Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Mol Cancer Res ; 6(6): 937-46, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18567798

ABSTRACT

Osteosarcoma is a primary malignant tumor of bone arising from primitive bone-forming mesenchymal cells and accounts for approximately 60% of malignant bone tumors. Our comparative genomic hybridization (CGH) studies have identified frequent amplification at 6p12-p21, 12q13-q15, and 17p11.2 in osteosarcoma. Of these amplified regions, 6p12-p21 is particularly interesting because of its association with progression and poor prognosis in patients with osteosarcoma. In an attempt to identify aberrantly expressed gene(s) mapping to the 6p12-p21 amplicon, a region-specific array was generated using 108 overlapping BAC and P1 clones covering a 28.8-Mb region at 0.26-Mb intervals. Based on array CGH analysis, the 6p amplicon was refined to 7.9 Mb between the clones RP11-91E11 and RP1-244F2 and 10 amplified clones, with possible target genes, were identified. To study the expression pattern of the target genes from the hotspot amplicon and known candidate genes from 6p12-21, we did quantitative reverse transcription-PCR analysis of MAPK14, MAPK13, CDKN1A, PIM1, MDGA1, BTB9, DNAH8, CCND3, PTK7, CDC5L, and RUNX2 on osteosarcoma patient samples and seven cell lines. The combined array CGH and quantitative reverse transcription-PCR analysis identified amplification and overexpression of CDC5L, CCND3, and RUNX2. We screened these three genes for protein expression by Western blotting and immunohistochemistry and detected overexpression of CDC5L. Furthermore, we used an in vivo assay to show that CDC5L possesses potential oncogenic activity. These results indicate that CDC5L, a cell cycle regulator important for the G2-M transition, is the most likely candidate oncogene for the 6p12-p21 amplicon found in osteosarcoma.


Subject(s)
Bone Neoplasms/genetics , Cell Cycle Proteins/genetics , Chromosomes, Human, Pair 6 , Gene Amplification , Osteosarcoma/genetics , RNA-Binding Proteins/genetics , Animals , Bone Neoplasms/metabolism , Cell Line, Tumor , Chromosome Mapping , Female , Genes, cdc , Humans , Male , Mice , NIH 3T3 Cells , Oncogenes , Osteosarcoma/metabolism , RNA, Messenger/metabolism
2.
Cancer Res ; 64(16): 5608-16, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15313898

ABSTRACT

The absence of p53 function increases risk for spontaneous tumorigenesis in the mammary gland. Hormonal stimulation enhances tumor risk in p53-null mammary epithelial cells as well as the incidence of aneuploidy. Aneuploidy appears in normal p53-null mammary epithelial cells within 5 weeks of hormone stimulation. Experiments reported herein assessed a possible mechanism of hormone-induced aneuploidy. Hormones increased DNA synthesis equally between wild-type (WT) and p53-null mammary epithelial cells. There were two distinct responses in p53-null cells to hormone exposure. First, Western blot analysis demonstrated that the levels of two proteins involved in regulating sister chromatid separation and the spindle checkpoint, Mad2 and separase (ESPL1) were increased in null compared with WT cells. In contrast, the levels of securin and Rad21 proteins were not increased in hormone-stimulated p53-null compared with WT cells. ESPL1 RNA was also increased in p53-null mouse mammary cells in vivo by 18 h of hormone stimulation and in human breast MCF7 cells in monolayer culture by 8 h of hormone stimulation. Furthermore, both promoters contained p53 and steroid hormone response elements. Mad2 protein was increased as a consequence of the absence of p53 function. The increase in Mad2 protein was observed also at the cellular level by immunohistochemistry. Second, hormones increased gene amplication in the distal arm of chromosome 2, as shown by comparative genomic hybridization. These results support the hypothesis that hormone stimulation acts to increase aneuploidy by several mechanisms. First, by increasing mitogenesis in the absence of the p53 checkpoint in G2, hormones allow the accumulation of cells that have experienced chromosome missegregation. Second, the absolute rate of chromosome missegregation may be increased by alterations in the levels of two proteins, separase and Mad2, which are important for maintaining chromosomal segregation and the normal spindle checkpoint during mitosis.


Subject(s)
Aneuploidy , Chromosomal Instability , Estrogens/pharmacology , Mammary Glands, Animal/physiology , Progesterone/pharmacology , Tumor Suppressor Protein p53/deficiency , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Line, Tumor , Endopeptidases/biosynthesis , Endopeptidases/genetics , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/physiology , Female , Mad2 Proteins , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Mice , Mice, Inbred BALB C , Repressor Proteins , Separase , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
3.
Oncogene ; 21(50): 7680-9, 2002 Oct 31.
Article in English | MEDLINE | ID: mdl-12400010

ABSTRACT

AP-1 transcription factors play a critical role in signal transduction pathways in many cells. We have investigated the role of AP-1 in controlling proliferative signals in breast cells, and have previously shown that AP-1 complexes are activated by peptide and steroid growth factors in both normal and malignant breast cells. In this study, we investigated the role of AP-1 in transducing proliferative signals induced by peptide and steroid growth factors. We used MCF-7 clones that express a specific inhibitor of AP-1, a dominant-negative cJun mutant (TAM67), under the control of an inducible promoter to investigate the role of AP-1 in regulating breast cancer growth. In the presence of doxycycline (Dox), the AP-1 inhibitor was not expressed, and the MCF-7 clones proliferated normally in response to serum stimulation. However, when Dox was withdrawn, TAM67 was expressed, AP-1 activity was inhibited, and serum-induced proliferation was blocked. We next investigated whether the mitogenic response to specific growth factors also requires AP-1. MCF-7 Tet-Off-TAM67 cells were grown in the presence of increasing concentrations of IGF-1, EGF, heregulin-beta, bFGF, or estrogen under un-induced and induced conditions. These studies showed that the AP-1 inhibitor completely blocked proliferation in response to the peptide growth factors (IGF-1, EGF, heregulin-beta, and bFGF), and partially blocked the response to estrogen. To investigate the effect of AP-1 blockade on in vivo tumor growth, we injected the MCF-7 Tet-Off TAM67 cells into nude mice receiving doxycycline to suppress the expression of the AP-1 inhibitor. After the mice developed tumors, they were randomized to either continue to receive Dox or not. In mice not receiving Dox, the expression of TAM67 was induced, and tumor growth was inhibited, while the tumors in mice receiving Dox continued to grow. Analysis of the tumors from these mice showed that the expression of TAM67 caused reduced proliferation of the breast cancer cells without inducing apoptosis. These results demonstrate that AP-1 blockade supresses mitogenic signals from multiple different peptide growth factors as well as estrogen, and inhibits the growth of MCF-7 breast cancer cells both in vitro and in vivo. These results suggest that novel agents specifically targeting AP-1 or its activating kinases could be promising agents for the treatment of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Signal Transduction , Transcription Factor AP-1/metabolism , Animals , Apoptosis/genetics , Breast Neoplasms/drug therapy , Cell Division/drug effects , Cell Division/physiology , Culture Media, Serum-Free/pharmacology , Estrogens/pharmacology , Female , Growth Substances/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Oncogene ; 33(48): 5511-5522, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-24276237

ABSTRACT

Separase, a protease encoded by the ESPL1 gene, cleaves the chromosomal cohesin during mitosis. Separase protein and transcripts are overexpressed in a wide range of human cancers. To investigate the physiological consequence of Separase overexpression in animals, we have generated a transgenic MMTV-Espl1 mouse model that overexpresses Separase protein in the mammary glands. MMTV-Espl1 mice in a C57BL/6 genetic background develop aggressive, highly aneuploid and estrogen receptor alpha-positive (ERα+) mammary adenocarcinomas with an 80% penetrance. The mammary tumors caused by overexpression of Separase, alone or combined with p53 heterozygosity, in mammary epithelium mimic several aspects of the most aggressive forms of human breast cancer, including high levels of genetic instability, cell cycle defects, poor differentiation, distant metastasis and metaplasia. Histopathologically, MMTV-Espl1 tumors are highly heterogeneous showing features of both luminal as well as basal subtypes of breast cancers, with aggressive disease phenotype. In addition to aneuploidy, Separase overexpression results in chromosomal instability (CIN) including premature chromatid separation (PCS), lagging chromosomes, anaphase bridges, micronuclei, centrosome amplification, multinucleated cells, gradual accumulation of DNA damage and progressive loss of tumor suppressors p53 and cadherin gene loci. These results suggest that Separase-overexpressing mammary cells are not only susceptible to chromosomal missegregation-induced aneuploidy but also other genetic instabilities including DNA damage and loss of key tumor suppressor gene loci, which in combination can initiate tumorigenesis and disease progression.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Separase/genetics , Adenocarcinoma/metabolism , Aneuploidy , Animals , Blotting, Western , Comparative Genomic Hybridization , Estrogen Receptor alpha/biosynthesis , Female , Fluorescent Antibody Technique , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse , Mice , Mice, Inbred C57BL , Mice, Transgenic , Separase/metabolism
5.
J Biomol Screen ; 19(6): 878-89, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24525869

ABSTRACT

Separase is an endopeptidase that cleaves cohesin subunit Rad21, facilitating the repair of DNA damage during interphase and the resolution of sister chromatid cohesion at anaphase. Separase activity is negatively regulated by securin and Cdk1-cyclin B in vivo. Separase overexpression is reported in a broad range of human tumors, and its overexpression in mouse models results in tumorigenesis. To elucidate further the mechanism of separase function and to test if inhibition of overexpressed separase can be used as a strategy to inhibit tumor-cell proliferation, small-molecule inhibitors of separase enzyme are essential. Here, we report a high-throughput screening for separase inhibitors (Sepins). We developed a fluorogenic separase assay using rhodamine 110-conjugated Rad21 peptide as substrate and screened a small-molecule compound library. We identified a noncompetitive inhibitor of separase called Sepin-1 that inhibits separase enzymatic activity with a half maximal inhibitory concentration (IC50) of 14.8 µM. Sepin-1 can inhibit the growth of human cancer cell lines and breast cancer xenograft tumors in mice by inhibiting cell proliferation and inducing apoptosis. The sensitivity to Sepin-1 in most cases is positively correlated to the level of separase in both cancer cell lines and tumors.


Subject(s)
Antineoplastic Agents/chemistry , Neoplasms/drug therapy , Neoplasms/enzymology , Separase/antagonists & inhibitors , Animals , Antibodies/chemistry , Apoptosis , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , DNA Damage , DNA-Binding Proteins , Disease Models, Animal , Female , High-Throughput Screening Assays , Humans , Hydrolysis , Inhibitory Concentration 50 , Maximum Tolerated Dose , Mice , Microscopy, Fluorescence , Neoplasm Transplantation , Neoplasms/pathology , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Peptides/chemistry , Phosphoproteins/chemistry , Rhodamines/chemistry
6.
PLoS One ; 6(7): e22167, 2011.
Article in English | MEDLINE | ID: mdl-21799785

ABSTRACT

BACKGROUND: Cohesin protease Separase plays a key role in faithful segregation of sister chromatids by cleaving the cohesin complex at the metaphase to anaphase transition. Homozygous deletion of ESPL1 gene that encodes Separase protein results in embryonic lethality in mice and Separase overexpression lead to aneuploidy and tumorigenesis. However, the effect of Separase haploinsufficiency has not been thoroughly investigated. METHODOLOGY/PRINCIPAL FINDINGS: Here we examined the effect of ESPL1 heterozygosity using a hypomorphic mouse model that has reduced germline Separase activity. We report that while ESPL1 mutant (ESPL1 (+/hyp)) mice have a normal phenotype, in the absence of p53, these mice develop spontaneous T- and B-cell lymphomas, and leukemia with a significantly shortened latency as compared to p53 null mice. The ESPL1 hypomorphic, p53 heterozygous transgenic mice (ESPL1(+/hyp), p53(+/-)) also show a significantly reduced life span with an altered tumor spectrum of carcinomas and sarcomas compared to p53(+/-) mice alone. Furthermore, ESPL1(+/hyp), p53(-/-) mice display significantly higher levels of genetic instability and aneuploidy in normal cells, as indicated by the abnormal metaphase counts and SKY analysis of primary splenocytes. CONCLUSIONS/SIGNIFICANCE: Our results indicate that reduced levels of Separase act synergistically with loss of p53 in the initiation and progression of B- and T- cell lymphomas, which is aided by increased chromosomal missegregation and accumulation of genomic instability. ESPL1(+/hyp), p53(-/-) mice provide a new animal model for mechanistic study of aggressive lymphoma and also for preclinical evaluation of new agents for its therapy.


Subject(s)
Aneuploidy , Cell Cycle Proteins/metabolism , Disease Progression , Endopeptidases/metabolism , Leukemia/pathology , Lymphoma, B-Cell/pathology , Lymphoma, T-Cell/pathology , Tumor Suppressor Protein p53/deficiency , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Proliferation , DNA Damage/genetics , Endopeptidases/deficiency , Endopeptidases/genetics , Female , Humans , Leukemia/enzymology , Leukemia/genetics , Longevity/genetics , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/genetics , Lymphoma, T-Cell/enzymology , Lymphoma, T-Cell/genetics , Mice , Neoplasm Metastasis , Phenotype , Separase , Thymus Neoplasms/enzymology , Thymus Neoplasms/genetics , Thymus Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL