Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Mol Biol Evol ; 31(12): 3282-301, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25205508

ABSTRACT

The caspases, a family of cysteine proteases, play multiple roles in apoptosis, inflammation, and cellular differentiation. Caspase-8 (Casp8), which was first identified in humans, functions as an initiator caspase in the apoptotic signaling mediated by cell-surface death receptors. To understand the evolution of function in the Casp8 protein family, casp8 orthologs were identified from a comprehensive range of vertebrates and invertebrates, including sponges and cnidarians, and characterized at both the gene and protein levels. Some introns have been conserved from cnidarians to mammals, but both losses and gains have also occurred; a new intron arose during teleost evolution, whereas in the ascidian Ciona intestinalis, the casp8 gene is intronless and is organized in an operon with a neighboring gene. Casp8 activities are near ubiquitous throughout the animal kingdom. Exogenous expression of a representative range of nonmammalian Casp8 proteins in cultured mammalian cells induced cell death, implying that these proteins possess proapoptotic activity. The cnidarian Casp8 proteins differ considerably from their bilaterian counterparts in terms of amino acid residues in the catalytic pocket, but display the same substrate specificity as human CASP8, highlighting the complexity of spatial structural interactions involved in enzymatic activity. Finally, it was confirmed that the interaction with an adaptor molecule, Fas-associated death domain protein, is also evolutionarily ancient. Thus, despite structural diversity and cooption to a variety of new functions, the ancient origins and near ubiquitous distribution of this activity across the animal kingdom emphasize the importance and utility of Casp8 as a central component of the metazoan molecular toolkit.


Subject(s)
Apoptosis , Caspase 8/genetics , Amino Acid Sequence , Animals , Annelida/genetics , Anthozoa/genetics , Base Sequence , Caspase 8/chemistry , Ciona intestinalis/genetics , Evolution, Molecular , Fish Proteins/genetics , Fishes/genetics , HeLa Cells , Humans , Molecular Sequence Data , Mytilus/genetics , Phylogeny , Planarians/genetics , Protein Conformation , Substrate Specificity
2.
Genes Cells ; 17(11): 875-96, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23025414

ABSTRACT

FADD is an adaptor protein that transmits apoptotic signals from death receptors. Additionally, FADD has been shown to play a role in various functions including cell proliferation. However, the physiological role of FADD during embryonic development remains to be delineated. Here, we show the novel roles FADD plays in development and the molecular mechanisms of these roles in Xenopus embryos. By whole-mount in situ hybridization and RT-PCR analysis, we observed that fadd is constantly expressed in early embryos. The upregulation or downregulation of FADD proteins by embryonic manipulation resulted in induction of apoptosis or size changes in the heart during development. Expression of a truncated form of FADD, FADDdd, which lacks pro-apoptotic activity, caused growth retardation of embryos associated with dramatic expressional fluctuations of genes that are regulated by NF-κB. Moreover, we isolated a homolog of mammalian cullin-4 (Cul4), a component of the ubiquitin E3 ligase family, as a FADDdd-interacting molecule in Xenopus embryos. Thus, our study shows that FADD has multiple functions in embryos; it plays a part in the regulation of NF-κB activation and heart formation, in addition to apoptosis. Furthermore, our findings provide new insights into how Cul4-based ligase is related to FADD signaling in embryogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Antigens, Differentiation/physiology , Apoptosis , Fas-Associated Death Domain Protein/physiology , Heart/embryology , NF-kappa B/metabolism , Receptors, Immunologic/physiology , Xenopus Proteins/physiology , Xenopus/embryology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Blastomeres/enzymology , Blastomeres/metabolism , Cullin Proteins/chemistry , Cullin Proteins/genetics , Cullin Proteins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Morpholinos/genetics , NF-kappa B/physiology , Peptide Fragments/chemistry , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Sequence Analysis, DNA , Sequence Deletion , Signal Transduction , Transcriptional Activation , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
3.
Sci Rep ; 12(1): 12363, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35859006

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) is a critical process by which cancer cells acquire malignant features. However, the molecular mechanism and functional implications of EMT and the mesenchymal-to-epithelial transition (MET) in tumor progression remain elusive. In this study, we established two aggressive cancer cell lines from the human oral cancer cell line SAS, mesenchymal-like SAS-m4 and epithelial-like SAS-δ. SAS-δ is a revertant cell obtained by inducing MET in SAS-m4. SAS-δ, but not SAS-m4, exhibited abnormal cell growth, including piled-up overgrowth and invasive tumor formation in the tongues of nude mice, suggesting that SAS-δ represented more advanced cancer cells than the parental SAS cells. EMT-related transcriptional factor SLUG is phosphorylated at T208 and partly stabilized by the Hippo pathway kinases, LATS1 and LATS2. Depletion of SLUG promoted the invasive activity of SAS-δ by increasing the protein levels of LATS1/2 and the proportion of the phosphorylated form among total SLUG protein. Our results suggest that the LATS1/2-SLUG axis regulates the transition of SAS cells to the advanced stage via repeated switching between EMT and MET. Therefore, an anti-SLUG-pT208 antibody would be valuable not alone as a malignant tumor marker antibody but also as a prognostic tool for patients with malignant disease.


Subject(s)
Mouth Neoplasms , Protein Serine-Threonine Kinases , Snail Family Transcription Factors , Animals , Humans , Mice , Cell Line, Tumor , Mice, Nude , Mouth Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Tumor Suppressor Proteins
4.
Cell Struct Funct ; 36(2): 209-22, 2011.
Article in English | MEDLINE | ID: mdl-21979235

ABSTRACT

Generation of iPS cells from mouse embryonic fibroblasts (MEF) was achieved using a BacMam transduction system containing a polycistronic plasmid expression vector for coincident and optimized expression of four defined reprogramming transcription factors. The sequences for Oct4, Klf4, Sox2 and c-Myc, were cloned as a fusion gene (OKSM) in a single open reading frame (ORF) via self-cleaving 2A peptides and expressed under the control of the CAG promoter. The transduction efficiency of primary MEF cells with BacMam particles carrying CAG-directed Venus reporter gene is 64-98%. After three successive transductions (at intervals of 3 days) of MEF cells with BacMam particles carrying a OKSM or OSKM cassette, the iPS cell colonies are observed in 15-24 days. A single transduction of MEF cells is also effective in generating sufficiently reprogrammed iPS cell lines. The iPS cell lines from colonies picked were positively stained by Nanog, SSEA-1 immunofluorescence and alkaline phosphatase substrate markers. The advantage of using the EOS-S(4+)-EmGFP reporter to identify sufficiently reprogrammed iPS cell lines is discussed by representing experimental results obtained with electroporated plasmids, such as a mixture of 2 tandem OS and KM plasmids and a polycistronic OKSM expression plasmid.


Subject(s)
Baculoviridae/genetics , Genetic Vectors/genetics , Induced Pluripotent Stem Cells/cytology , Animals , Cellular Reprogramming/genetics , Fibroblasts/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Open Reading Frames/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
5.
Biochem Biophys Res Commun ; 391(1): 621-6, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19932085

ABSTRACT

CD437, a synthetic retinoid, has a potent antitumor activity, in which an RAR-independent mechanism may be involved. Our previous study showed that CD437 transcriptionally upregulates the expression of thioredoxin-binding protein 2 (TBP2), leading to c-Jun N-terminal kinase 1 (JNK1)-mediated apoptosis. In the present study, we addressed the mechanism, by which CD437 induces TBP2 mRNA expression. CD437 efficiently caused the cell death of human osteosarcoma cells via apoptosis. CD437 also induced JNK1 activation through the upregulation of TBP2 mRNA, in consistent with our previous observation. A luciferase reporter assay for TBP2 promoter activation suggested that CD437-regulated TBP2 mRNA transcription requires the region between -400 and -300, which contains multiple possible ETS-binding sites. Finally, we demonstrated CD437-dependent recruitment of ETS1 transcription factor to this region by chromatin immunoprecipitation assay. These data suggest that ETS1 is involved in CD437-induced TBP2 mRNA expression in human osteosarcoma MG-63 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Carrier Proteins/genetics , Neoplasms/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Retinoids/pharmacology , Transcription, Genetic/drug effects , Cell Line, Tumor , Chromatin Immunoprecipitation , Humans , Promoter Regions, Genetic
6.
Cancers (Basel) ; 11(10)2019 Oct 14.
Article in English | MEDLINE | ID: mdl-31615015

ABSTRACT

Activation of the epidermal growth factor receptor (EGFR) pathway plays an important role in the progression of cancer and is associated with a poor prognosis in patients. The monoclonal antibody cetuximab, which displays EGFR extracellular domain-specific binding, has proven effective in the treatment of locally advanced disease and relapsed/metastatic disease. However, the effects of cetuximab are weaker than those of EGFR tyrosine kinase inhibitors (TKIs). This study investigates differences in the effects on cell growth of cetuximab and EGFR TKI AG1478 at the molecular level using oral squamous cell carcinoma (OSCC) cell lines. First, we found that there were EGFR-inhibitor-sensitive (EIS) and EGFR-inhibitor-resistant cell lines. The EIS cell lines expressed not only EGFR but also ErbB3, and both were clearly phosphorylated. The levels of phosphorylated ErbB3 were unaffected by cetuximab but were reduced by AG1478. EGFR ligand treatment increased the levels of phosphorylated EGFR but not phosphorylated ErbB3. Moreover, when EIS cell lines that were only capable of anchorage-dependent growth were grown in suspension, cell growth was suppressed and the levels of phosphorylated focal adhesion kinase (FAK), Src, and ErbB3 were significantly reduced. The levels of phosphorylated ErbB3 were unaffected by the FAK inhibitor PF573228, but were reduced by Src inhibition. Finally, combining cetuximab and a Src inhibitor produced an additive effect on the inhibition of EIS cell line growth.

7.
Oncotarget ; 10(10): 1014-1030, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30800215

ABSTRACT

Cancer stem cells (CSCs), which play important roles in tumor initiation and progression, are resistant to many types of therapies. However, the regulatory mechanisms underlying CSC-specific properties, including self-renewal, are poorly understood. Here, we found that LATS1/2, the core Hippo pathway-kinases, were highly expressed in the oral squamous cell carcinoma line SAS, which exhibits high capacity of CSCs, and that depletion of these kinases prevented SAS cells from forming spheres under serum-free conditions. Detailed examination of the expression and activation of LATS kinases and related proteins over a time course of sphere formation revealed that LATS1/2 were more highly expressed and markedly activated before initiation of self-renewal. Moreover, TAZ, SNAIL, CHK1/2, and Aurora-A were expressed in hierarchical, oscillating patterns during sphere formation, suggesting that the process consists of four sequential steps. Our results indicate that LATS1/2 trigger self-renewal of CSCs by regulating the Hippo pathway, the EMT, and cell division.

8.
Jpn Dent Sci Rev ; 54(2): 88-103, 2018 May.
Article in English | MEDLINE | ID: mdl-29755619

ABSTRACT

Human cancer tissues are heterogeneous in nature and become differentiated during expansion of cancer stem cells (CSCs). CSCs initiate tumorigenesis, and are involved in tumor recurrence and metastasis. Furthermore, data show that CSCs are highly resistant to anticancer drugs. Cetuximab, a specific anti-epidermal growth factor receptor (EGFR) monoclonal antibody, is used in cancer treatment. Although development of resistance to cetuximab is well recognized, the underlying mechanisms remain unclear. Lapatinib, a dual inhibitor of epidermal growth factor receptor (EGFR)/ErbB2, has antiproliferative effects and is used to treat patients with ErbB2-positive metastatic breast cancer. In this review, cetuximab and lapatinib-resistant oral squamous cell carcinoma (OSCC) cells proliferation and migration signal transduction passway is discussed by introducing our research.

9.
BMC Genomics ; 8: 141, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17540041

ABSTRACT

BACKGROUND: Death receptors on the cell surface and the interacting cytosolic molecules, adaptors and initiator caspases, are essential as core components of the extrinsic apoptotic signaling pathway. While the apoptotic machinery governing the extrinsic signaling pathway is well characterized in mammals, it is not fully understood in fish. RESULTS: We identified and characterized orthologs of mammalian Fas, FADD and caspase-8 that correspond to the death receptor, adaptor and initiator caspase, from the Medaka fish (Oryzias latipes). Medaka Fas, caspase-8 and FADD exhibited protein structures similar to that of their mammalian counterparts, containing a death domain (DD), a death effector domain (DED) or both. Functional analyses indicated that these molecules possess killing activity in mammalian cell lines upon overexpression or following activation by apoptotic stimuli, suggesting similar pro-apoptotic functions in the extrinsic pathway as those in mammals. Genomic sequence analysis revealed that the Medaka fas (tnfrsf6), fadd and caspase-8 (casp8) genes are organized in a similar genomic structure as the mammalian genes. Database search and phylogenetic analysis revealed that the fas gene, but not the fadd and casp8 genes, appear to be present only in vertebrates. CONCLUSION: Our results indicate that the core components necessary for the extrinsic apoptotic pathway are evolutionarily conserved in function and structure across vertebrate species. Based on these results, we presume the mechanism of apoptosis induction via death receptors was evolutionarily established during the appearance of vertebrates.


Subject(s)
Apoptosis , Caspase 8/genetics , Evolution, Molecular , Fas Ligand Protein/genetics , Fas-Associated Death Domain Protein/genetics , Oryzias/genetics , Receptors, Death Domain/genetics , Amino Acid Sequence , Animals , Base Sequence , Caspase 8/chemistry , Caspase 8/metabolism , Cells, Cultured , DNA, Complementary , Databases, Genetic , Embryo, Mammalian , Embryo, Nonmammalian , Exons , Expressed Sequence Tags , Fas Ligand Protein/chemistry , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/chemistry , Fas-Associated Death Domain Protein/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , Genome , HeLa Cells , Humans , Immunohistochemistry , Mice , Molecular Sequence Data , NIH 3T3 Cells , Open Reading Frames , Oryzias/metabolism , Phylogeny , Protein Structure, Tertiary , Receptors, Death Domain/chemistry , Receptors, Death Domain/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Signal Transduction
10.
Gene ; 396(1): 134-48, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17459614

ABSTRACT

Caspase-8, a member of the caspase family, plays an important role in apoptotic signal transduction in mammals. Here we report the identification and characterization of the caspase-8 (casp8) gene in the zebrafish Danio rerio. The zebrafish casp8 gene has a genomic organization similar to mammalian casp8 genes, consisting of 10 exons. By chromosome mapping, we found that casp8 maps on linkage group 6 (LG6), a zebrafish chromosome segment orthologous to the long arm of human Chr. 2, which carries CASP8. In contrast, the zebrafish casp10-like gene and the cflar gene separately localize on LG9 and LG11, respectively, and these genes form a cluster with CASP8 on the human chromosome. This chromosomal segregation is unique to fish but not other vertebrates. Furthermore, we examined the function of zebrafish Casp8 protein in mammalian cells, and showed that it has pro-apoptotic activity when overexpressed. In addition, this molecule was capable of transmitting apoptotic signals mediated through not only Fas but also the TNF receptor in mouse Casp8-deficient cells. Expression analysis showed that casp8 is maternally expressed, and transcripts continue to be present throughout embryogenesis and into larval stages. These results show that zebrafish casp8 has a structure and function similar to mammalian CASP8 orthologs, and our study suggests that the role of caspase-8 in the apoptotic signal pathway has been conserved over at least 450 million years of vertebrate evolution.


Subject(s)
Apoptosis , Biological Evolution , Caspase 8/metabolism , Zebrafish/genetics , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/metabolism , Caspase 8/chemistry , Caspase 8/genetics , Embryonic Development , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome/genetics , HeLa Cells , Humans , Mice , Molecular Sequence Data , Phylogeny , Physical Chromosome Mapping , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish/embryology
11.
Nucleic Acids Res ; 33(10): 3401-11, 2005.
Article in English | MEDLINE | ID: mdl-15951513

ABSTRACT

Promoters, including neither TATA box nor initiator, have been frequently found in testicular germ cell-specific genes in mice. These investigations imply that unique forms of the polymerase II transcription initiation machinery play a role in selective activation of germ cell-specific gene expression programs during spermatogenesis. However, there is little information about testis-specific core promoters, because useful germ cell culture system is not available. In this study, we characterize the regulatory region of the haploid-specific Oxct2b gene in detail by using in vivo transient transfection assay in combination with a transgenic approach, with electrophoretic mobility shift and chromatin immunoprecipitation assays. Expression studies using mutant constructs demonstrate that a 34 bp region, which extends from -49 to -16, acts as a core promoter in an orientation-dependent manner. This promoter region includes the cAMP-responsive element (CRE)-like sequence TGACGCAG, but contains no other motifs, such as a TATA box or initiator. The CRE-like element is indispensable for the core promoter activity, but not for activator in testicular germ cells, through the binding of a testis-specific CRE modulator transcription factor. These results indicate the presence of alternative transcriptional initiation machinery for cell-type-specific gene expression in testicular germ cells.


Subject(s)
Coenzyme A-Transferases/genetics , DNA-Binding Proteins/metabolism , Promoter Regions, Genetic , Response Elements , Testis/metabolism , Transcription Factors/metabolism , 5' Flanking Region , Animals , Base Sequence , Binding Sites , Coenzyme A-Transferases/biosynthesis , Cyclic AMP/metabolism , Cyclic AMP Response Element Modulator , Electroporation , Haploidy , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Sequence Alignment , Spermatids/metabolism , TATA Box , Transcription, Genetic , Transfection
12.
Oncol Rep ; 37(6): 3674-3680, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28440510

ABSTRACT

The activation of receptor tyrosine kinases (RTKs) results in cellular effects including cell proliferation, survival, migration and invasion; RTKs also play an important role in tumourigenesis. It has been reported that EGFR signalling controls the migration of oral squamous cell carcinoma (OSCC) SAS and HSC3 cells but not of HSC4 cells, although the proliferation of HSC4 cells is regulated by EGF/EGFR. In the present study, we investigated the roles of EGFR and the c-Met signalling pathway in cell migration via filopodia and lamellipodia formation, which may be prerequisites for migration. To explore the role of c-Met in cell migration, we inhibited c-Met RTK activity using the c-Met inhibitor SU11274 and activated c-Met using hepatocyte growth factor (HGF) in three OSCC cell lines HSC4, SAS and Ca9-22 and investigated migration potency using a wound healing assay. We showed that inhibition of c-Met significantly suppressed, and activation of c-Met significantly promoted, the migration of OSCC cells. Additionally, the migration of SAS and Ca9-22 cells was inhibited by the EGFR inhibitors AG1478 and cetuximab and promoted by EGF treatment. Moreover, migration potency was correlated with lamellipodia formation. Furthermore, western blot analyses demonstrated that SU11274 decreased and HGF increased lamellipodin protein levels as well as phosphorylated c-Met levels. Collectively, we demonstrated that c-Met signalling induced lamellipodia formation by upregulating lamellipodin, thereby promoting the migration of OSCC cells.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/genetics , Mouth Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cetuximab/administration & dosage , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Indoles/administration & dosage , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Piperazines/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pseudopodia/drug effects , Pseudopodia/genetics , Quinazolines/administration & dosage , Signal Transduction/drug effects , Sulfonamides/administration & dosage , Tyrphostins/administration & dosage
13.
Oncol Lett ; 13(2): 930-936, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28356980

ABSTRACT

Cell migration potency is essential in cancer metastasis and is often regulated by extracellular stimuli. Oral squamous cell carcinoma cell lines include those that are sensitive, as well as resistant, to the effects of the epidermal growth factor receptor (EGFR) inhibitor cetuximab on cell migration. In the present study, the molecular differences in the EGFR response to cell migration between the SAS cetuximab-sensitive and HSC4 cetuximab-resistant cell lines was examined. Treatment with the EGFR inhibitors AG1478 and cetuximab reduced the migration potency of SAS cells, but not HSC4 cells. The migration of the two cell lines was inhibited under serum-free culture conditions, and the addition of EGF to the serum-free medium promoted the migration of SAS cells, but not HSC4 cells. In addition, SAS cell migration was reduced by the mitogen-activated protein kinase kinase and protein kinase B (Akt) inhibitors PD98059 and MK2206, whereas HSC4 cell migration was only inhibited by MK2206. EGF induced an increase in extracellular signal-regulated kinase phosphorylation levels in HSC4 cells, and stimulated Akt phosphorylation levels in SAS cells. Furthermore, the staining of actin filaments with phalloidin was significantly increased by the inhibition of EGFR in SAS cells, but was not observed as altered in HSC4 cells. Conversely, the addition of EGF to the culture medium decreased the accumulation of actin filaments in SAS cells. The results suggest that the EGF-EGFR signaling pathway has an important role in SAS cell migration via the modulation of actin dynamics, and that HSC4 cell migration is regulated by a serum component other than EGFR.

14.
Nucleic Acids Res ; 31(16): 4797-804, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12907721

ABSTRACT

Methylation of CpG islands spanning promoter regions is associated with control of gene expression. However, it is considered that methylation of exonic CpG islands without promoter is not related to gene expression, because such exonic CpG islands are usually distant from the promoter. Whether methylation of exonic CpG islands near the promoter, as in the case of a CpG-rich intronless gene, causes repression of the promoter remains unknown. To gain insight into this issue, we investigated the distribution and methylation status of CpG dinucleotides in the mouse Tact1/Actl7b gene, which is intronless and expressed exclusively in testicular germ cells. The region upstream to the gene was poor in CpG, with CpG dinucleotides absent from the core promoter. However, a CpG island was found inside the open reading frame (ORF). Analysis of the methylation status of the Tact1/Actl7b gene including the 5'-flanking area demonstrated that all CpG sites were methylated in somatic cells, whereas these sites were unmethylated in the Tact1/Actl7b-positive testis. Trans fection experiments with in vitro-methylated constructs indicated that methylation of the ORF but not 5' upstream repressed Tact1/Actl7b promoter activity in somatic cells. Similar effects of ORF methylation on the promoter activity were observed in testicular germ cells. These are the first results indicating that methylation of the CpG island in the ORF represses its promoter in somatic cells and demethylation is necessary for gene expression in spermatogenic cells.


Subject(s)
CpG Islands/genetics , DNA Methylation , Gene Expression Regulation , Open Reading Frames/genetics , Proteins/genetics , Actins , Animals , Cytoskeletal Proteins , DNA/genetics , DNA/metabolism , Green Fluorescent Proteins , Introns/genetics , Luciferases/genetics , Luciferases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spermatids/cytology , Spermatids/metabolism , Testis/cytology , Testis/metabolism
15.
Oncol Rep ; 36(5): 3058-3064, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27633099

ABSTRACT

Lapatinib, a dual inhibitor of epidermal growth factor receptor (EGFR)/ErbB2, has antiproliferative effects and is used to treat patients with ErbB2-positive metastatic breast cancer. In the present study, we examined the effects of lapatinib on growth of oral and prostate cancer cells. Oral squamous cell carcinoma (OSCC) cell lines HSC3, HSC4 and Ca9-22 were sensitive to the antiproliferative effects of lapatinib in anchorage-dependent culture, but the OSCC cell lines KB and SAS and the prostate cancer cell line DU145 were resistant to lapatinib. Phosphorylation levels of EGFR in all cell lines decreased during lapatinib treatment in anchorage­dependent culture. Furthermore, the phosphorylation levels of ErbB2, ErbB3 and Akt and the protein levels of cyclin D1 were decreased by lapatinib treatment of HSC3, HSC4 and Ca9-22 cells. ErbB3 was not expressed and cyclin D1 protein levels were not altered by lapatinib treatment in KB, DU145 and SAS cells. The phosphorylation of ErbB2 and AKT was not affected by lapatinib in SAS cells and was not detected in KB and DU145 cells. Lapatinib-resistant cell lines exhibited sphere-forming ability, and SAS cells developed sensitivity to lapatinib during sphere formation. The phosphorylation levels of ErbB2 and AKT and protein levels of cyclin D2 increased during sphere formation of SAS cells and decreased with lapatinib treatment. In addition, sphere formation of SAS cells was inhibited by the AKT inhibitor MK2206. AKT phosphorylation and cyclin D2 levels in SAS spheres were decreased by MK2206 treatment. SAS cells expressed E-cadherin, but not vimentin and KB cells expressed vimentin, but not E-cadherin. DU145 cells expressed vimentin and E-cadherin. These results suggested that phosphorylation of EGFR and ErbB2 by cell detachment from the substratum induces the AKT pathway/cyclin D2-dependent sphere growth in SAS epithelial cancer stem-like cells, thereby rendering SAS spheres sensitive to lapatinib treatment.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Cyclin D1/biosynthesis , Cyclin D2/biosynthesis , Mouth Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/biosynthesis , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-3/biosynthesis , Cadherins/biosynthesis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D2/genetics , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lapatinib , Male , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Quinazolines/administration & dosage , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics
16.
Asian J Androl ; 18(4): 600-6, 2016.
Article in English | MEDLINE | ID: mdl-26208397

ABSTRACT

Basigin is a member of the immunoglobulin superfamily and plays various important roles in biological events including spermatogenesis. To examine the basigin molecular variants during spermatogenesis and sperm maturation in the mouse, immunoprecipitated basigin samples from testis and epididymal spermatozoa were analyzed by liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). The results demonstrated that basigin molecules from the testis and spermatozoa were separable into two major bands and that the differences in the molecular sizes were possibly because of an endoproteolytic cleavage. Since basigin is known to be a chaperone for the monocarboxylate transporter 1 (MCT1), the localization of basigin, MCT1 and MCT2 was examined during postnatal testicular development. Immunohistochemical studies showed different expression patterns of MCT1 and MCT2. MCT1 was localized on the surface of spermatogonia, spermatocytes, and spermatids. In contrast, MCT2 appeared on the principal piece of spermatozoa in the testis, where basigin was also observed. In mature epididymal spermatozoa, MCT2 was located on the midpiece, where basigin co-localized with MCT2 but not with MCT1. Furthermore, MCT2 was immunoprecipitated with basigin in mouse testes and sperm. These results suggest that basigin has a functional role as a binding partner with MCT2 in testicular and epididymal spermatozoa.


Subject(s)
Basigin/metabolism , Monocarboxylic Acid Transporters/metabolism , Spermatozoa/metabolism , Testis/metabolism , Animals , Epididymis/metabolism , Male , Mice , Sperm Maturation/physiology , Spermatogenesis/physiology , Symporters/metabolism , Tandem Mass Spectrometry
17.
Int J Oncol ; 47(6): 2165-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26497980

ABSTRACT

We have previously shown that growth of the oral squamous cell carcinoma cell line SAS, is resistant to cetuximab in monolayer culture conditions, even though epidermal growth factor receptor (EGFR) was phosphorylated, but the growth of SAS aggregates was sensitive to cetuximab. In the present study, we demonstrate differences in the EGFR signaling pathways utilized by SAS cells in monolayer and suspension cultures at the molecular level. Cetuximab treatment of SAS cells in monolayer cultures inhibits the phosphorylation of EGFR and ERK, and reduces the cell migratory potency, but not cell proliferation. AG1478 treatment reduces the phosphorylation of EGFR, ERK and AKT, and affects cell growth in monolayer cultures. The phosphorylation levels of EGFR and AKT are significantly higher in SAS cell aggregates compared to monolayer cultures. Treatment with cetuximab and AG1478 reduces the growth of SAS aggregates and eliminates the phosphorylation of EGFR and AKT. Furthermore, proliferation of SAS aggregates is also inhibited by LY294002 and MK2206, which are inhibitors of PI3K and AKT, respectively. In addition, treatment with the lipid raft disruptor filipin III reduced the phosphorylation levels of EGFR and Akt in SAS aggregates, but not in SAS monolayer culture. These results suggest the possibility that ligands in the serum stimulate the phosphorylation of EGFR localized in lipid rafts leading to PI3K-AKT activation, which results in the growth of SAS aggregates, therefore resulting in the sensitivity of SAS aggregates to cetuximab.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/pathology , Cell Culture Techniques/methods , Cetuximab/pharmacology , Drug Resistance, Neoplasm/physiology , Mouth Neoplasms/pathology , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Fluorescent Antibody Technique , Humans , Mouth Neoplasms/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
18.
Stem Cells Dev ; 24(2): 172-81, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25141025

ABSTRACT

The developmental fate of the multipotent neural crest (NC) is determined along with the neural axis in which NC cells are generated. Only the cranial NC can differentiate into mesectodermal derivatives such as osteoblasts, chondrocytes, and adipocytes in vivo. Here, we attempted to selectively differentiate mouse embryonic stem (ES) cells into cranial NC stem cells and propagate them to explore their developmental potential to differentiate into mesectodermal derivatives. Using aggregation cultures in feeder- and serum-free neural induction medium (NIM) without serum replacement and l-glutamine, we obtained NIM neurospheres composed of neuroepithelium. The NIM neurospheres expressed the rostral markers Otx1 and Otx2, but not nonrostral markers Hoxb4, Hoxb9, Lbx1, and TH, which characterize cranial neurospheres. Subsequently, AP2α, Sox9, p75, Snail, Slug, and Twist-positive NC cells were differentiated in 4-day adhesion cultures of cranial neurospheres. In addition, sphere clusters in adhesion cultures were differentiated into osteoblasts, while migrating cells were not. By taking advantage of the sphere-formation capability, we isolated and propagated NC stem cells from the sphere clusters and confirmed their multipotency. NC stem cells expressed NC and stem cell markers, and they maintained differentiation potency in the NC derivatives. These results show that cranial NC stem cells were obtained reproducibly and efficiently without special inducing factors, gene transfection, or fluorescence-activated cell sorting selection.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells , Neural Crest , Neural Stem Cells , Skull , Animals , Antigens, Differentiation/metabolism , Cell Separation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Mice , Neural Crest/cytology , Neural Crest/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Skull/cytology , Skull/embryology , Skull/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism
19.
Gene ; 298(2): 183-93, 2002 Oct 02.
Article in English | MEDLINE | ID: mdl-12426106

ABSTRACT

Ornithine decarboxylase antizyme 1 and 2 (OAZ1 and OAZ2) are expressed ubiquitously, and control the intracellular concentration of polyamines. Their testicular isoform, OAZt/Oaz3, is specifically expressed in differentiated haploid germ cells. We have identified and characterized the gene encoding OAZt in mice. The mouse OAZt gene contains, as does the human ortholog and paralogs, five exons and four introns. Comparison of the mouse OAZt with the human ortholog gene revealed that exon sizes are identical and nucleotide sequences in exons are highly homologous (83% identity). The major transcriptional start site was determined by primer extension assay. Promoter activity was confirmed by transgenic mouse assays, using the upstream region of the mouse OAZt gene fused to a EGFP reporter gene. The OAZt essential promoter located between -133 and +242, has two CREs and an Inr, and lacks a TATA box. These elements are conserved in the human ortholog but not in the paralogs, indicating that such a short upstream region including two CREs and Inr is sufficient to drive endogenous OAZt mRNA expression in the haploid testicular germ cells.


Subject(s)
Promoter Regions, Genetic/genetics , Proteins/genetics , Spermatozoa/metabolism , Testis/metabolism , 3T3 Cells , Animals , Base Sequence , DNA/chemistry , DNA/genetics , Exons , Female , Gene Expression , Genes/genetics , Green Fluorescent Proteins , Humans , Introns , Luciferases/genetics , Luciferases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Microscopy, Fluorescence , Molecular Sequence Data , Ornithine Decarboxylase/genetics , Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Analysis, DNA , Testis/cytology , Transcription Initiation Site , Transfection
20.
FEBS Lett ; 559(1-3): 159-64, 2004 Feb 13.
Article in English | MEDLINE | ID: mdl-14960325

ABSTRACT

The testicular isoform of the ornithine decarboxylase antizyme (OAZt) gene is expressed exclusively in the haploid spermatids of mice. The 357-bp region, which includes a TATA-less promoter and an untranslated region, is sufficient for OAZt gene expression in the spermatids of transgenic mice. In this study, in vivo transient transfection to living mouse testes was used to define the transcriptional regulatory elements of the OAZt gene promoter. We found that the 10-bp element that contains an initiator (Inr) plays a central role as the core promoter, in combination with a downstream element, while two cyclic adenosine monophosphate-responsive element (CRE)-like sites in the upstream region also contribute to promoter activity. The electrophoretic mobility shift assay showed binding of the testis-specific factors to these elements. Our results show that the in vivo DNA transfer technique enables detailed analysis of haploid germ cell-specific gene regulation in mice.


Subject(s)
Electroporation , Haploidy , Mice/genetics , Promoter Regions, Genetic/genetics , Proteins/genetics , Animals , Base Sequence , Genes, Regulator , Male , Molecular Sequence Data , Response Elements , Testis/cytology , Testis/metabolism , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL