Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Endocrinology ; 160(9): 2049-2060, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31184711

ABSTRACT

Although androgen deprivation therapy (ADT) and immunotherapy are potential treatment options in men with metastatic prostate cancer (CaP), androgen has conventionally been proposed to be a suppressor of the immune response. However, we herein report that DHT activates macrophages. When the murine macrophage cell line (RAW 264.7), human monocyte cell line (THP-1), and human peripheral blood monocytes were cultured with androgen-resistant CaP cell lines, DHT increased cytotoxicity of macrophages in a concentration-dependent manner. Further studies revealed that DHT induced M1 polarization and increased the expression levels of TNF-related apoptosis-inducing ligand (TRAIL) in macrophages and that this effect was abrogated when TRAIL was neutralized with a blocking antibody or small interfering RNA. Subsequent experiments demonstrated that induction of TRAIL expression was regulated by direct binding of androgen receptor to the TRAIL promoter region. Finally, an in vivo mouse study demonstrated that castration enhanced the growth of an androgen-resistant murine CaP tumor and that this protumorigenic effect of castration was blocked when macrophages were removed with clodronate liposomes. Collectively, these results demonstrate that DHT activates the cytotoxic activity of macrophages and suggest that immunotherapy may not be optimal when combined with ADT in CaP.


Subject(s)
Dihydrotestosterone/pharmacology , Macrophages/drug effects , Prostatic Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/physiology , Animals , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Humans , Immunotherapy , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/immunology , Receptors, Androgen/analysis
2.
Br J Cancer ; 118(5): 670-678, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29381686

ABSTRACT

BACKGROUND: Although the standard treatment for the patients with recurrent and metastatic prostate cancer (CaP) is androgen deprivation therapy, castration-resistant prostate cancer (CRPC) eventually emerges. Our previous report indicated that bone morphogenetic protein 6 (BMP6) induced CRPC via tumour-infiltrating macrophages. In a separate line of study, we have observed that the WNT5A/BMP6 loop in CaP bone metastasis mediates resistance to androgen deprivation in tissue culture. Simultaneously, we have reported that BMP6 induced castration resistance in CaP cells via tumour-infiltrating macrophages. Therefore, our present study aims to investigate the mechanism of WNT5A and its interaction with macrophages on CRPC. METHODS: Doxycycline inducible WNT5A overexpression prostate cancer cell line was used for detailed mechanical study. RESULTS: WNT5A was associated with increased expression of chemokine ligand 2 (CCL2) in the human CaP cell line, LNCaP. Mechanistically, this induction of CCL2 by WNT5A is likely to be mediated via the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signalling pathway. Our in vivo experiments demonstrated that the overexpression of WNT5A in LNCaP cells promoted castration resistance. Conversely, this resistance was inhibited with the removal of macrophages via clodronate liposomes. When patient-derived CaP LuCaP xenografts were analysed, high levels of WNT5A were correlated with increased levels of CCL2 and BMP6. In addition, higher levels of CCL2 and BMP6 were more commonly observed in intra-femoral transplanted tumours as compared to subcutaneous-transplanted tumours in the patient-derived PCSD1 bone-niche model. CONCLUSIONS: These findings collectively suggest that WNT5A may be a key gene that induces CRPC in the bone niche by recruiting and regulating macrophages through CCL2 and BMP6, respectively.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Chemokine CCL2/metabolism , Macrophages/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Up-Regulation , Wnt-5a Protein/metabolism , Aged , Animals , Cell Line, Tumor , Clodronic Acid/pharmacology , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Macrophages/metabolism , Male , Mice , Middle Aged , Neoplasm Transplantation , Prostatic Neoplasms, Castration-Resistant/pathology , Tissue Array Analysis
3.
Br J Cancer ; 116(7): 937-943, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28253524

ABSTRACT

BACKGROUND: Renal cell carcinoma (RCC) is one of the most lethal genitourinary cancers. The presence of androgen receptor (AR) in RCC has recently been shown to be associated with higher tumour stage irrespective of gender. Because the clinical context of androgens in female RCC patients is similar to that of prostate cancer patients undergoing androgen-deprivation therapy, mechanisms underlying the emergence of castration-resistant prostate cancer (CRPC) may be at play in AR-positive RCC cells. Therefore, we hypothesized that AR-positive RCC has intratumoral steroidogenesis and that anti-androgen therapy may result in tumour suppression. METHODS: Mice were injected with an AR-positive RCC cell line. When tumours became palpable, surgical castration was performed and tumour volume was measured. Using ELISA, the levels of intracellular testosterone and dihydrotesterone were measured in AR-positive human RCC cell lines. Lastly, male mice containing xenografts were treated with enzalutamide or abiraterone acetate (AA) for 3 weeks to measure tumour volume. RESULTS: We first observed in vivo that castration retards the growth of AR-positive RCC tumour xenograft in mice. Next, AR-positive human RCC cell lines and tissues were found to have elevated levels of testosterone and dihydrotestosterone and express key enzymes required for intracellular androgen biosynthesis. A mouse xenograft study with AR-positive RCC cell line using the commonly used anti-androgen therapies showed significant tumour suppression (P<0.01). CONCLUSIONS: Intracrine androgen biosynthesis is a potential source of androgen in AR-positive RCC and that the androgen signaling axis is a potential target of intervention in RCC.


Subject(s)
Androgens/biosynthesis , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms/metabolism , Abiraterone Acetate/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzamides , Blotting, Western , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Cell Proliferation/drug effects , Dihydrotestosterone/metabolism , Female , Humans , Immunoenzyme Techniques , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Male , Mice , Mice, Nude , Nitriles , Orchiectomy , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prognosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
J Immunol ; 192(12): 5703-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24821970

ABSTRACT

Kruppel-like factor (KLF) 13 is a transcription factor that positively regulates expression of the chemokine RANTES 3-5 d after activation of T cells. In this study, we document a key role for KLF13 in the expression of IL-4 in CD4(+) T cells. Gene expression analysis in activated T cells from Klf13(-/-) mice showed that IL-4, along with other Th2 cytokine genes, was downregulated when compared with cells from wild-type mice. The decreased levels of IL-4 were not associated with changes in expression of the Th2-inducing transcription factors GATA3 or c-Maf. Additional analysis revealed that KLF13 directly binds to IL-4 promoter regions and synergizes with c-Maf to positively regulate IL-4 expression. These results indicate that KLF13 is a positive regulator for differentiation of Th2 cells, as part of the transcriptional machinery that regulates IL-4 production in Th2 cells.


Subject(s)
Cell Cycle Proteins/immunology , Gene Expression Regulation/immunology , Interleukin-4/immunology , Kruppel-Like Transcription Factors/immunology , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-maf/immunology , Repressor Proteins/immunology , Th2 Cells/immunology , Animals , Cell Cycle Proteins/genetics , Gene Expression Regulation/genetics , Interleukin-4/genetics , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-maf/genetics , Repressor Proteins/genetics , Th2 Cells/cytology , Transcription, Genetic/genetics , Transcription, Genetic/immunology
5.
Prostate ; 74(2): 121-33, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24185914

ABSTRACT

INTRODUCTION. Overexpression of bone morphogenetic protein-6 (BMP-6) has been reported in human prostate cancer tissues. Previously we have demonstrated that BMP-6 enhances prostate cancer growth in mice and not in tissue culture. Herein, we have investigated the mechanism of BMP-6's pro-tumorigenic effect in prostate cancer. METHODS. Tramp C2 murine and LNCaP human prostate cancer cell lines were co-cultured with RAW 264.7 and THP-1 cells, respectively. IL-1a knockout mice were used to confirm the role of BMP-6/IL-1a loop in vivo. Lastly, conditional macrophage null mice cd11b-DTR was used. RESULTS. The results demonstrated that BMP-6 induced the expression of IL-1a in macrophages via a cross-talk between NF-kB1 p50 and Smad1. When endothelial cells were treated with conditioned media harvested from macrophages incubated with BMP-6, tube formation was detected. In the presence of IL-1a neutralizing antibody, endothelial tube formation was blocked. In vivo, tumor growth and neovascularization decreased significantly when BMP-6 was expressed in IL-1a knockout and conditional macrophage-null mice. CONCLUSIONS. Prostate cancer-derived BMP-6 stimulates tumor-associated macrophages to produce IL-1a through a crosstalk between Smad1 and NF-kB1; IL-1a, in turn, promotes angiogenesis and prostate cancer growth.


Subject(s)
Bone Morphogenetic Protein 6/physiology , Carcinogenesis/pathology , Interleukin-1alpha/physiology , Macrophages/pathology , Neovascularization, Pathologic/physiopathology , Prostatic Neoplasms/pathology , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Endothelium, Vascular/pathology , Humans , Interleukin-1alpha/deficiency , Interleukin-1alpha/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/physiology , Prostatic Neoplasms/blood supply , Signal Transduction/physiology , Smad1 Protein/physiology
6.
Cancer Res ; 73(12): 3604-14, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23633487

ABSTRACT

Dysregulated bone morphogenetic proteins (BMP) may contribute to the development and progression of renal cell carcinoma (RCC). Herein, we report that BMP-6 promotes the growth of RCC by interleukin (IL)-10-mediated M2 polarization of tumor-associated macrophages (TAM). BMP-6-mediated IL-10 expression in macrophages required Smad5 and STAT3. In human RCC specimens, the three-marker signature BMP-6/IL-10/CD68 was associated with a poor prognosis. Furthermore, patients with elevated IL-10 serum levels had worse outcome after surgery. Together, our results suggest that BMP-6/macrophage/IL-10 regulates M2 polarization of TAMs in RCC.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Carcinoma, Renal Cell/metabolism , Interleukin-10/metabolism , Kidney Neoplasms/metabolism , Macrophages/metabolism , Animals , Bone Morphogenetic Protein 6/genetics , Carcinoma, Renal Cell/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Clodronic Acid/pharmacology , Doxycycline/pharmacology , Humans , Immunoblotting , Interleukin-10/genetics , Kaplan-Meier Estimate , Kidney Neoplasms/pathology , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , Microscopy, Fluorescence , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Tumor Burden/drug effects
7.
Blood ; 120(8): 1658-67, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22797700

ABSTRACT

RANTES (CCL5) is a chemokine implicated in many human diseases. We previously showed that the transcription factor Kruppel-like factor 13 (KLF13) controls the late (3-5 days after activation) expression of RANTES in T lymphocytes and that KLF13 itself is translationally regulated through the 5'-untranslated region of its mRNA. Here, we show that KLF13 levels are further regulated by ubiquitination and degradation. KLF13 protein is undetectable in resting human T lymphocytes, but treatment with either proteosomal or lysosomal inhibitors increases KLF13 protein levels. Glycogen synthase kinase 3ß (GSK3ß)-mediated phosphorylation of KLF13 triggers the ubiquitination of KLF13 by the E3 ligase Fbw7γ, resulting in KLF13 protein degradation. Knockdown of either Fbw7γ or GSK3ß by small interfering RNA increases KLF13 expression in resting human T lymphocytes. In contrast, in murine T lymphocytes, KLF13 protein is abundant because of the absence of Fbw7γ. Treatment of unactivated human lymphocytes with lysosomal inhibitors stabilizes KLF13 protein, resulting in an increase of RANTES mRNA and protein. Taken together, these studies found that tightly regulated control of both synthesis and degradation allows rapid changes in the level of KLF13 in human T lymphocytes.


Subject(s)
Cell Cycle Proteins/metabolism , Chemokine CCL5/genetics , F-Box Proteins/metabolism , Gene Expression Regulation , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line , Cells, Cultured , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Humans , Kruppel-Like Transcription Factors/genetics , Mice , RNA Interference , RNA, Small Interfering/genetics , Repressor Proteins/genetics , T-Lymphocytes/cytology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
8.
Mol Cell Biochem ; 368(1-2): 147-53, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22707059

ABSTRACT

Smads are the secondary messengers of the transforming growth factor-ß (TGF-ß) signaling pathway. TGF-ß receptors phosphorylate the Receptor Smads (R-Smads) upon ligand binding; activated R-Smads translocate to the nucleus and function as transcription factors. Among the R-Smads, Smads 1, 5, and 8 mainly mediate signals in the bone morphogenetic proteins (BMPs) pathways, while Smads 2/3 mediate TGF-ß signaling. The regulation of Smads in the TGF-ß signal pathway has been well defined, but the relationship of Smads 1, 5, and 8 to the BMP pathways has been relatively understudied. To understand the specific regulation of BMP mediating Smads, we performed yeast two-hybrid screening using the Mad homology 2(MH2) domain of Smad8 as bait. In this screening, novel Smad-binding protein, CREBZF-a basic region-leucine zipper (bZIP) transcription factor-was identified. The interaction of CREBZF and Smads 1, 5, and 8 was confirmed by immunoprecipitation in a human prostate cancer cell line. Overexpression of CREBZF inhibited the promoter activity of BMP response element and abolished the cell growth inhibition induced by BMP-6. Thus, CREBZF inhibits the function of BMP-6 by interacting with Smads. The identification of this novel Smads-binding protein, among others will help us understand the modulation of BMP-signaling pathways.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Bone Morphogenetic Protein 6/biosynthesis , Promoter Regions, Genetic/physiology , Signal Transduction/physiology , Smad8 Protein/metabolism , Transforming Growth Factor beta/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Bone Morphogenetic Protein 6/genetics , Humans , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad8 Protein/genetics , Transforming Growth Factor beta/genetics , Two-Hybrid System Techniques
9.
Prostate ; 71(14): 1525-37, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21374653

ABSTRACT

BACKGROUND: Frequently associated with hormone refractory prostate cancer are neuroendocrine cells. Because these cells do not express androgen receptors and are castration-resistant, further understanding the mechanism of neuroendocrine differentiation (NED) of prostate cancer cells may yield novel intervention methods in hormone refractory prostate cancer. In this regard, the present study investigated the effect of macrophages on prostate cancer NED. METHODS: THP-1 and LNCaP or RAW264.7 and TRAMP-C2 cell line co-cultures were used to investigate NED-macrophage interactions. Also interleukin-6 (IL-6) knockout mice and macrophage-depleted mice were used to test NED in vivo. RESULTS: We found that co-culturing with THP-1 human monocytic cell line and RAW 264.7 murine macrophage cell line led to the NED of LNCaP and TRAMP-C2 prostate cancer cells, respectively. Specifically, the conditioned media of activated macrophages stimulated the expression of parathyroid hormone-related peptide (PTHrP), a marker of NED, in both LNCaP and TRAMP-C2 cells. Mechanistically, bone morphogenetic protein-6 (BMP-6) derived from prostate cancer cells increased the expression of IL-6 in macrophages. Subsequently, IL-6 induced the NED of prostate cancer cells. When this feedback loop was disrupted with neutralizing antibodies to either BMP-6 or IL-6, NED was no longer observed. In human prostate cancer tissues, neuroendocrine cells frequently co-localized with macrophages and BMP-6. In mice, the removal of IL-6 or macrophages blocked the BMP-6-induced NED of prostate cancer cells. CONCLUSIONS: Therefore, we propose that BMP-6 secreted by prostate cancer cells induces IL-6 expression in macrophages; IL-6, in turn, stimulates the NED of prostate cancer cells.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Interleukin-6/metabolism , Macrophages/pathology , Neuroendocrine Cells/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Coculture Techniques , Humans , Male , Mice, Knockout , Monocytes/cytology , Neuroendocrine Cells/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism
10.
J Biol Chem ; 285(50): 39401-8, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20889504

ABSTRACT

Unlike the prototype transforming growth factor-ß (TGF-ß), bone morphogenetic protein-6 (BMP-6) activates macrophages. Here, we report that BMP-6 induces the expression of IL-6 in macrophages. Using overexpression and knockdown experiments, we demonstrate that BMP receptor type II and activin-like kinase-2 are necessary for IL-6 induction by BMP-6. At the intracellular level, both Smad and p38 signaling pathways are required for the induction of IL-6. The cross-talk between the two pathways occurs at the level of transcription factor GATA4 and Smad 1/4. These results, taken together, demonstrate a novel BMP-6 signaling mechanism in which both the Smad and non-Smad pathways directly interact to activate the transcription of a target gene.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Interleukin-6/metabolism , Macrophages/metabolism , Signal Transduction , Activin Receptors, Type I/metabolism , Animals , Cell Line , GATA4 Transcription Factor/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Smad1 Protein/metabolism , Smad4 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Immunology ; 128(1 Suppl): e442-50, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19191909

ABSTRACT

Bone morphogenetic proteins (BMPs) are members of the transforming growth factor (TGF)-beta superfamily which regulates bone formation, haematopoiesis and development. While TGF-beta is known to be a negative regulator of the immune system, the effect of BMPs on the immune system is largely unknown. Herein, the effect of BMP-6 on the innate immune system was investigated using the murine macrophage cell line RAW 264.7. BMP-6 altered cellular morphology, inhibited cellular proliferation, increased the fraction of subG(1) phase cells, and decreased the fraction of cells in the S and G(2)M phases, without changing the percentage of apoptotic cells. In addition, BMP-6 induced expression of pro-inflammatory inducible nitric oxide synthase (iNOS) and the cytokine tumour necrosis factor (TNF)-alpha. Reverse transcription-polymerase chain reaction (RT-PCR) analysis demonstrated the expression of all three known type II BMP receptors [BMP-RII, activin (Act)-RIIA and Act-RIIB] and two of the three known type I receptors [activin receptor-like kinase 2 (ALK2) and ALK3]. Over-expression as well as knock-down studies using short hairpin RNA (shRNA) demonstrated that BMP-RII, ALK2 and ALK3 are the functional BMP-6 receptors in macrophages. Finally, the effect of BMP-6 was confirmed in murine peritoneal macrophages and the THP-1 human monocyte cell line. Taken together, these results demonstrate that BMP-6 regulates the proliferation and gene expression profile of macrophages.


Subject(s)
Bone Morphogenetic Protein 6/physiology , Bone Morphogenetic Protein Receptors, Type II/metabolism , Macrophages/immunology , Monocytes/immunology , Activin Receptors/metabolism , Animals , Apoptosis/immunology , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein Receptors, Type II/agonists , Cell Cycle/drug effects , Cell Cycle/immunology , Cell Line , Cell Proliferation/drug effects , Humans , Immunity, Innate , Macrophages/drug effects , Mice , Monocytes/drug effects , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/immunology , Signal Transduction , Transfection , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
12.
Carcinogenesis ; 29(10): 2019-24, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18684728

ABSTRACT

The green tea (Camellia sinensis) catechin, (-)-epigallocatechin-3-gallate (EGCG), has shown cancer-preventive activity in animal models. Previously, we have reported the bioavailability of EGCG in rats and mice. Here, we report that cotreatment of HT-29 human colon cancer cells with genistein (from soy) increased cytosolic EGCG by 2- to 5-fold compared with treatment with EGCG only. Inclusion of genistein, at non-cytotoxic concentrations, increased the growth inhibitory effects of EGCG against HT-29 cells (up to 2-fold at 20 microM genistein). Intragastric coadministration of EGCG (75 mg/kg) and genistein (200 mg/kg) to CF-1 mice resulted in an increase in plasma half-life (t(1/2) 148.7 +/- 16.4 versus 111.5 +/- 23.4 min) and exposure (AUC(0-->infinity) 183.9 +/- 20.2 versus 125.8 +/- 26.4 microg/ml x min) of EGCG. Cotreatment with genistein also increased the maximal concentration (C(max)), 6 h exposure (AUC(0-->360 min)), and t(1/2) of EGCG in the small intestine by 2.0-, 4.7- and 1.4-fold, respectively, compared with mice treated with EGCG only. Contrary to our expectations, the combination of 0.01% EGCG in the drinking fluid and 0.2% genistein in the diet enhanced intestinal tumorigenesis in male adenomatous polyposis coli (APC)(min/+) mice. This combination increased the multiplicity of tumors in the medial and distal small intestine: the largest increase was in tumors >2 mm in diameter (4.3-fold compared with controls). This study demonstrates that although genistein can enhance EGCG bioavailability and in vitro growth inhibitory activity, this combination enhances tumorigenesis in the APC(min/+) mouse. These results further show the need for careful cancer prevention studies in animal models and for caution when interpreting data from in vitro studies.


Subject(s)
Anticarcinogenic Agents/pharmacology , Catechin/analogs & derivatives , Genistein/pharmacology , Intestinal Neoplasms/prevention & control , Animals , Biological Availability , Catechin/pharmacokinetics , Catechin/pharmacology , Drug Interactions , HT29 Cells , Humans , Intestinal Neoplasms/pathology , Male , Mice
13.
Free Radic Res ; 41(7): 850-3, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17577746

ABSTRACT

Tea (Camellia sinensis) catechins have been studied for disease prevention. These compounds undergo oxidation and produce H(2)O(2). We have previously shown that holding tea solution or chewing tea leaves generates high salivary catechin levels. Herein, we examined the generation of H(2)O(2) in the oral cavity by green tea solution or leaves. Human volunteers holding green tea solution (0.1-0.6%) developed salivary H(2)O(2) with C(max) = 2.9-9.6 microM and AUC(0 --> infinity) = 8.5-285.3 microM min. Chewing 2 g green tea leaves produced higher levels of H(2)O(2) (C(max) = 31.2 microM, AUC(0 --> infinity) = 1290.9 microM min). Salivary H(2)O(2) correlated with catechin levels and with predicted levels of H(2)O(2) (C(max(expected)) = 36 microM vs C(max(determined)) = 31.2 microM). Salivary H(2)O(2) and catechin concentrations were similar to those that are biologically active in vitro. Catechin-generated H(2)O(2) may, therefore, have a role in disease prevention by green tea.


Subject(s)
Catechin/metabolism , Dental Caries/prevention & control , Hydrogen Peroxide/metabolism , Saliva/physiology , Tea , Humans , Mastication
14.
Free Radic Biol Med ; 42(8): 1211-21, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17382202

ABSTRACT

Garcinol, a polyisoprenylated benzophenone, from the Garcinia indica fruit rind, has been suggested to be an anti-inflammatory and anti-cancer agent. To explore the possible use of this redox-sensitive compound as a colon cancer preventive agent, we investigated the effects of garcinol and its oxidative derivatives, cambogin, garcim-1, and garcim-2, on the growth of HT-29 and HCT-116 colon cancer cells, as well as IEC-6 and INT-407 normal immortalized intestinal cells. Garcinol and its derivatives showed potent growth-inhibitory effects on all intestinal cells, showing IC50 of 3.2-21.4 microM after a 3-day treatment. Garcim-1 exhibited the strongest effect with IC50 of 3.2-5.9 microM. Garcinol was more effective in inhibiting growth of cancer cells than that of normal immortalized cells. Flow-cytometric analysis showed increased sub-G1 cells by treatment with garcinol and cambogin. Induction of apoptosis by garcinol and cambogin (2-10 microM) was also observed based on caspase-3 activation and enhanced annexin V staining. The inhibitory effect of garcinol on cell growth was much more pronounced in the absence of fetal bovine serum (FBS), decreasing IC50 to 1.5 from 11.8 microM in 72-h incubations and to 3 from 38 microM in 24-h incubations, possibly due to the binding of garcinol to FBS, which markedly reduced cellular levels of garcinol. Under these conditions, redox reactions seem not to be involved in the inhibition. In contrast to the inhibitory effect, low concentrations (<1 microM) of garcinol and cambogin stimulated the growth of both normal and cancer cells by 10-100%, and the activity seemed to be mediated by reactive oxygen species. In the presence of superoxide dismutase/catalase or N-acetyl cysteine, low concentrations of garcinol (<1 microM) decreased cell growth. Garcinol (0.5-1 microM) also increased the phosphorylation of extracellular signal-related kinase 1/2 and AKT and the level of survivin, and the effects were abolished in the presence of superoxide dismutase/catalase. Our results indicate that garcinol and its derivatives can inhibit intestinal cell growth, but low concentrations of garcinol can stimulate cell growth. It remains to be determined whether the currently observed stimulatory and inhibitory effects of garcinol on colon cell growth occur in vivo.


Subject(s)
Cell Cycle/drug effects , Cell Division/drug effects , Intestines/cytology , Terpenes/pharmacology , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Colonic Neoplasms , Humans , Intestines/drug effects , Kinetics , Plant Extracts/pharmacology
15.
Drug Metab Dispos ; 34(12): 2111-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16997910

ABSTRACT

(-)-Epigallocatechin-3-gallate (EGCG) is the widely studied catechin in green tea (Camellia sinensis). Previously, we have reported the low bioavailability of EGCG in rats and mice. As a means of improving the bioavailability of EGCG, we have prepared a peracetylated EGCG derivative (AcEGCG) and herein report its growth inhibitory activity and cellular uptake in vitro, as well as bioavailability in mice. AcEGCG exhibited enhanced growth inhibitory activity relative to EGCG in both KYSE150 human esophageal (IC50 = 10 versus 20 microM) and HCT116 human colon cancer cells (IC50 = 32 versus 45 microM). AcEGCG was rapidly converted to EGCG by HCT116 cells, and treatment of cells with AcEGCG resulted in a 2.8- to 30-fold greater intracellular concentration of EGCG as compared with treatment with EGCG. AcEGCG was also more potent than EGCG at inhibiting nitric oxide production (4.4-fold) and arachidonic acid release (2.0-fold) from lipopolysaccharide-stimulated RAW264.7 murine macrophages. Intragastric administration of AcEGCG to CF-1 mice resulted in higher bioavailability compared with administration of equimolar doses of EGCG. The plasma area under the curve from 0 to infinity (AUC0-->infinity) of total EGCG was 465.0 and 194.6 [(microg/ml) . min] from the administration of AcEGCG and EGCG, respectively. The t1/2 of EGCG was also increased following administration of AcEGCG compared with EGCG (441.0 versus 200.3 min). The AUC0-->infinity and t1/2 were also increased in small intestinal (2.8- and 4.3-fold, respectively) and colonic tissues (2.4- and 6.0-fold, respectively). These data suggest that acetylation represents a means of increasing the biological potency in vitro, increasing the bioavailability of EGCG in vivo, and may improve cancer-preventive activity.


Subject(s)
Acetates/pharmacology , Acetates/pharmacokinetics , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/pharmacokinetics , Catechin/analogs & derivatives , Acetates/chemical synthesis , Acetylation , Animals , Anticarcinogenic Agents/chemistry , Arachidonic Acid/antagonists & inhibitors , Biological Availability , Catechin/chemical synthesis , Catechin/chemistry , Catechin/pharmacokinetics , Catechin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Colon/metabolism , Esterases/metabolism , Humans , Intestine, Small/metabolism , Male , Mice , Mice, Inbred Strains , Nitric Oxide/antagonists & inhibitors
16.
Carcinogenesis ; 27(2): 278-86, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16093250

ABSTRACT

Garcinol, a polyisoprenylated benzophenone, from the fruit rind of Garcinia spp., has been shown to have anti-inflammatory and anticarcinogenic activities. To study its mechanism of action, we analyzed the effects of garcinol and its derivatives, including cambogin, garcim-1 and garcim-2, on arachidonic acid metabolism and nitric oxide (NO) synthesis in lipopolysaccharide (LPS)-stimulated RAW264.7 murine macrophages as well as in three intestinal cell lines. We also examined the effect of garcinol on cytosolic phospholipase A2 (cPLA2), cyclooxygenase-2 (COX-2), inducible NO synthase (iNOS), and related upstream signaling. At 1 microM, garcinol and its derivatives, added 1 h after LPS stimulation, significantly inhibited the release of arachidonic acid and its metabolites in macrophages; garcinol was the most effective, showing >50% inhibition. Similar inhibitory activity was also observed in intestinal cells, HT-29, HCT-116 and IEC-6 cells, showing 40-50% inhibition by 1 microM garcinol. In LPS-stimulated macrophages, garcinol inhibited the phosphorylation of cPLA2 without altering its protein level, and the effect was related to the inhibition of ERK1/2 phosphorylation. Garcinol inhibited NFkappaB activation and COX-2 expression only when it was added to the cells before LPS stimulation. Garcinol (1 microM) also significantly decreased iNOS expression and NO release from LPS-stimulated macrophages; this is probably due to the inhibition of the signal transducer and activator of transcription-1 (STAT-1), an upstream event in the activation of iNOS synthesis. The results suggest that garcinol modulates arachidonic acid metabolism by blocking the phosphorylation of cPLA2 and decreases iNOS protein level by inhibiting STAT-1 activation. These activities may contribute to the anti-inflammatory and anticarcinogenic actions of garcinol and its derivatives.


Subject(s)
Arachidonic Acid/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/analysis , Terpenes/pharmacology , Animals , Cytosol/chemistry , Intestines/cytology , Lipopolysaccharides/pharmacology , Macrophages , Mice , Phospholipases A/metabolism , Phospholipases A2 , Phosphorylation , Plant Extracts/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...