Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Cells ; 12(13)2023 06 23.
Article in English | MEDLINE | ID: mdl-37443730

ABSTRACT

Cell adhesion molecules, including integrins, cadherins, and claudins (CLDNs), are known to activate Src-family kinases (SFKs) that organize a variety of physiological and pathological processes; however, the underlying molecular basis remains unclear. Here, we identify the SFK members that are coupled with the CLDN6-adhesion signaling. Among SFK subtypes, BLK, FGR, HCK, and SRC were highly expressed in F9 cells and concentrated with CLDN6 along cell borders during epithelial differentiation. Immunoprecipitation assay showed that BLK and SRC, but not FGR or HCK, form a complex with CLDN6 via the C-terminal cytoplasmic domain. We also demonstrated, by pull-down assay, that recombinant BLK and SRC proteins directly bind to the C-terminal cytoplasmic domain of CLDN6 (CLDN6C). Unexpectedly, both recombinant SFK proteins recognized the CLDN6C peptide in a phosphotyrosine-independent manner. Furthermore, by comparing phenotypes of F9:Cldn6:Blk-/- and F9:Cldn6:Src-/- cells with those of wild-type F9 and F9:Cldn6 cells, we revealed that BLK and SRC are essential for CLDN6-triggered cellular events, namely epithelial differentiation and the expression of retinoid acid receptor target genes. These results indicate that selective SFK members appear to participate in the CLDN-adhesion signaling.


Subject(s)
Signal Transduction , src-Family Kinases , src-Family Kinases/metabolism , Cell Adhesion Molecules , Integrins , Receptors, Retinoic Acid , Claudins/genetics , Claudins/metabolism
2.
Anticancer Res ; 42(6): 2923-2930, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35641269

ABSTRACT

BACKGROUND/AIM: Liver X receptors (LXRs) are nuclear receptors with various functions, including the regulation of cholesterol metabolism, glucose homeostasis, and inflammation. We previously reported that LXR activation inhibits the growth of oral cancer cells by inducing cellular cholesterol efflux and that LXRß is expressed mainly in small-cell lung cancer (SCLC) tissues. SCLC is one of the most aggressive cancers, and identifying an effective therapeutic target molecule is desirable. Therefore, we investigated whether LXRß could be an effective target molecule for SCLC treatment through in vitro experiments. MATERIALS AND METHODS: We evaluated the influence of treatment with the LXR agonist T0901317 on cell proliferation and apoptosis in SCLC cell lines using cell viability, BrdU-ELISA, FACS, and western blot analyses. Moreover, the mechanism by which T0901317 inhibits SCLC cell proliferation was elucidated using qRT-PCR, western blot, a cholesterol quantification assay, and a genome editing technique. RESULTS: We showed that cultivated SCLC cells expressed LXRß and that an LXR agonist inhibited the proliferation of SCLC cells without toxicity to normal cells. Furthermore, the antitumoral effect of an LXR agonist on SCLC cells was attributed to the induction of ABCA1 by LXRß activation, resulting in an increase in cellular cholesterol efflux via ABCA1. CONCLUSION: The activation of LXRß up-regulates ABCA1 expression, causing cholesterol depletion in cancer cells. This mechanism could be a novel target strategy for SCLC.


Subject(s)
Lung Neoplasms , Orphan Nuclear Receptors , Cell Proliferation , Cholesterol/metabolism , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver X Receptors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Sulfonamides/pharmacology
3.
Am J Pathol ; 192(6): 847-861, 2022 06.
Article in English | MEDLINE | ID: mdl-35367201

ABSTRACT

Although recent reports have revealed the importance of the inactivation of both RB1 and TP53 in the transformation from lung adenocarcinoma into neuroendocrine carcinoma (NEC), the requirements for complete transformation into NEC have not been elucidated. To investigate alterations in the characteristics associated with the inactivation of RB1/TP53 and define the requirements for transformation into NEC cells, RB1/TP53 double-knockout A549 lung adenocarcinoma cells were established, and additional knockout of REST and transfection of ASCL1 and POU class 3 homeobox transcription factors (TFs) was conducted. More than 60 genes that are abundantly expressed in neural cells and several genes associated with epithelial-to-mesenchymal transition were up-regulated in RB1/TP53 double-knockout A549 cells. Although the expression of chromogranin A and synaptophysin was induced by additional knockout of REST (which mimics the status of most NECs), the expression of another neuroendocrine marker, CD56, and proneural TFs was not induced. However, coexpression of ASCL1 and POU3F4 in RB1/TP53/REST triple-knockout A549 cells induced the expression of not only CD56 but also other proneural TFs (NEUROD1 and insulinoma-associated 1) and induced NEC-like morphology. These findings suggest that the inactivation of RB1 and TP53 induces a state necessary for the transformation of lung adenocarcinoma into NEC and that further inactivation of REST and coexpression of ASCL1 and POU3F4 are the triggers for complete transformation into NEC.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Neuroendocrine , Lung Neoplasms , Neuroendocrine Cells , Small Cell Lung Carcinoma , Adenocarcinoma of Lung/pathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Humans , Infant, Newborn , Lung Neoplasms/pathology , Neuroendocrine Cells/metabolism , POU Domain Factors/metabolism , Retinoblastoma Binding Proteins , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics
4.
J Mol Histol ; 53(2): 437-448, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35094211

ABSTRACT

Endocrine secretory granules (ESGs) are morphological characteristics of endocrine/neuroendocrine cells and store peptide hormones/neurotransmitters. ESGs contain prohormones and ESG-related molecules, mainly chromogranin/secretogranin family proteins. However, the precise mechanism of ESG formation has not been elucidated. In this study, we experimentally induced ESGs in the non-neuroendocrine lung cancer cell line H1299. Since repressive element 1 silencing transcription factor (REST) and prospero homeobox 1 (PROX1) are closely associated with the expression of ESG-related molecules, we edited the REST gene and/or transfected PROX1 and then performed molecular biology, immunocytochemistry, and electron and immunoelectron microscopy assays to determine whether ESG-related molecules and ESGs were induced in H1299 cells. Although chromogranin/secretogranin family proteins were induced in H1299 cells by knockout of REST and the induction was accelerated by the PROX1 transgene, the ESGs could not be defined by electron microscopy. However, a small number of ESGs were detected in the H1299 cells lacking REST and expressing pro-opiomelanocortin (POMC) by electron microscopy. Furthermore, many ESGs were produced in the REST-lacking and PROX1- and POMC-expressing H1299 cells. These findings suggest that a lack of REST and the expression of genes related to ESG content are indispensable for ESG production and that PROX1 accelerates ESG production.Trial registration: Not applicable.


Subject(s)
Chromogranins , Genes, Homeobox , Chromogranins/genetics , Chromogranins/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Secretory Vesicles/metabolism , Transcription Factors/metabolism
5.
Mol Cancer Res ; 19(7): 1208-1220, 2021 07.
Article in English | MEDLINE | ID: mdl-33727343

ABSTRACT

Cell adhesion proteins not only maintain tissue integrity, but also possess signaling abilities to organize diverse cellular events in a variety of physiologic and pathologic processes; however, the underlying mechanism remains obscure. Among cell adhesion molecules, the claudin (CLDN) family is often aberrantly expressed in various cancers, but the biological relevance and molecular basis for this observation have not yet been established. Here, we show that high CLDN6 expression accelerates cellular proliferation and migration in two distinct human endometrial cancer cell lines in vitro. Using a xenograft model, we also revealed that aberrant CLDN6 expression promotes tumor growth and invasion in endometrial cancer tissues. The second extracellular domain and Y196/200 of CLDN6 were required to recruit and activate Src-family kinases (SFK) and to stimulate malignant phenotypes. Knockout and overexpression of ESR1 in endometrial carcinoma cells showed that the CLDN6-adhesion signal links to estrogen receptor α (ERα) to advance tumor progression. In particular, aberrant CLDN6-ERα signaling contributed to collective cell behaviors in the leading front of endometrial cancer cells. Importantly, we demonstrate that CLDN6/SFK/PI3K-dependent AKT and SGK (serum- and glucocorticoid-regulated kinase) signaling in endometrial cancer cells targets Ser518 in the human ERα to activate ERα transcriptional activity in a ligand-independent manner, thereby promoting tumor progression. Furthermore, CLDN6, at least in part, also regulated gene expression in an ERα-independent manner. IMPLICATIONS: The identification of this machinery highlights regulation of the transcription factors by cell adhesion to advance tumor progression.


Subject(s)
Cell Adhesion/genetics , Claudins/genetics , Endometrial Neoplasms/genetics , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , Signal Transduction/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Claudins/metabolism , Disease Progression , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Female , HEK293 Cells , Humans , Immediate-Early Proteins/metabolism , Mice, Inbred NOD , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transplantation, Heterologous , src-Family Kinases/metabolism
6.
J Pediatr Surg ; 56(9): 1668-1672, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33012558

ABSTRACT

BACKGROUND: Lymphatic malformation (LM) is a congenital disease caused by lymphatic vessel malformation. Although standard therapies for LMs are sclerotherapy and/or surgical excision, a new therapy using Japanese herbal medicine Eppikajutsuto (TJ-28) has been recently reported as clinically effective. We aimed to experimentally confirm the therapeutic effectiveness of TJ-28 for LMs. METHODS: LM lesions were generated in the mesentery and peritoneum of mice by intraperitoneal injection of Freund's incomplete adjuvant. Mice with LMs were treated by gavage or dietary administration of TJ-28 for 2 months. Formalin-fixed paraffin-embedded tissue sections of mesentery and peritoneum tissues were histologically and immunohistochemically examined by focusing on lymph nodes and perinodal lymph vessels. RESULTS: Multiple Freund's incomplete adjuvant-associated foreign-body granulomas were formed in the mesentery and peritoneum, resulting in congestion of lymph fluid and dilatation of lymph vessels. The numbers and sizes of lymph nodes were not significantly different between TJ-28-treated and control groups. However, the luminal areas of lymphatic vessels were reduced significantly in the TJ-28 treatment group by both gavage and dietary administrations. CONCLUSION: TJ-28 conspicuously reduced congestion of lymph fluid. This is the first histopathological evaluation of LM model mice to study the effectiveness of oral TJ-28 treatment.


Subject(s)
Lymphatic Abnormalities , Lymphatic Vessels , Pharmaceutical Preparations , Animals , Lymphatic Abnormalities/drug therapy , Mice , Plant Extracts
7.
Proc Natl Acad Sci U S A ; 116(49): 24600-24609, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31740618

ABSTRACT

Cell adhesion is essential for proper tissue architecture and function in multicellular organisms. Cell adhesion molecules not only maintain tissue integrity but also possess signaling properties that contribute to diverse cellular events such as cell growth, survival, differentiation, polarity, and migration; however, the underlying molecular basis remains poorly defined. Here we identify that the cell adhesion signal initiated by the tight-junction protein claudin-6 (CLDN6) regulates nuclear receptor activity. We show that CLDN6 recruits and activates Src-family kinases (SFKs) in second extracellular domain-dependent and Y196/200-dependent manners, and SFKs in turn phosphorylate CLDN6 at Y196/200. We demonstrate that the CLDN6/SFK/PI3K/AKT axis targets the AKT phosphorylation sites in the retinoic acid receptor γ (RARγ) and the estrogen receptor α (ERα) and stimulates their activities. Interestingly, these phosphorylation motifs are conserved in 14 of 48 members of human nuclear receptors. We propose that a similar link between diverse cell adhesion and nuclear receptor signalings coordinates a wide variety of physiological and pathological processes.


Subject(s)
Cell Adhesion/physiology , Claudins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cell Line , Claudins/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Regulation , Humans , Mice , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Domains , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction , Tyrosine/genetics , src-Family Kinases/metabolism , Retinoic Acid Receptor gamma
8.
Histol Histopathol ; 33(5): 497-505, 2018 May.
Article in English | MEDLINE | ID: mdl-29181837

ABSTRACT

Liver X receptors (LXRs) participate not only in maintaining cholesterol homeostasis but also in controlling cellular growth in many types of normal and tumor cells. We previously reported that LXRα was aberrantly expressed in human oral squamous cell carcinoma (HOSCC) tissues and cell lines, and that LXR stimulation led to significant reduction of proliferation of HOSCC cells via accelerating cholesterol efflux. Since LXRs and downstream proteins involved in cholesterol metabolism could be also applied as therapeutic targets in small cell lung carcinoma (SCLC) and pancreatic ductal adenocarcinoma (PDAC), we herein analyzed the distribution of LXR proteins in these refractory cancers as well as in normal human lung and pancreatic tissues. LXRß was observed in ciliated epithelial cells, bronchial gland epithelia, type II alveolar epithelia and alveolar macrophages of the lung, and was less expressed in bronchial basal cells and type I alveolar epithelia. In addition, LXRß was detected in epithelium of the pancreatic duct and acinar cells of the pancreas, and was weakly expressed in pancreatic islet cells. By contrast, LXRα expression was restricted to alveolar macrophages, and was not evident in any types of epithelial cells in the lung and pancreas. We also demonstrated that LXRß but not LXRα was abundantly expressed in nine cases of SCLC and twenty cases of PDAC tissues. These findings provide basic information for evaluating the efficacy of LXR-targeted treatment in SCLC and PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Small Cell/genetics , Gene Expression Regulation, Neoplastic/genetics , Liver X Receptors/biosynthesis , Liver X Receptors/genetics , Lung Neoplasms/genetics , Pancreatic Neoplasms/genetics , Aged , Aged, 80 and over , Antibody Specificity , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Drug Resistance, Neoplasm/genetics , Excitatory Postsynaptic Potentials , Female , Humans , Male , Middle Aged
9.
Oncotarget ; 8(55): 93382-93391, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29212157

ABSTRACT

Schizophrenia is thought to be caused by a combination of genetic and environmental factors; however, its pathogenesis remains largely unknown. Here, we focus on the endothelial tight-junction protein claudin-5 (CLDN5), because the CLDN5 gene is mapped to the schizophrenia-associated 22q11.2 deletion region, and a single nucleotide polymorphism in the CLDN5 locus is also linked to schizophrenia. We show, by RT-qPCR and immunohistochemistry, that the expressions of CLDN5 mRNA and protein are significantly increased and decreased, respectively, in the schizophrenic prefrontal cortex (PFC) compared with control PFC. These changes were not observed in the schizophrenic visual cortex (VC), and neither the density nor diameter of the CD34-positive microvessels was altered in the schizophrenic PFC or VC. Interestingly, protein kinase A (PKA) was activated in the microvascular and perivascular regions of the schizophrenic PFC, and the pPKA-positive microvascular endothelial cells occasionally exhibited focal loss of CLND5. Since we previously demonstrated that cAMP induced CLDN5 mRNA expression and size-selective loosening of the endothelial barrier in PKA-independent and -dependent manners, respectively, a similar mechanism could contribute to the discrepancy between mRNA and protein expression of CLDN5 in the schizophrenic PFC. Taken collectively, these findings provide novel insights into the pathophysiology of schizophrenia.

10.
Oncotarget ; 6(32): 33345-57, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26452260

ABSTRACT

Liver X receptors (LXRs) contribute not only to maintain cholesterol homeostasis but also to control cell growth. However, the molecular mechanisms behind the LXR-mediated anti-proliferative effects are largely unknown. Here we show, by immunohistochemistry, that LXRα and LXRß are differentially distributed in oral stratified squamous epithelia. By immunohistochemical and Western blot analyses, we also reveal that LXRα is abundantly expressed in human oral squamous cell carcinoma (HOSCC) tissues and cell lines. Cell counting, BrdU labeling and cell cycle assay indicated that LXR stimulation led to significant reduction of proliferation in HOSCC cells. Importantly, our study highlights, by using RNA interference, that the ATP-binding cassette transporter A1 (ABCA1)-accelerated cholesterol efflux is critical for the growth inhibitory action of LXRs in HOSCC cells. Moreover, we demonstrate that LXR activation reduces the growth of xenograft tumour of HOSCC cells in mice accompanied by the upregulation of ABCA1 expression and the decline of cholesterol levels in the tumour. These findings strongly suggested that targeting the LXR-regulated cholesterol transport, yielding in lowering intracellular cholesterol levels, could be a promising therapeutic option for certain types of cancers.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Carcinoma, Squamous Cell/pathology , Cell Proliferation/genetics , Cholesterol/metabolism , Mouth Neoplasms/pathology , Orphan Nuclear Receptors/physiology , ATP Binding Cassette Transporter 1/metabolism , Adult , Animals , Biological Transport/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Liver X Receptors , Male , Mice , Mice, SCID , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Rats , Rats, Wistar , Up-Regulation/genetics
11.
Pathol Int ; 62(4): 232-45, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22449227

ABSTRACT

Neural cell adhesion molecule 1 (NCAM1), synaptophysin (SYPT), and chromogranin A (CGA) are immunohistochemical markers for diagnosing lung neuroendocrine tumors (LNETs). However, the precise expression mechanisms have not been studied in enough detail. The purpose of the present study is to define the molecular mechanisms of NCAM1, SYPT, and CGA gene expressions, using cultivated lung cancer cells and focusing upon NeuroD1 (ND1), achaete-scute homolog-like 1 (ASCL1), and known transcription factors, repressor element 1 (RE1)-silencing transcription factor (REST) and c-AMP responsive element-binding protein (CREB). Promoter assays, chromatin immunoprecipitation, and transfection experiments revealed that ND1 activated NCAM1, that ASCL1 weakly upregulated SYPT expression, and that CGA expression was not regulated by ND1 or ASCL1. REST expression was restricted in non-small cell lung cancer (NSCLC) cells, and knockdown of REST could cause as much SYPT expression as in SCLC cells and weak CGA expression in NSCLC cells. However, CGA gene upregulation via CREB activation was not found in REST-lacking NSCLC cells, indicating the requirement of some additional mechanism for sufficient expression. These results suggest that NCAM1, SYPT and CGA expressions are differently regulated by neuroendocrine phenotype-specific transcription factors and provide a reason why NCAM1 and SYPT are frequently expressed in LNETs, irrespective of malignancy grade.


Subject(s)
CD56 Antigen/genetics , Carcinoma, Neuroendocrine/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Chromogranin A/genetics , Lung Neoplasms/genetics , Vesicular Transport Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD56 Antigen/metabolism , Carcinoma, Neuroendocrine/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Lung Neoplasms/pathology , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Synaptophysin , Transfection
12.
Exp Ther Med ; 2(4): 719-724, 2011 Jul.
Article in English | MEDLINE | ID: mdl-22977565

ABSTRACT

Malignant mesothelioma (MM) is an aggressive cancer with no effective treatment options. Enforced expression of the gap junction (GJ) component connexin 43 (Cx43) increases the sensitivity of MM cells to cisplatin. Bowman-Birk protease inhibitor (BBI) induces the restoration of Cx43 in several types of tumor cells. In this study, we examined the capability of BBI to enhance the cytotoxic effect of cisplatin in MM cells via the induction of Cx43. Human MM H28 cells were used. Cell viability was evaluated by a WST-1 assay and proteasomal activity was determined by fluorometric analysis. Protein and mRNA levels were determined by immunoblot analysis and real-time RT-PCR, respectively. GJ function mediated by Cx43 was evaluated using the scrape-loading method. BBI effectively inhibited H28 cell growth in a dose-dependent manner (200-400 µg/ml). In parallel with the growth inhibition, Cx43 levels (mRNA and protein) and GJ function were elevated by BBI treatment. Knockdown of BBI-induced Cx43 by an antisense nucleotide treatment almost cancelled the growth inhibition. BBI enhanced cisplatin-induced cytotoxicity in H28 cells, and down-regulation of Cx43 by the antisense nucleotide treatment abrogated the enhancing effect of BBI. The induction of Cx43 by BBI contributed to Src inactivation and subsequent induction of Bax. Furthermore, an Src inhibitor (SU6656) also enhanced cisplatin-induced cytotoxicity in H28 cells. These results suggest that BBI improves the cytotoxic efficacy of cisplatin in H28 cells via the inhibition of Src signaling.

13.
Pathol Int ; 61(1): 19-27, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21166939

ABSTRACT

Gene silencing by promoter hypermethylation plays an important role in molecular pathogenesis. We previously reported that insulin-like growth factor (IGF) binding protein-4 (IGFBP-4), which inhibits IGF-dependent growth, is expressed via early growth response-1 (EGR-1) and is often silenced in cultivated lung cancer cells. The purpose of the present study was to clarify clinicopathological factors associated with IGFBP-4 gene silencing in lung adenocarcinomas. Seventy-six surgically resected adenocarcinomas (20 well-, 35 moderately-, and 21 poorly-differentiated) were subjected to methylation-specific polymerase chain reaction (PCR) analysis for EGR-1-binding sites located in the IGFBP-4 promoter and immunohistochemistry for IGFBP-4, EGR-1, and Ki-67. Thirty-two adenocarcinomas (42%) revealed IGFBP-4 promoter hypermethylation, and the severity inversely correlated with the level of IGFBP-4 expression (P < 0.0001) and tumor differentiation (well versus poor, P = 0.0278; well/moderate versus poor, P = 0.0395). Furthermore, there was a negative correlation between Ki-67 labeling index and IGFBP-4 expression (P = 0.0361). These findings suggest that the expression of IGFBP-4 in adenocarcinoma cells in vivo is downregulated by epigenetic silencing in association with tumor differentiation, resulting in disruption of the mechanism of IGFBP-4-mediated growth inhibition.


Subject(s)
Adenocarcinoma/genetics , Gene Silencing , Insulin-Like Growth Factor Binding Protein 4/genetics , Lung Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Binding Sites , DNA Methylation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Humans , Immunohistochemistry , Insulin-Like Growth Factor Binding Protein 4/metabolism , Ki-67 Antigen/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Polymerase Chain Reaction , Promoter Regions, Genetic
14.
Am J Pathol ; 177(1): 70-83, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20489156

ABSTRACT

Vascular endothelial growth factor-A (VEGF-A) is crucial for angiogenesis, vascular permeability, and metastasis during tumor development. We demonstrate here that early growth response-1 (EGR-1), which is induced by the extracellular signal-regulated kinase (ERK) pathway activation, activates VEGF-A in lung cancer cells. Increased EGR-1 expression was found in adenocarcinoma cells carrying mutant K-RAS or EGFR genes. Hypoxic culture, siRNA experiment, luciferase assays, chromatin immunoprecipitation, electrophoretic mobility shift assays, and quantitative RT-PCR using EGR-1-inducible lung cancer cells demonstrated that EGR-1 binds to the proximal region of the VEGF-A promoter, activates VEGF-A expression, and enhances hypoxia inducible factor 1alpha (HIF-1alpha)-mediated VEGF-A expression. The EGR-1 modulator, NAB-2, was rapidly induced by increased levels of EGR-1. Pathology samples of human lung adenocarcinomas revealed correlations between EGR-1/HIF-1alpha and VEGF-A expressions and relative elevation of EGR-1 and VEGF-A expression in mutant K-RAS- or EGFR-carrying adenocarcinomas. Both EGR-1 and VEGF-A expression increased as tumors dedifferentiated, whereas HIF-1alpha expression did not. Although weak correlation was found between EGR-1 and NAB-2 expressions on the whole, NAB-2 expression decreased as tumors dedifferentiated, and inhibition of DNA methyltransferase/histone deacetylase increased NAB-2 expression in lung cancer cells despite no epigenetic alteration in the NAB-2 promoter. These findings suggest that EGR-1 plays important roles on VEGF-A expression in lung cancer cells, and epigenetic silencing of transactivator(s) associated with NAB-2 expression might also contribute to upregulate VEGF-A expression.


Subject(s)
Early Growth Response Protein 1/metabolism , Lung Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Decitabine , Early Growth Response Protein 1/genetics , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Genes, ras , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Promoter Regions, Genetic , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , Vascular Endothelial Growth Factor A/genetics
15.
Am J Pathol ; 175(3): 976-87, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19679880

ABSTRACT

Small cell lung cancer (SCLC) exhibits insulin-like growth factor-dependent growth. SCLC is the most aggressive among known in vivo lung cancers, whereas in vitro growth of SCLC is paradoxically slow as compared with that of non-SCLC (NSCLC). In this study, we demonstrate that SCLC cells overexpress insulin-like growth factor binding protein (IGFBP)-2 via NeuroD, a neuroendocrine cell-specific transcription factor. Chromatin immunoprecipitation, electrophoretic mobility shift, and IGFBP-2 promoter assays all revealed that NeuroD binds to the E-box in the 5'-untranslated region of IGFBP-2. A NeuroD transgene in both airway epithelial and NSCLC cells up-regulated the transcription of IGFBP-2 and retarded cell growth. Recombinant IGFBP-2 repressed the growth of both airway epithelial and NSCLC cells in a dose-dependent manner. A NeuroD-specific small interfering RNA repressed IGFBP-2 expression in SCLC, and neutralization of IGFBP-2 and an IGFBP-2-specific small interfering RNA increased SCLC cell growth. Pathological samples of SCLC also expressed IGFBP-2 abundantly, as compared with NSCLC, and showed only rare (8%) IGFBP-2 promoter methylation, whereas the IGFBP-2 promoter was methylated in 71% of adenocarcinomas and 29% of squamous cell carcinomas. These findings suggest that 1) SCLC has an IGFBP-2 overexpression mechanism distinct from NSCLC, 2) secreted IGFBP-2 contributes to the slow growth of SCLC in vitro, and 3) the epigenetic alterations in the IGFBP-2 promoter contribute to the striking differences in IGFBP-2 expression between SCLC and NSCLC in vivo.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Insulin-Like Growth Factor Binding Protein 2/metabolism , Lung Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Small Cell Lung Carcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Transformed , Cell Line, Tumor , DNA Methylation , DNA, Neoplasm/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/pharmacology , Lung/metabolism , Lung/pathology , Lung Neoplasms/pathology , Neurosecretory Systems/metabolism , Neurosecretory Systems/pathology , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Small Cell Lung Carcinoma/pathology , Transfection , Up-Regulation
16.
Life Sci ; 84(19-20): 650-6, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19232361

ABSTRACT

AIMS: Malignant mesothelioma is an aggressive cancer with no effective treatment options. A redox-silent analogue of alpha-tocotrienol, 6-O-carboxypropyl-alpha-tocotrienol (T3E) is a new potential anti-carcinogenic agent with less toxic effect on non-tumorigenic cells. Here, we evaluated the effect of T3E on killing of chemoresistant mesothelioma cell (H28). MAIN METHODS: The cytotoxic effect of T3E was evaluated by a WST-1 assay, and cell cycle and apoptosis analysis were done by FACS. Each signal molecule's activity was determined by protein array and immunoblot analysis. KEY FINDINGS: T3E effectively inhibited H28 cell growth at practical pharmacological concentrations (10-20 muM) without any effect on non-tumorigenic mesothelial cell (Met-5A). Inhibition of H28 cell growth by T3E mediated through G2/M arrest in cell cycle and induction of apoptosis. Protein array and immunoblot analyses revealed that T3E inhibited the activation of epidermal growth factor receptor (EGFR) via the inactivation of the Src family of protein tyrosine kinases (Src). However, the blockade of the EGFR signaling was not associated with the T3E-dependent H28 cell growth control. In addition to Src inactivation, T3E inhibited signal transduction and activation of transcription Stat3. A combination of an Src inhibitor, PP2, and a Stat3 inhibitor, AG490, induced G2/M arrest and enhanced apoptosis compared with PP2 alone. These results suggest that T3E suppresses H28 cell growth via the inhibition of Src activation and Src-independent Stat3 activation. SIGNIFICANCE: T3E can be a new effective therapeutic agent against chemoresistant mesothelioma cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Antioxidants/therapeutic use , Mesothelioma/drug therapy , Tocotrienols/chemistry , Antineoplastic Agents/chemistry , Antioxidants/chemistry , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Enzyme Activation , Humans , Mesothelioma/pathology , Oxidation-Reduction , Protein Array Analysis , STAT3 Transcription Factor/metabolism , Tocotrienols/pharmacology , Tocotrienols/therapeutic use , src-Family Kinases/metabolism
17.
Biochem Biophys Res Commun ; 365(4): 875-81, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-18042466

ABSTRACT

We have previously reported that a redox-silent analogue of alpha-tocotrienol (T3), 6-O-carboxypropyl-alpha-tocotrienol (T3E) shows more potential anti-carcinogenic property than T3 in a lung cancer cell (A549 cell). However, the mechanisms by which T3E exerts its potential anti-carcinogenic effect is still unclear. As tumor malignancy is associated with hypoxia adaptation, in this study, we examined whether T3E could suppress survival and invasion in A549 cells under hypoxia. Hypoxia treatment drastically-induced activation of the protein tyrosine kinase, Src, and its regulated signaling required for hypoxia adaptation of A549 tumor cells. The survival and invasion capacity of the tumor cells under hypoxia was suppressed by T3E via the inactivation of Src. More specifically, T3E-dependent inhibition of Src-induced Akt activation contributed to suppression of cell survival under hypoxia, and the reduction of fibrinolytic factors such as plasminogen activator-1(PAI-1) via the decrease of hypoxia-inducible factor-2alpha by T3E led to inhibition of hypoxic invasion. Overall these results suggest that T3E suppresses hypoxia adaptation of A549 cells by the inhibition in hypoxia-induced activation of Src signaling.


Subject(s)
Cell Survival/drug effects , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oxygen/metabolism , Tocotrienols/administration & dosage , Adaptation, Physiological/drug effects , Cell Hypoxia/drug effects , Cell Line, Tumor , Humans , Oxidation-Reduction/drug effects
18.
Cancer Lett ; 253(2): 249-57, 2007 Aug 18.
Article in English | MEDLINE | ID: mdl-17343982

ABSTRACT

Bowman-Birk protease inhibitor (BBI) from soybean acts as a potential chemopreventive agent in several types of tumors. However, the mechanism is still unclear. The present study was undertaken to estimate a mechanism of BBI-dependent negative growth control of human osteosarcoma cell (U2OS cell). BBI had negative growth control of the cells via induction of connexin (Cx) 43, a tumor suppressor gene in U2OS cells. This negative growth control by BBI was abrogated under down-regulation of Cx43 induced by a Cx43 antisense nucleotide treatment. It was also found that the BBI-dependent induction of Cx43 was due to elevation of Cx43 mRNA and stabilization of Cx43 protein. Especially, BBI-dependent inhibition of chymotrypsin-like activity in proteasome contributed to stabilization of Cx43 protein. These results suggest that a major negative growth effect of BBI is based on the restoration of Cx43 expression in U2OS cells.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/metabolism , Connexin 43/metabolism , Osteosarcoma/metabolism , Protease Inhibitors/pharmacology , Trypsin Inhibitor, Bowman-Birk Soybean/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Connexin 43/genetics , Humans , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL