Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Nutr Food Res ; 61(9)2017 09.
Article in English | MEDLINE | ID: mdl-28181403

ABSTRACT

SCOPE: Caffeic acid phenethyl ester (CAPE), a bioactive component of propolis, is considered as a new anti-cancer agent. Oral squamous cell carcinoma (OSCC) is the most common oral cancer with unsatisfying survival. N-myc downstream regulated family genes (NDRGs) involve in numerous physiological processes. We investigated the anti-cancer effect of CAPE on OSCC and related mechanisms. METHODS AND RESULTS: Cell proliferation assay, western blot, gene transfection and knockdown, and reporter assay were applied. We showed that CAPE attenuated OSCC cell proliferation and invasion in vitro, and safely and effectively inhibited OSCC cell growth in a xenograft animal model. CAPE treatment induced NDRG1, but not NDRG2 and NDRG3, expression in OSCC cells as determined by western blot, RT-qPCR, and reporter assay. The 5'-deletion assay demonstrated that CAPE increased NDRG1 promoter activity depending on the region of -128 to +46 of the 5'-flanking of NDRG1 gene. NDRG1 gene knockdown attenuated CAPE anti-growth effect on OSCC cells. CAPE activated mitogen-activated protein kinase (MAPK) signaling pathway. The extracellular signal regulated kinase (ERK) inhibitor (PD0325901) and ERK1 knockdown blocked CAPE-induced NDRG1 expression in OSCC cells. CONCLUSION: CAPE activated MAPK signaling pathway and increased NDRG1 expression through phosphorylation of ERK1/2 to repress OSCC cells growth.


Subject(s)
Caffeic Acids/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Cycle Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/physiology , Head and Neck Neoplasms/drug therapy , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/physiology , Mouth Neoplasms/drug therapy , Phenylethyl Alcohol/analogs & derivatives , Animals , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mouth Neoplasms/pathology , Phenylethyl Alcohol/pharmacology , Promoter Regions, Genetic , Squamous Cell Carcinoma of Head and Neck , Up-Regulation , Xenograft Model Antitumor Assays
2.
Int J Mol Sci ; 17(9)2016 Aug 31.
Article in English | MEDLINE | ID: mdl-27589737

ABSTRACT

Oral squamous cell carcinoma (OSCC) is a common malignancy with a growing worldwide incidence and prevalence. The N-myc downstream regulated gene (NDRG) family of NDRG1, 2, 3, and mammary serine protease inhibitor (Maspin) gene are well-known modulators in the neoplasia process. Current research has considered iron chelators as new anti-cancer agents; however, the anticancer activities of iron chelators and their target genes in OSCC have not been well investigated. We showed that iron chelators (Dp44mT, desferrioxamine (DFO), and deferasirox) all significantly inhibit SAS cell growth. Flow cytometry further indicated that Dp44mT inhibition of SAS cells growth was partly due to induction of G1 cell cycle arrest. Iron chelators enhanced expressions of NDRG1 and NDRG3 while repressing cyclin D1 expression in OSCC cells. The in vivo antitumor effect on OSCC and safety of Dp44mT were further confirmed through a xenograft animal model. The Dp44mT treatment also increased Maspin protein levels in SAS and OECM-1 cells. NDRG3 knockdown enhanced the growth of OECM-1 cells in vitro and in vivo. Our results indicated that NDRG3 is a tumor suppressor gene in OSCC cells, and Dp44mT could be a promising therapeutic agent for OSCC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Proliferation/drug effects , Iron Chelating Agents/pharmacology , Mouth Neoplasms/drug therapy , Thiosemicarbazones/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Iron Chelating Agents/therapeutic use , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Serpins/genetics , Serpins/metabolism , Thiosemicarbazones/therapeutic use
3.
Cancer Lett ; 375(1): 142-151, 2016 May 28.
Article in English | MEDLINE | ID: mdl-26965996

ABSTRACT

Prostate-derived Ets (E-twenty six) factor (PDEF), an epithelium-specific member of the Ets family of transcription factors, has been shown to play a role in suppressing the development of many epithelium-derived cancers such as prostate and breast cancer. It is not clear, however, whether PDEF is involved in the development or progression of bladder cancer. In a comparison between normal urothelium and bladder tumor tissue, we identified significant decreases of PDEF in the tumor tissue. Further, the immunohistochemistry assays indicated a significantly higher immunostaining of PDEF in low-grade bladder tumors. Additionally, the highly differentiated transitional-cell bladder carcinoma RT-4 cells expressed significantly more PDEF levels than the bladder carcinoma HT1376 and the T24 cells. Ectopic overexpression of PDEF attenuated proliferation, invasion, and tumorigenesis of bladder carcinoma cells in vitro and in vivo. PDEF enhanced the expression levels of mammary serine protease inhibitor (MASPIN), N-myc downstream regulated gene 1 (NDRG1), KAI1, and B-cell translocation gene 2 (BTG2). PDEF modulated epithelial-mesenchymal-transition (EMT) by upregulating E-cadherin expression and downregulating the expression of N-cadherin, SNAIL, SLUG, and vimentin, leading to lower migration and invasion abilities of bladder carcinoma cells. Filamentous actin (F-actin) polarization and remodeling were observed in PDEF-knockdown RT-4 cells. Our results suggest that PDEF gene expression is associated with the extent of bladder neoplasia and PDEF modulated the expressions of EMT-related genes. The induction of BTG2, NDRG1, MASPIN, and KAI1 gene expressions by PDEF may explain the inhibitory functions of PDEF on the proliferation, invasion, and tumorigenesis in bladder carcinoma cells.


Subject(s)
Carcinogenesis/metabolism , Epithelial-Mesenchymal Transition , Proto-Oncogene Proteins c-ets/metabolism , Urinary Bladder Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Proto-Oncogene Proteins c-ets/genetics , Tumor Burden , Urinary Bladder Neoplasms/pathology
4.
Sci Rep ; 5: 12870, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26249737

ABSTRACT

Growth differentiation factor-15 (GDF15), a member of the TGF-ß superfamily, affects tumor biology of certain cancers, but remains poorly understood in bladder cancer cells. This study determined the expression, regulation, function, and potential downstream target genes of GDF15 in bladder carcinoma cells. The transitional papilloma carcionoma cells (RT4) expressed higher levels of GDF15 as compared with the bladder carcinoma cells (HT1376 and T24). Treatments of recombinant human GDF15 (rhGDF15) reduced the proliferations of HT1376 and T24 cells. Expression of GDF15 was upregulated via DNA demethylation and p53. The cell proliferation, invasion, and tumorigenesis were reduced in ectopic overexpression of GDF15, while enhanced in GDF15 knockdown. The expressions of mammary serine protease inhibitor (MASPIN) and N-myc downstream-regulated family genes (NDRG1, NDRG2, and NDRG3) were upregulated by GDF15 overexpressions and rhGDF15 treatments in bladder carcinoma cells. GDF15 knockdown induced epithelial-mesenchymal transition (EMT) and F-actin polarization in HT1376 cells. Our results suggest that enhanced expressions of MASPIN and N-myc downstream-regulated family genes and the modulation of EMT may account for the inhibitory functions of GDF15 in the cell proliferation, invasion, and tumorigenesis of bladder carcinoma cells. The GDF15 should be considered as a tumor suppressor in human bladder carcinoma cells.


Subject(s)
Carcinogenesis/genetics , Cell Proliferation/genetics , Growth Differentiation Factor 15/genetics , Neoplasm Invasiveness/genetics , Tumor Suppressor Protein p53/genetics , Up-Regulation/genetics , Urinary Bladder Neoplasms/genetics , Actins/genetics , Carcinoma/genetics , Carcinoma/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA Methylation/genetics , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Transcriptional Activation/genetics , Transforming Growth Factor beta/genetics , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology
5.
Sci Rep ; 5: 8686, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25732125

ABSTRACT

WNT1 inducible signaling pathway protein 1 (WISP1) plays a key role in many cellular functions in a highly tissue-specific manner; however the role of WISP1 in breast cancer is still poorly understood. Here, we demonstrate that WISP1 acts as an oncogene in human breast cancer. We demonstrated that human breast cancer tissues had higher WISP1 mRNA expression than normal breast tissues and that treatment of recombinant WISP1 enhanced breast cancer cell proliferation. Further, ectopic expression of WISP1 increased the growth of breast cancer cells in vitro and in vivo. WISP1 transfection also induced epithelial-mesenchymal-transition (EMT) in MCF-7 cells, leading to higher migration and invasion. During this EMT-inducing process, E-cadherin was repressed and N-cadherin, snail, and ß-catenin were upregulated. Filamentous actin (F-actin) remodeling and polarization were also observed after WISP1 transfection into MCF-7 cells. Moreover, forced overexpression of WISP1 blocked the expression of NDRG1, a breast cancer tumor suppressor gene. Our study provides novel evidence that WISP1-modulated NDRG1 gene expression is dependent on a DNA fragment (-128 to +46) located within the human NDRG1 promoter. Thus, we concluded that WISP1 is a human breast cancer oncogene and is a potential therapeutic target.


Subject(s)
Breast Neoplasms/genetics , CCN Intercellular Signaling Proteins/genetics , Cell Transformation, Neoplastic/genetics , Proto-Oncogene Proteins/genetics , Actins/biosynthesis , Breast Neoplasms/pathology , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Female , Gene Expression , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , MCF-7 Cells , Oncogenes , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
6.
Cancer Lett ; 360(2): 310-8, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25721086

ABSTRACT

Oral squamous cell carcinoma (OSCC) is a well-known malignancy that accounts for the majority of oral cancers. B-cell translocation gene 2 (BTG2) is an important regulator of cell cycle dynamics in cancer cells. However, the role of BTG2 in OSCC cells and the influences of epigallocatechin-3-gallate (EGCG) on BTG2 gene expressions have not been well evaluated. The objectives of this study were to examine the effect of EGCG-induced BTG2 expression and the potential signal pathways involved. The (3)H-thymidine incorporation and Western-blot assays revealed cell proliferation was attenuated by EGCG via upregulation of BTG2 expression causing cell cycle G1 phase arrest in OSCC cells. BTG2 overexpression decreased tumor cell growth, while BTG2 knockdown illuminated the opposite effect in xenograft animal studies. Overexpressed BTG2 arrested the cell cycle at the G1 phase and downregulated protein expressions of cyclin A, cyclin D, and cyclin E. Western-blot assays indicated that EGCG induced phosphorylation of p38, JNK, and ERK. However, pretreatments with selective mitogen-activated protein kinase (MAPK) inhibitors, SB203580 (p38 inhibitor) and PD0325901 (ERK1/2 inhibitor), significantly suppressed the activation of EGCG on BTG2 expression. Our results indicate that EGCG attenuates cell proliferation of OSCC cells by upregulating BTG2 expression via p38 and ERK pathways.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Catechin/analogs & derivatives , Head and Neck Neoplasms/drug therapy , Immediate-Early Proteins/biosynthesis , MAP Kinase Signaling System/drug effects , Mouth Neoplasms/drug therapy , Tumor Suppressor Proteins/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Catechin/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immediate-Early Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck , Tumor Suppressor Proteins/genetics , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
7.
Prostate ; 75(6): 603-15, 2015 May.
Article in English | MEDLINE | ID: mdl-25560459

ABSTRACT

BACKGROUND: Liver X receptor (LXR) isoforms, LXRα and LXRß, have similar protein structures and ligands, but diverse tissue distribution. We used two synthetic, non-steroidal LXR agonists, T0901317 and GW3965, to investigate the effects of LXR agonist modulation on prostate specific antigen (PSA) via the expressions of androgen receptors (AR), LXRα, or LXRß, in prostate carcinoma cells. METHODS: LXRα- or LXRß-knockdown cells were transduced with specific shRNA lentiviral particles. LXRα and LXRß expressions were assessed by immunoblotting and RT-qPCR assays. Cell proliferation was determined by (3) H-thymidine incorporation assays. The effects of LXR agonists and epigallocatechin gallate (EGCG) on PSA expression were determined by ELISA, immunoblotting, or transient gene expression assays. RESULTS: Treatment with either T0901317 or GW3965 significantly attenuated cell proliferation of LNCaP cells. T0901317 treatment suppressed PSA expression while GW3965 treatment enhanced PSA expression. The increase of PSA promoter activity by GW3965 was dependent on the expression of AR. Either LXRα- or LXRß-knockdown did not affect the activation of androgen on PSA gene expression. However, as compared with mock knockdown-LNCaP cells, the LXRα-knockdown but not the LXRß-knockdown attenuated the effects of T0901317 and GW3965 on PSA expressions. The effect of GW3965 on PSA expression was blocked by the addition of EGCG. CONCLUSIONS: Our results indicate that T0901317 and GW3965 have divergent effects on PSA expressions. The effects of LXR agonists on PSA expression are LXRα-dependent and AR-dependent. EGCG blocks the inducing effect of GW3965 on PSA expression.


Subject(s)
Orphan Nuclear Receptors/agonists , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/drug therapy , Receptors, Androgen/physiology , Benzoates/pharmacology , Benzylamines/pharmacology , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line, Tumor , Cell Proliferation , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver X Receptors , Male , Orphan Nuclear Receptors/analysis , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Sulfonamides/pharmacology
8.
Cancer Lett ; 355(2): 242-52, 2014 Dec 28.
Article in English | MEDLINE | ID: mdl-25218595

ABSTRACT

Oral squamous cell carcinoma (OSCC) is the most common phenotype of oral cancer. N-myc downstream regulated gene 1 (NDRG1) is a modulator for cell proliferation, differentiation, and invasion. The role and function of NDRG1 in OSCC cells remain inconclusive. The (3)H-thymidine incorporation and in vitro matrigel invasion assays revealed NDRG1-knockdown significantly enhanced OSCC cell proliferation and invasion. Overexpressed NDRG1 arrested the cell cycle at the S-phase, thus attenuated cell proliferation in OECM-1 cells. The NDRG1-knockdown enhanced tumorigenesis of OECM-1 cells in the xenograft animal model. Western-blot and zymographic assays revealed that NDRG1 downregulated the gelatinase activities and protein levels of metalloproteinase-2 (MMP-2) and metalloproteinase-9 (MMP-9). NDRG1 modulated epithelial-mesenchymal transition (EMT) through upregulation of the E-cadherin expression, but downregulation of the N-cadherin, Vimentin, Snail-1, and Slug. Immunofluorescence staining indicated knockdown of NDRG1 enhanced F-actin expression and polymerization. Our results indicated NDRG1 attenuated OSCC cell growth in vitro and in vivo. The downregulation of EMT, MMP-2, and MMP-9 may explain the role of anti-invasion of NDRG1 in human OSCC cells. The experiments recognize that NDRG1 is an antitumor gene in OSCC cells.


Subject(s)
Carcinogenesis/pathology , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins/physiology , Head and Neck Neoplasms/pathology , Intracellular Signaling Peptides and Proteins/physiology , Mouth Neoplasms/pathology , Animals , Cadherins/genetics , Cadherins/metabolism , Carcinogenesis/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Neoplasm Invasiveness , Squamous Cell Carcinoma of Head and Neck , Up-Regulation
9.
Sci Rep ; 4: 5511, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24981574

ABSTRACT

Cisplatin is a widely used anti-cancer drug. The B-cell translocation gene 2 (BTG2) is involved in the cell cycle transition regulation. We evaluated the cisplatin effects on prostate cancer cell proliferation and the expressions of BTG2, p53, androgen receptor (AR) and prostate specific antigen (PSA) in prostate carcinoma, p53 wild-type LNCaP or p53-null PC-3, cells. Cisplatin treatments attenuated cell prostate cancer cell growth through inducing Go/G1 cell cycle arrest in lower concentration and apoptosis at higher dosage. Cisplatin treatments enhanced p53 and BTG2 expression, repressed AR and PSA expression, and blocked the activation of androgen on the PSA secretion in LNCaP cells. BTG2 knockdown in LNCaP cells attenuated cisplatin-mediated growth inhibition. Cisplatin enhanced BTG2 gene expression dependent on the DNA fragment located within -173 to -82 upstream of BTG2 translation initiation site in prostate cancer cells. Mutation of the p53 response element from GGGCAGAGCCC to GGGCACC or mutation of the NFκB response element from GGAAAGTCC to GGAAAGGAA by site-directed mutagenesis abolished the stimulation of cisplatin on the BTG2 promoter activity in LNCaP or PC-3 cells, respectively. Our results indicated that cisplatin attenuates prostate cancer cell proliferation partly mediated by upregulation of BTG2 through the p53-dependent pathway or p53-independent NFκB pathway.


Subject(s)
Cisplatin/administration & dosage , Immediate-Early Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Male , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Treatment Outcome , NF-kappaB-Inducing Kinase
10.
PLoS One ; 9(3): e93151, 2014.
Article in English | MEDLINE | ID: mdl-24664372

ABSTRACT

Interleukin-6 (IL-6), a multifunctional cytokine, contributes to proliferation or differentiation of prostate carcinoma cells in a highly cell type-specific manner. Celastrol (3-hydroxy-24-nor-2oxo-1(10),3,5,7-friedelatetrane-29-oic acid), also named as tripterine, is extracted from root of Chinese traditional herb Tripterygiumwilfordii Hook f with potent anti-inflammatory and anti-cancer activities. In this study, we evaluated the molecular mechanisms of celastrol on cell proliferation and IL-6 gene expression in prostate carcinoma cells. 3H-thymidine incorporation and flow cytometric analysis indicated that celastrol treatments arrested the cell cycle at the G0/G1 phase, thus attenuating cell proliferation in prostate carcinoma PC-3 cells; moreover, celastrol induced cell apoptosis at higher dosage. Knockdown of IL-6 attenuated the anti-proliferative effect of celastrol on PC-3 cells. Results from ELISA and 5'-deletion transient gene expression assays indicated that celastrol treatment decreased IL-6 secretion and gene expression, and this effect is dependent on the NF-κB response element within IL-6 promoter area since mutation of the NF-κB response element from AAATGTCCCATTTTCCC to AAATGTTACATTTTCCC by site-directed mutagenesis abolished the inhibition of celastrol on the IL-6 promoter activity. Celastrol also attenuated the activation of PMA and TNFα on the gene expression and secretion of IL-6 in PC-3 cells. Immunoblot assays revealed that celastrol treatment downregulated the expressions of IKKα, p50 and p65, supporting the 5'-deletion transient gene expression assay result that celastrol blocked IL-6 expression through the NF-κB pathway in PC-3 cells. For the first time, our results concluded that celastrol attenuates PC-3 cell proliferation via downregulation of IL-6 gene expression through the NF-κB-dependent pathway.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Interleukin-6/biosynthesis , NF-kappa B/metabolism , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Triterpenes/pharmacology , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Humans , Interleukin-6/genetics , Male , NF-kappa B/genetics , Neoplasm Proteins/genetics , Pentacyclic Triterpenes , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
11.
PLoS One ; 9(2): e89117, 2014.
Article in English | MEDLINE | ID: mdl-24586533

ABSTRACT

Camptothecin (CPT) and doxorubicin (DOX) have been demonstrated to have potent anti-tumor activity. The B-cell translocation gene 2 (BTG2) is involved in the regulation of cell cycle progression. We evaluated the molecular mechanisms of CPT and DOX on cell proliferation and the expressions of BTG2 and prostate specific antigen (PSA) in prostate carcinoma cells. Our results indicated that CPT or DOX treatments induced Go/G1 cell cycle arrest in LNCaP cells and apoptosis at higher dosage. Immunoblot and transient gene expression assay indicated that CPT or DOX treatments induced p53 and BTG2 gene expression, with the later effect dependent on the p53 response element within BTG2 promoter area since mutation of the p53 response element from GGGAAAGTCC to GGAGTCC or from GGCAGAGCCC to GGCACC by site-directed mutagenesis abolished the stimulation of CPT or DOX on the BTG2 promoter activity, which is also supported by our results that cotreatments of pifithrin-α, an inhibitor of p53 dependent transcriptional activation, blocked the induction of CPT or DOX on BTG2 gene expression. CPT or DOX also downregulated the protein expressions of androgen receptor (AR) and PSA. Transient gene expression assays suggested that CPT or DOX's attenuation of PSA promoter activity is dependent on both the androgen and p53 response elements within of the PSA promoter. Our results indicated that CPT and DOX attenuate cell proliferation via upregulation of BTG2 gene expression through the p53-dependent pathway. The CPT and DOX block the PSA gene expression by upregulation of p53 activity and downregulation of androgen receptor activity.


Subject(s)
Camptothecin/pharmacology , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Immediate-Early Proteins/genetics , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/pathology , Topoisomerase Inhibitors/pharmacology , Tumor Suppressor Proteins/genetics , Antineoplastic Agents/pharmacology , Base Sequence , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Promoter Regions, Genetic/genetics , Receptors, Androgen/metabolism , Tumor Suppressor Protein p53/metabolism
12.
Prostate ; 73(14): 1495-506, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23794209

ABSTRACT

BACKGROUND: Metallothioneins (MT1, MT2, MT3, and MT4) are regarded as modulators regulating a number of biological processes including cell proliferation, differentiation, and invasion. We determined the effects of androgen, cadmium, and arsenic on MT1/2 and MT3 in prostate carcinoma cells, and evaluated the functional effects of MT3 on cell proliferation, invasion, and tumorigenesis. METHODS: We determined the expression of MT1/2 and MT3 in prostate carcinoma cells by immunoblotting assays or real-time reverse transcription-polymerase chain reactions. The effects of ectopic MT3 overexpression or MT3-knockdown on cell proliferation, invasion, and tumorigenesis were determined by (3) H-thymidine incorporation, matrigel invasion, and murine xenograft studies. The effects of androgen, cadmium, and arsenic on target genes were assessed using immunoblotting and reporter assays. RESULTS: Androgen, cadmium, and arsenic treatments enhanced gene expression of MT1/2 and MT3 in prostate carcinoma LNCaP cells. Results of immunohistochemical staining indicated MT3 overexpression was found predominantly in the nuclear areas of PC-3 cells overexpressing MT3. Overexpression of MT3 significantly increased cell proliferation, invasion, and tumorigenic activities in PC-3 cells in vitro and in vivo. MT3 overexpression downregulated the gene expressions of N-myc downstream regulated gene 1 (Ndrg1) and maspin, and attenuated blocking effects of doxorubicin in PC-3 cells on cell proliferation. MT3-knockdown enhanced Ndrg1 and maspin expressions in LNCaP cells. CONCLUSIONS: The experiments indicate that MT3 is an androgen-upregulated gene, and promotes tumorigenesis of prostate carcinoma cells. The downregulation of Ndrg1 and maspin gene expressions appears to account for the enhancement of proliferative and invasive functions of MT3 in PC-3 cells.


Subject(s)
Carcinoma , Cell Cycle Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Metallothionein/genetics , Prostatic Neoplasms , Serpins/genetics , Androgens/metabolism , Animals , Arsenic/metabolism , Cadmium/metabolism , Carcinogenesis/genetics , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Genetic Enhancement , Humans , Male , Metallothionein 3 , Mice , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Signal Transduction/genetics , Up-Regulation
13.
Biomed Res Int ; 2013: 791212, 2013.
Article in English | MEDLINE | ID: mdl-23762858

ABSTRACT

Interleukin-6, a multifunctional cytokine, contributes to tumor cell proliferation and differentiation. However, the biological mechanisms that are affected by the expression of interleukin-6 in bladder cancer cells remain unclear. We evaluated the effects of interleukin-6 expression in human bladder carcinoma cells in vitro and in vivo. The results of interleukin-6-knockdown experiments in T24 cells and interleukin-6-overexpression experiments in HT1376 cells revealed that interleukin-6 reduced cell proliferation, migration, and invasion in vitro. Xenograft animal studies indicated that the overexpression of interleukin-6 downregulated tumorigenesis of bladder cells and that interleukin-6 knockdown reversed this effect. The results of RT-PCR, immunoblotting, and reporter assays indicated that the overexpression of interleukin-6 upregulated the expression of the mammary serine protease inhibitor (MASPIN), N-myc downstream gene 1 (NDRG1), and KAI1 proteins in HT1376 cells and that interleukin-6 knockdown reduced the expression of these proteins in T24 cells. In addition, results of immunoblotting assays revealed that interleukin-6 modulated epithelial-mesenchymal transitions by upregulating the expression of the E-cadherin, while downregulation N-cadherin and vimentin proteins. Our results suggest that the effects of interleukin-6 on the regulation of epithelial-mesenchymal transitions and the expressions of the MASPIN, NDRG1, and KAI1 genes attribute to the modulation of tumorigenesis in human bladder carcinoma cells.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Interleukin-6/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Animals , Cadherins/genetics , Cadherins/metabolism , Carcinogenesis/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kangai-1 Protein/genetics , Kangai-1 Protein/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Serpins/genetics , Serpins/metabolism , Urinary Bladder Neoplasms/genetics , Vimentin/metabolism
14.
J Mol Endocrinol ; 51(1): 131-41, 2013.
Article in English | MEDLINE | ID: mdl-23709747

ABSTRACT

Hypoxia induces metabolic alteration in cancer cells by stabilizing hypoxia-inducible factor 1α (HIF-1α (HIF1A)), which regulates the bioenergetic genes of glycolysis and lipid metabolic pathways. However, the target genes of hypoxia-induced metabolic alterations in the prostate remain uncertain. Mitochondrial aconitase (mACON) (ACONM) is an enzyme that is central to carbohydrate and energy metabolism and is responsible for the interconversion of citrate to isocitrate as part of the citric acid cycle in the human prostate. We evaluated the effects of the molecular mechanisms of hypoxia on mACON gene expression in PC-3 and LNCaP human prostate carcinoma cells. Immunoblotting assays revealed that hypoxia modulated mACON and lactate dehydrogenase A (LDHA) protein expression, while these effects were attenuated when HIF-1α was knocked down. Hypoxia induced fatty acid synthase (FASN) in PC-3 cells while hypoxia blocked FASN gene expression in LNCaP cells after 24-h incubation. Results of real-time RT-qPCR, immunoblotting, and transient gene expression assays revealed that hypoxia treatment or co-transfection with HIF-1α expression vector enhanced gene expression of mACON, implying that hypoxia modulated mACON at the transcriptional level. Hypoxia-induced mACON promoter activity is dependent on the DNA fragment located at -1013 to -842 upstream of the translation initiation site. l-mimosine, an iron chelator, stabilized HIF-1α but downregulated mACON gene expression, suggesting that iron chelation blocked the hypoxia-induced mACON gene expression. These results suggest that hypoxia dysregulates the expressions of LDHA, FASN, and mACON genes, and the hypoxia-induced mACON gene expression is via the HIF-1α-dependent and iron-dependent pathways in prostate carcinoma cells.


Subject(s)
Aconitate Hydratase/genetics , Carcinoma/genetics , Carcinoma/metabolism , Gene Expression Regulation, Neoplastic , Mitochondria/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Aconitate Hydratase/metabolism , Cell Hypoxia , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mitochondria/genetics
15.
J Mol Endocrinol ; 49(2): 153-63, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22872134

ABSTRACT

Growth differentiation factor-15 (GDF15), a member of the transforming growth factor-ß superfamily, is associated with human cancer progress. We evaluated the role GDF15 plays in tumorigenesis of prostate carcinoma PC-3 cells. Results from real-time RT-PCR and ELISA revealed that expression of GDF15 was approximately threefold higher in LNCaP cells than in PC-3 cells. Other prostate cell lines (PZ-HPV-7, CA-HPV-10, and DU145 cells) expressed extremely low levels of GDF15. Stable overexpression of GDF15 in PC-3 cells enhanced the degree of cell proliferation and invasion as shown in the (3)H-thymidine incorporation assay and in the Matrigel invasion assay respectively. Soft agar assays and xenograft animal studies indicated that overexpression of GDF15 in PC-3 cells increased tumorigenesis in vitro and in vivo. Results from RT-PCR, immunoblot, and reporter assays revealed that overexpression of GDF15 resulted in decreased expression of maspin and upregulation of interleukin-6 (IL6), matriptase, and N-myc downstream-regulated gene 1 (NDRG1) expression. Further studies revealed that overexpression of IL6 enhanced GDF15 expression in LNCaP cells while knockdown of IL6 blocked the expression of GDF15 in PC-3 cells, suggesting that expression of GDF15 is upregulated by IL6. This study demonstrated that expression of GDF15 induces cell proliferation, invasion, and tumorigenesis of prostate carcinoma PC-3 cells. The enhancement of tumorigenesis and invasiveness of prostate carcinoma cells that stably overexpress GDF15 may be caused by the dysregulation of maspin, matriptase, and IL6 gene expression. The expression of GDF15 and IL6 is controlled via a positive feedback loop in PC-3 cells.


Subject(s)
Growth Differentiation Factor 15/genetics , Interleukin-6/genetics , Prostatic Neoplasms/pathology , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Nude , Serine Endopeptidases/genetics , Serpins/genetics , Xenograft Model Antitumor Assays
16.
Am J Physiol Cell Physiol ; 302(4): C676-85, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22116304

ABSTRACT

L-Mimosine, an iron chelator and a prolyl 4-hydroxylase inhibitor, blocks many cancer cells at the late G1 phase. B-cell translocation gene 2 (Btg2) regulates the G1/S transition phases of the cell cycle. N-myc downstream regulated gene 1 (Ndrg1) is a differentiation-inducing gene upregulated by hypoxia. We evaluated the molecular mechanisms of L-mimosine on cell cycle modulation in PC-3 and LNCaP prostate carcinoma cells. The effect of L-mimosine on cell proliferation of prostate carcinoma cells was determined by the [3H]thymidine incorporation and flow cytometry assays. L-Mimosine arrested the cell cycle at the G1 phase in PC-3 cells and at the S phase in LNCaP cells, thus attenuating cell proliferation. Immunoblot assays indicated that hypoxia and L-mimosine stabilized hypoxia-inducible factor-1α (HIF-1α) and induced Btg2 and Ndrg1 protein expression, but downregulated protein levels of cyclin A in both PC-3 and LNCaP cells. L-Mimosine treatment decreased cyclin D1 protein in PC-3 cells, but not in LNCaP cells. Dimethyloxalylglycine, a pan-prolyl hydroxylase inhibitor, also induced Btg2 and Ndrg1 protein expression in LNCaP cells. The transient gene expression assay revealed that L-mimosine treatment or cotransfection with HIF-1α expression vector enhanced the promoter activities of Btg2 and Ndrg1 genes. Knockdown of HIF-1α attenuated the increasing protein levels of both Btg2 and Ndrg1 by hypoxia or L-mimosine in LNCaP cells. Our results indicated that hypoxia and L-mimosine modulated Btg2 and Ndrg1 at the transcriptional level, which is dependent on HIF-1α. L-Mimosine enhanced expression of Btg2 and Ndrg1, which attenuated cell proliferation of the PC-3 and LNCaP prostate carcinoma cells.


Subject(s)
Carcinoma/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Hypoxia/metabolism , Mimosine/pharmacology , Prostate/drug effects , Prostatic Neoplasms/metabolism , Transcriptional Activation/drug effects , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/pathology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Flow Cytometry , Humans , Hypoxia/genetics , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Organ Specificity , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Transfection , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
Int J Cancer ; 130(12): 2812-23, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-21780100

ABSTRACT

Luteolin is a polyphenolic flavone and has antitumor activity for many cancers. The prostate-derived Ets factor (PDEF), a novel epithelium-specific Ets transcription factor, acts as an androgen-independent transcriptional activator of the prostate-specific antigen (PSA) promoter. We determined the antitumor function of luteolin via upregulation of PDEF gene expression in human prostate carcinoma LNCaP cells. Results from flow cytometry and (3) H-thymidine incorporation assays revealed that luteolin treatments attenuated cell proliferation and arrested the cell cycle at the G1/S phase. High concentration of luteolin (30 µM) induced cell apoptosis. Immunoblot assays and enzyme linked immunosorbent assay revealed that luteolin treatment upregulated PDEF but downregulated androgen receptor (AR) gene expression, which decreased PSA gene expression in LNCaP cells. Results of immunoblot and transient gene expression assays revealed that luteolin treatments at proapoptosis dosage, enhanced gene expression of PDEF, B-cell translocation gene 2 (BTG2), N-myc downstream regulated gene 1 (NDRG1) and Maspin. Transient gene expression assays indicated that cotransfection of the PDEF expression vector enhanced the promoter activities of the BTG2, NDRG1 and Maspin genes. Stable overexpression of PDEF significantly induced BTG2, NDRG1 and Maspin gene expression, which markedly attenuated in vitro cell proliferation and invasion of LNCaP cells. The modulatory effect of luteolin on BTG2, NDRG1 and Maspin gene expression were attenuated when PDEF was knocked-down. These results suggest that luteolin blocks PSA gene expression by downregulation of AR expression. The enhancement of PDEF expression, which induced BTG2, NDRG1 and Maspin gene expression, could account for the function of luteolin for antiproliferation and anti-invasion in LNCaP cells.


Subject(s)
Luteolin/pharmacology , Prostate-Specific Antigen/biosynthesis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-ets/metabolism , Receptors, Androgen/biosynthesis , Apoptosis/drug effects , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Neoplasm Invasiveness , Promoter Regions, Genetic/drug effects , Prostate/metabolism , Prostate/pathology , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-ets/biosynthesis , Proto-Oncogene Proteins c-ets/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Serpins/biosynthesis , Serpins/genetics , Transcriptional Activation , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
18.
Mol Nutr Food Res ; 55(11): 1666-76, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21936051

ABSTRACT

SCOPE: Prostate-specific antigen (PSA) is a well-known marker for diagnosing and monitoring prostate cancer. Curcumin, a yellow curry pigment, has been reported to enhance androgen receptor (AR) degradation. We examined the effects of curcumin on increasing PSA expression by hypoxia and prolyl hydroxylase inhibitors, L-mimosine and dimethyloxalylglycine (DMOG), in human prostate carcinoma LNCaP cells. METHODS AND RESULTS: The 3H-thymidine incorporation assay revealed that either L-mimosine or DMOG treatments attenuated cell proliferation. Immunoblot and enzyme-linked immunosorbent assays (ELISA) indicated that both L-mimosine and DMOG have an effect similar to hypoxia, which stabilized hypoxia-inducible factor-1α (HIF-1α) and induced PSA gene expression. The results of the immunoblot and transient gene expression assays indicated that induction of the PSA expression by hypoxia is both HIF-1α- and AR-dependent. Immunoblot assays revealed that a curcumin treatment (10 µM) decreased the protein abundance of AR but did not significantly affect the protein levels of HIF-1α and vascular endothelial growth factor, which were induced by hypoxia. ELISA and transient gene expression assays indicated that curcumin blocked the activation of L-mimosine or DMOG treatment on PSA expression. CONCLUSIONS: These results indicate that curcumin blocked the enhanced effect of PSA expression by L-mimosine and DMOG that induce hypoxia condition.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma/drug therapy , Cell Hypoxia/drug effects , Curcumin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Amino Acids, Dicarboxylic/adverse effects , Amino Acids, Dicarboxylic/antagonists & inhibitors , Amino Acids, Dicarboxylic/pharmacology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Carcinoma/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Genes, Reporter/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Iron Chelating Agents/adverse effects , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Male , Mimosine/adverse effects , Mimosine/antagonists & inhibitors , Mimosine/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/metabolism , Recombinant Proteins/metabolism
19.
Anticancer Res ; 28(4A): 1969-76, 2008.
Article in English | MEDLINE | ID: mdl-18649734

ABSTRACT

A novel prostate cancer cell line (PC-J) was isolated from an androgen independent non-prostate specific antigen (non-PSA) producing carcinoma cell line. The homologous correlation between PC-J and PC-3 was determined by short tandem repeat analysis. The PSA promoter activity was detected by transient expression assay in the PC-J and LNCaP cells but not in androgen insensitive PC-3 cells. When the PC-J cells were cotransfected with androgen receptor, androgen receptor coactivators and PSA reporter vector cells, the reporter assays indicated that nuclear receptor coactivator 4 (NCOA4) but not androgen receptor activator 24 (ARA24) increased the sensitivity and maximum stimulation of dihydrotestosterone (DHT)-inducing PSA promoter activity. The RT-PCR assays revealed that the expression of several tumor markers, including interleukin-6, prostate stem cell antigen (PSCA), prostate epithelium-specific Ets transcription factor (PDEF) and matriptase, was lower in the PC-J cells than in the PC-3 cells. This cell model elucidated the regulation of PSA expression and enabled comparison of the gene profile at different stages of metastasis in prostatic carcinoma.


Subject(s)
Gene Expression Regulation, Neoplastic , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/genetics , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Gene Expression Profiling , Genetic Vectors/genetics , Humans , Luciferases/biosynthesis , Luciferases/genetics , Male , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Promoter Regions, Genetic , Prostate-Specific Antigen/biosynthesis , Prostatic Neoplasms/metabolism , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Transfection
20.
Anticancer Res ; 28(4A): 1977-83, 2008.
Article in English | MEDLINE | ID: mdl-18649735

ABSTRACT

The precise mechanisms of metastasis in prostatic cancer are still unknown. A subculture cell line (PC-J) was isolated from the metastasis human prostate cell line PC-3. In vitro cell proliferation, wound healing and invasion assays revealed that tumorigenesis and metastasis differed between PC-3 and PC-J cells. Eight weeks after nude mice were prostate-injected with PC-J and PC-3 cells, the PC-3 group had low tumor volume and exhibited metastasis whereas the PC-J group had high tumor volume and no metastasis. Subsequent RT-PCR and immunoblot assays indicated that matriptase was the putative metastatic gene. Overexpression of bikunin significantly reduced the gene expression of matriptase, which attenuated in vitro cell invasion in the PC-3 cells. In vitro and xenograft animal models indicated different metastatic characteristics between PC-3 and PC-J cells, suggesting that matriptase plays an important role in the metastasis of prostate cancer.


Subject(s)
Alpha-Globulins/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Serine Endopeptidases/biosynthesis , Alpha-Globulins/genetics , Animals , Cell Line, Tumor , Down-Regulation , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Serine Endopeptidases/genetics , Thymidine/metabolism , Transfection , Transplantation, Heterologous , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...