Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Invest ; 35(5): 301-312, 2017 May 28.
Article in English | MEDLINE | ID: mdl-28406729

ABSTRACT

We recently reported the existence of a physical interaction between the Myb-like transcription factor Dmp1 (Dmtf1) and p53 in which Dmp1 antagonized polyubiquitination of p53 by Mdm2 and promoted its nuclear localization. Dmp1 significantly stabilized p53-DNA complexes on promoters that contained p53-consensus sequences, which were either supershifted or disrupted with antibodies to Dmp1. Lysates from mice injected with doxorubicin showed that Dmp1 bound to p21Cip1, Bbc3, and Thbs1 gene regulatory regions in a p53-dependent fashion. Our data suggest that acceleration of DNA-binding of p53 by Dmp1 is a critical process for Dmp1 to increase the p53 function in Arf-deficient cells.


Subject(s)
Cell Nucleus/metabolism , DNA/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , A549 Cells , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Binding Sites , Cell Nucleus/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Doxorubicin/pharmacology , Genotype , Humans , Mice , Mice, Knockout , NIH 3T3 Cells , Phenotype , Promoter Regions, Genetic , Protein Binding , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Time Factors , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
2.
Cancer Res ; 72(7): 1740-50, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22331460

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.


Subject(s)
Cell Nucleus/metabolism , Transcription Factors/physiology , Tumor Suppressor Protein p53/physiology , Active Transport, Cell Nucleus , Animals , Binding Sites , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/physiology , Humans , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-mdm2/physiology , Transcription Factors/chemistry , Transcription, Genetic , Tumor Suppressor Protein p53/chemistry , Ubiquitination
3.
Clin Med Insights Oncol ; 5: 235-57, 2011.
Article in English | MEDLINE | ID: mdl-21836819

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.

4.
Cancer Res ; 70(22): 9084-94, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21062982

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.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Extracellular Matrix Proteins/metabolism , Phosphoproteins/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/metabolism , Extracellular Matrix Proteins/genetics , Female , Gene Expression , Humans , Male , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred Strains , Mice, Knockout , Molecular Sequence Data , Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Receptor, ErbB-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics
5.
Clin Med Insights Oncol ; 4: 15-34, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20567632

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.

6.
Int J Cancer ; 126(6): 1403-16, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-19816943

ABSTRACT

Dmp1 (Dmtf1) encodes a Myb-like transcription factor implicated in tumor suppression through direct activation of the Arf-p53 pathway. The human DMP1 gene is frequently deleted in non-small cell lung cancers, especially those that retain wild-type INK4a/ARF and/or p53. To identify novel genes that are regulated by Dmp1, transcriptional profiles of lung tissue from Dmp1-null and wild-type mice were generated using the GeneChip Microarray. Comparative analysis of gene expression changes between the two groups resulted in identification of numerous genes that may be regulated by Dmp1. Notably, amphiregulin (Areg), thrombospondin-1 (Tsp-1), JunB, Egr1, adrenomedullin (Adm), Bcl-3 and methyl-CpG binding domain protein 1 (Mbd1) were downregulated in the lungs from Dmp1-null mice while Gas1 and Ect2 genes were upregulated. These target genes were chosen for further analyses since they are involved in cell proliferation, transcription, angiogenesis/metastasis, apoptosis, or DNA methylation, and thus could account for the tumor suppressor phenotype of Dmp1. Dmp1 directly bound to the genomic loci of Areg, Tsp-1, JunB and Egr1. Significant upregulation or downregulation of the novel Dmp1 target genes was observed upon transient expression of Dmp1 in alveolar epithelial cells, an effect which was nullified by the inhibition of de novo mRNA synthesis. Interestingly, these genes and their protein products were significantly downregulated or upregulated in the lungs from Dmp1-heterozygous mice as well. Identification of novel Dmp1 target genes not only provides insights into the effects of Dmp1 on global gene expression, but also sheds light on the mechanism of haploid insufficiency of Dmp1 in tumor suppression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Early Growth Response Protein 1/genetics , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Proto-Oncogene Proteins c-jun/genetics , Thrombospondin 1/genetics , Transcription Factors/genetics , Amphiregulin , Animals , Blotting, Western , Chromatin Immunoprecipitation , Cluster Analysis , EGF Family of Proteins , Early Growth Response Protein 1/metabolism , Electrophoretic Mobility Shift Assay , Female , Gene Expression Profiling , Glycoproteins/metabolism , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Protein Binding , Proto-Oncogene Proteins c-jun/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondin 1/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism
7.
Expert Rev Mol Diagn ; 9(5): 423-40, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19580428

ABSTRACT

Mouse mammary tumor virus (MMTV) long terminal repeat (LTR)-driven transgenic mice are excellent models for breast cancer as they allow for the targeted expression of various oncogenes and growth factors in neoplastic transformation of mammary glands. Numerous MMTV-LTR-driven transgenic mouse models of breast cancer have been created in the past three decades, including MMTV-neu/ErbB2, cyclin D1, cyclin E, Ras, Myc, int-1 and c-rel. These transgenic mice develop mammary tumors with different latency, histology and invasiveness, reflecting the oncogenic pathways activated by the transgene. Recently, homologous sequences of the env gene of MMTV have been identified in approximately 40% of human breast cancers, but not in normal breast or other types of cancers, suggesting possible involvement of mammary tumor virus in human breast carcinogenesis. Accumulating evidence demonstrates the association of MMTV provirus with progesterone receptor, p53 mutations and advanced-stage breast cancer. Thus, the detection of MMTV-like sequences may have diagnostic value to predict the clinical outcome of breast cancer patients.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/diagnosis , Mammary Neoplasms, Animal/genetics , Mammary Tumor Virus, Mouse/genetics , Animals , Cell Transformation, Neoplastic , Genes, p53 , Humans , Mice , Mice, Transgenic , Models, Biological , Molecular Diagnostic Techniques , Receptors, Progesterone/genetics , Signal Transduction , Treatment Outcome
8.
Bioorg Med Chem ; 16(23): 10061-74, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18952446

ABSTRACT

Peptidyl alpha-hydroxylating monooxygenase (PHM) functions in vivo towards the biosynthesis of alpha-amidated peptide hormones in mammals and insects. PHM is a potential target for the development of inhibitors as drugs for the treatment of human disease and as insecticides for the management of insect pests. We show here that relatively simple ground state analogs of the PHM substrate hippuric acid (C(6)H(5)-CO-NH-CH(2)-COOH) inhibit the enzyme with K(i) values as low as 0.5microM. Substitution of sulfur atom(s) into the hippuric acid analog increases the affinity of PHM for the inhibitor. Replacement of the acetylglycine moiety, -CO-NH-CH(2)-COOH with an S-(thioacetyl)thioglycolic acid moiety, -CS-S-CH(2)-COOH, yields compounds with the highest PHM affinity. Both S-(2-phenylthioacetyl)thioglycolate and S-(4-ethylthiobenzoyl)thioglycolic acid inhibit the proliferation of cultured human prostate cancer cells at concentrations >100-fold excess of their respective K(i) values. Comparison of K(i) values between mammalian PHM and insect PHM shows differences in potency suggesting that a PHM-based insecticide with limited human toxicity can be developed.


Subject(s)
Enzyme Inhibitors/chemistry , Hippurates/chemistry , Hippurates/pharmacology , Insecticides/chemistry , Mixed Function Oxygenases/antagonists & inhibitors , Multienzyme Complexes/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Hippurates/chemical synthesis , Humans , Inhibitory Concentration 50 , Insecticides/metabolism , Insecticides/pharmacology , Mixed Function Oxygenases/metabolism , Models, Molecular , Multienzyme Complexes/metabolism , Rats , Structure-Activity Relationship , Tumor Cells, Cultured
9.
Clin Med Oncol ; 2: 209-19, 2008.
Article in English | MEDLINE | ID: mdl-21892281

ABSTRACT

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 (hDMP1) 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, hDMP1 is clearly involved in human carcinogenesis, and tumors with hDMP1 deletion may constitute a discrete disease entity.

10.
J Immunol ; 179(12): 8009-15, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18056340

ABSTRACT

We have previously demonstrated that the NKR repertoire is profoundly disrupted by SHIP deficiency. This repertoire disruption is characterized by receptor dominance where inhibitory signals from 2B4 repress killing of complex targets expressing MHC class I and activating ligands. In this study, we examine the molecular basis of receptor dominance in SHIP-/- NK cells. In this study, we show that in SHIP-/- NK cells there is a pronounced bias toward the 2B4 long isoform. We have also characterized signaling molecules recruited to 2B4 in SHIP-/- NK cells. Interestingly, we find that approximately 10- to 16-fold more Src homology region 2 domain-containing phosphatase 1 (SHP1) is recruited to 2B4 in SHIP-/- NK cells when compared with wild type. Consistent with SHP1 overrecruitment, treatment with sodium orthovanadate or a novel inhibitor with micromolar activity against SHP1 restores the ability of SHIP-/- NK cells to kill Rae1+ RMA and M157+ targets. These findings define the molecular basis for hyporesponsiveness by SHIP-deficient NK cells.


Subject(s)
Antigens, CD/metabolism , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Membrane Glycoproteins/metabolism , Phosphoric Monoester Hydrolases/deficiency , Receptors, Immunologic/metabolism , Animals , Antigens, CD/analysis , Enzyme Inhibitors/pharmacology , Inositol Polyphosphate 5-Phosphatases , Killer Cells, Natural/enzymology , Membrane Glycoproteins/analysis , Mice , Mice, Mutant Strains , Phosphoric Monoester Hydrolases/genetics , Protein Isoforms/metabolism , Protein Transport , Receptors, Immunologic/analysis , Signaling Lymphocytic Activation Molecule Family , Vanadates/pharmacology , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...