Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Oncogene ; 33(40): 4837-46, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-24186199

ABSTRACT

p53 is an established tumor suppressor that can activate the transcription of multiple target genes. Recent evidence suggests that p53 may contribute to the regulation of cell invasion and migration. In this study, we show that the forkhead box transcription factor FOXF1 is a novel target of the p53 family because FOXF1 is upregulated by p53, TAp73 and TAp63. We show that FOXF1 is induced upon DNA damage in a p53-dependent manner. Furthermore, we identified a response element located within the FOXF1 gene that is responsive to wild-type p53, TAp73ß and TAp63γ. The ectopic expression of FOXF1 inhibited cancer cell invasion and migration, whereas the inactivation of FOXF1 stimulated cell invasion and migration. We also show that FOXF1 regulates the transcriptional activity of E-cadherin (CDH1) by acting on its FOXF1 consensus binding site located upstream of the E-cadherin gene. Collectively, our results show that FOXF1 is a p53 family target gene, and our data suggest that FOXF1 and p53 form a portion of a regulatory transcriptional network that appears to have an important role in cancer cell invasion and migration.


Subject(s)
Cell Movement , Forkhead Transcription Factors/genetics , Tumor Suppressor Protein p53/physiology , Antigens, CD , Base Sequence , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , HEK293 Cells , Humans , Neoplasm Invasiveness , Response Elements , Transcription, Genetic , Up-Regulation
2.
Cancer Gene Ther ; 19(11): 749-56, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22956039

ABSTRACT

The p53 tumor suppressor belongs to a gene family that includes two other structurally and functionally related members: p73 and p63. The regulation of p53 activity differs significantly from that of p73 and p63. To enhance the tumor suppressive activity of p53, we constructed six recombinant adenoviruses that encode hybrid proteins with three functional domains derived from either p53 or TAp63γ. The potency of these hybrid molecules in suppressing tumorigenesis was evaluated using in vitro and in vivo models. Of the hybrid molecules tested, one hybrid named p63-53O was the most potent activator of apoptosis in human cancer cells. The p63-53O hybrid is composed of the transcriptional activation domain and DNA-binding domain of TAp63γ and the oligomerization domain of p53. The p63-53O hybrid efficiently transactivated p53AIP1. Moreover, silencing of p53AIP1 partially abolished the apoptotic response to p63-53O in human cancer cells. The p53-p63 hybrid molecule is a novel potent anti-proliferative agent for the treatment of cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, p53 , Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/genetics , Neoplasms/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
3.
Oncogene ; 28(29): 2643-53, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19448676

ABSTRACT

The mitotic checkpoint gene CHFR (checkpoint with forkhead and ring finger domains) is silenced in various human cancers by promoter hypermethylation, suggesting that CHFR is a tumor suppressor. Here, we show that CHFR functions as a negative regulator of the nuclear factor-kappaB (NF-kappaB) pathway. Expression of CHFR inhibited NF-kappaB reporter activity, whereas knockdown of CHFR activated reporter activity. These activities are independent of its RING finger domain. Furthermore, we found that CHFR physically interacts with p65 in cells. Electrophoretic mobility shift assays (EMSAs) and ELISA-based NF-kappaB-binding assays showed that CHFR negatively regulated transcriptional activity of p65. In addition, our data show that interleukin (IL)-8 is significantly downregulated by CHFR, and that the migration of human endothelial cells is suppressed in culture medium conditioned from CHFR-expressing cancer cells. Using a xenograft model, we show that neovascularization is suppressed by adenovirus-mediated transfer of CHFR. These results indicate that expression of CHFR markedly reduces the expression of IL-8 through the inhibition of NF-kappaB. As the NF-kappaB signaling pathway plays a critical role in the development and progression of cancer, our findings show the functional relationship between epigenetic alteration and inflammation/angiogenesis in human cancer cells, thereby showing several potential targets for therapeutic intervention.


Subject(s)
Cell Cycle Proteins/metabolism , Gene Expression Regulation, Neoplastic , Interleukin-8/genetics , Neoplasm Proteins/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/genetics , Transcription Factor RelA/antagonists & inhibitors , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , DNA/metabolism , Down-Regulation , Electrophoretic Mobility Shift Assay , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Neoplasm Proteins/genetics , Poly-ADP-Ribose Binding Proteins , Transcription, Genetic , Ubiquitin-Protein Ligases , Xenograft Model Antitumor Assays
4.
Br J Cancer ; 98(6): 1147-56, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18283316

ABSTRACT

Although mutation of APC or CTNNB1 (beta-catenin) is rare in breast cancer, activation of Wnt signalling is nonetheless thought to play an important role in breast tumorigenesis, and epigenetic silencing of Wnt antagonist genes, including the secreted frizzled-related protein (SFRP) and Dickkopf (DKK) families, has been observed in various tumours. In breast cancer, frequent methylation and silencing of SFRP1 was recently documented; however, altered expression of other Wnt antagonist genes is largely unknown. In the present study, we found frequent methylation of SFRP family genes in breast cancer cell lines (SFRP1, 7 out of 11, 64%; SFRP2, 11 out of 11, 100%; SFRP5, 10 out of 11, 91%) and primary breast tumours (SFRP1, 31 out of 78, 40%; SFRP2, 60 out of 78, 77%; SFRP5, 55 out of 78, 71%). We also observed methylation of DKK1, although less frequently, in cell lines (3 out of 11, 27%) and primary tumours (15 out of 78, 19%). Breast cancer cell lines express various Wnt ligands, and overexpression of SFRPs inhibited cancer cell growth. In addition, overexpression of a beta-catenin mutant and depletion of SFRP1 using small interfering RNA synergistically upregulated transcriptional activity of T-cell factor/lymphocyte enhancer factor. Our results confirm the frequent methylation and silencing of Wnt antagonist genes in breast cancer, and suggest that their loss of function contributes to activation of Wnt signalling in breast carcinogenesis.


Subject(s)
Breast Neoplasms/genetics , Epigenesis, Genetic , Eye Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Wnt Proteins/physiology , Adaptor Proteins, Signal Transducing , Cell Line, Tumor , DNA Methylation , Female , Gene Silencing , Genes, Tumor Suppressor , Humans
5.
Oncogene ; 26(32): 4699-713, 2007 Jul 12.
Article in English | MEDLINE | ID: mdl-17297461

ABSTRACT

Activation of Wnt signaling has been implicated in gastric tumorigenesis, although mutations in APC (adenomatous polyposis coli), CTNNB1 (beta-catenin) and AXIN are seen much less frequently in gastric cancer (GC) than in colorectal cancer. In the present study, we investigated the relationship between activation of Wnt signaling and changes in the expression of secreted frizzled-related protein (SFRP) family genes in GC. We frequently observed nuclear beta-catenin accumulation (13/15; 87%) and detected the active form of beta-catenin in most (12/16; 75%) GC cell lines. CpG methylation-dependent silencing of SFRP1, SFRP2 and SFRP5 was frequently seen among GC cell lines (SFRP1, 16/16, 100%; SFRP2, 16/16, 100%; SFRP5, 13/16, 81%) and primary GC specimens (SFRP1, 42/46, 91%; SFRP2, 44/46, 96%; SFRP5, 30/46, 65%), and treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine rapidly restored SFRP expression. Ectopic expression of SFRPs downregulated T-cell factor/lymphocyte enhancer factor transcriptional activity, suppressed cell growth and induced apoptosis in GC cells. Analysis of global expression revealed that overexpression of SFRP2 repressed Wnt target genes and induced changes in the expression of numerous genes related to proliferation, growth and apoptosis in GC cells. It thus appears that aberrant SFRP methylation is one of the major mechanisms by which Wnt signaling is activated in GC.


Subject(s)
Carcinoma/genetics , Epigenesis, Genetic , Proto-Oncogene Proteins/genetics , Stomach Neoplasms/genetics , Wnt Proteins/genetics , Carcinoma/chemistry , Cell Line, Tumor , CpG Islands , DNA Methylation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Proto-Oncogene Proteins/analysis , Signal Transduction , Stomach Neoplasms/chemistry , TCF Transcription Factors/antagonists & inhibitors
6.
Br J Cancer ; 94(6): 914-21, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16479257

ABSTRACT

Transcription factor 2 gene (TCF2) encodes hepatocyte nuclear factor 1beta (HNF1beta), a transcription factor associated with development and metabolism. Mutation of TCF2 has been observed in renal cell cancer, and by screening aberrantly methylated genes, we have now identified TCF2 as a target for epigenetic inactivation in ovarian cancer. TCF2 was methylated in 53% of ovarian cancer cell lines and 26% of primary ovarian cancers, resulting in loss of the gene's expression. TCF2 expression was restored by treating cells with a methyltransferase inhibitor, 5-aza-2'deoxycitidine (5-aza-dC). In addition, chromatin immunoprecipitation showed deacetylation of histone H3 in methylated cells and, when combined with 5-aza-dC, the histone deacetylase inhibitor trichostatin A synergistically induced TCF2 expression. Epigenetic inactivation of TCF2 was also seen in colorectal, gastric and pancreatic cell lines, suggesting general involvement of epigenetic inactivation of TCF2 in tumorigenesis. Restoration of TCF2 expression induced expression of HNF4alpha, a transcriptional target of HNF1beta, indicating that epigenetic silencing of TCF2 leads to alteration of the hepatocyte nuclear factor network in tumours. These results suggest that TCF2 is involved in the development of ovarian cancers and may represent a useful target for their detection and treatment.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/biosynthesis , Hepatocyte Nuclear Factor 1-beta/metabolism , Ovarian Neoplasms/genetics , Base Sequence , DNA Methylation , Epigenesis, Genetic , Female , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gene Expression Profiling , Humans , Molecular Sequence Data , Ovarian Neoplasms/pathology , Tumor Cells, Cultured
7.
Cancer Gene Ther ; 13(4): 385-92, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16244591

ABSTRACT

Glioblastomas are the most common primary brain tumors in adults. These tumors exhibit a high degree of vascularization, and malignant progression from astrocytoma to glioblastoma is often accompanied by increased angiogenesis and the upregulation of vascular endothelial growth factor and its receptors. In this study, we investigated the in vivo antiangiogenic and antitumor effects of brain-specific angiogenesis inhibitor 1 (BAI1) using human glioblastoma cell lines. Glioblastoma cells were transduced with an adenoviral vector encoding BAI1 (AdBAI1), and Northern and Western blot analyses, respectively, demonstrated BAI1 mRNA and protein expression in the transduced tumor cells. Using an in vivo neovascularization assay, we found that angiogenesis surrounding AdBAI1-transduced glioblastoma cells transplanted into transparent skinfold chambers of SCID mice was significantly impaired compared to control treated cells. Additionally, in vivo inoculation with AdBAI1 of established subcutaneous or intracerebral transplanted tumors significantly impaired tumor growth and promoted increased mouse survival. Morphologically, the tumors exhibited signs of impaired angiogenesis, such as extensive necrosis and reduced intratumoral vascular density. Taken together, these data strongly indicate that BAI1 may be an excellent gene therapy candidate for the treatment of brain tumors, especially human glioblastomas.


Subject(s)
Angiogenic Proteins/biosynthesis , Brain Neoplasms/blood supply , Glioblastoma/blood supply , Neovascularization, Pathologic/therapy , Adenoviridae/genetics , Angiogenic Proteins/genetics , Animals , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Genetic Therapy , Genetic Vectors , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Mice , Mice, SCID , Neoplasm Transplantation , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled , Transduction, Genetic
8.
Oncogene ; 25(12): 1733-42, 2006 Mar 16.
Article in English | MEDLINE | ID: mdl-16278676

ABSTRACT

Epigenetic alterations like DNA methylation and the resulting inactivation of cancer-related genes often contribute to the development of various cancers. To identify the genes that are silenced by aberrant methylation in renal cell carcinoma (RCC), we subjected two RCC lines to methylated CpG island amplification/representational difference analysis. This identified 27 CpG islands. Combined bisulfite restriction analysis of these CpG islands in primary RCC cases revealed that four were methylated in a tumor-specific manner. One of these was identified as the human homeo-box gene B13 (HOXB13) gene, but the remaining three CpG islands were not associated with known genes. The methylation frequencies of HOXB13 in primary RCC samples and lines were 30 and 73%, respectively. The methylation status of HOXB13 correlated with the loss of its expression both in RCC lines and primary tumors, and methyltransferase inhibitor treatment induced the recovery of its expression. Exogenous expression of HOXB13 in RCC cells that lacked endogenous HOXB13 expression suppressed colony formation and induced apoptotic features. Furthermore, HOXB13 methylation correlated positively with tumor grade and microvessel invasion. These results suggest that HOXB13 is a novel candidate tumor suppressor gene in RCC and that its inactivation may play an important role in both RCC tumorigenesis and progression.


Subject(s)
Carcinoma, Renal Cell/genetics , Epigenesis, Genetic , Genes, Tumor Suppressor , Homeodomain Proteins/genetics , Kidney Neoplasms/genetics , Apoptosis/physiology , Cell Line, Tumor , CpG Islands , DNA Methylation , Gene Silencing , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction
9.
Carcinogenesis ; 27(1): 53-63, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16081516

ABSTRACT

p63 is critical for squamous development and exists as multiple isotypes of two subclasses, TA and DeltaN. DeltaNp63 isotypes can antagonize transcription by TAp63 and p53, and are highly expressed in squamous cell cancers. Using mouse keratinocytes as a biological model of squamous epithelium, we show that multiple p63 isotypes, DeltaN- and TA-containing, are expressed and differentially modulated during in vitro murine keratinocyte differentiation. DeltaNp63alpha declines with Ca2+-induced differentiation, while a smaller DeltaN-form, DeltaNp63s, persists, suggesting unique functions of the two DeltaN-forms. To investigate the impact of dysregulated p63 expression that is observed in cancers and to define the biological contribution of the different domains of the p63 isotypes, DeltaNp63alpha, DeltaNp63p40, TAp63alpha, TAp63gamma or beta-galactosidase were overexpressed in primary murine keratinocytes. Microarray, RT-PCR and western blot analyses revealed that overexpression of DeltaNp63p40, which lacks the entire alpha-tail present in DeltaNp63alpha, permits expression of a full panel of differentiation markers. This is in contrast to overexpression of the full-length DeltaNp63alpha, which blocks induction of keratin 10, loricrin and filaggrin. These findings support a role for the alpha-tail of DeltaNp63alpha in blocking differentiation-specific gene expression. Overexpression of either TAp63 isotype permits keratin 10 and loricrin expression, thus the alpha-terminus requires the cooperation of the DeltaN domain in blocking early differentiation. However, both TA isotypes block filaggrin induction. The DeltaN-terminus is sufficient to maintain keratinocytes in a proliferative state, as both DeltaN forms block Ca2+-mediated p21WAF1 induction and S-phase arrest, while sustaining elevated PCNA levels. No alteration in cell cycle regulation was observed in keratinocytes overexpressing TAp63alpha or TAp63gamma. Clarifying the functional distinctions between p63 isotypes and domains will help to elucidate how their dysregulation impacts tumor biology and may suggest novel therapeutic strategies for modulating behavior of tumor cells with altered expression of p53 family members.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Gene Expression Regulation , Keratinocytes/cytology , Keratinocytes/metabolism , Phosphoproteins/metabolism , Trans-Activators/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , Calcium/metabolism , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Cell Proliferation , Filaggrin Proteins , Genes, Tumor Suppressor , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Keratin-10/genetics , Keratin-10/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Papilloma/chemically induced , Papilloma/metabolism , Papilloma/pathology , Phosphoproteins/genetics , Polymerase Chain Reaction , Proliferating Cell Nuclear Antigen/metabolism , Protein Isoforms , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , S Phase , Sequence Deletion , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Trans-Activators/genetics , beta-Galactosidase/metabolism
10.
Br J Cancer ; 92(6): 1165-72, 2005 Mar 28.
Article in English | MEDLINE | ID: mdl-15756280

ABSTRACT

Hypoxia is a key factor contributing to the progression of human neoplasias and to the development of resistance to chemotherapy. BNIP3 is a proapoptotic member of the Bcl-2 protein family involved in hypoxia-induced cell death. We evaluated the expression and methylation status of BNIP3 gene to better understand the role of epigenetic alteration of its expression in haematopoietic tumours. Methylation of the region around the BNIP3 transcription start site was detected in four acute lymphocytic leukaemia, one multiple myeloma and one Burkitt lymphoma cell lines, and was closely associated with silencing the gene. That expression of BNIP3 was restored by treatment with 5-aza2'-deoxycytidine (5-aza-dC), a methyltransferase inhibitor, which confirmed the gene to be epigenetically inactivated by methylation. Notably, re-expression of BNIP3 using 5-aza2-dC also restored hypoxia-mediated cell death in methylated cell lines. Acetylation of histone H3 in the 5' region of the gene, which was assessed using chromatin immunoprecipitation assays, correlated directly with gene expression and inversely with DNA methylation. Among primary tumours, methylation of BNIP3 was detected in five of 34 (15%) acute lymphocytic leukaemias, six of 35 (17%) acute myelogenous leukaemias and three of 14 (21%) multiple myelomas. These results suggest that aberrant DNA methylation of the 5' CpG island and histone deacetylation play key roles in silencing BNIP3 expression in haematopoietic tumours.


Subject(s)
DNA Methylation , Gene Silencing , Hematologic Neoplasms/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , Base Sequence , Cell Line, Tumor , CpG Islands , Humans , Molecular Sequence Data
12.
Br J Cancer ; 90(4): 844-52, 2004 Feb 23.
Article in English | MEDLINE | ID: mdl-14970863

ABSTRACT

By presenting immunogenic peptides at the cell surface, major histocompatibility complex (MHC) class II molecules play a key role in the control of adaptive immune responses. Whether expressed constitutively or induced by interferon-gamma, expression of MHC class II molecules is regulated via coactivator class II transactivator (CIITA); moreover, suppression of their expression is one mechanism by which cancer cells escape host immunity. In this study, we surveyed the relationship between the expression of one MHC class II antigen, HLA-DR, and its coactivators in a group of haematopoietic cell lines, and explored the role of the aberrant DNA methylation in silencing HLA-DR expression. Among 26 cell lines studied, HLA-DR expression was lost from eight T-cell and two myeloid leukaemia cell lines, and this loss was closely associated with suppression of CIITA-PIV expression. Notably, nine of the 10 cell lines that lost CIITA-PIV expression showed methylation of the gene's 5' CpG island. Thus, DNA methylation is believed to inhibit the expression of MHC class II molecules in haematopoietic tumour cells by silencing its coactivator, CIITA-PIV. Furthermore, methylation of CIITA-PIV was detected in seven of 32 primary acute myeloid leukaemia specimens, indicating that epigenetic alteration is not a cell line-specific phenomenon. Collectively, these data suggest that, by suppressing expression of MHC class II molecules, epigenetic inactivation of CIITA provides a survival advantage to a subset of haematopoietic tumours.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Methylation , Gene Expression Regulation, Neoplastic , HLA-DR Antigens/biosynthesis , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Interferon-gamma/pharmacology , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Acetylation , Cell Survival , Flow Cytometry , Genes, MHC Class II , HLA-DR Antigens/immunology , Histones/metabolism , Humans , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
13.
Br J Cancer ; 86(11): 1817-23, 2002 Jun 05.
Article in English | MEDLINE | ID: mdl-12087472

ABSTRACT

Death-associated protein kinase is a positive regulator of programmed cell death induced by interferon gamma. To investigate the role of epigenetic inactivation of death-associated protein kinase in gastrointestinal cancer, we examined the methylation status of the 5' CpG island of the death-associated protein kinase gene. Methylation of the 5' CpG island was detected in 3 of 9 colorectal and 3 of 17 gastric cancer cell lines, while among primary tumours, it was detected in 4 of 28 (14%) colorectal and 4 of 27 (15%) gastric cancers. By contrast, methylation of the edge of the CpG island was detected in virtually every sample examined. Death-associated protein kinase expression was diminished in four cell lines that showed dense methylation of the 5' CpG island, and treatment with 5-aza-2'-deoxycitidine, a methyltransferase inhibitor, restored gene expression. Acetylation of histones H3 and H4 in the 5' region of the gene was assessed by chromatin immunoprecipitation and was found to correlate directly with gene expression and inversely with DNA methylation. Thus, aberrant DNA methylation and histone deacetylation of the 5' CpG island, but not the edge of the CpG island, appears to play a key role in silencing death-associated protein kinase expression in gastrointestinal malignancies.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/genetics , Colorectal Neoplasms/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Deacetylases/metabolism , Stomach Neoplasms/genetics , Apoptosis Regulatory Proteins , Base Sequence , Colorectal Neoplasms/enzymology , DNA Primers , Death-Associated Protein Kinases , Dinucleoside Phosphates/genetics , Gene Expression Regulation, Enzymologic , Humans , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/enzymology , Tumor Cells, Cultured
14.
J Invest Dermatol ; 117(4): 914-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11676832

ABSTRACT

Melanoma cells rarely contain mutant p53 and hardly undergo apoptosis by wild-type p53. By using recombinant adenoviruses that express p53 or p53-related p51A or p73beta, we tested their apoptotic activities in melanoma cells. Yeast functional assay revealed a mutation of p53 at the 258th codon (AAA [K] instead of GAA [E]) in one cell line, 70W, out of six human melanoma cell lines analyzed (SK-mel-23, SK-mel-24, SK-mel-118, TXM18, 70W, and G361). Adenovirus-mediated transfer of p53, p51A, and/or p73beta suppressed growth and induced apoptotic DNA fragmentation of SK-mel-23, SK-mel-118, and 70W cells. Interestingly, p51A induced DNA fragmentation in them more significantly than p53 and p73beta. By Western blotting we analyzed levels of apoptosis-related proteins in cells expressing p53 family members. Apoptotic Bax and antiapoptotic Bcl-2 were not significantly upregulated or downregulated by expression of p53, p51A, or p73beta, except for p53-expressing 70W cells, which contained a larger amount of Bax protein than LacZ-expressing cells. Activation of caspase-3 was demonstrated only in p51A-expressing SK-mel-118 cells. We show here that p51A can mediate apoptosis in both wild-type and mutant p53-expressing melanoma cells more significantly than p53 and p73beta. It is also suggested that in melanoma cells (i) cellular target protein(s) other than Bcl-2 and Bax might be responsible for induction of p51A-mediated apoptosis and (ii) caspase-3 is not always involved in the apoptosis by p53 family members.


Subject(s)
Apoptosis , Melanoma/physiopathology , Tumor Suppressor Protein p53/pharmacology , Adenoviridae/genetics , Caspase 3 , Caspases/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/pharmacology , Enzyme Activation , Gene Transfer Techniques , Genetic Vectors , Homeostasis , Humans , Melanoma/pathology , Multigene Family , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein
15.
Gene Ther ; 8(18): 1401-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11571580

ABSTRACT

p53 gene therapy is being tested clinically for the treatment of human cancer, however, some cancer models (in vivo and in vitro) are resistant to p53. To explore the potential use of two p53 homologues, p73 and p51/p63, in cancer gene therapy, we introduced p53, p73 and p51/p63 into colorectal cancer cell lines via adenoviral vectors, and compared their effects on cell growth. Among 10 cell lines tested, six cell lines displayed a similar response following transduction of p53, p73beta or p51A/p63gamma; two lines underwent cell-cycle arrest, three lines exhibited apoptosis and one line showed no-effect following transduction. The effect on cell-cycle progression was variable in the other four cell lines. Interestingly, three cell lines were resistant to p53-mediated apoptosis, including two lines having endogenous wild-type p53 alleles, but underwent apoptosis after transduction of p73beta or p51A/p63gamma. Similar to p53, transduction of p51A/p63gamma induced extensive apoptosis when combined with adriamycin or X-radiation in SW480 cells, which are normally resistant to apoptosis. Transduction of p73beta and p51A/p63gamma also reduced the tumorigenicity of two colorectal cancer cells in vivo. These results suggest that adenovirus-mediated p73beta and p51A/p63gamma transfer are potential novel approaches for the treatment of human cancers, particularly for tumors that are resistant to p53 gene therapy.


Subject(s)
Adenoviridae/genetics , Apoptosis/genetics , Colorectal Neoplasms/therapy , Genes, p53 , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Membrane Proteins , Animals , Antineoplastic Agents/pharmacology , Carcinogenicity Tests , Colorectal Neoplasms/pathology , DNA-Binding Proteins/genetics , Doxorubicin/pharmacology , Genes, Tumor Suppressor , Humans , Mice , Mice, Nude , Nuclear Proteins/genetics , Phosphoproteins/genetics , Trans-Activators/genetics , Transcription Factors , Transduction, Genetic/methods , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Proteins
16.
Jpn J Cancer Res ; 92(4): 475-81, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11346471

ABSTRACT

We investigated the effect of p53 status on involvement of caspase-3 activation in cell death induced by X-irradiation, using rat embryonic fibroblasts (REFs) transduced with a temperature-sensitive mutant (mt) p53 gene. Cells with wild-type (wt) p53 showed greater resistance to X-irradiation than cells with mt p53. In cells with wt p53, X-irradiation-induced apoptosis was not inhibited by the caspase-3 inhibitor acetyl-L-aspartyl-L-methionyl-L-glutaminyl-L-aspartyl-aldehyde (Ac-DMQD-CHO) and caspase-3 activity was not elevated following X-irradiation, although induction of p53 and p21 / WAF-1 protein was observed. In contrast, irradiated cells with mt p53 showed 89% inhibition of cell death with Ac-DMQD-CHO and 98% inhibition with the antioxidant N-acetyl-L-cysteine (NAC). In cells with mt p53, caspase-3 activity was increased approximately 5 times beyond baseline activity at 24 h after irradiation. This increase was almost completely inhibited by NAC. However, inhibition of caspase-3 by Ac-DMQD-CHO failed to decrease production of reactive oxygen species by cells with mt p53. Differential involvement of caspase-3 is a reason for differences in sensitivity to X-irradiation in cells with different p53 status. Caspase-3 activation appears to occur downstream from generation of reactive oxygen species occurring independently of wt p53 during X-irradiation-induced cell death.


Subject(s)
Apoptosis/radiation effects , Caspases/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3 , Caspase Inhibitors , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Genes, p53/genetics , Mutation , Oligopeptides/pharmacology , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/radiation effects , Transduction, Genetic , Tumor Suppressor Protein p53/biosynthesis
17.
Tumour Biol ; 22(2): 123-30, 2001.
Article in English | MEDLINE | ID: mdl-11125285

ABSTRACT

Previous studies reported that mutation of the adenomatous polyposis coli (APC) gene was not observed in the majority of gastric cancers. To evaluate the role of the APC/beta-catenin/Tcf pathway, we analyzed mutations in the beta-catenin gene and the accumulation of beta-catenin protein in gastric carcinomas. An interstitial deletion spanning exon 3 of the beta-catenin gene was observed in 1 of 13 gastric cancer cell lines. No missense mutation was found in these 13 cell lines. Nuclear and/or cytoplasmic localization of beta-catenin was observed in 16 of 70 primary gastric carcinomas by immunohistochemistry, while we found no mutations in exon 3 in 35 carcinoma tissues available for PCR amplification. Our findings suggest that somatic mutations of the beta-catenin gene are rare in human gastric carcinomas and that accumulation of normal beta-catenin protein in a subset of gastric cancers may be due to other mechanisms of its activation.


Subject(s)
Carcinoma/genetics , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , DNA Mutational Analysis , Mutation , Stomach Neoplasms/genetics , Trans-Activators , Adenomatous Polyposis Coli Protein , Carcinoma/metabolism , Carcinoma/pathology , Cell Nucleus/metabolism , Cytoplasm/metabolism , Epithelial Cells/metabolism , Exons , Gene Deletion , Humans , Immunohistochemistry , Models, Genetic , Polymerase Chain Reaction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Cells, Cultured , beta Catenin
18.
Cell ; 102(6): 849-62, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11030628

ABSTRACT

Through direct cloning of p53 binding sequences from human genomic DNA, we have isolated a novel gene, designated p53AIP1 (p53-regulated Apoptosis-Inducing Protein 1), whose expression is inducible by wild-type p53. Ectopically expressed p53AIP1, which is localized within mitochondria, leads to apoptotic cell death through dissipation of mitochondrial A(psi)m. We have found that upon severe DNA damage, Ser-46 on p53 is phosphorylated and apoptosis is induced. In addition, substitution of Ser-46 inhibits the ability of p53 to induce apoptosis and selectively blocks expression of p53AIP1. Our results suggest that p53AIP1 is likely to play an important role in mediating p53-dependent apoptosis, and phosphorylation of Ser-46 regulates the transcriptional activation of this apoptosis-inducing gene.


Subject(s)
Apoptosis/physiology , Proteins/genetics , Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma , Animals , Apoptosis Regulatory Proteins , Breast Neoplasms , COS Cells , Carcinoma, Non-Small-Cell Lung , Cloning, Molecular , Colorectal Neoplasms , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA Damage/physiology , Fibroblasts/cytology , Gene Expression Regulation, Neoplastic , Humans , In Situ Nick-End Labeling , Lung Neoplasms , Mitochondria/metabolism , Molecular Sequence Data , Mutagenesis/physiology , Osteosarcoma , Phosphorylation , Protein Binding/physiology , Serine/metabolism , Signal Transduction/genetics , Skin/cytology , Transcriptional Activation/physiology , Tumor Cells, Cultured
19.
Jpn J Cancer Res ; 91(6): 612-5, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10874213

ABSTRACT

Recent studies demonstrated that a single guanine insertion polymorphism in a matrix metalloprotease-1 promoter created an Ets binding site and affected the elevation of the transcriptional level of matrix metalloproteinase-1 (MMP-1). Furthermore, in tumor cell lines derived from melanoma and breast cancer, the incidence of the 2G / 2G genotype was significantly higher than that in the normal population. To evaluate the contribution of this polymorphism in endometrial carcinomas, we genotyped 100 endometrial carcinomas and then analyzed immunoexpression of MMP-1 in these carcinomas. We found that endometrial carcinoma patients showed a significantly higher rate of 1G / 2G or 2G / 2G genotype than control individuals, and that tumors containing the 2G allele(a) expressed MMP-1 protein more frequently than those with 1G / 1G genotype. Therefore, the single nucleotide polymorphism at the MMP-1 promoter affected the expression level of the MMP-1 protein, which may result in the association with more aggressive character in endometrial carcinoma. Our result suggests that the presence of 2G polymorphism at the MMP-1 promoter may be one of the risk factors for the development and / or progression of endometrial carcinoma.


Subject(s)
Endometrial Neoplasms/genetics , Matrix Metalloproteinase 1/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Base Sequence , DNA, Neoplasm/genetics , Female , Genotype , Guanine/physiology , Humans , Polymerase Chain Reaction
20.
Jpn J Cancer Res ; 91(4): 395-8, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10804287

ABSTRACT

Recently, an activating mutation of the SRC gene has been implicated in about one-tenth of advanced colon cancers. The SRC 531 mutation results in truncation of SRC directly C-terminal to the regulatory Tyr 530 and appears to activate the Tyr 530. To investigate whether mutation of SRC plays an important role in the development and progression of gynecological tumors, we performed mutational analysis of the entire coding region of SRC in 70 ovarian carcinomas, 68 endometrial carcinomas and 3 endometrial stromal sarcomas by means of polymerase chain reaction-single strand conformation polymorphism (PCR-SSCP) followed by nucleotide sequencing and restriction fragment length polymorphism (RFLP) analysis. We found one truncated mutation at codon 531 (Gln to Stop) in an endometrial carcinoma. However, we found no mutation of this gene in ovarian carcinoma or endometrial stromal sarcoma. Our results suggest that mutation of SRC may be implicated in a small proportion of endometrial carcinomas.


Subject(s)
Endometrial Neoplasms/genetics , Genes, src , Mutation , Adult , Female , Humans , Polymorphism, Restriction Fragment Length , Polymorphism, Single-Stranded Conformational
SELECTION OF CITATIONS
SEARCH DETAIL
...