancer Res 63: 4978–4983
    1. Struewing JP, Hartge P, Wacholder S, Baker SM, Berlin M, McAdams M, Timmerman MM, Brody LC, Tucker MA (1997) The risk of cancer associated with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med 336: 1401–1408
    1. Sun S, Han J, Ralph Jr WM, Chandrasekaran A, Liu K, Auborn KJ, Carter TH (2004) Endoplasmic reticulum stress as a correlate of cytotoxicity in human tumor cells exposed to diindolylmethane in vitro. Cell Stress Chaperones 9: 76–87
    1. Thompson D, Easton DF, Breast Cancer Linkage Consortium (2002) Cancer incidence in BRCA1 mutation carriers. J Natl Cancer Inst 94: 1358–1365
    1. Vissac-Sabatier C, Bignon YJ, Bernard-Gallon DJ (2003) Effects of the phytoestrogens genistein and daidzein on BRCA2 tumor suppressor gene expression in breast cell lines. Nutr Cancer 45: 247–255
    1. Wang TT, Sathyamoorthy N, Phang JM (1996) Molecular effects of genistein on estrogen receptor mediated pathways. Carcinogenesis 17: 271–275
    1. Wang XZ, Harding HP, Zhang Y, Jolicoeur EM, Kuroda M, Ron D (1998) Cloning of mammalian Ire1 reveals diversity in the ER stress responses. EMBO J 17: 5708–5817
    1. Wilson CA, Ramos L, Villasenor MR, Anders KH, Press MF, Clarke K, Karlan B, Chen JJ, Scully R, Livingston D, Zuch RH, Kanter MH, Cohen S, Calzone FJ, Slamon DJ (1999) Localization of human BRCA1 and its loss in high-grade, non-inherited breast carcinomas. Nat Genet 21: 236–340
    1. Xiong J, Fan S, Meng Q, Schramm L, Wang C, Bouzahza B, Zhou J, Zafonte B, Goldberg ID, Haddad BR, Pestell RG, Rosen EM (2003) BRCA1 inhibition of telomerase activity in cultured cells. Mol Cell Biol 23: 8668–8690
    1. Yamamoto K, Yoshida H, Kokame K, Kaufman RJ, Mori K (2004) Differential contributions of ATF6 and XBP1 to the activation of endoplasmic reticulum stress-responsive cis-acting elements ERSE, UPRE and ERSE-II. J Biochem (Tokyo) 136: 343–350
    1. Yeh S, Hu YC, Rahman M, Lin HK, Hsu CL, Ting HJ, Kang HY, Chang C (2000) Increase of androgen-induced cell death and androgen receptor transactivation by BRCA1 in prostate cancer cells. Proc Natl Acad Sci USA 97: 11256–11261
    1. Yoshida H, Haze K, Yanagi H, Yura T, Mori K (1998) Identification of the cis-acting endoplasmic reticulum stress response element responsible for transcriptional induction of mammalian glucose-regulated proteins. J Biol Chem 273: 33741–33749

Source: PubMed

Подписаться