Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Experimental Therapeutics, Molecular Targets, and Chemical Biology

Regulation of Prostaglandin Metabolism by Calcitriol Attenuates Growth Stimulation in Prostate Cancer Cells

Jacqueline Moreno, Aruna V. Krishnan, Srilatha Swami, Larisa Nonn, Donna M. Peehl and David Feldman
Jacqueline Moreno
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Aruna V. Krishnan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Srilatha Swami
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Larisa Nonn
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Donna M. Peehl
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
David Feldman
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/0008-5472.CAN-05-1435 Published September 2005
  • Article
  • Info & Metrics
  • PDF
Loading

Abstract

Calcitriol exhibits antiproliferative and prodifferentiation effects in prostate cancer. Our goal is to further define the mechanisms underlying these actions. We studied established human prostate cancer cell lines and primary prostatic epithelial cells and showed that calcitriol regulated the expression of genes involved in the metabolism of prostaglandins (PGs), known stimulators of prostate cell growth. Calcitriol significantly repressed the mRNA and protein expression of prostaglandin endoperoxide synthase/cyclooxygenase-2 (COX-2), the key PG synthesis enzyme. Calcitriol also up-regulated the expression of 15-hydroxyprostaglandin dehydrogenase, the enzyme initiating PG catabolism. This dual action was associated with decreased prostaglandin E2 secretion into the conditioned media of prostate cancer cells exposed to calcitriol. Calcitriol also repressed the mRNA expression of the PG receptors EP2 and FP, providing a potential additional mechanism of suppression of the biological activity of PGs. Calcitriol treatment attenuated PG-mediated functional responses, including the stimulation of prostate cancer cell growth. The combination of calcitriol with nonsteroidal anti-inflammatory drugs (NSAIDs) synergistically acted to achieve significant prostate cancer cell growth inhibition at ∼2 to 10 times lower concentrations of the drugs than when used alone. In conclusion, the regulation of PG metabolism and biological actions constitutes a novel pathway of calcitriol action that may contribute to its antiproliferative effects in prostate cells. We propose that a combination of calcitriol and nonselective NSAIDs might be a useful chemopreventive and/or therapeutic strategy in men with prostate cancer, as it would allow the use of lower concentrations of both drugs, thereby reducing their toxic side effects.

  • Calcitriol
  • prostaglandins
  • 15-PGDH
  • COX-2
  • NSAIDs

Introduction

In the United States, prostate cancer remains the most common solid tumor malignancy in men, causing ∼30,000 deaths in 2005 ( 1). Effective treatment options include surgery and radiation therapy. The main treatment strategy for advanced prostate cancer involves androgen deprivation therapy to which patients initially respond very well. However, most patients eventually fail this therapy and frequently develop metastatic disease. Current research on prostate cancer aims to identify new agents that would prevent and/or inhibit its progression.

1,25-Dihydroxyvitamin D3 (calcitriol), the active form of vitamin D, is the major regulator of calcium and phosphate homeostasis in bone, kidney, and intestine ( 2). However, calcitriol has also been shown to exhibit antiproliferative and prodifferentiation effects in many normal and malignant cells including prostate cancer cells ( 3– 10). There are multiple mechanisms underlying the antiproliferative effects of calcitriol, which vary between target cells ( 10). These include cell cycle arrest ( 9, 11) and the induction of apoptosis ( 12). Several genes that mediate these growth regulatory effects have been identified to be the molecular targets of calcitriol action, such as p21, p27, bcl-2, and insulin-like growth factor binding protein-3 (IGFBP-3; refs. 5– 14). We recently did cDNA microarray analyses to more fully characterize the spectrum of genes regulated by calcitriol in prostate cells ( 15, 16). Among the newly identified genes regulated by calcitriol, we found two genes which play a key role in prostaglandin (PG) metabolism: the prostaglandin endoperoxide synthase-2 or cyclooxygenase (COX)-2 and the NAD+-dependent 15-hydroxyprostaglandin dehydrogenase (15-PGDH). PGs are synthesized from free arachidonic acid ( 17) by COXs. There are two well-characterized COX isoforms: COX-1, a constitutive form of the enzyme, and COX-2, an inducible form of the enzyme. PGs are implicated in the initiation and progression of many malignancies including prostate cancer ( 18– 20). Tumor cells with elevated COX-2 levels are highly resistant to apoptosis, show increased angiogenic potential, and exert suppressive effects on host immunity ( 19, 20). Nonsteroidal anti-inflammatory drugs (NSAIDs), known inhibitors of both COX-1 and COX-2 enzymatic activity, are under intense investigation to prevent and/or treat malignancies ( 19, 21). 15-PGDH, which mediates the catalytic inactivation of PGs by converting them to the corresponding keto derivatives, has been found to be down-regulated in some cancers ( 22, 23) and has recently been regarded as a tumor suppressor gene ( 24).

In the current study, we investigated the regulation of COX-2 and 15-PGDH by calcitriol in the androgen-dependent LNCaP and androgen-independent PC-3 human prostate cancer cell lines as well as in primary prostatic epithelial cells derived from normal and cancerous human prostate tissue. Calcitriol reduced the expression of COX-2 and increased that of 15-PGDH. Calcitriol treatment of prostate cancer cells decreased the concentration of prostaglandin E2 (PGE2) secreted into the conditioned medium. In addition, calcitriol also decreased the expression of the mRNA of PG receptors EP2 and FP. Our data indicate that these mechanisms led to the attenuation of PG-mediated functional responses by calcitriol, including the suppression of PG stimulation of cell growth. Further, our study showed that the combination of calcitriol and NSAIDs exhibited synergistic growth inhibition, suggesting that the combination might be a useful therapeutic and/or chemopreventive strategy in prostate cancer.

Materials and Methods

Materials

PGE2, prostaglandin F2α (PGF2α), arachidonic acid, NS-398, and SC-58125 were obtained from Cayman Chemical Co. (Ann Arbor, MI). Calcitriol was a gift from Leo Pharma A/S (Ballerup, Denmark). All stock solutions were made in 100% ethanol and stored at −20°C. Tissue culture media were obtained from Mediatech, Inc. (Herndon, VA). Tissue culture supplements and fetal bovine serum (FBS) were obtained from Life Technologies, Inc. (Grand Island, NY).

Methods

Cell culture. LNCaP (ATCC no. CRL-1740) and PC-3 (ATCC no. CRL-1435) cells were grown in RPMI 1640 supplemented with 5% FBS, 100 IU/mL of penicillin, and 100 μg/mL streptomycin (Life Technologies). Cells were maintained at 37°C with 5% CO2 in a humidified incubator. Primary cells were derived from radical prostatectomy specimens from men undergoing surgery for prostate cancer treatment. None of the patients had received prior therapy for prostate cancer. The normal cell strains (E-PZ-1 to -3) were derived from peripheral zone tissue with no histologic evidence of cancer in adjacent sections. The cancer cell strains used, E-CA-1 (Gleason grade 3/3), E-CA-2 (Gleason grade 4/5), and E-CA-3 (Gleason grade 4/3), were derived from adenocarcinoma specimens. Primary cell cultures were established from the prostate tissue samples and propagated in culture as we have previously described ( 25).

Cell proliferation assay. Prostate cancer cells were seeded at an initial density of 1.5 × 105 cells/well in six-well tissue culture plates and allowed to attach overnight in RPMI 1640 with 5% FBS. Cell cultures were shifted to medium containing 2% FBS and treated in triplicate over the next 6 days with either 0.1% ethanol vehicle or the indicated concentrations of drugs. Fresh media and the drugs were replenished every other day. At the end of the treatment, the cells were collected by gentle scraping and subjected to lysis in 0.2 N NaOH. Cell proliferation was assessed by the determination of DNA content ( 26).

RNA isolation and real-time reverse transcription-PCR. Total RNA was isolated from vehicle or drug-treated cells by the Chomczynski method using Trizol reagent (Invitrogen, Life Technologies, Inc., Carlsbad, CA) as previously described ( 15). The yield and purity of isolated RNA were checked by UV spectrophotometry. Five micrograms of total RNA were used in reverse transcription reactions using the SuperScript III first strand synthesis kit (Invitrogen). Gene expression in vehicle or calcitriol-treated cells was determined by real-time PCR using the reverse transcription product and gene-specific primers. The reactions were carried out with the DyNamo SYBR Green qPCR kit (Finnzymes, Oy, M.J. Research, Reno, NV) in a 20 μL reaction volume containing gene-specific primers (10 pmol). All real-time PCR reactions were done in duplicate according to the following program: incubation at 72°C for 5 minutes, incubation at 95°C for 5 minutes, and 40 cycles of 94°C for 20 seconds, 58°C for 15 seconds, and 72°C for 20 seconds. PCR products were subjected to agarose gel electrophoresis to determine the purity and size of the amplified products ( 27). Real-time PCR was carried out using an Opticon 2 DNA engine (M.J. Research). Relative changes in mRNA expression levels were assessed by the 2−ΔΔC(T) method ( 28). Changes in mRNA expression of the different genes were normalized to that of TATA binding protein (TBP) gene or glyceraldehyde-3-phosphate dehydrogenase (GAPDH) gene. The primers used were as follows: COX-2: 5′-GATACTCAGGCAGAGATGATCTACCC-3′ (sense), 5′-AGACCAGGCACCAGACCAAAGA-3′ (antisense); 15-PGDH: 5′-GACTCTGTTCATCCAGTGCG-3′ (sense), 5′-CCTTCACCTCCATTTTGCTTACTC-3′ (antisense); c-fos: 5′-GAATAAGATGGCTGCAGCCAAATGCCGCAA-3′ (sense), 5′-CAGTCAGATCAAGGGAAGCCACAGACATCT-3′ (antisense; ref. 29); EP2: 5′-GTGCTGACAAGGCACTTCATGT-3′ (sense), 5′-TGTTCCTCCAAAGGCCAAGTAC-3′ (antisense); FP: 5′-GCACATTGATGGGCAACTAGAA-3′ (sense), 5′-GCACCTATCATTGGCATGTAGCT-3′ (antisense); TBP: 5′-CACTCACAGACTCTCACAACTGC-3′ (sense), 5′-GTGGTTCGTGGCTCTCTTATC-3′ (antisense); GAPDH: 5′-GCCTCAAGATCATCAGCA-3′ (sense), 5′-GTTGCTGTAGCCAAATTC-3′ (antisense).

Measurement of prostaglandin E2 secretion. Subconfluent LNCaP cells were treated with vehicle or calcitriol for 48 hours. Conditioned media were collected and secreted PGE2 levels were quantitated using a PGE2 monoclonal enzyme immunoassay kit (Cayman Chemical) according to the protocol of the manufacturer.

Western blot analysis. Cell lysates were prepared from vehicle or calcitriol-treated cells by lysis with a buffer containing 50 mmol/L Tris-HCl, 1 mmol/L EDTA, and 1.6 mmol/L CHAPS (Sigma-Aldrich, St. Louis, MO) supplemented with a protease inhibitor cocktail (Complet, Roche Diagnostics GmbH, Mannheim, Germany). Lysates were incubated at 4°C for 20 minutes and centrifuged at 10,000 × g for 1 minute to sediment particulate material. The protein concentration of the supernatant was measured by the Bradford method ( 30). Proteins were separated in either 10% NuPAGE gels in MOPS-SDS running buffer (Invitrogen) or 10% polyacrylamide Tris-Tricine (Sigma-Aldrich) gels depending on the size of the protein to be detected. After electrophoresis, proteins were transferred onto nitrocellulose membranes by electroblotting. The COX-2 monoclonal (1:1000 dilution) and 15-PGDH polyclonal antibodies (1:250 dilution) used in our study were purchased from Cayman Chemicals. β-Actin monoclonal antibody (dilution 1:500) was obtained from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). Membranes were incubated with the appropriate primary antibodies followed by incubations with a secondary antibody to immunoglobulin G conjugated to horseradish peroxidase (Cell Signaling Technology, Inc., Beverly, MA). Immunoreactive bands were visualized using the enhanced chemiluminescence Western blot detection system (Amersham, Piscataway, NJ) according to the instructions of the manufacturer. The blots were also probed for the expression of β-actin as a control. COX-2 protein was visualized as a ∼70 kDa immunoreactive band. 15-PGDH protein was visualized as a ∼29 kDa immunoreactive band. Immunoreactive bands were scanned by densitometry (HP Scanjet 7400C) and quantified using Bio-Rad software (Bio-Rad, Hercules, CA). COX-2 or 15-PGDH signals were normalized to β-actin levels in each sample.

Results

We previously showed by cDNA microarray analysis that calcitriol regulated the expression of two of the key genes involved in PG metabolism (i.e., COX-2 and 15-PGDH) in LNCaP human prostate cancer cells ( 15) and 15-PGDH in primary normal prostatic epithelial cells ( 16). In the present study, we extended these observations to include an evaluation of calcitriol effects on the expression of these two genes at both the mRNA and protein levels in LNCaP and PC-3 cells. In addition, we also examined the effects of calcitriol in primary prostatic epithelial cell strains derived from normal prostate as well as prostate adenocarcinoma specimens.

Down-regulation of cyclooxygenase-2 expression by calcitriol. Real-time reverse transcription-PCR (RT-PCR) analysis showed significant decreases in COX-2 mRNA levels in both androgen-dependent LNCaP (∼70% inhibition) and androgen-independent PC-3 cells (∼45% inhibition) due to calcitriol treatment ( Fig. 1A ). Although both LNCaP and PC-3 prostate cancer cells have been shown to express COX-2 protein ( 31), we found that PC-3 cells exhibited higher basal levels of COX-2 protein expression when compared with LNCaP cells (not shown). We therefore used PC-3 cells to assess the effect of calcitriol on COX-2 protein expression. Figure 1B shows that the addition of 10 nmol/L calcitriol to PC-3 cultures for 48 hours reduced COX-2 protein level to ∼60% of control, with 100 nmol/L calcitriol having no further effect.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Calcitriol regulates COX-2 and 15-PGDH expression in prostate cancer cell lines. A, calcitriol decreases COX-2 mRNA levels. Subconfluent cultures of LNCaP and PC-3 cells were treated with 0.1% ethanol (Con) or 10 nmol/L calcitriol (Cal) for 24 hours and total RNA was extracted. COX-2 mRNA levels were determined by real-time RT-PCR as described in Materials and Methods and were normalized to TBP mRNA levels in the same samples. COX-2/TBP ratio shown as a percent of control set at 100%; columns, mean from five experiments; bars, SE. B, calcitriol decreases COX-2 protein levels. Subconfluent cultures of PC-3 cells were treated with 0.1% ethanol (Con) or 10 or 100 nmol/L calcitriol (Cal) for 48 hours. Fifty micrograms of total protein were subjected to Western blot analysis as described in Materials and Methods. Representative Western blot. The densitometric units of COX-2 immunoreactive bands were normalized to the densitometric units of the corresponding β-actin bands. Results expressed as the ratio of the control set at 1. C, calcitriol increases 15-PGDH mRNA levels. Cells were treated and processed as described in A. 15-PGDH/TBP ratio in calcitriol-treated cells given as a percent of control set at 100%; columns, mean from five experiments; bars, SE. D, calcitriol increases 15-PGDH protein levels. LNCaP cells were treated as in B. The densitometric units of 15-PGDH immunoreactive bands were normalized to the densitometric units of the corresponding β-actin bands. Results expressed as the ratio of the control set at 1; columns, mean of three experiments; bars, SE. *, P < 0.05; **, P < 0.01, when compared with control.

Up-regulation of 15-hydroxyprostaglandin dehydrogenase expression by calcitriol. We examined the effect of calcitriol on 15-PGDH mRNA levels in LNCaP and PC-3 cells. Our data show that 10 nmol/L calcitriol increased 15-PGDH mRNA expression by ∼3.6-fold in LNCaP cells and by ∼3-fold in PC-3 cells ( Fig. 1C). We found that the basal protein expression of 15-PGDH varied between different cell lines with the LNCaP exhibiting appreciable levels of the 15-PGDH protein whereas barely detectable levels were seen in PC-3 cells. Therefore, we examined the effect of calcitriol on 15-PGDH protein expression in LNCaP cells and found a dose-dependent increase in 15-PGDH protein levels in response to calcitriol treatment ( Fig. 1D).

Calcitriol effects on cyclooxygenase-2 and 15-hydroxyprostaglandin dehydrogenase mRNA levels in primary prostatic epithelial cells. We extended our analysis to include calcitriol effects on primary cultures of prostatic epithelial cells derived from normal prostate as well as adenocarcinoma specimens removed at surgery. Real-time RT-PCR analysis showed considerable decreases (55-90%) in COX-2 mRNA levels in two of the three normal primary cell strains tested (E-PZ-1 and E-PZ-3) after 24 hours of calcitriol treatment ( Fig. 2A ). In all three cancer-derived primary cultures (E-CA-1 to -3) significant reductions (∼48-60%) in COX-2 mRNA levels were seen at an earlier time point, after 6 hours of calcitriol treatment, and the down-regulatory effect was lost by 24 hours except in the case of E-CA-2 ( Fig. 2B). Figure 2C and D shows the calcitriol-induced changes in 15-PGDH mRNA in primary prostatic cells. In the normal primary cells, calcitriol treatment caused appreciable increases in 15-PGDH mRNA in two of the three strains tested. The time course of this effect showed minor differences. In E-PZ-1 and E-PZ-2 cells significant increases (∼2- to 18-fold) were achieved at the end of 6 and 24 hours, respectively ( Fig. 2C). In two of three of the cancer-derived primary cultures (E-CA-2 and -3), significant increases (∼2- to 5-fold) were seen at the end of 24 hours ( Fig. 2D). In general, the magnitude of COX-2 mRNA down-regulation as well as 15-PGDH mRNA increase was more pronounced in the primary cells derived from normal prostatic tissue when compared with both the cancer-derived primary cells and the established prostate cancer cell lines.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Calcitriol regulates the expression of COX-2 and 15-PGDH mRNA in primary prostatic epithelial cells. Primary cultures of prostatic epithelial cells derived from the peripheral zone of normal prostate tissue (E-PZ-1 to -3) or adenocarcinoma (E-CA-1 to -3) were treated with 0.1% ethanol (Con) or with 10 nmol/L calcitriol (Cal) for 6 or 24 hours. Total RNA was extracted and COX-2 and 15-PGDH mRNA levels were quantitated by real-time RT-PCR using gene-specific primers as described in Materials and Methods. COX-2 and 15-PGDH mRNA levels were normalized to GAPDH mRNA levels and are given as a percent of control set at 100%. Columns, mean from three experiments; bars, SE. Effect of calcitriol on COX-2 mRNA in three different normal primary epithelial cell strains (A) and in cancer-derived cell strains (B). Changes in 15-PGDH mRNA in normal cell strains (C) and in cancer-derived cell strains (D). *, P < 0.05; **, P < 0.01; ***, P < 0.001, when compared with control.

Effect of calcitriol on prostaglandin levels. As a result of the dual action of calcitriol to down-regulate the expression of PG synthesizing COX-2 and increase the PG catabolizing 15-PGDH, we expected a reduction in PG production and secretion in prostate cancer cells treated with calcitriol. We measured the levels of PGE2 in the conditioned media from LNCaP cells treated with various concentrations of calcitriol for 48 hours. Figure 3A shows that the addition of calcitriol caused a significant reduction in PGE2 secretion with the maximal decrease (∼34%) seen with 100 nmol/L calcitriol.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

A, calcitriol decreases PGE2 levels. Subconfluent cultures of LNCaP cells were treated with 0.1% ethanol (Con) or with the indicated concentrations of calcitriol (Cal) for 48 hours. Conditioned media from control and calcitriol-treated cultures were collected and PGE2 levels were determined using an enzyme immunoassay kit (Materials and Methods). Columns, mean from three experiments; bars, SE. *, P < 0.05. B, changes due to calcitriol treatment in EP2 mRNA. LNCaP cells were grown to subconfluence and treated with vehicle (0.1% ethanol; Con) or 10 nmol/L calcitriol (Cal) for 24 hours. Total RNA was extracted and analyzed for the mRNA expression of EP2 by real-time RT-PCR using gene-specific primers as described in Materials and Methods. EP2 mRNA levels were normalized to the TBP mRNA levels. Values given as a percent of control set at 100%; columns, mean from three experiments; bars, SE. *, P < 0.05, when compared with control. C, changes due to calcitriol treatment in FP mRNA. LNCaP cells were treated and processed as in B for the mRNA expression of FP by real-time RT-PCR. FP mRNA levels were normalized to the TBP mRNA levels. Values are given as a percent of control set at 100%; columns, mean from three experiments; bars, SE. *, P < 0.05, when compared with control. D, calcitriol inhibits PG-mediated induction of c-fos mRNA. Subconfluent cultures of PC-3 cells were transferred to serum-free RPMI 1640 containing 0.1% ethanol vehicle or 10 nmol/L calcitriol during 48 hours (pretreatment). Following the pretreatment, the cultures were exposed for 30 minutes to arachidonic acid (AA; 3 μmol/L) added to the culture medium. The cell cultures were then scraped, RNA was isolated, and c-fos mRNA levels were determined by real-time RT-PCR as described in Materials and Methods. c-fos mRNA levels were normalized to TBP mRNA levels and are given as a percent of control set at 100%. Columns, mean from three experiments; bars, SE. *, P < 0.05, when compared with control; +, P < 0.05, when compared with arachidonic acid.

Effects of calcitriol on prostaglandin receptor expression. Prostate cancer cells have been shown to express the PGE receptor subtypes EP2 and EP4 ( 29). We examined the effects of calcitriol on the expression of the PGE2 receptor isoforms EP1, EP2, EP3, and EP4, and the PGF2α receptor FP. LNCaP cells treated with 10 nmol/L calcitriol for 24 hours showed a significant (∼45%) down-regulation of EP2 mRNA ( Fig. 3B). We did not detect any changes in the levels of EP1, EP3, or EP4 mRNA following calcitriol treatment (not shown). FP mRNA levels were also down-regulated (∼33% decrease) by calcitriol ( Fig. 3C).

Inhibition of prostaglandin-mediated induction of c-fos mRNA by calcitriol. Because calcitriol modulated the levels of biologically active PGs as well as PG receptor expression, we examined its effect on a PG-mediated functional response, (i.e., the induction of the immediate-early gene c-fos; ref. 29). As serum is a potent inducer of c-fos expression ( 32), we conducted the experiment under serum-free conditions using PC-3 cells. Unlike LNCaP, PC-3 cells could be briefly maintained in serum-free media for calcitriol pretreatment and subsequent treatment with the PG precursor arachidonic acid. PC-3 cells were pretreated with vehicle or 10 nmol/L calcitriol for 48 hours followed by a brief (30 minutes) exposure to exogenous arachidonic acid (3 μmol/L) directly added to the culture medium. RNA was then isolated and the induction of c-fos mRNA was determined as an indicator of the biological activity of PGs endogenously synthesized from arachidonic acid. As shown in Fig. 3D, in vehicle pretreated cells arachidonic acid exposure resulted in a significant induction (∼2.5-fold) of c-fos mRNA levels after 30 minutes. Calcitriol pretreatment completely abrogated the induction of c-fos mRNA due to arachidonic acid addition. Calcitriol pretreatment by itself caused a minor increase in c-fos mRNA levels when compared with vehicle pretreated cells, which was not statistically significant.

Effects of calcitriol on prostaglandin-mediated growth stimulation. We examined the effect of calcitriol on the stimulation of prostate cancer cell growth by exogenous PG addition as well as by endogenous PGs derived from the substrate arachidonic acid added to the culture medium. We treated LNCaP and PC-3 cells with arachidonic acid (3 μmol/L), PGE2, or PGF2α (10 μmol/L each) in the absence or presence of 10 nmol/L calcitriol. Our results revealed a moderate but significant growth stimulation by arachidonic acid and exogenous PGs in both LNCaP ( Fig. 4A ) and PC-3 cells ( Fig. 4B). Calcitriol had a marked growth inhibitory action when given alone. In addition, calcitriol blocked the growth stimulation due to endogenous PGs derived from the added arachidonic acid as well as exogenous PG addition ( Fig. 4A and B).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Calcitriol abrogates the growth stimulatory effects of arachidonic acid (AA) and exogenous PGs. LNCaP (A) and PC-3 (B) were treated with arachidonic acid (3 μmol/L), PGE2 (10 μmol/L), or PGF2α (10 μmol/L) individually or in combination with 10 nmol/L calcitriol (Cal) for 6 days. Cell growth was determined by measurement of DNA content as described in Materials and Methods. DNA contents are given as percentage of control value set at 100%, which was equivalent to 12.3 ± 1.2 μg/well for LNCaP cells and 19.3 ± 1.7 μg/well for PC-3 cells. Columns, mean from six experiments; bars, SE. *, P < 0.05, when compared with control; ++, P < 0.01, when compared with arachidonic acid, PGE2, or PGF2α alone.

Synergistic inhibition of prostate cancer cell growth by calcitriol and nonsteroidal anti-inflammatory drugs. We next examined the combined effect of calcitriol and NSAIDs, which are potent inhibitors of COX enzyme activity. We tested a number of both COX-2–selective and nonselective NSAIDs including NS-398, SC-58125, flufenamic acid, sulindac sulfide, indomethacin, naproxen, and ibuprofen. Figure 5A to D illustrates the effect on prostate cancer cell growth of calcitriol alone or in combination with the NSAIDs that exhibited the best growth inhibitory effect when used at a reduced dose. We show the effects of calcitriol alone and in combination with the COX-2–selective NSAIDs SC-58125 on the growth of LNCaP cells ( Fig. 5A) and NS-398 on the growth of PC-3 cells ( Fig. 5B). In LNCaP cells, calcitriol by itself had a modest effect (∼20%) at 1 nmol/L but caused significant growth inhibition (∼40%) at 10 nmol/L ( Fig. 5A). The addition of the COX-2–specific inhibitor SC-58125 by itself had a modest effect on cell growth (∼20% inhibition), which was not statistically significant, at the concentration tested (5 μmol/L). The combination of 1 nmol/L calcitriol with SC-58125, however, had a more pronounced inhibitory effect (∼73% growth inhibition with the combination versus ∼20% inhibition with the individual agents), indicating a synergistic interaction between these two drugs to inhibit cell growth. SC-58125 also enhanced the growth inhibition seen with the higher concentration of calcitriol (∼80% inhibition with the combination versus ∼40% inhibition with 10 nmol/L calcitriol alone). Similar synergistic growth inhibitory effects were evident in PC-3 cells treated with a combination of calcitriol and the COX-2–selective inhibitor NS-398 ( Fig. 5B). NS-398, when used alone at 7.5 μmol/L, did not affect the growth of PC-3 cells. However, it enhanced the growth inhibition seen with both 1 and 10 nmol/L calcitriol (∼60% inhibition with the combination versus ∼20% inhibition with 1 nmol/L calcitriol alone, and ∼75% inhibition with the combination versus ∼40% inhibition with 10 nmol/L calcitriol alone).

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Synergistic inhibition of prostate cancer cell growth by calcitriol and NSAIDs. LNCaP or PC-3 cells were treated with 0.1% ethanol vehicle (Con) or 10 nmol/L calcitriol (Cal) in the presence and absence of the indicated NSAID. Cell growth was determined by measuring the DNA content as indicated in Materials and Methods. DNA contents are given as percentage of control value set at 100%. A, LNCaP cells treated with a combination of calcitriol (Cal) and COX-2–specific NSAID SC-58125 (5 μmol/L). 100% DNA content = 10.15 ± 1.22 μg/well. B, PC-3 treated with calcitriol (Cal) in the presence and absence of the COX-2–selective NSAID NS-398 (7.5 μmol/L). 100% DNA content = 17.42 ± 1.93 μg/well. C, LNCaP cells treated with calcitriol (Cal) in the presence and absence of the nonselective NSAID naproxen (Nap; 200 μmol/L). 100% DNA content = 9.22 ± 0.5 μg/well. D, PC-3 cells treated with calcitriol (Cal) in the presence and absence of the nonselective NSAID ibuprofen (Ibu; 150 μmol/L). 100% DNA content = 21.7 ± 0.9 μg/well. Columns, mean from six experiments; bars, SE. *, P < 0.05; **, P < 0.01, when compared with control. ++, P < 0.01, when compared with 1 or 10 nmol/L Cal alone.

The growth inhibitory effect of calcitriol was similarly enhanced when combined with nonselective NSAIDs that inhibit the enzymatic activity of both COX-1 and COX-2. The nonselective NSAID naproxen at 200 μmol/L did not inhibit the growth of LNCaP cells ( Fig. 5C). However, it enhanced the growth inhibition seen with 1 and 10 nmol/L calcitriol (∼65% inhibition with the combination versus ∼48% inhibition with 10 nmol/L calcitriol alone). Similarly, in PC-3 cells ( Fig. 5D), the nonselective NSAID ibuprofen at 150 μmol/L enhanced the growth inhibitory effect of calcitriol (∼74% inhibition with the combination versus ∼40% inhibition with 10 nmol/L calcitriol alone) whereas it did not affect cell growth when used alone at this concentration.

Based on extensive dose-response analysis (not shown), we calculated the interaction index (γ) using an isobolar method ( 33) for each drug combination. This analysis indicated a synergistic (superadditive) effect. The data suggested that ∼2 to 10 times lower concentration of each drug is needed when used in combination to achieve the same degree of growth inhibition as achieved by the individual drugs.

Discussion

Calcitriol acts by multiple pathways to inhibit the proliferation of prostate cancer cells ( 5– 14). Our study shows that the regulation of PG metabolism is a novel and additional pathway by which calcitriol may exert its antiproliferative actions in prostate cancer cells. We have shown that calcitriol regulates biologically active PG levels and PG actions by three mechanisms: (a) the suppression of COX-2 expression, (b) the up-regulation of 15-PGDH expression, and (c) the reduction of EP2 and FP PG receptor mRNA expression. We propose that these three effects act together to effectively inhibit the stimulation of prostate cancer cell proliferation by endogenously derived PGs as well as PGs added exogenously. Because PGs have been shown to promote prostate cell growth, inhibit apoptosis, and stimulate prostate cancer progression ( 18– 20), we postulate that these effects of calcitriol to reduce PG actions significantly contribute to the anticancer effects of the hormone in prostate cancer.

The transformation of arachidonic acid into PGs and thromboxanes in mammalian cells is catalyzed by the enzyme COX, which has two well-characterized isoforms. COX-1 is constitutively expressed and is involved in housekeeping functions ( 17, 34). COX-2 is an immediate-early gene that is induced by a variety of growth promoting stimuli such as serum and growth factors, tumor promoters, cytokines, and proinflammatory agents ( 17, 34), and is regarded as an oncogene ( 24). COX-2 is overexpressed in various cancers including some, but not all, prostate cancers ( 18, 21). Inhibitors of COX-2 activity have been shown to suppress prostate cancer cell growth both in vivo and in vitro ( 31, 35, 36). Our results show the significant repression of COX-2 mRNA expression by calcitriol in prostate cancer cell lines as well as in primary prostatic epithelial cells and also a reduction in COX-2 protein levels in prostate cancer cell lines, suggesting that COX-2 is a calcitriol target gene.

PGE2 and PGF2α are rapidly catabolized in vivo into their biologically inactive 13,14-dihydro-15-keto metabolites by a two-step process carried out sequentially. The first step is initiated by the reversible oxidation of their 15(S)-hydroxyl group by the enzyme 15-PGDH ( 37). 15-PGDH is widely expressed in many mammalian tissues ( 38) and has been shown to be modulated by several hormones and factors ( 37– 39), indicating the potential importance of the regulation of this enzyme. In LNCaP cells, 15-PGDH expression is up-regulated by androgens, interleukin-6, and the cyclic AMP inducer forskolin in a protein kinase A–dependent manner ( 40, 41). We now show that calcitriol is an important regulator of 15-PGDH expression in prostate cancer cells. The partial repression of COX-2 mRNA expression and the increase in 15-PGDH mRNA expression are also seen in primary prostatic epithelial cells derived from normal prostate, suggesting that these calcitriol effects are not restricted to malignant prostate cells. 15-PGDH expression has been shown to be decreased in many cancers ( 22, 23, 42). Calcitriol has also been shown to increase the expression of 15-PGDH in neonatal monocytes ( 43), where it exhibits prodifferentiation effects. 15-PGDH, which physiologically antagonizes COX-2, has recently been described as a putative oncogene antagonist that functions as a tumor suppressor in colon cancer by Yan et al. ( 24) who found that 15-PGDH was universally expressed in normal colon specimens but was routinely absent or severely reduced in cancer specimens. More importantly, stable transfection of a 15-PGDH expression vector into cancer cells greatly reduced the ability of the cells to form tumors and/or slowed tumor growth in nude mice. The authors concluded that 15-PGDH suppressed the effects of the oncogene COX-2 and exhibited an additional effect to inhibit angiogenesis in vivo ( 24). Our present study shows calcitriol-mediated suppression of the oncogene COX-2 and an increase in the expression of the putative tumor suppressor 15-PGDH in prostate cells, suggesting that calcitriol may play an important role in the chemoprevention of prostate cancer.

As a result of its dual action to modulate COX-2 and 15-PGDH expression, we expected calcitriol to reduce the levels of PGs in prostate cancer cells. This indeed was the case as shown by the decrease in PGE2 levels in the conditioned media from LNCaP cells following calcitriol treatment. Calcitriol regulation of PGE2 synthesis and secretion has been also reported in growth plate chondrocytes ( 44), in monocytes ( 43, 45), and in interleukin-1β–stimulated rheumatoid synovial fibroblasts ( 46). The effects of calcitriol on PG synthesis and signaling in these target cells seem to be related to the rapid nongenomic actions of calcitriol ( 47).

PGs exert their myriad effects through G-protein coupled membrane receptors which activate different signal transduction pathways ( 48). Prostate cancer cells have been shown to express the PGE receptor subtypes EP2 and EP4 ( 29). Interestingly, our study shows that calcitriol decreases the mRNA expression of the PGE2 and PGF2α receptor subtypes EP2 and FP, providing yet another mechanism for the suppression of the biological activity of PGs by calcitriol. In a recent study examining the changes in gene expression profile in the kidney of vitamin D receptor (VDR) knockout mice, Li et al. ( 49) report increases in the expression of EP3 and FP genes in VDR−/− kidneys, suggesting that calcitriol may also regulate the expression of PG receptors in kidney. Our study indicates that calcitriol not only decreases the concentration of PGs but may also inhibit the biological activity of these reduced PG levels by repressing of EP2 and FP receptor mRNA expression in prostate cancer cells.

Chen and Hughes-Fulford ( 29) have shown that arachidonic acid increases the expression of the immediate-early gene c-fos by undergoing a COX-2–mediated conversion to PGE2, binding of PGE2 to EP2/EP4 receptors, and subsequent activation of the protein kinase A pathway, which leads to the expression of growth-related genes. PGE2 has also been to shown to up-regulate the gene expression of its own synthesizing enzyme COX-2 in prostate cancer cells, thereby completing a positive feedback loop ( 31, 50). We therefore examined the effect of calcitriol treatment on the induction of c-fos and cell growth by arachidonic acid in prostate cancer cells and found that calcitriol abolished c-fos induction and growth stimulation by arachidonic acid. Our interpretation of these observations is that they reflect both the effect of calcitriol to decrease endogenous synthesis of PGs due to COX-2 suppression and the ability of calcitriol to attenuate the biological activity of the PGs due to 15-PGDH up-regulation and EP and FP receptor down-regulation. The suppression by calcitriol of the growth stimulation by exogenous PG addition is probably due to its ability to enhance PG catabolism through the up-regulation of 15-PGDH expression as well as PG receptor down-regulation.

NSAIDs are known inhibitors of COX activity and have been shown to exhibit growth-suppressive effects in in vivo and in vitro models of prostate cancer ( 19, 35, 36, 50– 52). The growth inhibitory and proapoptotic actions of NSAIDs are due to their ability to inhibit cyclooxygenase activity to a large degree, although in recent years mechanisms independent of COX-2 inhibition are also believed to play a role ( 52). Our data show that the combination of calcitriol with COX-2–selective, as well as nonselective NSAIDs, acts synergistically to reduce the growth of prostate cancer cells. Our hypothesis is that the action of calcitriol at the genomic level to reduce COX-2 expression decreases the levels of COX-2 protein and allows the use of lower concentrations of NSAIDs to inhibit COX-2 enzyme activity, resulting in the enhanced growth inhibition seen with the combination. The potential use of NSAIDs as chemopreventive or therapeutic agents for a variety of malignancies, including prostate cancer, is being intensely investigated ( 20, 21, 51, 53). We propose that a combination of calcitriol and NSAID might be a useful therapeutic strategy in prostate cancer. The clinical use of NSAIDs has recently become controversial because of the cardiovascular complications associated with the use of high doses of COX-2–selective NSAIDs for prolonged periods of time ( 54, 55). In comparison with the COX-2–selective inhibitors, the use of a nonselective NSAID such as naproxen has been shown to be associated with decreased cardiovascular adverse effects ( 56). As shown by our study, an enhancement of growth inhibition is seen when calcitriol is combined with nonselective NSAIDs such as naproxen and ibuprofen. The clinical utility of the calcitriol combination with a nonselective NSAID is therefore worthy of evaluation, especially because the combination allows the use of lower concentrations of calcitriol and the NSAIDs, thereby improving the safety profile of the NSAIDs.

In conclusion, calcitriol acts by three separate mechanisms: decreasing COX-2 expression, increasing 15-PGDH expression, and reducing PG receptor mRNA levels. We believe that these actions contribute to suppress the proliferative stimulus provided by PGs in prostate cancer cells. The regulation of PG metabolism and biological actions constitutes an additional novel pathway of calcitriol action mediating its antiproliferative effects in prostate cells. We propose that a combination of calcitriol and a nonselective NSAID, such as naproxen, might be a useful therapeutic and/or chemopreventive strategy in prostate cancer, as it would achieve greater efficacy and allow the use of lower concentrations of both drugs, thereby reducing their toxic side effects.

Acknowledgments

Grant support: NIH grants DK42482 and CA 092238 (D. Feldman) and Department of Defense grant PC040120 (J. Moreno).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • Received April 27, 2005.
  • Accepted June 14, 2005.
  • ©2005 American Association for Cancer Research.

References

  1. ↵
    Jemal A, Murray T, Ward E, et al. Cancer statistics, 2005. CA Cancer J Clin 2005; 55: 10–30.
    OpenUrlCrossRefPubMed
  2. ↵
    Feldman D, Malloy PJ, Gross C. Vitamin D: biology, actions and clinical implications. In: Marcus R, Feldman D, Kelsey J, editors. Osteoporosis. 2nd ed, Vol. 1. San Diego: Academic Press; 2001. p. 257–303.
  3. ↵
    Peehl DM, Skowronski RJ, Leung GK, Wong ST, Stamey TA, Feldman D. Antiproliferative effects of 1,25-dihydroxyvitamin D3 on primary cultures of human prostatic cells. Cancer Res 1994; 54: 805–10.
    OpenUrlAbstract/FREE Full Text
  4. Feldman D, Skowronski RJ, Peehl DM. Vitamin D and prostate cancer. Adv Exp Med Biol 1995; 375: 53–63.
    OpenUrlPubMed
  5. ↵
    Miller GJ. Vitamin D and prostate cancer: biologic interactions and clinical potentials. Cancer Metastasis Rev 1998; 17: 353–60.
    OpenUrlCrossRefPubMed
  6. Konety BR, Getzenberg RH. Vitamin D and prostate cancer. Urol Clin North Am 2002; 29: 95–106.
    OpenUrlCrossRefPubMed
  7. Krishnan AV, Peehl DM, Feldman D. The role of vitamin D in prostate cancer. Recent Results Cancer Res 2003; 164: 205–21.
    OpenUrlCrossRefPubMed
  8. Krishnan AV, Peehl DM, Feldman D. Inhibition of prostate cancer growth by vitamin D: regulation of target gene expression. J Cell Biochem 2003; 88: 363–71.
    OpenUrlCrossRefPubMed
  9. ↵
    Stewart LV, Weigel NL. Vitamin D and prostate cancer. Exp Biol Med (Maywood) 2004; 229: 277–84.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Krishnan AV, Peehl DM, Feldman D. Vitamin D and prostate cancer. In: Feldman D, Pike JW, Glorieux F, editors. Vitamin D. 2nd ed. San Diego: Academic Press; 2005. p. 1679–707.
  11. ↵
    Zhuang SH, Burnstein KL. Antiproliferative effect of 1α,25-dihydroxyvitamin D3 in human prostate cancer cell line LNCaP involves reduction of cyclin-dependent kinase 2 activity and persistent G1 accumulation. Endocrinology 1998; 139: 1197–207.
    OpenUrlCrossRefPubMed
  12. ↵
    Blutt SE, McDonnell TJ, Polek TC, Weigel NL. Calcitriol-induced apoptosis in LNCaP cells is blocked by overexpression of Bcl-2. Endocrinology 2000; 141: 10–7.
    OpenUrlCrossRefPubMed
  13. Boyle BJ, Zhao XY, Cohen P, Feldman D. Insulin-like growth factor binding protein-3 mediates 1 α,25-dihydroxyvitamin d(3) growth inhibition in the LNCaP prostate cancer cell line through p21/WAF1. J Urol 2001; 165: 1319–24.
    OpenUrlCrossRefPubMed
  14. ↵
    Freedman LP. Transcriptional targets of the vitamin D3 receptor-mediating cell cycle arrest and differentiation. J Nutr 1999; 129: 581–6S.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Krishnan AV, Shinghal R, Raghavachari N, Brooks JD, Peehl DM, Feldman D. Analysis of vitamin D-regulated gene expression in LNCaP human prostate cancer cells using cDNA microarrays. Prostate 2004; 59: 243–51.
    OpenUrlCrossRefPubMed
  16. ↵
    Peehl DM, Shinghal R, Nonn L, et al. Molecular activity of 1,25-dihydroxyvitamin D3 in primary cultures of human prostatic epithelial cells revealed by cDNA microarray analysis. J Steroid Biochem Mol Biol 2004; 92: 131–41.
    OpenUrlCrossRefPubMed
  17. ↵
    Vane JR, Bakhle YS, Botting RM. Cyclooxygenases 1 and 2. Annu Rev Pharmacol Toxicol 1998; 38: 97–120.
    OpenUrlCrossRefPubMed
  18. ↵
    Badawi AF. The role of prostaglandin synthesis in prostate cancer. BJU Int 2000; 85: 451–62.
    OpenUrlCrossRefPubMed
  19. ↵
    Hussain T, Gupta S, Mukhtar H. Cyclooxygenase-2 and prostate carcinogenesis. Cancer Lett 2003; 191: 125–35.
    OpenUrlCrossRefPubMed
  20. ↵
    Basler JW, Piazza GA. Nonsteroidal anti-inflammatory drugs and cyclooxygenase-2 selective inhibitors for prostate cancer chemoprevention. J Urol 2004; 171: S59–62; discussion S62–3.
    OpenUrlCrossRefPubMed
  21. ↵
    Dubois RN, Abramson SB, Crofford L, et al. Cyclooxygenase in biology and disease. FASEB J 1998; 12: 1063–73.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Backlund MG, Mann JR, Holla VR, et al. 15-Hydroxyprostaglandin dehydrogenase is down-regulated in colorectal cancer. J Biol Chem 2004; 280: 3217–23.
  23. ↵
    Ding Y, Tong M, Liu S, Moscow JA, Tai HH. NAD+-linked 15-hydroxyprostaglandin dehydrogenase (15-PGDH) behaves as a tumor suppressor in lung cancer. Carcinogenesis 2005; 26: 65–72.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Yan M, Rerko RM, Platzer P, et al. 15-Hydroxyprostaglandin dehydrogenase, a COX-2 oncogene antagonist, is a TGF-β-induced suppressor of human gastrointestinal cancers. Proc Natl Acad Sci U S A 2004; 101: 17468–73.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Peehl DM. Human prostatic epithelial cells. In: Freshney RI, Freshney MG, editors. Culture of epithelial cells. New York: Wiley-Liss, Inc.; 2002. p. 159–80.
  26. ↵
    Burton K. A study of the conditions and mechanism of the diphenylamine reaction for the colorimetric estimation of deoxyribonucleic acid. Biochem J 1956; 62: 315–23.
    OpenUrlFREE Full Text
  27. ↵
    Sambrook J, Russell DW. Molecular cloning: a laboratory manual, 3rd ed. Cold Spring Harbor: Cold Spring Harbor Laboratory Press; 2001.
  28. ↵
    Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−ΔΔC(T)) method. Methods 2001; 25: 402–8.
    OpenUrlCrossRefPubMed
  29. ↵
    Chen Y, Hughes-Fulford M. Prostaglandin E2 and the protein kinase A pathway mediate arachidonic acid induction of c-fos in human prostate cancer cells. Br J Cancer 2000; 82: 2000–6.
    OpenUrlCrossRefPubMed
  30. ↵
    Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976; 72: 248–54.
    OpenUrlCrossRefPubMed
  31. ↵
    Tjandrawinata RR, Hughes-Fulford M. Up-regulation of cyclooxygenase-2 by product-prostaglandin E2. Adv Exp Med Biol 1997; 407: 163–70.
    OpenUrlCrossRefPubMed
  32. ↵
    Curran T, Bravo R, Muller R. Transient induction of c-fos and c-myc in an immediate consequence of growth factor stimulation. Cancer Surv 1985; 4: 655–81.
    OpenUrlPubMed
  33. ↵
    Tallarida RJ. The interaction index: a measure of drug synergism. Pain 2002; 98: 163–8.
    OpenUrlCrossRefPubMed
  34. ↵
    Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem 2000; 69: 145–82.
    OpenUrlCrossRefPubMed
  35. ↵
    Narayanan BA, Narayanan NK, Pittman B, Reddy BS. Regression of mouse prostatic intraepithelial neoplasia by nonsteroidal anti-inflammatory drugs in the transgenic adenocarcinoma mouse prostate model. Clin Cancer Res 2004; 10: 7727–37.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Srinath P, Rao PN, Knaus EE, Suresh MR. Effect of cyclooxygenase-2 (COX-2) inhibitors on prostate cancer cell proliferation. Anticancer Res 2003; 23: 3923–8.
    OpenUrlPubMed
  37. ↵
    Ensor C, Tai HH. 15-Hydroxyprostaglandin dehydrogenase. J Lipid Mediat Cell Signal 1995; 12: 313–9.
    OpenUrlCrossRefPubMed
  38. ↵
    Tai HH, Ensor CM, Tong M, Zhou H, Yan F. Prostaglandin catabolizing enzymes. Prostaglandins Other Lipid Mediat 2002; 68–9: 483–93.
    OpenUrl
  39. ↵
    Greenland KJ, Jantke I, Jenatschke S, Bracken KE, Vinson C, Gellersen B. The human NAD+-dependent 15-hydroxyprostaglandin dehydrogenase gene promoter is controlled by Ets and activating protein-1 transcription factors and progesterone. Endocrinology 2000; 141: 581–97.
    OpenUrlCrossRefPubMed
  40. ↵
    Tong M, Tai HH. Induction of NAD(+)-linked 15-hydroxyprostaglandin dehydrogenase expression by androgens in human prostate cancer cells. Biochem Biophys Res Commun 2000; 276: 77–81.
    OpenUrlCrossRefPubMed
  41. ↵
    Tong M, Tai HH. Synergistic induction of the nicotinamide adenine dinucleotide-linked 15-hydroxyprostaglandin dehydrogenase by an androgen and interleukin-6 or forskolin in human prostate cancer cells. Endocrinology 2004; 145: 2141–7.
    OpenUrlCrossRefPubMed
  42. ↵
    Liu S, Stromberg A, Tai HH, Moscow JA. Thiamine transporter gene expression and exogenous thiamine modulate the expression of genes involved in drug and prostaglandin metabolism in breast cancer cells. Mol Cancer Res 2004; 2: 477–87.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Pichaud F, Roux S, Frendo JL, et al. 1,25-dihydroxyvitamin D3 induces NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase in human neonatal monocytes. Blood 1997; 89: 2105–12.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Boyan BD, Sylvia VL, Dean DD, Del Toro F, Schwartz Z. Differential regulation of growth plate chondrocytes by 1α,25-(OH)2D3 and 24R,25-(OH)2D3 involves cell-maturation-specific membrane-receptor-activated phospholipid metabolism. Crit Rev Oral Biol Med 2002; 13: 143–54.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    Zarrabeitia MT, Riancho JA, Amado JA, Olmos JM, Gonzalez-Macias J. Effect of calcitriol on the secretion of prostaglandin E2, interleukin 1, and tumor necrosis factor α by human monocytes. Bone 1992; 13: 185–9.
    OpenUrlPubMed
  46. ↵
    Tetlow LC, Woolley DE. Effects of 1 α,25dihy droxyvitaminD3 on matrix metalloproteinase expression by rheumatoid synovial cells and articular chondrocytes in vitro. Ann N Y Acad Sci 1999; 878: 615–8.
    OpenUrlCrossRefPubMed
  47. ↵
    Norman AW, Okamura WH, Bishop JE, Henry HL. Update on biological actions of 1α,25(OH)2-vitamin D3 (rapid effects) and 24R,25(OH)2-vitamin D3. Mol Cell Endocrinol 2002; 197: 1–13.
    OpenUrlCrossRefPubMed
  48. ↵
    Hata AN, Breyer RM. Pharmacology and signaling of prostaglandin receptors: multiple roles in inflammation and immune modulation. Pharmacol Ther 2004; 103: 147–66.
    OpenUrlCrossRefPubMed
  49. ↵
    Li X, Zheng W, Li YC. Altered gene expression profile in the kidney of vitamin D receptor knockout mice. J Cell Biochem 2003; 89: 709–19.
    OpenUrlCrossRefPubMed
  50. ↵
    Tjandrawinata RR, Dahiya R, Hughes-Fulford M. Induction of cyclo-oxygenase-2 mRNA by prostaglandin E2 in human prostatic carcinoma cells. Br J Cancer 1997; 75: 1111–8.
    OpenUrlCrossRefPubMed
  51. ↵
    Pruthi RS, Derksen E, Gaston K. Cyclooxygenase-2 as a potential target in the prevention and treatment of genitourinary tumors: a review. J Urol 2003; 169: 2352–9.
    OpenUrlCrossRefPubMed
  52. ↵
    Song X, Lin HP, Johnson AJ, et al. Cyclooxygenase-2, player or spectator in cyclooxygenase-2 inhibitor-induced apoptosis in prostate cancer cells. J Natl Cancer Inst 2002; 94: 585–91.
    OpenUrlAbstract/FREE Full Text
  53. ↵
    Rao CV, Reddy BS. NSAIDs and chemoprevention. Curr Cancer Drug Targets 2004; 4: 29–42.
    OpenUrlCrossRefPubMed
  54. ↵
    Bresalier RS, Sandler RS, Quan H, et al. Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N Engl J Med 2005; 352: 1092–102.
    OpenUrlCrossRefPubMed
  55. ↵
    Solomon SD, McMurray JJ, Pfeffer MA, et al. Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N Engl J Med 2005; 352: 1071–80.
    OpenUrlCrossRefPubMed
  56. ↵
    Bombardier C, Laine L, Reicin A, et al. Comparison of upper gastrointestinal toxicity of rofecoxib and naproxen in patients with rheumatoid arthritis. VIGOR Study Group. N Engl J Med 2000; 343: 1520–8.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
Cancer Research: 65 (17)
September 2005
Volume 65, Issue 17
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Regulation of Prostaglandin Metabolism by Calcitriol Attenuates Growth Stimulation in Prostate Cancer Cells
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Regulation of Prostaglandin Metabolism by Calcitriol Attenuates Growth Stimulation in Prostate Cancer Cells
Jacqueline Moreno, Aruna V. Krishnan, Srilatha Swami, Larisa Nonn, Donna M. Peehl and David Feldman
Cancer Res September 1 2005 (65) (17) 7917-7925; DOI: 10.1158/0008-5472.CAN-05-1435

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Regulation of Prostaglandin Metabolism by Calcitriol Attenuates Growth Stimulation in Prostate Cancer Cells
Jacqueline Moreno, Aruna V. Krishnan, Srilatha Swami, Larisa Nonn, Donna M. Peehl and David Feldman
Cancer Res September 1 2005 (65) (17) 7917-7925; DOI: 10.1158/0008-5472.CAN-05-1435
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • References
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • MBP-1 Inhibits Breast Cancer Metastasis
  • Decitabine Sensitivity in Testicular Cancer
  • Drug-Induced Regulation of FA/BRCA Gene Expression
Show more Experimental Therapeutics, Molecular Targets, and Chemical Biology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement