Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
3.
Oncogene ; 36(39): 5522-5531, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28553954

ABSTRACT

Malignant mesothelioma (MM) is an aggressive tumor commonly caused by asbestos exposure after a long latency. Focal adhesion kinase (FAK) inhibitors inhibit the cell growth of Merlin-deficient MM cells; however, their clinical efficacy has not been clearly determined. The aim of this study was to evaluate the growth inhibitory effect of the FAK inhibitor VS-4718 on MM cell lines and identify biomarkers for its efficacy. Although most Merlin-deficient cell lines were sensitive to VS-4718 compared with control MeT-5A cells, a subset of these cell lines exhibited resistance to this drug. Microarray and qRT-PCR analyses using RNA isolated from Merlin-deficient MM cell lines revealed a significant correlation between E-cadherin mRNA levels and VS-4718 resistance. Merlin- and E-cadherin-negative Y-MESO-22 cells underwent apoptosis upon treatment with a low concentration of VS-4718, whereas Merlin-negative, E-cadherin-positive Y-MESO-9 cells did not undergo VS-4718-induced apoptosis. Furthermore, E-cadherin knockdown in Merlin-negative MM cells significantly sensitized cells to VS-4718 and induced apoptotic cell death upon VS-4718 treatment. Together, our results suggest that E-cadherin serves as a predictive biomarker for molecular target therapy with FAK inhibitors for patients with mesothelioma and that its expression endows MM cells with resistance to FAK inhibitors.


Subject(s)
Cadherins/biosynthesis , Focal Adhesion Kinase 1/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mesothelioma/drug therapy , Mesothelioma/metabolism , Neurofibromin 2/deficiency , Protein Kinase Inhibitors/pharmacology , Antigens, CD , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Gene Expression , Humans , Lung Neoplasms/pathology , Mesothelioma/pathology , Mesothelioma, Malignant , Neurofibromin 2/metabolism , Signal Transduction
4.
Oncogene ; 34(1): 73-83, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-24336325

ABSTRACT

Malignant mesothelioma (MM) is one of the most aggressive neoplasms usually associated with asbestos exposure and is highly refractory to current therapeutic modalities. MMs show frequent activation of a transcriptional coactivator Yes-associated protein (YAP), which is attributed to the neurofibromatosis type 2 (NF2)-Hippo pathway dysfunction, leading to deregulated cell proliferation and acquisition of a malignant phenotype. However, the whole mechanism of disordered YAP activation in MMs has not yet been well clarified. In the present study, we investigated various components of the NF2-Hippo pathway, and eventually found that MM cells frequently showed downregulation of LIM-domain protein AJUBA, a binding partner of large tumor suppressor type 2 (LATS2), which is one of the last-step kinases of the NF2-Hippo pathway. Although loss of AJUBA expression was independent of the alteration status of other Hippo pathway components, MM cell lines with AJUBA inactivation showed a more dephosphorylated (activated) level of YAP. Immunohistochemical analysis showed frequent downregulation of AJUBA in primary MMs, which was associated with YAP constitutive activation. We found that AJUBA transduction into MM cells significantly suppressed promoter activities of YAP-target genes, and the suppression of YAP activity by AJUBA was remarkably canceled by knockdown of LATS2. In connection with these results, transduction of AJUBA-expressing lentivirus significantly inhibited the proliferation and anchorage-independent growth of the MM cells that harbored ordinary LATS family expression. Taken together, our findings indicate that AJUBA negatively regulates YAP activity through the LATS family, and inactivation of AJUBA is a novel key mechanism in MM cell proliferation.


Subject(s)
Gene Expression Regulation, Neoplastic , LIM Domain Proteins/metabolism , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Cytoplasm/metabolism , Hippo Signaling Pathway , Humans , Immunohistochemistry , Lentivirus/genetics , Mesothelioma, Malignant , Neurofibromin 2/metabolism , Phenotype , Phosphoproteins/metabolism , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factors , YAP-Signaling Proteins
5.
Oncogene ; 34(13): 1629-40, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-24769899

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly invasive cancer with a poor prognosis. Although microRNA (miRNA) transcripts have a crucial role in carcinogenesis and development, little information is known regarding the aberrant DNA methylation of miRNAs in PDAC. Using methylated DNA immunoprecipitation-chip analysis, we found that miR-615-5p was hypermethylated in its putative promoter region, which silenced its expression in PDAC cell lines. In addition, the overexpression of miR-615-5p in pancreatic cancer cells suppressed cell proliferation, migration and invasion. Insulin-like growth factor 2 (IGF2) is an imprinted gene, and its abnormal expression contributes to tumor growth. Here, we identified IGF2 as a target of miR-615-5p using a luciferase reporter assay. IGF2 upregulation in PDAC tissues was not correlated with a loss of imprinting but was inversely correlated with miR-615-5p downregulation. In addition, miR-615-5p suppressed pancreatic cancer cell proliferation, migration and invasion by directly targeting IGF2, and this effect could be reversed by co-transfection with IGF2. Furthermore, the stable overexpression of miR-615-5p inhibited tumor growth in vivo and was correlated with IGF2 expression. Using RNA sequencing, we further identified miR-615-5p as potentially targeting other genes, such as the proto-oncogene JUNB, and interfering with the insulin signaling pathway. Taken together, our results demonstrate that miR-615-5p was abnormally downregulated in PDAC cells due to promoter hypermethylation, which limited its inhibition of IGF2 and other target genes, thereby contributing to tumor growth, invasion and migration. These data demonstrate a novel and important role of miR-615-5p as a tumor suppressor in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , DNA Methylation , Epigenesis, Genetic , Genes, Tumor Suppressor , MicroRNAs/physiology , Pancreatic Neoplasms/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Insulin-Like Growth Factor II/genetics , Neoplasm Invasiveness , Pancreatic Neoplasms/pathology , Proto-Oncogene Mas , Sequence Analysis, RNA
6.
Oncogene ; 32(13): 1724-34, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-22614011

ABSTRACT

Hepatocellular carcinoma (HCC) typically relies on angiogenesis for its malignant behavior, including growth and metastasis. Vasohibin 2 (VASH2) was previously identified as an angiogenic factor, but its role in tumorigenesis is unknown. Using quantitative PCR and western blot analyses, we found that VASH2 is overexpressed in HCC cells and tissues. Using chromatin immunoprecipitation, we detected histone modifications at the putative VASH2 promoter, with increased H3K4 trimethylation and H3 acetylation and decreased H3K27 trimethylation, suggesting that epigenetic mechanisms are responsible for the deregulated VASH2 transcription in HCC. Knockdown of VASH2 via siRNA inhibited the proliferation of the hepatoma cell lines by delaying cell cycle progression and increasing apoptosis. Importantly, we found VASH2 secreted in the culture supernatant, and co-expression of its secretory chaperone small vasohibin-binding protein (SVBP) further enhanced VASH2 secretion. The supernatant from HepG2 cells expressing VASH2 enhanced the proliferation, migration and tube formation of human umbilical vein endothelial cells, and knockdown of VASH2 significantly inhibited these effects. In an in vivo study using a nude mouse model, we found that exogenous VASH2 significantly contributed to tumor growth, microvessel density and hemoglobin concentration in the tumors. Further analyses showed that the VASH2-mediated increase in the transcription of fibroblast growth factor-2, vascular endothelial growth factor and vasohibin 1 may be the mechanism underlying these effects. Taken together, these data indicate that VASH2 is abnormally expressed in HCC cells as a result of histone modifications and that VASH2 contributes to the angiogenesis in HCC via an SVBP-mediated paracrine mechanism. These results indicate a novel and important role for VASH2 in HCC angiogenesis and malignant transformation.


Subject(s)
Angiogenic Proteins/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Neovascularization, Pathologic/genetics , Angiogenic Proteins/antagonists & inhibitors , Angiogenic Proteins/metabolism , Angiogenic Proteins/physiology , Animals , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cells, Cultured , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Small Interfering/pharmacology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Xenograft Model Antitumor Assays
7.
Oncogene ; 32(37): 4427-35, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23045273

ABSTRACT

Despite initial dramatic response, epidermal growth factor receptor (EGFR) mutant lung cancer patients always acquire resistance to EGFR-tyrosine kinase inhibitors (TKIs). Gatekeeper T790M mutation in EGFR is the most prevalent genetic alteration underlying acquired resistance to EGFR-TKI, and EGFR mutant lung cancer cells are reported to be addictive to EGFR/Akt signaling even after acquired T790M mutation. Here, we focused on Akt kinase-interacting protein1 (Aki1), a scaffold protein of PI3K (phosphoinositide 3-kinase)/PDK1 (3-phosphoinositide-dependent protein kinase)/Akt that determines receptor signal selectivity for non-mutated EGFR, and assessed its role in EGFR mutant lung cancer with or without gatekeeper T790M mutation. Cell line-based assays showed that Aki1 constitutively associates with mutant EGFR in lung cancer cells with (H1975) or without (PC-9 and HCC827) T790M gatekeeper mutation. Silencing of Aki1 induced apoptosis of EGFR mutant lung cancer cells. Treatment with Aki1 siRNA dramatically inhibited growth of H1975 cells in a xenograft model. Moreover, silencing of Aki1 further potentiated growth inhibitory effect of new generation EGFR-TKIs against H1975 cells in vitro. Aki1 was frequently expressed in tumor cells of EGFR mutant lung cancer patients (53/56 cases), including those with acquired resistance to EGFR-TKI treatment (7/7 cases). Our data suggest that Aki1 may be a critical mediator of survival signaling from mutant EGFR to Akt, and may therefore be an ideal target for EGFR mutant lung cancer patients, especially those with acquired EGFR-TKI resistance due to EGFR T790M gatekeeper mutation.


Subject(s)
DNA-Binding Proteins/metabolism , ErbB Receptors/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Disease Models, Animal , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gene Expression , Gene Knockdown Techniques , Humans , Ligands , Lung Neoplasms/pathology , Mice , Protein Binding , Protein Kinase Inhibitors/pharmacology , Transplantation, Heterologous
8.
Oncogene ; 31(49): 5117-22, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-22286761

ABSTRACT

Malignant mesothelioma (MM) shows frequent inactivation of the neurofibromatosis type 2 (NF2) -tumor-suppressor gene. Recent studies have documented that the Hippo signaling pathway, a downstream cascade of Merlin (a product of NF2), has a key role in organ size control and carcinogenesis by regulating cell proliferation and apoptosis. We previously reported that MMs show overexpression of Yes-associated protein (YAP) transcriptional coactivator, the main downstream effector of the Hippo signaling pathway, which results from the inactivation of NF2, LATS2 and/or SAV1 genes (the latter two encoding core components of the mammalian Hippo pathway) or amplification of YAP itself. However, the detailed roles of YAP remain unclear, especially the target genes of YAP that enhance MM cell growth and survival. Here, we demonstrated that YAP-knockdown inhibited cell motility, invasion and anchorage-independent growth as well as cell proliferation of MM cells in vitro. We analyzed genes commonly regulated by YAP in three MM cell lines with constitutive YAP-activation, and found that the major subsets of YAP-upregulating genes encode cell cycle regulators. Among them, YAP directly induced the transcription of CCND1 and FOXM1, in cooperation with TEAD transcription factor. We also found that knockdown of CCND1 and FOXM1 suppressed MM cell proliferation, although the inhibitory effects were less evident than those of YAP knockdown. These results indicate that constitutive YAP activation in MM cells promotes cell cycle progression giving more aggressive phenotypes to MM cells.


Subject(s)
Cell Cycle/genetics , Mesothelioma/genetics , Mesothelioma/pathology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cyclin D1/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Nuclear Proteins/genetics , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcriptome , Up-Regulation
9.
Oncogene ; 29(13): 1941-51, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20101228

ABSTRACT

Malignant pleural mesothelioma (MPM) is resistant to chemotherapy and thus shows a dismal prognosis. Osteopontin (OPN), a secreted noncollagenous and phosphoprotein, is suggested to be involved in the pathogenesis of MPM. However, the precise role of OPN, especially in the multidrug resistance of MPM, remains to be elucidated. We therefore established stable transfectants (ACC-MESO-1/OPN), which constitutively express OPN, to determine its role in the chemoresistance observed in MPM. The introduction of the OPN gene provides MPM cells with upregulated multidrug resistance through the mechanism of enhanced hyaluronate (HA) binding. The expression of CD44 variant isoforms, which inhibit HA binding, significantly decreased in ACC-MESO-1/OPN cells in comparison to control transfectants. Interestingly, the inhibition of the HA-CD44 interaction abrogated multidrug resistance in the ACC-MESO-1/OPN, thus suggesting the involvement of the surviving signal emanating from the HA-CD44 interaction. An enhanced level of the p-Akt in ACC-MESO-1/OPN cells was observed, and was diminished by CD44 siRNA. Inhibition of the Akt phosphorylation increased in number of the cells underwent apoptosis induced by NVB, VP-16 and GEM. Collectively, these results indicate that OPN is strongly involved in multidrug resistance by enhancing the CD44 binding to HA.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Hyaluronic Acid/pharmacology , Mesothelioma/pathology , Osteopontin/metabolism , Pleural Neoplasms/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronic Acid/metabolism , Mesothelioma/metabolism , Pleural Neoplasms/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
10.
Oncogene ; 26(27): 4025-31, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17213806

ABSTRACT

We previously established a highly metastatic subline, LNM35, from the NCI-H460 lung cancer cell line, and demonstrated upregulation of a novel gene, CLCP1 (CUB, LCCL-homology, coagulation factor V/VIII homology domains protein), in LNM35 and lung cancer specimens. In this study, we focused on the potential roles of that gene in cancer metastasis. First, we established stable LNM35 RNAi clones, in which CLCP1 expression was suppressed by RNAi, and found that their motility was significantly reduced, although growth rates were not changed. Next, in vitro selection of a phage display library demonstrated that a phage clone displaying a peptide similar to a sequence within the Sema domain of semaphorin 4B (SEMA4B) interacted with LNM35. Immunoprecipitation experiments confirmed interaction of CLCP1 with SEMA4B, regulation of CLCP1 protein by ubiquitination and proteasome degradation enhanced in the presence of SEMA4B. These results are the first to indicate that CLCP1 plays a role in cell motility, whereas they also showed that at least one of its ligands is SEMA4B and that their interaction mediates proteasome degradation by CLCP1. Although the physiological role of the interaction between CLCP1 and SEMA4B remains to be investigated, this novel gene may become a target of therapy to inhibit metastasis of lung cancers.


Subject(s)
Cell Movement/physiology , Membrane Proteins/physiology , Semaphorins/metabolism , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cysteine Proteinase Inhibitors/pharmacology , Humans , Immunoblotting , Immunoprecipitation , Leupeptins/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Oligopeptides/genetics , Oligopeptides/metabolism , Peptide Library , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Binding/drug effects , RNA Interference , Semaphorins/genetics , Transfection , Tunicamycin/pharmacology , Ubiquitin/metabolism
11.
Diabet Med ; 23(8): 894-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16911628

ABSTRACT

AIMS: The aldose reductase (AR) gene, a rate-limiting enzyme of the polyol pathway, has been investigated as a candidate gene in determining susceptibility to diabetic microangiopathy. However, the association of the AR gene with diabetic macroangiopathy has not been investigated. Therefore, the present study was conducted to determine whether genetic variations of AR may determine susceptibility to diabetic macroangiopathy. METHODS: There were 378 Type 2 diabetic patients enrolled in this study. A single nucleotide polymorphism in the promoter region (C-106T) was genotyped and the AR protein content of erythrocytes measured by ELISA. RESULTS: There were no significant differences in genotypic or allelic distribution in patients with or without ischaemic heart diseases, but there was a significant increase in the frequency of the CT + TT genotype and T allele in patients with stroke (P = 0.019 and P = 0.012). The erythrocyte AR protein content was increased in patients with the CT and TT genotype compared with those with the CC genotype. After adjustment for age, duration of diabetes, body mass index, systolic blood pressure, HbA1c, and serum creatinine, triglycerides, and total cholesterol in multivariate logistic-regression models, the association between this AR genotype and stroke remained significant. CONCLUSIONS: Our results suggest that the CT or TT genotype of the AR gene might be a genetic marker of susceptibility to stroke in Type 2 diabetic patients. This observation might contribute to the development of strategies for the prevention of stroke in Type 2 diabetic patients.


Subject(s)
Aldehyde Reductase/genetics , Asian People/genetics , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Genetic/genetics , Adult , Aged , Aged, 80 and over , Genetic Predisposition to Disease/ethnology , Humans , Male , Middle Aged
12.
Kyobu Geka ; 58(4): 337-40, 2005 Apr.
Article in Japanese | MEDLINE | ID: mdl-15828258

ABSTRACT

We report a case of a 27-year-old woman with primary neurogenous sarcoma of the lung. She had no symptoms but an abnormal shadow of the right lower lung field on the chest X-ray. Chest computed tomography (CT) revealed a well defined round mass, 20 mm in maximum diameter, at the right S9. Pathological study of the specimen obtained by CT-guided percutaneous needle biopsy showed undefferentiated carcinoma. Positron emission tomography (PET) disclosed intensely increased uptake of fluoro-2-deoxy-D-glucose (FDG) at the lung lesion without other abnormal uptakes. The patient underwent right lower lobectomy of the lung and mediastinal lymph nodes dissection. Results from immunohistological study yielded a definitive diagnosis of neurogenous sarcoma. Postoperative course was uneventful, and there has been no evidence of recurrence and metastasis for more than a year after the surgery. Reported cases of primary neurogenous sarcoma of the lung are reviewed.


Subject(s)
Lung Neoplasms/surgery , Neurofibrosarcoma/surgery , Pneumonectomy , Adult , Female , Humans , Lung Neoplasms/diagnostic imaging , Lymph Node Excision , Neurofibrosarcoma/diagnostic imaging , Pneumonectomy/methods , Tomography, X-Ray Computed
13.
Thorax ; 58(10): 892-900, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14514947

ABSTRACT

It has been hypothesised that clinically evident lung cancers have accumulated many different genetic or epigenetic abnormalities in oncogenes and/or tumour suppressor genes. This notion has important clinical ramifications. Recent developments in our knowledge of the molecular biology of lung cancer are reviewed, with particular reference to genetic abnormalities in tumour suppressor gene inactivation and overactivity of growth promoting oncogenes. These changes lead to the "hallmarks of lung cancer". These hallmarks are the new rational targets for early detection, prevention, and treatment of lung cancer.


Subject(s)
Lung Neoplasms/genetics , Apoptosis/genetics , DNA Methylation , Genes, Tumor Suppressor , Growth Substances/genetics , Humans , Molecular Biology , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neovascularization, Pathologic/genetics , Proto-Oncogenes/genetics , Telomerase/genetics
14.
Proc Natl Acad Sci U S A ; 98(24): 13954-9, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11717452

ABSTRACT

Semaphorins SEMA3B and its homologue SEMA3F are 3p21.3 candidate tumor suppressor genes (TSGs), the expression of which is frequently lost in lung cancers. To test the TSG candidacy of SEMA3B and SEMA3F, we transfected them into lung cancer NCI-H1299 cells, which do not express either gene. Colony formation of H1299 cells was reduced 90% after transfection with wild-type SEMA3B compared with the control vector. By contrast, only 30-40% reduction in colony formation was seen after the transfection of SEMA3F or SEMA3B variants carrying lung cancer-associated single amino acid missense mutations. H1299 cells transfected with wild-type but not mutant SEMA3B underwent apoptosis. We found that lung cancers (n = 34) always express the neuropilin-1 receptor for secreted semaphorins, whereas 82% expressed the neuropilin-2 receptor. Because SEMA3B and SEMA3F are secreted proteins, we tested conditioned medium from COS-7 cells transfected with SEMA3B and SEMA3F and found that medium from wild-type SEMA3B transfectants reduced the growth of several lung cancer lines 30-90%, whereas SEMA3B mutants or SEMA3F had little effect in the same assay. Sequencing of sodium bisulfite-treated DNA showed dense methylation of CpG sites in the SEMA3B 5' region of lung cancers not expressing SEMA3B but no methylation in SEMA3B-expressing tumors. These results are consistent with SEMA3B functioning as a TSG, the expression of which is inactivated frequently in lung cancers by allele loss and promoter region methylation.


Subject(s)
Apoptosis , Chromosomes, Human, Pair 3 , Genes, Tumor Suppressor/physiology , Lung Neoplasms/physiopathology , Membrane Glycoproteins/genetics , Animals , Base Sequence , COS Cells , Cell Division , Chlorocebus aethiops , CpG Islands , Culture Media, Conditioned , DNA Methylation , DNA, Complementary , Gene Expression , Humans , Molecular Sequence Data , Mutagenesis , Nerve Tissue Proteins/biosynthesis , Neuropilin-1 , Semaphorins , Transfection , Tumor Cells, Cultured
15.
Oncogene ; 20(43): 6258-62, 2001 Sep 27.
Article in English | MEDLINE | ID: mdl-11593436

ABSTRACT

Recently we identified FUS1 as a candidate tumor suppressor gene (TSG) in the 120 kb 3p21.3 critical region contained in nested lung and breast cancer homozygous deletions. Mutation of FUS1 is infrequent in lung cancers which we have confirmed in 40 other primary lung cancers. In addition, we found no evidence for FUS1 promoter region methylation. Because haploinsufficiency or low expression of Fus1 may play a role in lung tumorigenesis, we tested the effect of exogenously induced overexpression of Fus1 protein and found 60-80% inhibition of colony formation for non-small cell lung cancer lines NCI-H1299 (showing allele loss for FUS1) and NCI-H322 (containing only a mutated FUS1 allele) in vitro. By contrast, a similar level of expression of a tumor-acquired mutant form of FUS1 protein did not significantly suppress colony formation. Also, induced expression of Fus1 under the control of an Ecdysone regulated promoter decreased colony formation 75%, increased the doubling time twofold, and arrested H1299 cells in G1. In conclusion, our data are consistent with the hypothesis that FUS1 may function as a 3p21.3 TSG, warranting further studies of its function in the pathogenesis of human cancers.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 3 , Fungal Proteins/metabolism , Homozygote , Lung Neoplasms/genetics , Alleles , Blotting, Western , Cell Division , DNA Methylation , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Models, Genetic , Mutation , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Tumor Cells, Cultured
16.
Prenat Diagn ; 21(8): 634-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11536261

ABSTRACT

We report a Japanese boy who died at Day 28 of life because of severe carbamoyl phosphate synthetase I (CPS1) deficiency that was proven by enzyme assay. By analysis of cDNA and genomic DNA, he was shown to be a compound heterozygote with two point mutations of the CPS1 gene, 840G>C leading to an aberrant splicing and 1123C>T (predicting Q375X). The 840G>C was a mutation described in another Japanese family. Since his parents carried each mutation heterozygously, we performed prenatal diagnosis at 16 weeks of his mother's next gestation by multiplex PCR and melting curve analysis in a single capillary containing two-color fluorescent (LC-Red 640 and LC-Red 705) probes on LightCycler. We analyzed genomic DNA extracted from amniotic cells and found that the fetus was homozygous for the wild-type alleles. At term a healthy girl was born without hyperammonemia.


Subject(s)
Carbamoyl-Phosphate Synthase I Deficiency Disease/diagnosis , Membrane Proteins/genetics , Schizosaccharomyces pombe Proteins , Carbamoyl-Phosphate Synthase I Deficiency Disease/genetics , DNA Primers , Diagnosis, Differential , Female , Glucosyltransferases , Humans , Infant, Newborn , Male , Polymerase Chain Reaction , Pregnancy , Pregnancy Trimester, Second , Prenatal Diagnosis
17.
Cancer Gene Ther ; 8(6): 421-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11498762

ABSTRACT

CD40-CD40 ligand (CD40L) interaction is an important costimulatory signaling pathway in the crosstalk between T cells and antigen-presenting cells. This receptor-ligand system is known to be essential in eliciting strong cellular immunity. Here we demonstrate that murine lung cancer cells (3LLSA) transduced with the CD40L gene (3LLSA-CD40L) were rejected in syngeneic C57BL/6 mice, but grew in CD40-deficient mice to the same extent as control tumor cells. Immunohistochemical study showed that inflammatory cells, including CD4+, CD8+ T cells and NK cells, infiltrated into the inoculated 3LLSA-CD40L tumor tissue. Inoculation of 3LLSA-CD40L cells into mice resulted in the induction of 3LLSA-specific cytotoxic T-cell immunity, and the growth of parental 3LLSA tumors was inhibited when 3LLSA cells were inoculated into C57BL/6 mice mixed with 3LLSA-CD40L cells or when they were rechallenged 4 weeks after 3LLSA-CD40L cells were rejected. Furthermore, co-inoculation of interferon (IFN)-gamma-transduced cells (3LLSA-IFNgamma) with 3LLSA-CD40L cells enhanced the antitumor immunity efficiently in vivo. These results indicate that the in vivo priming with CD40L- and IFN-gamma gene-transduced lung cancer cells is a promising strategy for inducing antitumor immunity in the treatment of lung cancer.


Subject(s)
CD40 Ligand/genetics , Cancer Vaccines , Genetic Therapy/methods , Interferon-gamma/genetics , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Transduction, Genetic , Animals , DNA, Complementary/metabolism , Immunohistochemistry , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Recombinant Proteins/metabolism , Spleen/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Time Factors , Transfection , Tumor Cells, Cultured
18.
Oncogene ; 20(31): 4249-57, 2001 Jul 12.
Article in English | MEDLINE | ID: mdl-11464291

ABSTRACT

The beta-catenin gene (CTNNB1) has been shown to be genetically mutated in various human malignancies. To determine whether the beta-catenin gene is responsible for oncogenesis in thoracic malignancies, we searched for the mutation in 166 lung cancers (90 primary tumors and 76 cell lines), one blastoma and 10 malignant mesotheliomas (two primary tumors and eight cell lines). Among the lung cancers, including 43 small cell lung cancers (SCLCs) and 123 non-small cell lung cancers (NSCLCs), we identified four alterations in exon 3, which is the target region of mutation for stabilizing beta-catenin. One primary adenocarcinoma had a somatic mutation from C to G, leading to an amino acid substitution from Ser to Cys at codon 37. Among the cell lines, SCLC NCI-H1092 had a mutation from A to G, leading to an Asp to Gly substitution at codon 6, NSCLC HCC15 had a mutation from C to T, leading to a Ser to Phe substitution at codon 45, and NSCLC NCI-H358 had a mutation from A to G, leading to a Thr to Ala substitution at codon 75. One blastoma also had a somatic mutation from C to G, leading to a Ser to Cys substitution at codon 37. Among the 10 malignant mesotheliomas, we identified a homozygous deletion in the NCI-H28 cell line. Cloning of the rearranged fragment from NCI-H28 indicated that all the exons except exon 1 of the beta-catenin gene are deleted and that the deletion junction is 13 kb downstream from exon 1. Furthermore, Northern blot analysis of 26 lung cancer and eight mesothelioma cell line RNAs detected ubiquitous expression of the beta-catenin messages except NCI-H28, although Western blot analysis showed that relatively less amounts of protein products were expressed in some of lung cancer cell lines. Our findings suggest that the beta-catenin gene is infrequently mutated in lung cancer and that the NCI-H28 homozygous deletion of the beta-catenin gene might indicate the possibility of a new tumor suppressor gene residing in this region at 3p21.3, where various types of human cancers show frequent allelic loss.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 3 , Cytoskeletal Proteins/genetics , Homozygote , Lung Neoplasms/genetics , Mesothelioma/genetics , Mutation , Trans-Activators , Base Sequence , DNA, Neoplasm , Exons , Gene Rearrangement , Humans , Molecular Sequence Data , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL