104 results on '"Kazushi Inoue"'
Search Results
2. Recent Progress in Mouse Models for Tumor Suppressor Genes and its Implications in Human Cancer
- Author
-
Kazushi Inoue, Elizabeth A. Fry, and Pankaj Taneja
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2013
3. Dmp1α inhibits HER2/neu-induced mammary tumorigenesis.
- Author
-
Elizabeth A Fry, Pankaj Taneja, Dejan Maglic, Sinan Zhu, Guangchao Sui, and Kazushi Inoue
- Subjects
Medicine ,Science - Abstract
Our recent study shows a pivotal role of Dmp1 in quenching hyperproliferative signals from HER2 to the Arf-p53 pathway as a safety mechanism to prevent breast carcinogenesis. To directly demonstrate the role of Dmp1 in preventing HER2/neu-driven oncogenic transformation, we established Flag-Dmp1α transgenic mice (MDTG) under the control of the mouse mammary tumor virus (MMTV) promoter. The mice were viable but exhibited poorly developed mammary glands with markedly reduced milk production; thus more than half of parous females were unable to support the lives of new born pups. The mammary glands of the MDTG mice had very low Ki-67 expression but high levels of Arf, Ink4a, p53, and p21(Cip1), markers of senescence and accelerated aging. In all strains of generated MDTG;neu mice, tumor development was significantly delayed with decreased tumor weight. Tumors from MDTG;neu mice expressed Flag-Dmp1α and Ki-67 in a mutually exclusive fashion indicating that transgenic Dmp1α prevented tumor growth in vivo. Genomic DNA analyses showed that the Dmp1α transgene was partially lost in half of the MDTG;neu tumors, and Western blot analyses showed Dmp1α protein downregulation in 80% of the cases. Our data demonstrate critical roles of Dmp1 in preventing mammary tumorigenesis and raise the possibility of treating breast cancer by restoring Dmp1α expression.
- Published
- 2013
- Full Text
- View/download PDF
4. Positive and negative regulation of prostate stem cell antigen expression by Yin Yang 1 in prostate epithelial cell lines.
- Author
-
Shuai Tang, Meenu Mishra, Donna P Frazier, Miranda L Moore, Kazushi Inoue, Rajendar Deora, Guangchao Sui, and Purnima Dubey
- Subjects
Medicine ,Science - Abstract
Prostate cancer is influenced by epigenetic modification of genes involved in cancer development and progression. Increased expression of Prostate Stem Cell Antigen (PSCA) is correlated with development of malignant human prostate cancer, while studies in mouse models suggest that decreased PSCA levels promote prostate cancer metastasis. These studies suggest that PSCA has context-dependent functions, and could be differentially regulated during tumor progression. In the present study, we identified the multi-functional transcription factor Yin Yang 1 (YY1) as a modulator of PSCA expression in prostate epithelial cell lines. Increased YY1 levels are observed in prostatic intraepithelial neoplasia (PIN) and advanced disease. We show that androgen-mediated up-regulation of PSCA in prostate epithelial cell lines is dependent on YY1. We identified two direct YY1 binding sites within the PSCA promoter, and showed that the upstream site inhibited, while the downstream site, proximal to the androgen-responsive element, stimulated PSCA promoter activity. Thus, changes in PSCA expression levels in prostate cancer may at least partly be affected by cellular levels of YY1. Our results also suggest multiple roles for YY1 in prostate cancer which may contribute to disease progression by modulation of genes such as PSCA.
- Published
- 2012
- Full Text
- View/download PDF
5. Transgenic and Knockout Mice Models to Reveal the Functions of Tumor Suppressor Genes
- Author
-
Pankaj Taneja, Sinan Zhu, Dejan Maglic, Eizabeth A. Fry, Robert D. Kendig, and Kazushi Inoue
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Cancer is caused by multiple genetic alterations leading to uncontrolled cell proliferation through multiple pathways. Malignant cells arise from a variety of genetic factors, such as mutations in tumor suppressor genes (TSGs) that are involved in regulating the cell cycle, apoptosis, or cell differentiation, or maintenance of genomic integrity. Tumor suppressor mouse models are the most frequently used animal models in cancer research. The anti-tumorigenic functions of TSGs, and their role in development and differentiation, and inhibition of oncogenes are discussed. In this review, we summarize some of the important transgenic and knockout mouse models for TSGs, including Rb, p53, Ink4a/Arf, Brca1/2 , and their related genes.
- Published
- 2011
- Full Text
- View/download PDF
6. Classical and Novel Prognostic Markers for Breast Cancer and their Clinical Significance
- Author
-
Pankaj Taneja, Dejan Maglic, Fumitake Kai, Sinan Zhu, Robert D. Kendig, A. Fry Elizabeth, and Kazushi Inoue
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The use of biomarkers ensures breast cancer patients receive optimal treatment. Established biomarkers such as estrogen receptor (ER) and progesterone receptor (PR) have been playing significant roles in the selection and management of patients for endocrine therapy. HER2 is a strong predictor of response to trastuzumab. Recently, the roles of ER as a negative and HER2 as a positive indicator for chemotherapy have been established. Ki67 has traditionally been recognized as a poor prognostic factor, but recent studies suggest that measurement of Ki67-positive cells during treatment will more effectively predict treatment efficacy for both anti-hormonal and chemotherapy. p53 mutations are found in 20–35% of human breast cancers and are associated with aggressive disease with poor clinical outcome when the DNA-binding domain is mutated. The utility of cyclin D1 as a predictor of breast cancer prognosis is controversial, but cyclin D1b overexpression is associated with poor prognosis. Likewise, overexpression of the low molecular weight form of cyclin E1 protein predicts poor prognosis. Breast cancers from BRCA1/2 carriers often show high nuclear grades, negativity to ER/PR/HER2, and p53 mutations, and thus, are associated with poor prognosis. The prognostic values of other molecular markers, such as p14 ARF , TBX2/3, VEGF in breast cancer are also discussed. Careful evaluation of these biomarkers with current treatment modality is required to determine whether their measurement or monitoring offer significant clinical benefits.
- Published
- 2010
- Full Text
- View/download PDF
7. Signal Transduction Involving the Dmp1 Transcription Factor and its Alteration in Human Cancer
- Author
-
Takayuki Sugiyama, Donna P. Frazier, Pankaj Taneja, Robert D. Kendig, Rachel L. Morgan, Lauren A. Matise, Sarah J. Lagedrost, and Kazushi Inoue
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Summary Dmp1 (cyclin D-interacting myb-like protein 1; also called Dmtf1) is a transcription factor that has been isolated in a yeast two-hybrid screen through its binding property to cyclin D2. Dmp1 directly binds to and activates the Arf promoter and induces Arf-p53-dependent cell cycle arrest in primary cells. D-type cyclins usually inhibit Dmp1-mediated transcription in a Cdk-independent fashion; however, Dmp1 shows synergistic effects with D-cyclins on the Arf promoter. Ras or Myc oncogene-induced tumor formation is accelerated in both Dmp1 +/- and Dmp1 -/- mice with no significant differences between Dmp1 +/- and Dmp1 -/- . Thus, Dmp1 is haplo-insufficient for tumor suppression. Tumors from Dmp1 -/- or Dmp1 +/- mice often retain wild-type Arf and p53 , suggesting that Dmp1 is a physiological regulator of the Arf-p53 pathway. The Dmp1 promoter is activated by oncogenic Ras-Raf signaling, while it is repressed by physiological mitogenic stimuli, overexpression of E2F proteins, and genotoxic stimuli mediated by NF-κB. The human DMP1 gene (h DMP1 ) is located on chromosome 7q21 and is hemizygously deleted in approximately 40% of human lung cancers, especially those that retain normal INK4a/ARF and P53 loci. Thus, h DMP1 is clearly involved in human carcinogenesis, and tumors with h DMP1 deletion may constitute a discrete disease entity.
- Published
- 2008
- Full Text
- View/download PDF
8. Supplementary Figure 3 from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Author
-
Eitan Shaulian, Kazushi Inoue, Shira Anzi, Keren Saadon, and Orli Yogev
- Abstract
Supplementary Figure 3 from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 1 from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Author
-
Eitan Shaulian, Kazushi Inoue, Shira Anzi, Keren Saadon, and Orli Yogev
- Abstract
Supplementary Figure 1 from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figures 1-12 from Dmp1 Physically Interacts with p53 and Positively Regulates p53′s Stability, Nuclear Localization, and Function
- Author
-
Kazushi Inoue, Guangchao Sui, Sarah J. Lagedrost, Elizabeth A. Fry, Rachel L. Morgan, Takayuki Sugiyama, Dejan Maglic, Fumitake Kai, Robert D. Kendig, and Donna P. Frazier
- Abstract
PDF file - 786K
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Methods from Dmp1 Physically Interacts with p53 and Positively Regulates p53′s Stability, Nuclear Localization, and Function
- Author
-
Kazushi Inoue, Guangchao Sui, Sarah J. Lagedrost, Elizabeth A. Fry, Rachel L. Morgan, Takayuki Sugiyama, Dejan Maglic, Fumitake Kai, Robert D. Kendig, and Donna P. Frazier
- Abstract
PDF file - 49K
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Methods, Figures 1-6 from Critical Roles of DMP1 in Human Epidermal Growth Factor Receptor 2/neu-Arf-p53 Signaling and Breast Cancer Development
- Author
-
Kazushi Inoue, Mark C. Willingham, Donna P. Frazier, Robert D. Kendig, Takayuki Sugiyama, Fumitake Kai, Dejan Maglic, and Pankaj Taneja
- Abstract
Supplementary Methods, Figures 1-6 from Critical Roles of DMP1 in Human Epidermal Growth Factor Receptor 2/neu-Arf-p53 Signaling and Breast Cancer Development
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figure 5 from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Author
-
Eitan Shaulian, Kazushi Inoue, Shira Anzi, Keren Saadon, and Orli Yogev
- Abstract
Supplementary Figure 5 from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure 2 from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Author
-
Eitan Shaulian, Kazushi Inoue, Shira Anzi, Keren Saadon, and Orli Yogev
- Abstract
Supplementary Figure 2 from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Published
- 2023
- Full Text
- View/download PDF
15. Data from Critical Roles of DMP1 in Human Epidermal Growth Factor Receptor 2/neu-Arf-p53 Signaling and Breast Cancer Development
- Author
-
Kazushi Inoue, Mark C. Willingham, Donna P. Frazier, Robert D. Kendig, Takayuki Sugiyama, Fumitake Kai, Dejan Maglic, and Pankaj Taneja
- Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression stimulates cell growth in p53-mutated cells while it inhibits cell proliferation in those with wild-type p53, but the molecular mechanism is unknown. The Dmp1 promoter was activated by HER2/neu through the phosphatidylinositol-3′-kinase-Akt-NF-κB pathway, which in turn stimulated Arf transcription. Binding of p65 and p52 subunits of NF-κB was shown to the Dmp1 promoter and that of Dmp1 to the Arf promoter on HER2/neu overexpression. Both Dmp1 and p53 were induced in premalignant lesions from mouse mammary tumor virus-neu mice, and mammary tumorigenesis was significantly accelerated in both Dmp1+/− and Dmp1−/− mice. Selective deletion of Dmp1 and/or overexpression of Tbx2/Pokemon was found in >50% of wild-type HER2/neu carcinomas, although the involvement of Arf, Mdm2, or p53 was rare. Tumors from Dmp1+/−, Dmp1−/−, and wild-type neu mice with hemizygous Dmp1 deletion showed significant downregulation of Arf and p21Cip1/WAF1, showing p53 inactivity and more aggressive phenotypes than tumors without Dmp1 deletion. Notably, endogenous hDMP1 mRNA decreased when HER2 was depleted in human breast cancer cells. Our study shows the pivotal roles of Dmp1 in HER2/neu-p53 signaling and breast carcinogenesis. Cancer Res; 70(22); 9084–94. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
16. Data from Dmp1 Physically Interacts with p53 and Positively Regulates p53′s Stability, Nuclear Localization, and Function
- Author
-
Kazushi Inoue, Guangchao Sui, Sarah J. Lagedrost, Elizabeth A. Fry, Rachel L. Morgan, Takayuki Sugiyama, Dejan Maglic, Fumitake Kai, Robert D. Kendig, and Donna P. Frazier
- Abstract
The transcription factor Dmp1 is a Ras/HER2-activated haplo-insufficient tumor suppressor that activates the Arf/p53 pathway of cell-cycle arrest. Recent evidence suggests that Dmp1 may activate p53 independently of Arf in certain cell types. Here, we report findings supporting this concept with the definition of an Arf-independent function for Dmp1 in tumor suppression. We found that Dmp1 and p53 can interact directly in mammalian cells via the carboxyl-terminus of p53 and the DNA-binding domain of Dmp1. Expression of Dmp1 antagonized ubiquitination of p53 by Mdm2 and promoted nuclear localization of p53. Dmp1–p53 binding significantly increased the level of p53, independent of the DNA-binding activity of Dmp1. Mechanistically, p53 target genes were activated synergistically by the coexpression of Dmp1 and p53 in p53−/−;Arf−/− cells, and genotoxic responses of these genes were hampered more dramatically in Dmp1−/− and p53−/− cells than in Arf−/− cells. Together, our findings identify a robust new mechanism of p53 activation mediated by direct physical interaction between Dmp1 and p53. Cancer Res; 72(7); 1740–50. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figure 4 from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Author
-
Eitan Shaulian, Kazushi Inoue, Shira Anzi, Keren Saadon, and Orli Yogev
- Abstract
Supplementary Figure 4 from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Published
- 2023
- Full Text
- View/download PDF
18. Data from DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Author
-
Eitan Shaulian, Kazushi Inoue, Shira Anzi, Keren Saadon, and Orli Yogev
- Abstract
The dynamic behavior of the nucleolus plays a role in the detection of and response to DNA damage of cells. Two nucleolar proteins, p14ARF/p19ARF and B23, were shown to translocate out of the nucleolus after exposure of cells to DNA-damaging agents. This translocation affects multiple cellular functions, such as DNA repair, proliferation, and survival. In this study, we identify a pathway and scrutinize the mechanisms leading to the translocation of these proteins after exposure of cells to DNA-damaging agents. We show that redistribution of B23 and p19ARF after the exposure to genotoxic stress occurs preferentially when the c-Jun-NH2-kinase (JNK) pathway is activated and is inhibited when the JNK pathway is impaired. The stress-induced translocation of alternative reading frame (ARF) is JNK dependent and mediated by two activator proteins, c-Jun and JunB. Thr91 and Thr93 of c-Jun are required for the translocation, but the transcriptional activity of c-Jun is dispensable. Instead, c-Jun interacts with B23 in a dose-dependent manner. c-Jun itself is excluded from the nucleolus in a JNK-dependent manner. Hence, we suggest that c-Jun translocates B23 and ARF from the nucleolus after JNK activation by means of protein interactions. In senescent cells, JNK activity and c-Jun levels are reduced concomitantly with ARF nucleolar accumulation, and UV radiation does not cause the translocation of ARF. [Cancer Res 2008;68(5):1398–406]
- Published
- 2023
- Full Text
- View/download PDF
19. Mechanisms regulating DMTF1β/γ expression and their functional interplay with DMTF1α
- Author
-
Kuida Chen, Tianqi Xu, Dangdang Li, Jialiang Li, Ke Shi, Jingru Hu, Guangyue Li, Guangchao Sui, Tianxin Li, and Kazushi Inoue
- Subjects
0301 basic medicine ,Gene isoform ,Cancer Research ,cyclin D binding myb-like transcription factor 1 ,Tumor suppressor gene ,Carcinogenesis ,Cyclin D ,Breast Neoplasms ,Biology ,alternative splicing ,03 medical and health sciences ,Transactivation ,breast cancer ,0302 clinical medicine ,Cell Line, Tumor ,Gene expression ,Humans ,Protein Isoforms ,Promoter Regions, Genetic ,Transcription factor ,Cell Proliferation ,Cell Nucleus ,Alternative splicing ,isoforms ,Articles ,Cell cycle ,Cell biology ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,gene expression ,biology.protein ,Female ,Transcription Factors - Abstract
The cyclin D binding myb‑like transcription factor 1 (DMTF1), a haplo‑insufficient tumor suppressor gene, has 3 alternatively spliced mRNA isoforms encoding DMTF1α, β and γ proteins. Previous studies have indicated a tumor suppressive role of DMTF1α and the oncogenic activity of DMTF1β, while the function of DMTF1γ remains largely undetermined. In the present study, the mechanisms regulating DMTF1 isoform expression were investigated and the functional interplay of DMTF1β and γ with DMTF1α was characterized. It was found that specific regions of DMTF1β and γ transcripts can impair their mRNA integrity or stability, and thus reduce protein expression levels. Additionally, DMTF1β and γ proteins exhibited a reduced stability compared to DMTF1α and all 3 DMTF1 isoforms were localized in the nuclei. Two basic residues, K52 and R53, in the DMTF1 isoforms determined their nuclear localization. Importantly, both DMTF1β and γ could associate with DMTF1α and antagonize its transactivation of the ARF promoter. Consistently, the ratios of both DMTF1β/α and γ/α were significantly associated with a poor prognoses of breast cancer patients, suggesting oncogenic roles of DMTF1β and γ isoforms in breast cancer development.
- Published
- 2020
- Full Text
- View/download PDF
20. c-MYB and DMTF1 in Cancer
- Author
-
Kazushi Inoue and Elizabeth A. Fry
- Subjects
0301 basic medicine ,Cancer Research ,animal structures ,Cyclin D ,Translocation, Genetic ,Article ,Small Molecule Libraries ,Proto-Oncogene Proteins c-myb ,03 medical and health sciences ,Transactivation ,0302 clinical medicine ,Cyclin D2 ,Neoplasms ,medicine ,Transcriptional regulation ,Humans ,MYB ,Promoter Regions, Genetic ,Transcription factor ,Hemizygote ,Oncogene ,biology ,ADP-Ribosylation Factors ,Chemistry ,General Medicine ,Oligonucleotides, Antisense ,medicine.disease ,Survival Analysis ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Leukemia ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Transcription Factors - Abstract
The c-Myb gene encodes a transcription factor that regulates cell proliferation, differentiation, and apoptosis through protein-protein interaction and transcriptional regulation of signaling pathways. The protein is frequently overexpressed in human leukemias, breast cancers, and other solid tumors suggesting that it is a bona fide oncogene. c-MYB is often overexpressed by translocation in human tumors with t(6;7)(q23;q34) resulting in c-MYB-TCRβ in T cell ALL, t(X;6)(p11;q23) with c-MYB-GATA1 in acute basophilic leukemia, and t(6;9)(q22–23;p23–24) with c-MYB-NF1B in adenoid cystic carcinoma. Antisense oligonucleotides to c-MYB were developed to purge bone marrow cells to eliminate tumor cells in leukemias. Recently small molecules that inhibit c-MYB activity have been developed to disrupt its interaction with p300. The Dmp1 (cyclin D binding myb-like protein 1; Dmtf1) gene was isolated through its virtue for binding to cyclin D2. It is a transcription factor that has a Myb-like repeat for DNA binding. The Dmtf1 protein directly binds to the Arf promoter for transactivation and physically interacts with p53 to activate the p53 pathway. The gene is hemizygously deleted in 35–42% of human cancers, and is associated with longer survival. The significances of aberrant expression of c-MYB and DMTF1 proteins in human cancers and their clinical significances are discussed.
- Published
- 2019
- Full Text
- View/download PDF
21. The oncogenicity of tumor-derived mutant p53 is enhanced by the recruitment of PLK3
- Author
-
Shilpa Singh, Brad Windle, Sumitra Deb, Elizabeth A. Fry, Jolene J. Windle, W. Andrew Yeudall, Kazushi Inoue, Swati Palit Deb, Catherine A. Vaughan, Steven R. Grossman, Mark A. Subler, and Raghavendra Pillappa
- Subjects
0301 basic medicine ,Transcriptional Activation ,Lung Neoplasms ,Carcinogenesis ,Science ,Mutant ,General Physics and Astronomy ,Mice, Transgenic ,Oncogenicity ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,PLK3 ,Serine ,03 medical and health sciences ,Transactivation ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Protein Domains ,Cell Line, Tumor ,Spheroids, Cellular ,medicine ,Animals ,Humans ,Gene Knock-In Techniques ,Phosphorylation ,Tumour-suppressor proteins ,Lung ,Mutation ,Multidisciplinary ,Chemistry ,Tumor Suppressor Proteins ,General Chemistry ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,Mechanisms of disease ,030220 oncology & carcinogenesis ,Gain of Function Mutation ,Female ,Lung cancer ,Tumor Suppressor Protein p53 - Abstract
p53 mutations with single amino acid changes in cancer often lead to dominant oncogenic changes. Here, we have developed a mouse model of gain-of-function (GOF) p53-driven lung cancer utilizing conditionally active LSL p53-R172H and LSL K-Ras-G12D knock-in alleles that can be activated by Cre in lung club cells. Mutation of the p53 transactivation domain (TAD) (p53-L25Q/W26S/R172H) eliminating significant transactivation activity resulted in loss of tumorigenicity, demonstrating that transactivation mediated by or dependent on TAD is required for oncogenicity by GOF p53. GOF p53 TAD mutations significantly reduce phosphorylation of nearby p53 serine 20 (S20), which is a target for PLK3 phosphorylation. Knocking out PLK3 attenuated S20 phosphorylation along with transactivation and oncogenicity by GOF p53, indicating that GOF p53 exploits PLK3 to trigger its transactivation capability and exert oncogenic functions. Our data show a mechanistic involvement of PLK3 in mutant p53 pathway of oncogenesis., The mechanisms of how gain-of-function (GOF) mutant p53 drives carcinogenesis are unclear. Here, the authors show that a GOF mutant p53 requires its transactivation capability to induce mouse lung tumors and this is dependent on PLK3 phosphorylation of GOF mutant p53.
- Published
- 2021
22. Survival of Lung Cancer Patients Dependent on the LOH Status for DMP1, ARF, and p53
- Author
-
Fumitake Kai, Robert A. Kratzke, Elizabeth A. Fry, Gloria E Niehans, and Kazushi Inoue
- Subjects
Male ,0301 basic medicine ,Oncology ,p53 ,Lung Neoplasms ,Loss of Heterozygosity ,Favorable prognosis ,YY1 ,lcsh:Chemistry ,Loss of heterozygosity ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Cyclin D1 ,lcsh:QH301-705.5 ,Spectroscopy ,ARF ,General Medicine ,Prognosis ,University hospital ,Computer Science Applications ,030220 oncology & carcinogenesis ,Female ,DMTF1 ,medicine.medical_specialty ,Locus (genetics) ,Article ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,Breast cancer ,Internal medicine ,medicine ,Humans ,Physical and Theoretical Chemistry ,Adverse effect ,Lung cancer ,Molecular Biology ,neoplasms ,INK4a ,Cyclin-Dependent Kinase Inhibitor p16 ,business.industry ,Organic Chemistry ,medicine.disease ,Survival Analysis ,respiratory tract diseases ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Tumor Suppressor Protein p53 ,business ,Transcription Factors - Abstract
Lung cancer is the leading cause of cancer deaths in the world, and accounts for more solid tumor deaths than any other carcinomas. The prognostic values of DMP1, ARF, and p53-loss are unknown in lung cancer. We have conducted survival analyses of non-small cell lung cancer (NSCLC) patients from the University of Minnesota VA hospital and those from the Wake Forest University Hospital. Loss of Heterozygosity (LOH) for hDMP1 was found in 26 of 70 cases (37.1%), that of the ARF/INK4a locus was found in 33 of 70 (47.1%), and that of the p53 locus in 43 cases (61.4%) in the University of Minnesota samples. LOH for hDMP1 was associated with favorable prognosis while that of p53 predicted worse prognosis. The survival was much shorter for ARF-loss than INK4a-loss, emphasizing the importance of ARF in human NSCLC. The adverse effect of p53 LOH on NSCLC patients&rsquo, survival was neutralized by simultaneous loss of the hDMP1 locus in NSCLC and breast cancer, suggesting the possible therapy of epithelial cancers with metastatic ability.
- Published
- 2020
- Full Text
- View/download PDF
23. Oncogenic and tumor-suppressive mouse models for breast cancer engaging HER2/neu
- Author
-
Elizabeth A. Fry, Pankaj Taneja, and Kazushi Inoue
- Subjects
0301 basic medicine ,Genetically modified mouse ,Cancer Research ,biology ,medicine.disease_cause ,Molecular biology ,HER2/neu ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cyclin D1 ,Oncology ,Trastuzumab ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,medicine ,Cancer research ,skin and connective tissue diseases ,Carcinogenesis ,neoplasms ,Monoclonal antibody therapy ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
The human c-ErbB2 (HER2) gene is amplified in ∼20% of human breast cancers (BCs), but the protein is overexpressed in ∼30% of the cases indicating that multiple different mechanisms contribute to HER2 overexpression in tumors. It has long been used as a molecular marker of BC for subcategorization for the prediction of prognosis and determination of therapeutic strategies. In comparison to ER(+) BCs, HER2-positive BCs are more invasive, but the patients respond to monoclonal antibody therapy with trastuzumab or tyrosine kinase inhibitors at least at early stages. To understand the pathophysiology of HER2-driven carcinogenesis and test HER2-targeting therapeutic agents in vivo, numerous mouse models have been created that faithfully reproduce HER2(+) BCs in mice. They include MMTV-neu (active mutant or wild type, rat neu or HER2) models, neu promoter-driven neuNT-transgenic mice, neuNT-knock-in mice at the neu locus and doxycycline-inducible neuNT-transgenic models. HER2/neu activates the Phosphatidylinositol-3 kinase-AKT-NF-κB pathway to stimulate the mitogenic cyclin D1/Cdk4-Rb-E2F pathway. Of note, overexpression of HER2 also stimulates the cell autonomous Dmp1-Arf-p53 tumor suppressor pathway to quench oncogenic signals to prevent the emergence of cancer cells. Hence tumor development by MMTV-neu mice was dramatically accelerated in mice that lack Dmp1, Arf or p53 with invasion and metastasis. Expressions of neuNT under the endogenous promoter underwent gene amplification, closely recapitulating human HER2(+) BCs. MMTV-HER2 models have been shown to be useful to test humanized monoclonal antibodies to HER2. These mouse models will be useful for the screening of novel therapeutic agents against BCs with HER2 overexpression.
- Published
- 2016
- Full Text
- View/download PDF
24. Novel Molecular Markers for Breast Cancer
- Author
-
Kazushi Inoue and Elizabeth A. Fry
- Subjects
0301 basic medicine ,Microbiology (medical) ,Immunology ,B-Myb ,Estrogen receptor ,Review ,Bioinformatics ,lcsh:RC254-282 ,Metastasis ,DMP1β ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Progesterone receptor ,medicine ,Immunology and Allergy ,cyclin E ,metastasis ,Aromatase ,Twist ,epithelial–mesenchymal transition (EMT) ,therapy ,biology ,business.industry ,Cancer ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,In vitro ,3. Good health ,030104 developmental biology ,Hormone receptor ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,prognosis ,business ,DMTF1 - Abstract
The use of molecular biomarkers assures that breast cancer (BC) patients receive optimal treatment. Established biomarkers, such as estrogen receptor, progesterone receptor, HER2, and Ki67, have been playing significant roles in the subcategorization of BC to predict the prognosis and decide the specific therapy to each patient. Antihormonal therapy using 4-hydroxytamoxifen or aromatase inhibitors have been employed in patients whose tumor cells express hormone receptors, while monoclonal antibody to HER2 has been administered to HER2-positive BCs. Although new therapeutic agents have been developed in the past few decades, many patients still die of the disease due to relapse; thus, novel molecular markers that predict therapeutic failure and those that can be targets for specific therapy are expected. We have chosen four of such molecules by reviewing recent publications, which are cyclin E, B-Myb, Twist, and DMP1β. The oncogenicity of these molecules has been demonstrated in vivo and/or in vitro through studies using transgenic mice or siRNAs, and their expressions have been shown to be associated with shortened overall or disease-free survival of BC patients. The former three molecules have been shown to accelerate epithelial-mesenchymal transition that is often associated with cancer stem cell-ness and metastasis; all these four can be novel therapeutic targets as well. Thus, large prospective studies employing immunohistochemistry will be needed to establish the predictive values of these molecules in patients with BC.
- Published
- 2016
25. Aberrant splicing of the DMP1-ARF-MDM2-p53 pathway in cancer
- Author
-
Elizabeth A. Fry and Kazushi Inoue
- Subjects
0301 basic medicine ,Cancer Research ,Tumor suppressor gene ,Alternative splicing ,Biology ,medicine.disease ,medicine.disease_cause ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,RNA splicing ,Cancer cell ,medicine ,Cancer research ,Neoplastic transformation ,Carcinogenesis ,Gene - Abstract
Alternative splicing (AS) of mRNA precursors is a ubiquitous mechanism for generating numerous transcripts with different activities from one genomic locus in mammalian cells. The gene products from a single locus can thus have similar, dominant-negative or even opposing functions. Aberrant AS has been found in cancer to express proteins that promote cell growth, local invasion and metastasis. This review will focus on the aberrant splicing of tumor suppressor/oncogenes that belong to the DMP1-ARF-MDM2-p53 pathway. Our recent study shows that the DMP1 locus generates both tumor-suppressive DMP1α (p53-dependent) and oncogenic DMP1β (p53-independent) splice variants, and the DMP1β/α ratio increases with neoplastic transformation of breast epithelial cells. This process is associated with high DMP1β protein expression and shorter survival of breast cancer (BC) patients. Accumulating pieces of evidence show that ARF is frequently inactivated by aberrant splicing in human cancers, demonstrating its involvement in human malignancies. Splice variants from the MDM2 locus promote cell growth in culture and accelerate tumorigenesis in vivo. Human cancers expressing these splice variants are associated with advanced stage/metastasis, and thus have negative clinical impacts. Although they lack most of the p53-binding domain, their activities are mostly dependent on p53 since they bind to wild-type MDM2. The p53 locus produces splice isoforms that have either favorable (β/γ at the C-terminus) or negative impact (Δ40, Δ133 at the N-terminus) on patients' survival. As the oncogenic AS products from these loci are expressed only in cancer cells, they may eventually become targets for molecular therapies.
- Published
- 2016
- Full Text
- View/download PDF
26. Tumor suppression by the EGR1, DMP1, ARF, p53, and PTEN Network
- Author
-
Elizabeth A. Fry and Kazushi Inoue
- Subjects
0301 basic medicine ,Cancer Research ,endocrine system ,biology ,Chemistry ,Regulator ,EGR1 ,General Medicine ,Article ,law.invention ,Cell biology ,03 medical and health sciences ,Transactivation ,030104 developmental biology ,0302 clinical medicine ,Oncology ,stomatognathic system ,law ,Transcription (biology) ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Suppressor ,PTEN ,Transcription factor - Abstract
Recent studies have indicated that EGR1 is a direct regulator of tumor suppressors including TGFβ1, PTEN, and p53. The Myb-like transcription factor Dmp1 is a physiological regulator of the Arf-p53 pathway through transactivation of the Arf promoter and physical interaction of p53. The Dmp1 promoter has binding sites for Egr proteins, and Egr1 is a target for Dmp1. Crosstalks between p53 and PTEN have been reported. The Egr1-Dmp1-Arf-p53-Pten pathway displays multiple modes of interaction with each other, suggesting the existence of a functional network of tumor suppressors that maintain normal cell growth and prevent the emergence of incipient cancer cells.
- Published
- 2018
27. Aberrant expression of p16
- Author
-
Kazushi, Inoue and Elizabeth A, Fry
- Subjects
neoplasms ,Article - Abstract
The ARF and INK4a genes are located in the same CDKN2a locus, both showing its tumor suppressive activity. ARF has been shown to detect potentially harmful oncogenic signals, making incipient cancer cells undergo senescence or apoptosis. INK4a, on the other hand, responds to signals from aging in a variety of tissues including islets of Langerhans, neuronal cells, and cancer stem cells in general. It also detects oncogenic signals from incipient cancer cells to induce them senescent to prevent neoplastic transformation. Both of these genes are inactivated by gene deletion, promoter methylation, frame shift, and aberrant splicing although mutations changing the amino acid sequences affect only the latter. Recent studies indicated that polycomb gene products EZH2 and BMI1 repressed p16INK4a expression in primary cells, but not in cells deficient for pRB protein function. It was also reported that that p14ARF inhibits the stability of the p16INK4a protein in human cancer cell lines and mouse embryonic fibroblasts through its interaction with regenerating islet-derived protein 3γ. Overexpression of INK4a is associated with better prognosis of cancer when it is associated with human papilloma virus infection. However, it has a worse prognostic value in other tumors since it is an indicator of pRB loss. The p16INK4a tumor suppressive protein can thus be used as a biomarker to detect early stage cancer cells as well as advanced tumor cells with pRB inactivation since it is not expressed in normal cells.
- Published
- 2018
28. Aberrant Expression of p14ARF in Human Cancers: A New Biomarker?
- Author
-
Elizabeth A. Fry and Kazushi Inoue
- Subjects
0301 basic medicine ,biology ,Article ,Ubiquitin ligase ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,p14arf ,BMI1 ,CDKN2A ,Cancer cell ,biology.protein ,Cancer research ,Nuclear protein ,E2F ,Transcription factor ,030217 neurology & neurosurgery - Abstract
The ARF and INK4a genes are located on the CDKN2a locus, both showing tumor suppressive activity. ARF has been shown to monitor potentially harmful oncogenic signalings, making early stage cancer cells undergo senescence or programmed cell death to prevent cancer. Conversely, INK4a detects both aging and incipient cancer cell signals, and thus these two gene functions are different. The efficiency of detection of oncogenic signals is more efficient for the for the former than the latter in the mouse system. Both ARF and INK4a genes are inactivated by gene deletion, promoter methylation, frame shift, aberrant splicing although point mutations for the coding region affect only the latter. Recent studies show the splicing alterations that affect only ARF or both ARF and INK4a genes suggesting that ARF is inactivated in human tumors more frequently than what was previously thought. The ARF gene is activated by E2Fs and Dmp1 transcription factors while it is repressed by Bmi1, Tbx2/3, Twist1, and Pokemon nuclear proteins. It is also regulated at protein levels by Arf ubiquitin ligase named ULF, MKRN1, and Siva1. The prognostic value of ARF overexpression is controversial since it is induced in early stage cancer cells to eliminate pre-malignant cells (better prognosis); however, it may also indicate that the tumor cells have mutant p53 associated with worse prognosis. The ARF tumor suppressive protein can be used as a biomarker to detect early stage cancer cells as well as advanced stage tumors with p53 inactivation.
- Published
- 2018
29. Translocations involving ETS family proteins in human cancer
- Author
-
Elizabeth A. Fry, Ali Mallakin, and Kazushi Inoue
- Subjects
0301 basic medicine ,Chimeric gene ,Biology ,medicine.disease_cause ,TMPRSS2 ,Article ,03 medical and health sciences ,ETV6 ,030104 developmental biology ,0302 clinical medicine ,ETS1 ,030220 oncology & carcinogenesis ,FLI1 ,medicine ,Cancer research ,Carcinogenesis ,Gene ,Transcription factor - Abstract
The ETS transcription factors regulate expression of genes involved in normal cell development, proliferation, differentiation, angiogenesis, and apoptosis, consisting of 28 family members in humans. Dysregulation of these transcription factors facilitates cell proliferation in cancers, and several members participate in invasion and metastasis by activating certain gene transcriptions. ETS1 and ETS2 are the founding members of the ETS family and regulate transcription by binding to ETS sequences. Three chimeric genes involving ETS genes have been identified in human cancers, which are EWS-FLI1 in Ewing’s sarcoma, TMPRSS2-ERG in prostate cancer, and ETV6-RUNX1 in acute lymphocytic leukemia. Although these fusion transcripts definitely contribute to the pathogenesis of the disease, the impact of these fusion transcripts on patients’ prognosis is highly controversial. In the present review, the roles of ETS protein translocations in human carcinogenesis are discussed.
- Published
- 2018
- Full Text
- View/download PDF
30. Biochemical characterization of ratfish (Chimaera monstrosa) liver oil; cytotoxic and antineoplastic evaluation in cancer cell lines
- Author
-
Ali Mallakin, Pankaj Taneja, Hans-Uwe Dahms, Natalie Anne Ward, and Kazushi Inoue
- Subjects
Cytotoxic T cell ,Chimera (mythology) ,Biology ,Cancer cell lines ,Molecular biology - Published
- 2018
- Full Text
- View/download PDF
31. Aberrant expression of ETS1 and ETS2 proteins in cancer
- Author
-
Elizabeth A. Fry and Kazushi Inoue
- Subjects
Telomerase ,Cell growth ,Wild type ,General Medicine ,Biology ,medicine.disease_cause ,Article ,Cell biology ,ETS1 ,Transcription (biology) ,medicine ,Carcinogenesis ,Gene ,Transcription factor - Abstract
The ETS transcription factors regulate expression of genes involved in normal cell development, proliferation, differentiation, angiogenesis, and apoptosis, consisting of 28 family members in humans. Dysregulation of these transcription factors facilitates cell proliferation in cancers, and several members participate in invasion and metastasis by activating gene transcription. ETS1 and ETS2 are the founding members of the ETS family and regulate transcription by binding to ETS sequences. They are both involved in oncogenesis and tumor suppression depending on the biological situations used. The essential roles of ETS proteins in human telomere maintenance have been suggested, which have been linked to creation of new Ets binding sites. Recently, preferential binding of ETS2 to gain-of-function mutant p53 and ETS1 to wild type p53 (WTp53) has been suggested, raising the tumor promoting role for the former and tumor suppressive role for the latter. The oncogenic and tumor suppressive functions of ETS1 and 2 proteins have been discussed.
- Published
- 2018
- Full Text
- View/download PDF
32. Aberrant Splicing of Estrogen Receptor, HER2, and CD44 Genes in Breast Cancer
- Author
-
Elizabeth A. Fry and Kazushi Inoue
- Subjects
lcsh:QH426-470 ,Estrogen receptor ,Review ,medicine.disease_cause ,Bioinformatics ,Biochemistry ,03 medical and health sciences ,Exon ,alternative splicing ,0302 clinical medicine ,breast cancer ,HER2 ,Genetics ,Medicine ,metastasis ,CD44 ,Gene ,030304 developmental biology ,0303 health sciences ,biology ,business.industry ,Alternative splicing ,3. Good health ,stem cell ,lcsh:Genetics ,tumorigenesis ,030220 oncology & carcinogenesis ,RNA splicing ,biology.protein ,prognosis ,Signal transduction ,business ,Carcinogenesis ,signal transduction ,estrogen receptor - Abstract
Breast cancer (BC) is the most common cause of cancer-related death among women under the age of 50 years. Established biomarkers, such as hormone receptors (estrogen receptor [ER]/progesterone receptor) and human epidermal growth factor receptor 2 (HER2), play significant roles in the selection of patients for endocrine and trastuzumab therapies. However, the initial treatment response is often followed by tumor relapse with intrinsic resistance to the first-line therapy, so it has been expected to identify novel molecular markers to improve the survival and quality of life of patients. Alternative splicing of pre-messenger RNAs is a ubiquitous and flexible mechanism for the control of gene expression in mammalian cells. It provides cells with the opportunity to create protein isoforms with different, even opposing, functions from a single genomic locus. Aberrant alternative splicing is very common in cancer where emerging tumor cells take advantage of this flexibility to produce proteins that promote cell growth and survival. While a number of splicing alterations have been reported in human cancers, we focus on aberrant splicing of ER, HER2, and CD44 genes from the viewpoint of BC development. ERα36, a splice variant from the ER1 locus, governs nongenomic membrane signaling pathways triggered by estrogen and confers 4-hydroxytamoxifen resistance in BC therapy. The alternative spliced isoform of HER2 lacking exon 20 (Δ16HER2) has been reported in human BC; this isoform is associated with transforming ability than the wild-type HER2 and recapitulates the phenotypes of endocrine therapy-resistant BC. Although both CD44 splice isoforms ( CD44s, CD44v) play essential roles in BC development, CD44v is more associated with those with favorable prognosis, such as luminal A subtype, while CD44s is linked to those with poor prognosis, such as HER2 or basal cell subtypes that are often metastatic. Hence, the detection of splice variants from these loci will provide keys to understand the pathogenesis, predict the prognosis, and choose specific therapies for BC.
- Published
- 2015
33. DMP1β, a splice isoform of the tumour suppressorDMP1locus, induces proliferation and progression of breast cancer
- Author
-
Guangchao Sui, Ali Mallakin, Daniel B. Stovall, Elizabeth A. Fry, Mark C. Willingham, J. Mark Cline, Pankaj Taneja, Kazushi Inoue, Dejan Maglic, and David L. Caudell
- Subjects
Gene isoform ,Genetically modified mouse ,Gene knockdown ,Pathology ,medicine.medical_specialty ,Cell growth ,Alternative splicing ,Transdifferentiation ,Biology ,medicine.disease ,Pathology and Forensic Medicine ,Breast cancer ,Cancer cell ,Cancer research ,medicine - Abstract
Our recent work has indicated that the DMP1 locus on 7q21, encoding a haplo-insufficient tumour suppressor, is hemizygously deleted at a high frequency in breast cancer. The locus encodes DMP1α protein, an activator of the p53 pathway leading to cell cycle arrest and senescence, and two other functionally undefined isoforms, DMP1β and DMP1γ. In this study, we show that the DMP1 locus is alternatively spliced in ∼30% of breast cancer cases with relatively decreased DMP1α and increased DMP1β expression. RNA-seq analyses of a publicly available database showed significantly increased DMP1β mRNA in 43-55% of human breast cancers, dependent on histological subtypes. Similarly, DMP1β protein was found to be overexpressed in ∼60% of tumours relative to their surrounding normal tissue. Importantly, alteration of DMP1 splicing and DMP1β overexpression were associated with poor clinical outcomes of the breast cancer patients, indicating that DMP1β may have a biological function. Indeed, DMP1β increased proliferation of non-tumourigenic mammary epithelial cells and knockdown of endogenous DMP1 inhibited breast cancer cell growth. To determine DMP1β's role in vivo, we established MMTV-DMP1β transgenic mouse lines. DMP1β overexpression was sufficient to induce mammary gland hyperplasia and multifocal tumour lesions in mice at 7-18 months of age. The tumours formed were adenosquamous carcinomas with evidence of transdifferentiation and keratinized deposits. Overall, we identify alternative splicing as a mechanism utilized by cancer cells to modulate the DMP1 locus through diminishing DMP1α tumour suppressor expression, while simultaneously up-regulating the tumour-promoting DMP1β isoform.
- Published
- 2015
- Full Text
- View/download PDF
34. Haploinsufficient tumor suppressor genes
- Author
-
Kazushi, Inoue and Elizabeth A, Fry
- Subjects
Article - Abstract
Haploinsufficiency of tumor suppressor genes (TSGs) indicates that the reduced levels of proteins in cells that lack one allele of the genomic locus results in the inability of the cell to execute normal cellular functions contributing to tumor development. Representative cases of haploinsufficient TSGs are p27Kip1, p53, DMP1, NF1, and PTEN. Tumor development is significantly accelerated in both mice with homozygous and heterozygous gene deletion, with expression of the wild type allele in the latter. Newly characterized TSGs such as AML1, EGR1, TGFβR1/2, and SMAD4 have also shown haploid insufficiency for tumor suppression. This phenotype has typically been demonstrated in gene knockout mouse models, but analyses of human samples have been conducted in some cases. Recent studies suggest collaboration of multiple haploinsufficient TSGs in 5q-, 7q-, and 8q- syndromes, which is called compound haploinsufficiency. Although ARF is a classical TSG, it also belongs to this category since Arf+/− accelerates tumor development when both alleles for Ink4a are inactivated. Haploid insufficiency of Arf was also reported in myeloid leukemogenesis in the presence of inv(16). In case of p53, p53+/− cells achieve only ~25% of p53 mRNA and protein levels as compared to those in wild type, which could explain the mechanism. TGFβR1+/− collaborates with ApcMin+/− in colorectal cancer development; TGFβR2+/− and Smad4+/− collaborates with K-Ras mutation in pancreatic ductal adenocarcinomagenesis, demonstrating the synergism of haploinsufficient TSGs and other oncogenic events. These TSGs can be targets for activation therapy in cancer since they retain a functional allele even in tumor cells.
- Published
- 2017
35. Cooperation between Dmp1 Loss and Cyclin D1 Overexpression in Breast Cancer
- Author
-
George Kulik, Guangchao Sui, Sinan Zhu, Elizabeth A. Fry, Ryan T. Mott, Pankaj Taneja, and Kazushi Inoue
- Subjects
G2 Phase ,Carcinogenesis ,Cyclin D ,Cyclin A ,Cyclin B ,Mitosis ,Apoptosis ,Breast Neoplasms ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cyclin D1 ,stomatognathic system ,medicine ,Animals ,Humans ,Neoplastic transformation ,Neoplasm Metastasis ,Promoter Regions, Genetic ,Cyclin-Dependent Kinase Inhibitor p16 ,030304 developmental biology ,0303 health sciences ,Mammary Neoplasms, Experimental ,Regular Article ,Fibroblasts ,medicine.disease ,3. Good health ,Gene Expression Regulation, Neoplastic ,Mammary Tumor Virus, Mouse ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,biology.protein ,Female ,Cyclin A2 ,Transcription Factors - Abstract
Cyclin D1 is a component of the core cell-cycle machinery and is frequently overexpressed in breast cancer. It physically interacts with the tumor suppressor Dmp1 that attenuates the oncogenic signals from Ras and HER2 by inducing Arf/p53-dependent cell-cycle arrest. Currently, the biological significance of Dmp1-cyclin D1 interplay in breast cancer has not been determined. Here, we show that cyclin D1 bound to Dmp1 to activate both Arf and Ink4a promoters and, consequently, induced apoptosis or G2/M cell-cycle delay in normal cells to protect them from neoplastic transformation. The cyclin D1-induced Ink4a/Arf gene expression was dependent on Dmp1 because the induction was not detected in Dmp1-deficient or DMP1-depleted cells. Arf/Ink4a expression was increased in pre-malignant mammary glands from Dmp1(+/+);MMTV-cyclin D1 and Dmp1(+/+);MMTV-D1T286A mice but significantly down-regulated in those from Dmp1-deficient mice. Selective Dmp1 deletion was found in 21% of the MMTV-D1 and D1T286A mammary carcinomas, and the Dmp1 heterozygous status significantly accelerated mouse mammary tumorigenesis with reduced apoptosis and increased metastasis. Overall, our study reveals a pivotal role of combined Dmp1 loss and cyclin D1 overexpression in breast cancer.
- Published
- 2013
- Full Text
- View/download PDF
36. Clinical applications of mouse models for breast cancer engaging HER2/neu
- Author
-
Pankaj Taneja, Elizabeth A. Fry, and Kazushi Inoue
- Subjects
0301 basic medicine ,Oncology ,Genetically modified mouse ,medicine.medical_specialty ,medicine.drug_class ,medicine.medical_treatment ,Lapatinib ,Monoclonal antibody ,HER2/neu ,Article ,03 medical and health sciences ,0302 clinical medicine ,Trastuzumab ,Internal medicine ,medicine ,skin and connective tissue diseases ,neoplasms ,Monoclonal antibody therapy ,biology ,business.industry ,Immunotherapy ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,business ,Tyrosine kinase ,medicine.drug - Abstract
Human c-ErbB2 (HER2) has long been used as a marker of breast cancer (BC) for sub-categorization for the prediction of prognosis, and determination of therapeutic strategies. HER2 overexpressing BCs are more invasive/metastatic; but patients respond to monoclonal antibody therapy with trastuzumab or tyrosine kinase inhibitors, at least at early stages. To date, numerous mouse models that faithfully reproduce HER2(+) BCs have been created in mice. We recently reviewed different mouse models of BC overexpressing wild type or mutant neu driven by MMTV, neu, or doxycycline-inducible promoters. These mice have been used to demonstrate the histopathology, oncogenic signaling pathways initiated by aberrant overexpression of HER2 in the mammary epithelium, and interaction between oncogenes and tumor suppressor genes at molecular levels. In this review, we focus on their clinical applications. They can be used to test the efficacy of HER(2) inhibitors before starting clinical trials, characterize the tumor-initiating cells that could be the cause of relapse after therapy as well as to analyze the molecular mechanisms of therapeutic resistance targeting HER2. MMTV-human ErbB2 (HER2) mouse models have recently been established since the monoclonal antibody to HER2 (trastuzumab; Herceptin®) does not recognize the rat neu protein. It has been reported that early intervention with HER2 monoclonal antibody would be beneficial for preventing mammary carcinogenesis. MDA-7/IL-24 as well as naturally-occurring chemicals have also been tested using MMTV-neu models. Recent studies have shown that MMTV-neu models are useful to develop vaccines to HER2 for immunotherapy. The mouse models employing HER2/neu will be essential for future antibody or drug screenings to overcome resistance to trastuzumab or HER(2)-specific tyrosine kinase inhibitors.
- Published
- 2016
37. Dmp1 Physically Interacts with p53 and Positively Regulates p53′s Stability, Nuclear Localization, and Function
- Author
-
Takayuki Sugiyama, Rachel L. Morgan, Elizabeth A. Fry, Kazushi Inoue, Guangchao Sui, Dejan Maglic, Fumitake Kai, Donna P. Frazier, Sarah J. Lagedrost, and Robert D. Kendig
- Subjects
Cancer Research ,Cell type ,Transcription, Genetic ,Active Transport, Cell Nucleus ,Article ,Mice ,stomatognathic system ,Proto-Oncogene Proteins c-mdm2 ,Ubiquitin ,medicine ,Animals ,Humans ,Binding site ,Transcription factor ,Cells, Cultured ,Cyclin-Dependent Kinase Inhibitor p16 ,Cell Nucleus ,Binding Sites ,biology ,Ubiquitination ,Molecular biology ,Cell biology ,Mice, Inbred C57BL ,Cell nucleus ,medicine.anatomical_structure ,Oncology ,biology.protein ,Mdm2 ,Tumor Suppressor Protein p53 ,Nuclear localization sequence ,Transcription Factors - Abstract
The transcription factor Dmp1 is a Ras/HER2-activated haplo-insufficient tumor suppressor that activates the Arf/p53 pathway of cell-cycle arrest. Recent evidence suggests that Dmp1 may activate p53 independently of Arf in certain cell types. Here, we report findings supporting this concept with the definition of an Arf-independent function for Dmp1 in tumor suppression. We found that Dmp1 and p53 can interact directly in mammalian cells via the carboxyl-terminus of p53 and the DNA-binding domain of Dmp1. Expression of Dmp1 antagonized ubiquitination of p53 by Mdm2 and promoted nuclear localization of p53. Dmp1–p53 binding significantly increased the level of p53, independent of the DNA-binding activity of Dmp1. Mechanistically, p53 target genes were activated synergistically by the coexpression of Dmp1 and p53 in p53−/−;Arf−/− cells, and genotoxic responses of these genes were hampered more dramatically in Dmp1−/− and p53−/− cells than in Arf−/− cells. Together, our findings identify a robust new mechanism of p53 activation mediated by direct physical interaction between Dmp1 and p53. Cancer Res; 72(7); 1740–50. ©2012 AACR.
- Published
- 2012
- Full Text
- View/download PDF
38. Critical Roles of DMP1 in Human Epidermal Growth Factor Receptor 2/neu-Arf-p53 Signaling and Breast Cancer Development
- Author
-
Fumitake Kai, Takayuki Sugiyama, Mark C. Willingham, Pankaj Taneja, Robert D. Kendig, Donna P. Frazier, Dejan Maglic, and Kazushi Inoue
- Subjects
Cancer Research ,Mammary tumor ,Tumor suppressor gene ,Cell growth ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,stomatognathic system ,Oncology ,Downregulation and upregulation ,Cancer cell ,medicine ,Cancer research ,Signal transduction ,skin and connective tissue diseases ,Carcinogenesis - Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression stimulates cell growth in p53-mutated cells while it inhibits cell proliferation in those with wild-type p53, but the molecular mechanism is unknown. The Dmp1 promoter was activated by HER2/neu through the phosphatidylinositol-3′-kinase-Akt-NF-κB pathway, which in turn stimulated Arf transcription. Binding of p65 and p52 subunits of NF-κB was shown to the Dmp1 promoter and that of Dmp1 to the Arf promoter on HER2/neu overexpression. Both Dmp1 and p53 were induced in premalignant lesions from mouse mammary tumor virus-neu mice, and mammary tumorigenesis was significantly accelerated in both Dmp1+/− and Dmp1−/− mice. Selective deletion of Dmp1 and/or overexpression of Tbx2/Pokemon was found in >50% of wild-type HER2/neu carcinomas, although the involvement of Arf, Mdm2, or p53 was rare. Tumors from Dmp1+/−, Dmp1−/−, and wild-type neu mice with hemizygous Dmp1 deletion showed significant downregulation of Arf and p21Cip1/WAF1, showing p53 inactivity and more aggressive phenotypes than tumors without Dmp1 deletion. Notably, endogenous hDMP1 mRNA decreased when HER2 was depleted in human breast cancer cells. Our study shows the pivotal roles of Dmp1 in HER2/neu-p53 signaling and breast carcinogenesis. Cancer Res; 70(22); 9084–94. ©2010 AACR.
- Published
- 2010
- Full Text
- View/download PDF
39. Emerging Roles of DMP1 in Lung Cancer
- Author
-
Kazushi Inoue, Rachel L. Morgan, Pankaj Taneja, Takayuki Sugiyama, and Donna P. Frazier
- Subjects
Cancer Research ,Lung Neoplasms ,Cell ,Biology ,medicine.disease_cause ,Article ,law.invention ,stomatognathic system ,Transcription (biology) ,law ,medicine ,Animals ,Humans ,Lung cancer ,Transcription factor ,Extracellular Matrix Proteins ,Lung ,Phosphoproteins ,medicine.disease ,Cell biology ,medicine.anatomical_structure ,Oncology ,ras Proteins ,Cancer research ,Suppressor ,Signal transduction ,Carcinogenesis ,Signal Transduction - Abstract
The Ras-activated transcription factor DMP1 can stimulate Arf transcription to promote p53-dependent cell arrest. One recent study deepens the pathophysiologic significance of this pathway in cancer, first, by identifying DMP1 losses in human lung cancers that lack ARF/p53 mutations, and second, by demonstrating that Dmp1 deletions in the mouse are sufficient to promote K-ras–induced lung tumorigenesis via mechanisms consistent with a disruption of Arf/p53 suppressor function. These findings prompt further investigations of the prognostic value of DMP1 alterations in human cancers and the oncogenic events that can cooperate with DMP1 inactivation to drive tumorigenesis. [Cancer Res 2008;68(12):4487–90]
- Published
- 2008
- Full Text
- View/download PDF
40. DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway
- Author
-
Orli Yogev, Eitan Shaulian, Kazushi Inoue, Keren Saadon, and Shira Anzi
- Subjects
Threonine ,Cancer Research ,MAP Kinase Kinase 4 ,Ultraviolet Rays ,Nucleolus ,DNA repair ,DNA damage ,Chromosomal translocation ,Biology ,Models, Biological ,Humans ,Phosphorylation ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Activator (genetics) ,Kinase ,JNK Mitogen-Activated Protein Kinases ,Nuclear Proteins ,Molecular biology ,Transport protein ,Gene Expression Regulation, Neoplastic ,Protein Transport ,Oncology ,Nucleophosmin ,Cell Nucleolus ,DNA Damage - Abstract
The dynamic behavior of the nucleolus plays a role in the detection of and response to DNA damage of cells. Two nucleolar proteins, p14ARF/p19ARF and B23, were shown to translocate out of the nucleolus after exposure of cells to DNA-damaging agents. This translocation affects multiple cellular functions, such as DNA repair, proliferation, and survival. In this study, we identify a pathway and scrutinize the mechanisms leading to the translocation of these proteins after exposure of cells to DNA-damaging agents. We show that redistribution of B23 and p19ARF after the exposure to genotoxic stress occurs preferentially when the c-Jun-NH2-kinase (JNK) pathway is activated and is inhibited when the JNK pathway is impaired. The stress-induced translocation of alternative reading frame (ARF) is JNK dependent and mediated by two activator proteins, c-Jun and JunB. Thr91 and Thr93 of c-Jun are required for the translocation, but the transcriptional activity of c-Jun is dispensable. Instead, c-Jun interacts with B23 in a dose-dependent manner. c-Jun itself is excluded from the nucleolus in a JNK-dependent manner. Hence, we suggest that c-Jun translocates B23 and ARF from the nucleolus after JNK activation by means of protein interactions. In senescent cells, JNK activity and c-Jun levels are reduced concomitantly with ARF nucleolar accumulation, and UV radiation does not cause the translocation of ARF. [Cancer Res 2008;68(5):1398–406]
- Published
- 2008
- Full Text
- View/download PDF
41. Dmp1 and tumor suppression
- Author
-
Donna P. Frazier, Ali Mallakin, and Kazushi Inoue
- Subjects
Cancer Research ,Cyclin D ,Cyclin A ,Cyclin B ,Article ,Mice ,stomatognathic system ,Cyclin D2 ,Cyclin-dependent kinase ,Cyclins ,Genetics ,Animals ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Cyclin-Dependent Kinase Inhibitor p16 ,Cyclin ,biology ,Tumor Suppressor Proteins ,Cell cycle ,Cyclin-Dependent Kinases ,Genes, ras ,biology.protein ,Cancer research ,Tumor Suppressor Protein p53 ,E2F1 Transcription Factor ,Cyclin A2 ,Transcription Factors - Abstract
Dmp1 (cyclin D binding myb-like protein 1; also called Dmtf1) is a transcription factor that was isolated in a yeast two-hybrid screen through its binding property to cyclin D2. Although it was initially predicted to be involved in the cyclin D-Rb pathway, overexpression of Dmp1 in primary cells induces cell cycle arrest in an Arf, p53-dependent fashion. Dmp1 is a unique Arf regulator, the promoter of which is activated by oncogenic Ras-Raf signaling. Dmp1 expression is repressed by physiological mitogenic stimuli as well as by overexpressed E2F proteins; thus, it is a novel marker of cells that have exited from the cell cycle. Spontaneous and oncogene-induced tumor formation is accelerated in both Dmp1(+/-) and Dmp1(-/-) mice; the Dmp1(+/-) tumors often retain and express the wild-type allele; thus, Dmp1 is haplo-insufficient for tumor suppression. Tumors from Dmp1(+/-) and Dmp1(-/-) mice often retain wild-type Arf and p53, suggesting that Dmp1 is a physiological regulator of the Arf-p53 pathway. The human DMP1 (hDMP1) gene is located on chromosome 7q21, the locus of which is often deleted in myeloid leukemia and also in some types of solid tumors. Post-translational modification of Dmp1 and its role in human malignancy remain to be investigated.
- Published
- 2007
- Full Text
- View/download PDF
42. Aberrant splicing of the DMP1-ARF-MDM2-p53 pathway in cancer
- Author
-
Kazushi, Inoue and Elizabeth A, Fry
- Subjects
Gene Expression Regulation, Neoplastic ,Alternative Splicing ,Extracellular Matrix Proteins ,Humans ,Breast Neoplasms ,Female ,Proto-Oncogene Proteins c-mdm2 ,Tumor Suppressor Protein p53 ,Phosphoproteins ,Cyclin-Dependent Kinase Inhibitor p16 ,Article ,Cell Proliferation ,Signal Transduction - Abstract
Alternative splicing of mRNA precursors is a ubiquitous mechanism for generating numerous transcripts with different activities from one genomic locus in mammalian cells. The gene products from a single locus can thus have similar, dominant-negative, or even opposing functions. Aberrant alternative splicing has been found in cancer to express proteins that promote cell growth, local invasion, and metastasis. This review will focus on the aberrant splicing of tumor suppressor/oncogenes that belong to the DMP1-ARF-MDM2-p53 pathway. Our recent study shows that the DMP1 locus generates both tumor-suppressive DMP1α (p53-dependent) and oncogenic DMP1β (p53-independent) splice variants, and the DMP1β/α ratio increases with neoplastic transformation of breast epithelial cells. This process is associated with high DMP1β protein expression and shorter survival of breast cancer (BC) patients. Accumulating pieces of evidence show that ARF is frequently inactivated by aberrant splicing in human cancers, demonstrating its involvement in human malignancies. Splice variants from the MDM2 locus promote cell growth in culture and accelerate tumorigenesis in vivo. Human cancers expressing these splice variants are associated with advanced stage/metastasis, and thus have negative clinical impacts. Although they lack most of the p53-binding domain, their activities are mostly dependent on p53 since they bind to wild type MDM2. The p53 locus produces splice isoforms that have either favorable (β/γ at the C-terminus) or negative impact (Δ40, Δ133 at the N-terminus) on patients' survival. Since the oncogenic alternative splicing products from these loci are expressed only in cancer cells, they may eventually become targets for molecular therapies.
- Published
- 2015
43. Transcription factors that interact with p53 and Mdm2
- Author
-
Kazushi, Inoue, Elizabeth A, Fry, and Donna P, Frazier
- Subjects
Gene Expression Regulation, Neoplastic ,Transcriptional Activation ,Carcinogenesis ,Neoplasms ,Animals ,Humans ,Proto-Oncogene Proteins c-mdm2 ,Tumor Suppressor Protein p53 ,Article ,Transcription Factors - Abstract
The tumor suppressor p53 is activated upon cellular stresses such as DNA damage, oncogene activation, hypoxia, which transactivates sets of genes that induce DNA repair, cell cycle arrest, apoptosis, or autophagy, playing crucial roles in the prevention of tumor formation. The central regulator of the p53 pathway is Mdm2 which inhibits transcriptional activity, nuclear localization, and protein stability. More than 30 cellular p53-binding proteins have been isolated and characterized including Mdm2, Mdm4, p300, BRCA1/2, ATM, ABL, and 53BP-1/2. Most of them are nuclear proteins; however, not much is known about p53-binding transcription factors. In this review, we focus on transcription factors that directly interact with p53/Mdm2 through direct binding including Dmp1, E2F1, YB-1, and YY1. Dmp1 and YB-1 bind only to p53 while E2F1 and YY1 bind to both p53 and Mdm2. Dmp1 has been shown to bind to p53 and block all the known functions for Mdm2 on p53 inhibition, providing a secondary mechanism for tumor suppression in Arf-null cells. Although E2F1-p53 binding provides a checkpoint mechanism to silence hyperactive E2F1, YB-1 or YY1 interaction with p53 subverts the activity of p53, contributing to cell cycle progression and tumorigenesis. Thus, the modes and consequences for each protein-protein interaction vary from the viewpoint of tumor development and suppression.
- Published
- 2015
44. Induction of Transcriptionally Active Jun Proteins Regulates Drug-induced Senescence
- Author
-
Orli Yogev, Eitan Shaulian, Kazushi Inoue, and Shira Anzi
- Subjects
Aphidicolin ,Senescence ,Aging ,Transcription, Genetic ,Proto-Oncogene Proteins c-jun ,JUNB ,Cyclin D ,Blotting, Western ,Antineoplastic Agents ,Apoptosis ,Kidney ,Polymerase Chain Reaction ,Biochemistry ,Mice ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Animals ,Humans ,Hydroxyurea ,Cyclin D1 ,Molecular Biology ,Cells, Cultured ,Cyclin-Dependent Kinase Inhibitor p16 ,Protein kinase C ,biology ,Kinase ,Cell Cycle ,Cell Biology ,Fibroblasts ,Cell cycle ,Transcription Factor AP-1 ,Gene Expression Regulation ,chemistry ,Cancer research ,biology.protein ,Proto-Oncogene Proteins c-fos - Abstract
The drug hydroxyurea (HU) is used for cancer therapy and treatment of sickle cell anemia. It inhibits cell cycle progression by blocking DNA synthesis and drives cells to undergo apoptosis or enter senescence. We demonstrate here that HU induces the expression of two AP-1 proteins, c-Jun and JunB, which exert antagonistic effects on the cell cycle. Moreover, the induction of c-Jun is observed following treatment with two other drugs that inhibit the cell cycle in S phase, aphidicolin and camptothecin. The induction of c-Jun, which promotes cell cycle progression, up-regulates expression of cyclin D after exposure of cells to HU. Deficiency in c-jun prevents elevation of cyclin D expression and extends entrance into HU-induced senescence but also renders cells more resistant to HU-dependent apoptosis. The induction of c-Jun is independent of JNK activity, and additionally, of c-Jun autoregulatory activity but is inhibited upon inhibition of protein kinase C activity. Therefore, we suggest that c-Jun activity prevents drug-induced senescence. Conversely, the JunB target gene, tumor suppressor p16(INK4a), a cyclin-dependent kinase inhibitor essential for the induction of drug-induced senescence, is also up-regulated by HU in a JunB-dependent manner. Constitutive expression of JunB up-regulates p16(INK4a) and increases the sensitivity of mouse fibroblasts to drug-induced-senescence. Thus, we suggest that in contrast to c-Jun, JunB drives cells to enter HU-dependent senescence. The effect of HU treatment, which regulates the intricate web of AP-1 transcription, depends on the balance between c-Jun and JunB activities.
- Published
- 2006
- Full Text
- View/download PDF
45. Convergence of Itch-induced ubiquitination with MEKK1-JNK signaling in Th2 tolerance and airway inflammation
- Author
-
Michael Croft, Chris Elly, Chun Yang, Shahram Salek-Ardakani, K. Venuprasad, Yun-Cai Liu, Jun-Li Luo, Michael Karin, Min Gao, Yohsuke Harada, and Kazushi Inoue
- Subjects
CD4-Positive T-Lymphocytes ,MAP Kinase Kinase 4 ,Ubiquitin-Protein Ligases ,Oncogene Protein p65(gag-jun) ,MAP Kinase Kinase Kinase 1 ,Mice, Transgenic ,Ubiquitin-Activating Enzymes ,Immune tolerance ,Mice ,Th2 Cells ,Immune system ,Antigen ,Immune Tolerance ,Animals ,Bronchitis ,Cells, Cultured ,Clonal Anergy ,biology ,Clonal anergy ,General Medicine ,Flow Cytometry ,Ubiquitin ligase ,Mice, Inbred C57BL ,Tolerance induction ,Mitogen-activated protein kinase ,Immunology ,biology.protein ,Signal transduction ,Research Article ,Signal Transduction - Abstract
The immune system is capable of mounting robust responses against invading pathogens but refrains from attacking self. Many studies have focused on tolerance induction of Th1 cells, whose failure results in development of autoimmune diseases. However, the molecular mechanisms governing tolerance induction in Th2 cells and its relation to allergic responses remain unclear. Here we used both in vivo and in vitro protocols to demonstrate that Th2 cells either containing a mitogen and extracellular kinase kinase 1 (MEKK1) mutant or lacking JNK1 or the E3 ubiquitin ligase Itch cannot be tolerized. In a mouse allergic model, injection of high-dose tolerizing antigen failed to block the development of airway inflammation in Itch-/- mice. This study suggests that MEKK1-JNK signaling regulates Itch E3 ligase-mediated tolerogenic process in Th2 cells. These findings have therapeutic implications for allergic diseases.
- Published
- 2006
- Full Text
- View/download PDF
46. Alterations of p63 and p73 in Human Cancers
- Author
-
Elizabeth A. Fry and Kazushi Inoue
- Subjects
Mice, Knockout ,Gene isoform ,Transcription, Genetic ,Tumor suppressor gene ,Oncogene ,RNA Splicing ,Tumor Suppressor Proteins ,Membrane Proteins ,Nuclear Proteins ,Tumor Protein p73 ,Biology ,Article ,DNA-Binding Proteins ,Mice ,Tumor progression ,Neoplasms ,Cancer research ,Animals ,Humans ,Gene family ,Gene ,Gene knockout - Abstract
p53 and its related genes, p63 and p73 constitute the p53 gene family. While p53 is the most frequently mutated gene in human tumors, p63 and p73 are rarely mutated or deleted in cancers. Many studies have reported p63/p73 overexpression in human cancers while others showed that a loss of p63/p73 is associated with tumor progression and metastasis. Thus, whether p63 or p73 is a tumor suppressor gene or an oncogene has been a matter of debate. This controversy has been attributed to the existence of multiple splicing isoforms with distinct functions; the full-length TA isoform of p63 has structural and functional similarity to wild-type p53, whereas the ΔNp63 acts primarily in dominant-negative fashion against all family members of p53. Differential activities of TA and ΔN isoforms have been shown in vivo by creating isoform-specific gene knockout mice. All p53, p63, p73 proteins bind to and activate target genes with p53-response elements; p63 also binds to distinct p63-response elements and regulate expression of specific target genes involved in skin, limb, and craniofacial development. Interestingly, several studies have shown that both p63 and p73 are involved in cellular response to cancer therapy and others have indicated that both of these molecules are required for p53-induced apoptosis, suggesting functional interplay among p53 family proteins. Consistent with these findings, aberrant splicing that result in ΔNp63 or ΔNp73 overexpression are frequently found in human cancers, and is associated with poor clinical outcomes of patients in the latter. Thus immunohistochemical staining of tumor specimen with ΔNp73-specific antibody might have diagnostic values in cancer clinics.
- Published
- 2014
- Full Text
- View/download PDF
47. Disruption of the ARF transcriptional activator DMP1 facilitates cell immortalization, Ras transformation, and tumorigenesis
- Author
-
Martine F. Roussel, Jerold E. Rehg, Masashi Adachi, Charles J. Sherr, Kazushi Inoue, John L. Cleveland, and Renren Wen
- Subjects
Cell cycle checkpoint ,Tumor suppressor gene ,Embryo ,Biology ,medicine.disease_cause ,Molecular biology ,Embryonic stem cell ,Cell biology ,stomatognathic system ,Cell culture ,Genetics ,biology.protein ,medicine ,Mdm2 ,Carcinogenesis ,Transcription factor ,Developmental Biology - Abstract
The DMP1 transcription factor induces the ARF tumor suppressor gene in mouse fibroblasts, leading to cell cycle arrest in a p53-dependent manner. We disrupted sequences encoding the DNA-binding domain of DMP1 in mouse embryonic stem cells and derived animals lacking the functional protein. DMP1-null animals are small at birth, and males develop more slowly than their wild-type littermates. Some adult animals exhibit seizures and/or obstuctive uropathy, each of unknown cause. The growth of explantedDMP1-null mouse embryo fibroblasts (MEFs) is progressively retarded as cells are passaged in culture on defined transfer protocols; but, unlike the behavior of normal cells, p19ARF, Mdm2, and p53 levels remain relatively low and DMP1-null MEFs do not senesce. Whereas the establishment of cell lines from MEFs is usually always accompanied by either p53 or ARF loss of function, continuously passaged DMP1-null cells readily give rise to established 3T3 and 3T9 cell lines that retain wild-type ARFand functional p53 genes. Early-passage DMP1-null cells, like MEFs from either ARF-null or p53-null mice, can be morphologically transformed by oncogenic Ha-Ras (Val-12) alone. Splenic lymphocytes harvested from both DMP1-null andARF-null mice exhibit enhanced proliferative responses in long-term cultures when stimulated to divide with antibody to CD3 and interleukin-2. Although only 1 of 40 DMP1-null animals spontaneously developed a tumor in the first year of life, neonatal treatment with dimethylbenzanthracene or ionizing radiation induced tumors of various histologic types that were not observed in similarly treated DMP1+/+ animals. Karyotypic analyses of MEFs and lymphomas from DMP1-null animals revealed pseudodiploid chromosome numbers, consistent with the retention of wild-type p53. Together, these data suggest that ARF function is compromised, but not eliminated, in animals lacking functional DMP1.
- Published
- 2000
- Full Text
- View/download PDF
48. Regulation of the CD13/Aminopeptidase N Gene by DMP1, a Transcription Factor Antagonized by D-Type Cyclins
- Author
-
Kazushi Inoue, Linda H. Shapiro, and Charles J. Sherr
- Subjects
animal structures ,Molecular Sequence Data ,CD13 Antigens ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Mice ,Transactivation ,stomatognathic system ,Cyclin-dependent kinase ,Cyclin D ,Cyclins ,Consensus sequence ,Animals ,Binding site ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,DNA Primers ,Cyclin ,Base Sequence ,biology ,Promoter ,3T3 Cells ,Cell Biology ,Molecular biology ,biology.protein ,Cyclin-dependent kinase 6 ,Transcription Factors - Abstract
The binding of the Myb-like DMP1 transcription factor to DNA consensus sequences [CCCG(G/T)ATGT] in artificial promoters is antagonized by D-type cyclins with no requirement for their catalytic partners, cyclin-dependent kinase (CDK) 4 and CDK6. The subset of DMP1 binding sites containing the GGA core can bind Ets family transcription factors Ets-1 and Ets-2. Screening of a series of natural promoters revealed that the CD13/aminopeptidase N (APN; EC 3.4.11.2) promoter could bind and be activated by DMP1. Activation of CD13/APN required both the intact DNA binding and transactivation domains of DMP1 and was inhibited by D-type cyclins, but not by cyclins A, B, C, or H, in a CDK-independent manner. CD13/APN is transactivated by a cooperative interaction between c-Myb bound to its cognate site and Ets-1 tethered to one of three GGA core-containing sites located 30-50 base pairs downstream. DMP1 binds to one of the Ets binding sites (designated Ets C) and synergizes with c-Myb in activating CD13/APN expression. Analysis of nuclear lysates from KG1a early myeloid cells using an oligonucleotide probe containing only the DMP1/Ets C binding site indicated that endogenous DMP1 and a putative Ets family member bind this element in vivo. DMP1-DNA complexes were significantly more stable than those containing the Ets factor. These data indicate that two different Myb family proteins collaborate in regulating APN gene expression and point to a role for DMP1 in normal myeloid cell development.
- Published
- 1998
- Full Text
- View/download PDF
49. Wilms' Tumor Gene (WT1) Competes With Differentiation-Inducing Signal in Hematopoietic Progenitor Cells
- Author
-
Eui Ho Kim, Hiroya Tamaki, Hiroyasu Ogawa, Akihiro Tsuboi, Tetsu Akiyama, Tadamitsu Kishimoto, Yoshihiro Oka, Kazushi Inoue, Haruo Sugiyama, Yusuke Oji, Toshihiro Soma, Manabu Kawakami, and Toyoshi Tatekawa
- Subjects
STAT3 Transcription Factor ,congenital, hereditary, and neonatal diseases and abnormalities ,Genes, Wilms Tumor ,Tumor suppressor gene ,Neutrophils ,Cellular differentiation ,Immunology ,Biology ,urologic and male genital diseases ,Biochemistry ,Cell Line ,Mice ,Granulocyte Colony-Stimulating Factor ,Animals ,Humans ,Progenitor cell ,WT1 Proteins ,Interleukin 3 ,Regulation of gene expression ,urogenital system ,Gene Transfer Techniques ,Cell Differentiation ,Cell Biology ,Hematology ,Transfection ,Hematopoietic Stem Cells ,female genital diseases and pregnancy complications ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Trans-Activators ,Cancer research ,Signal transduction ,Stem cell ,Signal Transduction ,Transcription Factors - Abstract
The WT1 gene is a tumor-suppressor gene that was isolated as a gene responsible for Wilms' tumor, a childhood kidney neoplasm. We have previously reported that the WT1 gene is strongly expressed in leukemia cells with an increase in its expression levels at relapse and an inverse correlation between its expression levels and prognosis, thus making it a novel tumor marker for leukemic blast cells. Furthermore, WT1 antisense oligomers have been found to inhibit the growth of leukemic cells. These results strongly suggested the involvement of the WT1 gene in human leukemogenesis. The present study was performed to prove our hypothesis that the WT1 gene plays a key role in leukemogenesis and performs an oncogenic function in hematopoietic progenitor cells, rather than a tumor-suppressor gene function. 32D cl3, an interleukin-3-dependent myeloid progenitor cell line, differentiates into mature neutrophils in response to granulocyte colony-stimulating factor (G-CSF). However, when transfected wild-type WT1 gene was constitutively expressed in 32D cl3, the cells stopped differentiating and continued to proliferate in response to G-CSF. As for signal transduction mediated by G-CSF receptor (G-CSFR), Stat3alpha was constitutively activated in wild-type WT1-infected 32D cl3 in response to G-CSF, whereas, in WT1-uninfected 32D cl3, activation of Stat3alpha was only transient. However, most interesting was the fact that G-CSF stimulation resulted in constitutive activation of Stat3beta only in wild-type WT1-infected 32D cl3, but not in WT1-uninfected 32D cl3. Thus, WT1 expression constitutively activated both Stat3alpha and Stat3beta. A transient activation of Stat1 was detected in both wild-type WT1-infected and uninfected 32D cl3 after G-CSF stimulation, but no difference in its activation was found. No activation of MAP kinase was detected in both wild-type WT1-infected and uninfected 32D cl3 after G-CSF stimulation. These results demonstrated that WT1 expression competed with the differentiation-inducing signal mediated by G-CSFR and constitutively activated Stat3, resulting in the blocking of differentiation and subsequent proliferation. Therefore, the data presented here support our hypothesis that the WT1 gene plays an essential role in leukemogenesis and performs an oncogenic function in hematopoietic progenitor cells and represent the first demonstration of an important role of the WT1 gene in signal transduction in hematopoietic progenitor cells.
- Published
- 1998
- Full Text
- View/download PDF
50. Gene Expression and Cell Cycle Arrest Mediated by Transcription Factor DMP1 Is Antagonized by D-Type Cyclins through a Cyclin-Dependent-Kinase-Independent Mechanism
- Author
-
Charles J. Sherr and Kazushi Inoue
- Subjects
Transcriptional Activation ,Cell cycle checkpoint ,Recombinant Fusion Proteins ,Gene Expression ,Spodoptera ,Cell Line ,Mice ,Cyclin D1 ,stomatognathic system ,Cyclin-dependent kinase ,Proto-Oncogene Proteins ,Animals ,Humans ,Cell Growth and Development ,Molecular Biology ,Cyclin ,Binding Sites ,biology ,Cyclin-dependent kinase 4 ,Cell Cycle ,Cyclin-dependent kinase 2 ,Chromosome Mapping ,Cyclin-Dependent Kinase 4 ,3T3 Cells ,DNA ,Cell Biology ,DNA-binding domain ,Cell cycle ,Molecular biology ,Cyclin-Dependent Kinases ,biology.protein ,Cell Division ,Transcription Factors - Abstract
A novel 761-amino-acid transcription factor, DMP1, contains a central DNA binding domain that includes three imperfect myb repeats flanked by acidic transactivating domains at the amino and carboxyl termini. D-type cyclins associate with a region of the DMP1 DNA binding domain immediately adjacent to the myb repeats to form heteromeric complexes which detectably interact neither with cyclin-dependent kinase 4 (CDK4) nor with DNA. The segment of D-type cyclins required for its interaction with DMP1 falls outside the "cyclin box," which contains the residues predicted to contact CDK4. Hence, D-type cyclin point mutants that do not interact with CDK4 can still bind to DMP1. Enforced coexpression of either of three D-type cyclins (D1, D2, or D3) with DMP1 in mammalian cells canceled its ability to activate gene expression. This property was not shared by cyclins A, B, C, or H; did not depend upon CDK4 or CDK2 coexpression; was not subverted by a mutation in cyclin D1 that prevents its interaction with CDK4; and was unaffected by inhibitors of CDK4 catalytic activity. Introduction of DMP1 into mouse NIH 3T3 fibroblasts inhibited entry into S phase. Cell cycle arrest depended upon the ability of DMP1 to bind to DNA and to transactivate gene expression and was specifically antagonized by coexpression of D-type cyclins, including a D1 point mutant that does not bind to CDK4. Taken together, these findings suggest that DMP1 induces genes that inhibit S phase entry and that D-type cyclins can override DMP1-mediated growth arrest in a CDK-independent manner.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.