Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Cell Death Dis ; 15(8): 599, 2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39155279

ABSTRACT

Oral squamous cell carcinoma (OSCC) is a prevalent cancer worldwide, exhibiting unique regional prevalence. Despite advancements in diagnostics and therapy, the 5-year survival rate for patients has seen limited improvement. A deeper understanding of OSCC pathogenesis, especially its molecular underpinnings, is essential for improving detection, prevention, and treatment. In this context, noncoding RNAs, such as circular RNAs (circRNAs), have gained recognition as crucial regulators and potential biomarkers in OSCC progression. Our study highlights the discovery of previously uncharacterized circRNAs, including a SNX5 gene-derived circRNA, circSNX5, through deep sequencing of OSCC patient tissue transcriptomes. We established circSNX5's tumor-specific expression and its strong correlation with patient survival using structure-specific and quantitative PCR analyses. In vitro and in vivo experiments underscored circSNX5 RNA's regulatory role in cancer growth and metastasis. Further, our omics profiling and functional assays revealed that ADAM10 is a critical effector in circSNX5-mediated cancer progression, with circSNX5 maintaining ADAM10 expression by sponging miR-323. This novel circRNA-miRNA-mRNA regulatory axis significantly contributes to oral cancer progression and malignancy. Moreover, we discovered that circSNX5 RNA is produced via noncanonical sequential back-splicing of pre-mRNA, a process negatively regulated by the RNA-binding protein STAU1. This finding adds a new dimension to our understanding of exonic circRNA biogenesis in the eukaryotic transcriptome. Collectively, our findings offer a detailed mechanistic dissection and functional interpretation of a novel circRNA, shedding light on the role of the noncoding transcriptome in cancer biology and potentially paving the way for innovative therapeutic strategies.


Subject(s)
Mouth Neoplasms , RNA, Circular , Sorting Nexins , Humans , RNA, Circular/genetics , RNA, Circular/metabolism , Sorting Nexins/metabolism , Sorting Nexins/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , Mice , Mice, Nude , MicroRNAs/metabolism , MicroRNAs/genetics , Male , Female , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism
2.
Medicina (Kaunas) ; 59(11)2023 Oct 26.
Article in English | MEDLINE | ID: mdl-38003945

ABSTRACT

Background and Objectives: Urinary tract stones have long been a common ailment afflicting the population, with a high incidence and a wide distribution across different age groups. Effectively preventing the occurrence of urinary tract stones is of paramount importance. The primary aim of this study is to investigate the correlations between individual characteristics, water consumption habits, dietary habits, exercise habits, and the occurrence of urinary tract stones in a Taiwanese population. Materials and Methods: This study is cross-sectional research conducted over one month in 2022. One hundred eligible urinary stone cases were recruited through physician screening at outpatient clinics, and an additional one hundred samples from surgical outpatients without urinary tract stones were included as the control group. A questionnaire survey was employed to gather information on demographic variables, dietary habits, water consumption, and exercise habits of the cases. Descriptive statistics, chi-square tests, and logistic regression were used to explore the impact of relevant factors on urinary stone formation. Results: The analysis results revealed that among the demographic variables, males exhibited a higher risk of contracting urinary tract stones than females, and the majority of cases fell within the 40- to 49-year-old age group. Unhealthy lifestyle habits such as smoking and betel nut chewing also demonstrated a higher susceptibility to urinary tract stones. A logistic regression analysis showed that individuals who engaged in physical activity more than three times per week and those with inadequate water intakes had a higher risk of developing urinary tract stones. Conclusions: There is a close relationship between lifestyle and urinary tract stones. It is recommended that individuals continue to hydrate adequately during exercise.


Subject(s)
Urinary Calculi , Male , Female , Humans , Adult , Middle Aged , Cross-Sectional Studies , Urinary Calculi/epidemiology , Urinary Calculi/etiology , Diet/adverse effects , Risk Factors , Life Style
3.
J Control Release ; 148(3): 292-302, 2010 Dec 20.
Article in English | MEDLINE | ID: mdl-20854857

ABSTRACT

Colorectal cancer (CRC) is the third most common malignancy and the fourth most frequent cause of cancer deaths worldwide. Ligand-mediated diagnosis and targeted therapy would have vital clinical applications in cancer treatment. In this study, an orthotopic model of colorectal cancer was established in mice. In vivo phage library selection was then utilized to isolate peptides specifically recognizing the vasculature of colorectal cancer tissues. A phage (termed TCP-1 phage) was isolated by this manner and it homed to the colorectal cancer tissues by 11- to 94-fold more than other organs. Chemical synthetic peptide (CTPSPFSHC, termed TCP-1) displayed by TCP-1 phage inhibited the homing ability of the phage to the tumor mass when co-injected intravenously with the TCP-1 phage into mice with colon cancer. Meanwhile, immunostaining analysis indicated that TCP-1 phage and peptide localized in the vasculature of the colorectal cancer tissue, but not of normal tissues. Moreover, TCP-1 peptide bound to blood vessels of surgical tissue samples of human colorectal cancer. After intravenous injection of FITC-labeled TCP-1 into the tumor-bearing mice for 20h, there was a strong fluorescent signal in the tumors but not other tissues when observed under blue light. In addition, TCP-1 conjugated with a pro-apoptotic peptide specifically induced apoptosis of tumor-associated blood vessels in vivo. The data define a novel peptide TCP-1 as an effective agent for imaging detection and drug delivery for colorectal cancer.


Subject(s)
Colorectal Neoplasms/blood supply , Colorectal Neoplasms/diagnosis , Drug Carriers/chemistry , Peptide Library , Peptides/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Drug Carriers/chemical synthesis , Fluorescein/administration & dosage , Fluorescein-5-isothiocyanate/chemistry , Humans , Male , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Peptides/chemical synthesis
4.
Autophagy ; 6(2): 228-38, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20087064

ABSTRACT

The ubiquitin-proteasome system and macroautophagy are two complementary pathways for protein degradation. Emerging evidence suggests that proteasome inhibition might be a promising approach for the treatment of cancer. In this study, we show that proteasome inhibitor MG-132 suppressed gastric cancer cell proliferation and induced macroautophagy. The induction of macroautophagy was evidenced by the formation of LC3(+) autophagosomes and the accumulation of acidic vesicular organelles and autolysosomes and was accompanied by the suppression of mammalian target of rapamycin complex 1 activity. Abolition of macroautophagy by knockdown of Class III phosphatidylinositol-3 kinase Vps34 or ATG5/7 sensitized gastric cancer cells to the antiproliferative effect of MG-132 by promoting G(2)/M cell cycle arrest. In addition, MG-132 increased ERK phosphorylation whose inhibition by MEK inhibitor significantly enhanced the antiproliferative effect of proteasome inhibition. To conclude, this study demonstrates that macroautophagy and ERK phosphorylation serve as protective mechanisms to counteract the antiproliferative effect of proteasome inhibition. This discovery may have implications for the application of proteasome-directed therapy for the treatment of cancer.


Subject(s)
Autophagy/physiology , Cell Proliferation/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Proteasome Inhibitors , Stomach Neoplasms/physiopathology , Animals , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Leupeptins/pharmacology , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases , Vacuoles/drug effects , Vacuoles/metabolism
5.
Biochem Pharmacol ; 78(9): 1224-32, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19591809

ABSTRACT

Resveratrol, a naturally occurring polyphenolic antioxidant, is a compound holding promise for cancer chemoprevention. Previous studies suggest that 2,3',4,5'-tetramethoxy-trans-stilbene (TMS) and 3,4,4',5,-tetramethoxy-trans-stilbene (MR-4), both of which are derivatives of resveratrol, are potent apoptosis-inducing agents with clinical potential. In this study, we chemically synthesized 2,3',4,4',5'-pentamethoxy-trans-stilbene (PMS), the hybrid molecule of TMS and MR-4, and determined its effects on colon cancer growth. When compared with its parent compounds, PMS displayed more potent in vitro anti-mitogenic effect on colon cancer cells (Caco-2, HT-29 and SW1116). Moreover, PMS inhibited tumor growth in vivo in a colon cancer xenograft model. In this connection, PMS strongly induced apoptosis in HT-29 cells as evidenced by increased PARP cleavage, DNA fragmentation, and accumulation of sub-G(1) population. Further mechanistic analysis revealed that PMS enhanced the polymerization of microtubules, which was followed by G(2)/M mitotic arrest and caspase-dependent apoptosis. The activation of caspases-3, -7, -8, and -9 was involved in PMS-induced apoptosis with concomitant down-regulation of the pro-survival PI3K/Akt signaling. Collectively, these data suggest that PMS is a potent inducer of apoptosis via targeting microtubules and may merit investigation as a potential chemoprophylactic and therapeutic agent for colon cancer.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/pathology , Microtubules/drug effects , Stilbenes/pharmacology , Animals , Blotting, Western , Caspases/metabolism , Cell Division , Cell Line, Tumor , Colonic Neoplasms/enzymology , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , G2 Phase , Humans , Mice , Mice, Nude , Poly(ADP-ribose) Polymerases/metabolism
6.
Int J Cancer ; 125(11): 2540-6, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19623651

ABSTRACT

Overexpression of cyclooxygenase-2 (COX-2) and elevation of its derivative prostaglandin E(2) (PGE(2)) are implicated in human esophageal squamous cell carcinoma. The expression of c-Myc, an oncogenic transcription factor, is also upregulated in this malignant disease. This study sought to elucidate whether a functional connection exists between COX-2/PGE(2) and c-Myc in esophageal squamous cell carcinoma. Results showed that PGE(2) substantially increased the proliferation of cultured esophageal squamous cell carcinoma cells. In this regard, PGE(2) substantially increased the mRNA and protein expression of c-Myc and its association with the binding partner Max. Knockdown of c-Myc by RNA interference also significantly attenuated PGE(2)-induced cell proliferation. Further, mechanistic study revealed that PGE(2) increased the protein stability and nuclear accumulation of c-Myc via phosphorylation on serine 62 in an extracellular signal regulated kinase (ERK)-dependent manner. To this end, ERK activation by PGE(2) was completely abolished by protein kinase C (PKC) inhibitors. Moreover, the effect of PGE(2) on c-Myc expression was mimicked by EP2 receptor agonist. In addition, knockdown of EP2 receptor by EP2 siRNA attenuated PGE(2)-induced c-Myc expression. Collectively, our findings suggest that PGE(2) upregulates c-Myc via the EP2/PKC/ERK pathway. This study sheds new light on the carcinogenic mechanism of PGE(2) in esophageal squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Proliferation , Dinoprostone/pharmacology , Esophageal Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cyclooxygenase 2/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Oxytocics/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP2 Subtype , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
7.
Biochem Biophys Res Commun ; 386(4): 598-601, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19540199

ABSTRACT

Protein homeostasis relies on a balance between protein synthesis and protein degradation. The ubiquitin-proteasome system is a major catabolic pathway for protein degradation. In this respect, proteasome inhibition has been used therapeutically for the treatment of cancer. Whether inhibition of protein degradation by proteasome inhibitor can repress protein translation via a negative feedback mechanism, however, is unknown. In this study, proteasome inhibitor MG-132 lowered the proliferation of colon cancer cells HT-29 and SW1116. In this connection, MG-132 reduced the phosphorylation of mammalian target of rapamycin (mTOR) at Ser2448 and Ser2481 and the phosphorylation of its downstream targets 4E-BP1 and p70/p85 S6 kinases. Further analysis revealed that MG-132 inhibited protein translation as evidenced by the reductions of (35)S-methionine incorporation and polysomes/80S ratio. Knockdown of raptor, a structural component of mTOR complex 1, mimicked the anti-proliferative effect of MG-132. To conclude, we demonstrate that the inhibition of protein degradation by proteasome inhibitor represses mTOR signaling and protein translation in colon cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/enzymology , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Protein Kinases/metabolism , Protein Synthesis Inhibitors/pharmacology , Adaptor Proteins, Signal Transducing , Humans , Phosphorylation/drug effects , Polyribosomes/drug effects , Polyribosomes/metabolism , Proteasome Inhibitors , Protein Biosynthesis/drug effects , Proteins/genetics , Regulatory-Associated Protein of mTOR , TOR Serine-Threonine Kinases
8.
Biochem Biophys Res Commun ; 382(2): 451-6, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19289106

ABSTRACT

Macroautophagy is a process by which cytoplasmic content and organelles are sequestered by double-membrane bound vesicles and subsequently delivered to lysosomes for degradation. Macroautophagy serves as a major intracellular pathway for protein degradation and as a pro-survival mechanism in time of stress by generating nutrients. In the present study, bafilomycin A(1), a vacuolar type H(+)-ATPase inhibitor, suppresses macroautophagy by preventing acidification of lysosomes in colon cancer cells. Diminished macroautophagy was evidenced by the accumulation of undegraded LC3 protein. Suppression of macroautophagy by bafilomycin A(1) induced G(0)/G(1) cell cycle arrest and apoptosis which were accompanied by the down-regulation of cyclin D(1) and cyclin E, the up-regulation of p21(Cip1) as well as cleavages of caspases-3, -7, -8, and -9 and PARP. Further investigation revealed that bafilomycin A(1) increased the phosphorylation of ERK, JNK, and p38. In this regard, p38 inhibitor partially reversed the anti-proliferative effect of bafilomycin A(1). To conclude, inhibition of macroautophagy by bafilomycin A(1) lowers G(1)-S transition and induces apoptosis in colon cancer cells. Our results not only indicate that inhibitors of macroautophagy may be used therapeutically to inhibit cancer growth, but also delineate the relationship between macroautophagy and apoptosis.


Subject(s)
Apoptosis , Autophagy/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Macrolides/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoplasmic Vesicles/drug effects , Humans , Microtubule-Associated Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism
9.
Mol Pharmacol ; 75(6): 1364-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19264847

ABSTRACT

Doxorubicin is a chemotherapeutic drug widely used for the treatment of advanced esophageal squamous cell carcinoma. However, its efficacy is usually limited by the development of multidrug resistance (MDR), which has been linked to the up-regulation of P-glycoprotein (P-gp) in cancer cells. Conventional nonsteroidal anti-inflammatory drugs and cyclooxygenase 2 (COX-2)-selective inhibitors have been demonstrated to overcome MDR in some cancer cells. Here we sought to elucidate the effect of COX inhibitors on doxorubicin-induced cytotoxicity in relation to P-gp function in human esophageal squamous cell carcinoma cells. Among the five tested COX inhibitors [indomethacin, 4-[5-(4-chlorophenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]-benzenesulfonamide (SC236), 5-(4-chlorophenyl)-1-(4-methoxyphenyl)-3-trifluorom-ethylpyrazole (SC560), nimesulide, and N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide (NS398)], all of which substantially suppressed prostaglandin E(2) (PGE(2)) production to a similar extent, only the nonselective COX inhibitor indomethacin and the COX-2-selective inhibitor SC236 enhanced cytotoxic effects of doxorubicin on HKESC-1 and HKESC-2 cells. Moreover, these effects could not be reversed by the addition of PGE(2). Knockdown of COX-2 by small interference RNA also failed to mimic the enhancing effects of indomethacin or SC236, implicating that their action is COX- and PGE(2)-independent. To this end, we observed that indomethacin and SC236 directly functioned as noncompetitive inhibitors of P-gp, which were manifested as a reduction of P-gp ATPase activity. Collectively, these findings suggest that the direct inhibitory effects of indomethacin and SC236 on P-gp may contribute to their ability to increase the intracellular retention of doxorubicin and thus enhance its cytotoxicity. The combination of indomethacin or SC236 with doxorubicin may have significant potential clinical application, especially in the circumvention of P-gp-mediated MDR in cancer cells.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Indomethacin/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Adenosine Triphosphatases/antagonists & inhibitors , Carcinoma, Squamous Cell , Cell Line, Tumor , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/pharmacology , Down-Regulation , Drug Resistance, Neoplasm , Drug Synergism , Esophageal Neoplasms , Gene Knockdown Techniques , Humans , NF-kappa B/antagonists & inhibitors , RNA, Small Interfering/genetics
10.
Biochem Biophys Res Commun ; 382(1): 79-84, 2009 Apr 24.
Article in English | MEDLINE | ID: mdl-19258012

ABSTRACT

Evolving evidence supports that cyclooxygenase-1 (COX-1) takes part in colon carcinogenesis. The effects of COX-1 inhibition on colon cancer cells, however, remains obscured. In this study, we demonstrate that COX-1 inhibitor sc-560 inhibited colon cancer cell proliferation with concomitant G(0)/G(1)-phase cell cycle arrest. The anti-proliferative effect was associated with down-regulation of c-Fos, cyclin E(2) and E(2)F-1 and up-regulation of p21(Waf1/Cip1) and p27(Kip1). In addition, sc-560 induced macroautophagy, an emerging mechanism of tumor suppression, as evidenced by the formation of LC3(+) autophagic vacuoles, enhanced LC3 processing, and the accumulation of acidic vesicular organelles and autolysosomes. In this connection, 3-methyladenine, a Class III phosphoinositide 3-kinase inhibitor, significantly abolished the formation of LC3(+) autophagic vacuoles and the processing of LC3 induced by sc-560. To conclude, this study reveals the unreported relationship between COX-1 and proliferation/macroautophagy of colon cancer cells.


Subject(s)
Autophagy , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Cyclooxygenase 1/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Autophagy/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase Inhibitors/pharmacology , Humans , Microtubule-Associated Proteins/metabolism , Pyrazoles/pharmacology , Vacuoles/metabolism
11.
Biochem Biophys Res Commun ; 374(2): 258-63, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18638451

ABSTRACT

The ubiquitin-proteasome system (UPS) and lysosome-dependent macroautophagy (autophagy) are two major intracellular pathways for protein degradation. Blockade of UPS by proteasome inhibitors has been shown to activate autophagy. Recent evidence also suggests that proteasome inhibitors may inhibit cancer growth. In this study, the effect of a proteasome inhibitor MG-132 on the proliferation and autophagy of cultured colon cancer cells (HT-29) was elucidated. Results showed that MG-132 inhibited HT-29 cell proliferation and induced G(2)/M cell cycle arrest which was associated with the formation of LC3(+) autophagic vacuoles and the accumulation of acidic vesicular organelles. MG-132 also increased the protein expression of LC3-I and -II in a time-dependent manner. In this connection, 3-methyladenine, a Class III phosphoinositide 3-kinase inhibitor, significantly abolished the formation of LC3(+) autophagic vacuoles and the expression of LC3-II but not LC3-I induced by MG-132. Taken together, this study demonstrates that inhibition of proteasome in colon cancer cells lowers cell proliferation and activates autophagy. This discovery may shed a new light on the novel function of proteasome in the regulation of autophagy and proliferation in colon cancer cells.


Subject(s)
Autophagy , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Proteasome Inhibitors , Adenine/analogs & derivatives , Adenine/pharmacology , Autophagy/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasmic Vesicles/metabolism , G2 Phase/drug effects , Humans , Leupeptins/pharmacology , Microtubule-Associated Proteins/biosynthesis , Vacuoles/metabolism
12.
J Pharmacol Exp Ther ; 327(1): 258-67, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18583546

ABSTRACT

The use of nonsteroidal anti-inflammatory drugs is associated with a lower risk for esophageal squamous cell carcinoma, in which overexpression of cyclooxygenase-2 (COX-2) is frequently reported. Prostaglandin E(2) (PGE(2)), a COX-2-derived eicosanoid, is implicated in the promotion of cancer growth. However, the precise role of PGE(2) in the disease development of esophageal squamous cell carcinoma remains elusive. In this study, we investigated the effect of PGE(2) on the proliferation of cultured esophageal squamous cell carcinoma cells (HKESC-1). Results showed that HKESC-1 cells expressed all four series of prostaglandin (EP) receptors, namely, EP1 to EP4 receptors. In this regard, PGE(2) and the EP2 receptor agonist (+/-)-15-deoxy-16S-hydroxy-17-cyclobutyl PGE(1) methyl ester (butaprost) markedly increased HKESC-1 cell proliferation. Moreover, the mitogenic effect of PGE(2) was significantly attenuated by RNA interference-mediated knockdown of the EP2 receptor, indicating that this receptor mediated the mitogenic effect of PGE(2). In this connection, PGE(2) and butaprost induced phosphorylation of extracellular signal-regulated kinases 1/2 (Erk1/2), whose down-regulation by RNA interference significantly attenuated PGE(2)-induced cell proliferation. In addition, PGE(2) and butaprost increased c-Fos expression and activator protein 1 (AP-1) transcriptional activity, which were abolished by the mitogen-activated protein kinase/Erk kinase inhibitor 1,4-diamino-2,3-dicyano-1,4-bis(o-aminophenylmercapto)-butadiene ethanolate (U0126). AP-1-binding inhibitor curcumin also partially reversed the mitogenic effect of PGE(2). Taken together, these data demonstrate for the first time that the EP2 receptor mediates the mitogenic effect of PGE(2) in esophageal squamous cell carcinoma via activation of the Erk/AP-1 pathway. This study supports the growth-promoting action of PGE(2) in esophageal squamous cell carcinoma and the potential application of EP2 receptor antagonists in the treatment of this disease.


Subject(s)
Carcinoma, Squamous Cell/pathology , Dinoprostone/pharmacology , Esophageal Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Mitogens/pharmacology , Receptors, Prostaglandin E/physiology , Transcription Factor AP-1/metabolism , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Butadienes/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase 1/analysis , Cyclooxygenase 2/analysis , Dinoprostone/metabolism , Enzyme Activation , Humans , Nitriles/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/analysis , Receptors, Prostaglandin E, EP2 Subtype
SELECTION OF CITATIONS
SEARCH DETAIL