Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Eur Arch Otorhinolaryngol ; 272(9): 2227-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24964769

ABSTRACT

Recent studies have shed considerable light into schwannomas. To date, only merlin has been identified as a hallmark or pathogenesis of both sporadic and NF2-related schwannomas. Here, we show, by immunoblot and immunohistochemical analyses of 58 sporadic vestibular schwannomas, that upregulation of p53 was observed in 90 % of tumors examined. No p53 mutations were found in 12 % tumors analyzed. Expression of p14ARF was negative in 95 % of tumors, while overexpression of MDM2 was found in all specimens. Aberrant DNA hypermethylation of the p14ARF promoter was observed in three of seven tumors examined (43 %), associated with remarkably decreased mRNA levels. The very high degree of concordance in the aberrations of the p14ARF/MDM2/p53 pathway in this tumor may be considered to be a new player in the pathogenesis of sporadic vestibular schwannomas. Moreover, expression of p21 (waf1) was negative in all tumors, suggesting that the aberration of this pathway is associated with greater attenuation of p21-mediated signals that are critical for growth inhibition. These data are in agreement with the model in RT-4 rat schwannoma cells: i.e., overexpression of ARF was associated with accumulation of p21 expression both at protein and mRNA levels. ShRNA knock-down of p53 expression attenuated p21-mediated increase in cellular arrest in the G1-phase, suggesting that p14ARF regulates p21 protein levels through a p53-dependent pathway. Thus, this study reveals a high degree of concordance in the aberrations of the p14ARF/MDM2/p53 pathway with the development of sporadic vestibular schwannomas.


Subject(s)
Neuroma, Acoustic/genetics , Neuroma, Acoustic/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Protein p53/metabolism , Adult , DNA Methylation , Female , Humans , Immunohistochemistry , Male , Middle Aged , Polymerase Chain Reaction , Promoter Regions, Genetic , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/genetics , Up-Regulation
2.
Cancer Cell ; 9(5): 379-90, 2006 May.
Article in English | MEDLINE | ID: mdl-16697958

ABSTRACT

Histiocytic sarcoma (HS) is a rare malignant proliferation of histiocytes of uncertain molecular pathogenesis. Here, genetic analysis of coincident loss of Pten and Ink4a/Arf tumor suppressors in the mouse revealed a neoplastic phenotype dominated by a premalignant expansion of biphenotypic myelolymphoid cells followed by the development of HS. Pten protein loss occurred only in the histiocytic portion of tumors, suggesting a stepwise genetic inactivation in the generation of HS. Similarly, human HS showed genetic or epigenetic inactivation of PTEN, p16(INK4A), and p14(ARF), supporting the relevance of this genetically engineered mouse model of HS. These genetic and translational observations establish a cooperative role of Pten and Ink4a/Arf in the development of HS and provide mechanistic insights into the pathogenesis of human HS.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Histiocytic Disorders, Malignant/pathology , Lymphocytes/immunology , Myeloid Cells/immunology , PTEN Phosphohydrolase/metabolism , Sarcoma/pathology , Tumor Suppressor Protein p14ARF/metabolism , Animals , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Histiocytic Disorders, Malignant/immunology , Homeostasis , Humans , Immunophenotyping , Methylation , Mice , Mutation/genetics , PTEN Phosphohydrolase/deficiency , Proto-Oncogene Proteins c-akt/metabolism , Sarcoma/immunology , Tumor Suppressor Protein p14ARF/deficiency
3.
Nature ; 433(7023): 278-85, 2005 Jan 20.
Article in English | MEDLINE | ID: mdl-15662416

ABSTRACT

Aberrant transcriptional repression through chromatin remodelling and histone deacetylation has been postulated to represent a driving force underlying tumorigenesis because histone deacetylase inhibitors have been found to be effective in cancer treatment. However, the molecular mechanisms by which transcriptional derepression would be linked to tumour suppression are poorly understood. Here we identify the transcriptional repressor Pokemon (encoded by the Zbtb7 gene) as a critical factor in oncogenesis. Mouse embryonic fibroblasts lacking Zbtb7 are completely refractory to oncogene-mediated cellular transformation. Conversely, Pokemon overexpression leads to overt oncogenic transformation both in vitro and in vivo in transgenic mice. Pokemon can specifically repress the transcription of the tumour suppressor gene ARF through direct binding. We find that Pokemon is aberrantly overexpressed in human cancers and that its expression levels predict biological behaviour and clinical outcome. Pokemon's critical role in cellular transformation makes it an attractive target for therapeutic intervention.


Subject(s)
Cell Transformation, Neoplastic , Down-Regulation/genetics , Proto-Oncogenes/physiology , Repressor Proteins/metabolism , Tumor Suppressor Protein p14ARF/genetics , Animals , Apoptosis , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16 , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Mas , Proto-Oncogenes/genetics , Repressor Proteins/genetics , Substrate Specificity , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Protein p14ARF/metabolism
4.
Proc Natl Acad Sci U S A ; 104(26): 10968-73, 2007 Jun 26.
Article in English | MEDLINE | ID: mdl-17576930

ABSTRACT

Inactivation of the p53 pathway represents the most common molecular defect of human cancer. But in the setting of melanoma, a highly aggressive and invariably fatal malignancy in its advanced disseminated form, mutation/deletion of p53 is relatively rare, whereas its positive regulator ARF is often lost. Here, we show that genetic deficiency in Arf but not p53 facilitates rapid development of melanoma in a genetically engineered mouse model. This difference is accounted for, at least in part, by the unanticipated observation that, unlike fibroblasts, senescence control in melanocytes is strongly regulated by Arf and not p53. Moreover, oncogenic NRAS collaborates with deficiency in Arf, but not p53, to fully transform melanocytes. Our data demonstrate that ARF and p53, although linked in a common pathway, suppress tumorigenesis through distinct, lineage-dependent mechanisms and suggest that ARF helps restrict melanoma progression by executing the oncogene-induced senescence program in benign nevi. Thus, therapeutics designed to restore wild-type p53 function may be insufficient to counter melanoma and other malignancies in which ARF holds p53-independent tumor suppressor activity.


Subject(s)
Cellular Senescence , Melanoma/pathology , Proto-Oncogene Proteins p21(ras)/physiology , Tumor Suppressor Protein p14ARF/physiology , Tumor Suppressor Protein p53 , Animals , Cell Transformation, Neoplastic , Disease Models, Animal , Melanocytes/pathology , Melanoma/etiology , Mice , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Protein p14ARF/genetics
5.
Neurochem Res ; 34(12): 2055-66, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19856100

ABSTRACT

Primary malignant brain cancer, one of the most deadly diseases, has a high rate of recurrence after treatment. Studies in the past several years have led to the hypothesis that the root of the recurrence may be brain tumor stem cells (BTSCs), stem-like subpopulation of cells that are responsible for propagating the tumor. Current treatments combining surgery and chemoradiotherapy could not eliminate BTSCs because these cells are highly infiltrative and possess several properties that can reduce the damages caused by radiation or anti-cancer drugs. BTSCs are similar to NSCs in molecular marker expression and multi-lineage differentiation potential. Genetic analyses of Drosophila CNS neoplasia, mouse glioma models, and human glioma tissues have revealed a link between increased NSC self-renewal and brain tumorigenesis. Furthermore, data from various rodent models of malignant brain tumors have provided compelling evidence that multipotent NSCs and lineage-restricted neural progenitor cells (NPCs) could be the cell origin of brain tumors. Thus, the first event of brain tumorigenesis might be the occurrence of oncogenic mutations in the stem cell self-renewal pathway in an NSC or NPC. These mutations convert the NSC or NPC to a BTSC, which then initiates and sustains the growth of the tumor. The self-renewal of BTSCs is controlled by several evolutionarily conserved signaling pathways and requires an intact vascular niche. Targeting these pathways and the vascular niche could be a principle in novel brain tumor therapies aimed to eliminate BTSCs.


Subject(s)
Brain Neoplasms/pathology , Neoplastic Stem Cells/pathology , Neural Stem Cells/physiology , Animals , Astrocytes/pathology , Brain Neoplasms/therapy , Cell Dedifferentiation , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Drosophila , Glial Fibrillary Acidic Protein , Glioma/etiology , Glioma/pathology , Humans , Intermediate Filament Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nestin , Signal Transduction , Tumor Suppressor Protein p14ARF/deficiency
6.
Biomolecules ; 9(3)2019 03 04.
Article in English | MEDLINE | ID: mdl-30836703

ABSTRACT

The CDKN2a/ARF locus expresses two partially overlapping transcripts that encode two distinct proteins, namely p14ARF (p19Arf in mouse) and p16INK4a, which present no sequence identity. Initial data obtained in mice showed that both proteins are potent tumor suppressors. In line with a tumor-suppressive role, ARF-deficient mice develop lymphomas, sarcomas, and adenocarcinomas, with a median survival rate of one year of age. In humans, the importance of ARF inactivation in cancer is less clear whereas a more obvious role has been documented for p16INK4a. Indeed, many alterations in human tumors result in the elimination of the entire locus, while the majority of point mutations affect p16INK4a. Nevertheless, specific mutations of p14ARF have been described in different types of human cancers such as colorectal and gastric carcinomas, melanoma and glioblastoma. The activity of the tumor suppressor ARF has been shown to rely on both p53-dependent and independent functions. However, novel data collected in the last years has challenged the traditional and established role of this protein as a tumor suppressor. In particular, tumors retaining ARF expression evolve to metastatic and invasive phenotypes and in humans are associated with a poor prognosis. In this review, the recent evidence and the molecular mechanisms of a novel role played by ARF will be presented and discussed, both in pathological and physiological contexts.


Subject(s)
Adenocarcinoma/metabolism , Lymphoma/metabolism , Sarcoma/metabolism , Tumor Suppressor Protein p14ARF/chemistry , Tumor Suppressor Protein p14ARF/metabolism , Adenocarcinoma/genetics , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Lymphoma/genetics , Mice , Mice, Knockout , Sarcoma/genetics , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Protein p14ARF/genetics
7.
Oncogene ; 26(7): 1088-97, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-16909113

ABSTRACT

The molecular pathogenesis of pleomorphic xanthoastrocytoma (PXA), a rare astrocytic brain tumor with a relatively favorable prognosis, is still poorly understood. We characterized 50 PXAs by comparative genomic hybridization (CGH) and found the most common imbalance to be loss on chromosome 9 in 50% of tumors. Other recurrent losses affected chromosomes 17 (10%), 8, 18, 22 (4% each). Recurrent gains were identified on chromosomes X (16%), 7, 9q, 20 (8% each), 4, 5, 19 (4% each). Two tumors demonstrated amplifications mapping to 2p23-p25, 4p15, 12q13, 12q21, 21q21 and 21q22. Analysis of 10 PXAs with available high molecular weight DNA by high-resolution array-based CGH indicated homozygous 9p21.3 deletions involving the CDKN2A/p14(ARF)/CDKN2B loci in six tumors (60%). Interphase fluorescence in situ hybridization to tissue sections confirmed the presence of tumor cells with homozygous 9p21.3 deletions. Mutational analysis of candidate genes on 9q, PTCH and TSC1, revealed no mutations in PXAs with 9q loss and no evidence of TSC1 promoter methylation. However, PXAs consistently showed low TSC1 transcript levels. Taken together, our study identifies loss of chromosome 9 as the most common chromosomal imbalance in PXAs and suggests important roles for homozygous CDKN2A/p14(ARF)/CDKN2B deletion as well as low TSC1 mRNA expression in these tumors.


Subject(s)
Astrocytoma/genetics , Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Deletion , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Proteins/deficiency , Adolescent , Adult , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p15/deficiency , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Female , Homozygote , Humans , Male , Middle Aged , RNA, Messenger/biosynthesis , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
8.
Cancer Res ; 66(5): 2608-15, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16510579

ABSTRACT

Nevi and melanomas correlate to childhood and intermittent solar UV exposure, xeroderma pigmentosum patients run increased risk, and p16(Ink4a) expression is often lost in malignant progression. To ascertain the effect of these risk factors, pigmented hairless Ink4a/Arf-, Xpa- knockout mice were subjected to various combinations of neonatal [7,12-dimethylbenz(a)anthracene (DMBA) or UVB exposure] and adult treatments (12-O-tetradecanoylphorbol-13-acetate or subacute daily UVB exposure or intermittent overexposure). Nevi occurred earliest, grew largest, and were most numerous in mice exposed to DMBA followed by intermittent UVB overexposure [effect of six minimal edemal doses (MED), 1 x /2 weeks > 4 MED 1 x /wk]. Neonatal UV exposure enhanced nevus induction but lost its effect after 200 days. The Xpa(-/-) mice proved exquisitely sensitive to UV-driven nevus induction, indicating the involvement of pyrimidine dimer DNA lesions, but Xpa(+/+) mice developed many more nevi (>40 per mouse) at high UV dosages not tolerated by Xpa(-/-) mice. Ink4a/Arf(-/-) mice developed most skin tumors faster, but surprisingly developed nevi slower than their heterozygous counterparts especially after neonatal UV exposure. Despite raising >1,600 nevi, only six melanomas arose in our experiments with Ink4a/Arf knockout mice (five of which in Xpa(+/+) mice at high UV dosages). In contrast to human nevi, these nevi lacked hotspot mutations in Braf or Ras genes, possibly explaining the lack of malignant progression in the Ink4a/Arf(-/-) mice. Hence, although our experiments did not effectively emulate human melanoma, they provided clear evidence that intermittent UV overexposure strongly stimulates and the Ink4a/Arf(-/-) genotype may actually impair nevus development.


Subject(s)
Cocarcinogenesis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Neoplasms, Radiation-Induced/etiology , Nevus/etiology , Skin Neoplasms/etiology , Tumor Suppressor Protein p14ARF/genetics , Xeroderma Pigmentosum Group A Protein/genetics , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/genetics , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Melanoma, Experimental/etiology , Melanoma, Experimental/genetics , Mice , Mice, Knockout , Neoplasms, Radiation-Induced/chemically induced , Neoplasms, Radiation-Induced/genetics , Nevus/genetics , Sarcoma/etiology , Sarcoma/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Tumor Suppressor Protein p14ARF/deficiency , Ultraviolet Rays
9.
Cancer Res ; 66(15): 7473-81, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16885344

ABSTRACT

Glioblastoma multiforme is the most common and lethal form of primary brain cancer. Diagnosis of this advanced glioma has a poor prognosis due to the ineffectiveness of current therapies. Aberrant expression of receptor tyrosine kinases (RTK) in glioblastoma multiformes is suggestive of their role in initiation and maintenance of these tumors of the central nervous system. In fact, ectopic expression of the orphan RTK ROS is a frequent event in human brain cancers, yet the pathologic significance of this expression remains undetermined. Here, we show that a glioblastoma-associated, ligand-independent rearrangement product of ROS (FIG-ROS) cooperates with loss of the tumor suppressor gene locus Ink4a;Arf to produce glioblastomas in the mouse. We show that this FIG-ROS-mediated tumor formation in vivo parallels the activation of the tyrosine phosphatase SH2 domain-containing phosphatase-2 (SHP-2) and a phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling axis in tumors and tumor-derived cell lines. We have established a fully penetrant preclinical model for adult onset of glioblastoma multiforme in keeping with major genetic events observed in the human disease. These findings provide novel and important insights into the role of ROS and SHP-2 function in solid tumor biology and set the stage for preclinical testing of targeted therapeutic approaches.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Astrocytoma/enzymology , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Enzyme Activation , Glioblastoma/enzymology , Glioblastoma/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Protein Phosphatase 2 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Signal Transduction , TOR Serine-Threonine Kinases , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Protein p14ARF/genetics , src Homology Domains
10.
Mol Cells ; 41(5): 381-389, 2018 May 31.
Article in English | MEDLINE | ID: mdl-29665672

ABSTRACT

ARF is a tumor suppressor protein that has a pivotal role in the prevention of cancer development through regulating cell proliferation, senescence, and apoptosis. As a factor that induces senescence, the role of ARF as a tumor suppressor is closely linked to the p53-MDM2 axis, which is a key process that restrains tumor formation. Thus, many cancer cells either lack a functional ARF or p53, which enables them to evade cell oncogenic stress-mediated cycle arrest, senescence, or apoptosis. In particular, the ARF gene is a frequent target of genetic and epigenetic alterations including promoter hyper-methylation or gene deletion. However, as many cancer cells still express ARF, pathways that negatively modulate transcriptional or post-translational regulation of ARF could be potentially important means for cancer cells to induce cellular proliferation. These recent findings of regulators affecting ARF protein stability along with its low levels in numerous human cancers indicate the significance of an ARF post-translational mechanism in cancers. Novel findings of regulators stimulating or suppressing ARF function would provide new therapeutic targets to manage cancer- and senescence-related diseases. In this review, we present the current knowledge on the regulation and alterations of ARF expression in human cancers, and indicate the importance of regulators of ARF as a prognostic marker and in potential therapeutic strategies.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p18/physiology , Neoplasm Proteins/physiology , Tumor Suppressor Protein p14ARF/physiology , Apoptosis , Cell Division , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p18/deficiency , Cyclin-Dependent Kinase Inhibitor p18/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, p16 , Humans , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Protein Stability , Proto-Oncogene Proteins c-mdm2/physiology , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p53/physiology , Ubiquitination
11.
Oncogene ; 25(21): 3023-31, 2006 May 18.
Article in English | MEDLINE | ID: mdl-16407836

ABSTRACT

Analysis of the INK4A/ARF locus in human T-ALL patients revealed frequent deletions in exon 2, the exon common to both p16(INK4A) and p14(ARF). Other studies have described selective deletion of exon 1beta of p14(ARF) or methylation of the p16(INK4A) promoter. Therefore, it is unclear from these studies whether loss of p16(INK4A) and/or p14(ARF) contributes to the development of T-ALL. To elucidate the relative contribution of the ink4a/arf locus to T-cell leukemogenesis, we mated our tal1 transgenic mice to ink4a/arf-/-, p16(ink4a)-/-, and p19(arf)-/- mice and generated tal1/ink4a/arf+/-, tal1/p16(ink4a)+/-, and tal1/p19(arf)+/- mice. Each of these mice developed T-cell leukemia rapidly, indicating that loss of either p16(ink4a) or p19(arf) cooperates with Tal1 to induce leukemia in mice. Preleukemic studies reveal that Tal1 expression stimulates entry into the cell cycle and thymocyte apoptosis in vivo. Interestingly, mice expressing a DNA-binding mutant of Tal1 do not exhibit increases in S phase cells. The S phase induction is accompanied by an increase in thymocyte apoptosis in tal1 transgenic mice. Whereas apoptosis is reduced to wild-type levels in tal1/ink4a/arf-/- mice, S phase induction remains unaffected. Thus, Tal1 stimulates cell cycle entry independent of the ink4a/arf locus, but its ability to induce apoptosis is Ink4a/Arf-dependent.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p16/physiology , Gene Deletion , Leukemia-Lymphoma, Adult T-Cell/genetics , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p14ARF/physiology , Animals , Antigens, CD/analysis , Antigens, Neoplasm/analysis , Apoptosis/genetics , Cell Cycle/genetics , Cyclin-Dependent Kinase Inhibitor p16/deficiency , DNA/metabolism , Exons/genetics , Gene Expression Regulation , Gene Expression Regulation, Leukemic , Genes, p16 , Glycoproteins/physiology , Immunophenotyping , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Preleukemia/genetics , Preleukemia/metabolism , Protein Binding , Protein Structure, Tertiary , Receptor, Notch1/physiology , S Phase/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1 , T-Lymphocyte Subsets/pathology , Transcription Factors/physiology , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Protein p14ARF/genetics , Wnt Proteins/physiology
12.
Oncogene ; 25(29): 4110-5, 2006 Jul 06.
Article in English | MEDLINE | ID: mdl-16491120

ABSTRACT

In contrast to other cell cycle inhibitors, the tumor suppressor p16Ink4a is not detectable or expressed at very low levels in embryonic and adult mouse tissues, and therefore it has often been considered as a specialized checkpoint protein that does not participate in the control of normal cell cycle progression. However, Ink4a-/- mice possess increased thymus size and cellularity, thus suggesting the involvement of p16(Ink4a) in the control of thymocyte proliferation. In this study, we found increased numbers of CD8 and CD4 T lymphocytes in thymus and spleen from Ink4a-/- mice. Unexpectedly, this was not related to an increase in T-cell division rates, which were similar in lymphoid organs of Ink4a-/- and wild-type mice. In contrast, T-cell apoptosis rates were significantly decreased in thymus and spleen from Ink4a-/- mice. Moreover, whereas p16Ink4a-deficient and wild-type T cells were equally sensitive to Fas or TCR-mediated apoptosis, the former were clearly more resistant to apoptosis induced by oxidative stress or gamma irradiation. Our results indicate that p16Ink4a function is associated with T-cell apoptosis, and subsequently contributes to the control of T-cell population size in lymphoid organs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Thymus Gland/immunology , Tumor Suppressor Protein p14ARF/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Apoptosis/radiation effects , CD4-Positive T-Lymphocytes/cytology , Cell Division/genetics , Cell Division/immunology , Cell Division/radiation effects , Cell Survival/genetics , Cell Survival/immunology , Cell Survival/radiation effects , Cyclin-Dependent Kinase Inhibitor p16 , Gamma Rays , Mice , Mice, Knockout , Organ Size/immunology , Organ Size/radiation effects , Oxidative Stress/genetics , Oxidative Stress/immunology , Oxidative Stress/radiation effects , Receptors, Antigen, T-Cell/immunology , Thymus Gland/cytology , Thymus Gland/embryology , Tumor Suppressor Protein p14ARF/deficiency , fas Receptor/immunology
13.
Mol Cell Biol ; 22(15): 5308-18, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12101227

ABSTRACT

It has been proposed that the E2F1 transcription factor serves as a link between the Rb/E2F proliferation pathway and the p53 apoptosis pathway by inducing the expression of p19ARF, a protein that regulates p53 stability. We find that although p19ARF contributes to p53 accumulation in response to E2F expression, p19ARF is not required for E2F1-mediated apoptosis. E2F1 can signal p53 phosphorylation in the absence of p19ARF, similar to the observed modifications to p53 in response to DNA damage. These modifications are not observed in the absence of p19ARF following expression of E2F2, an E2F family member that does not induce apoptosis in mouse embryo fibroblasts but can induce p19ARF and p53 protein expression. p53 modification is found to be crucial for E2F1-mediated apoptosis, and this apoptosis is compromised when E2F1 is coexpressed with a p53 mutant lacking many N- and C-terminal phosphorylation sites. Additionally, E2F1-mediated apoptosis is abolished in the presence of caffeine, an inhibitor of phosphatidylinositol 3-kinase-related kinases that phosphorylate p53. These findings suggest that p53 phosphorylation is a key step in E2F1-mediated apoptosis and that this modification can occur in the absence of p19ARF.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins , DNA-Binding Proteins , Nuclear Proteins , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Cyclin-Dependent Kinase Inhibitor p16 , E2F Transcription Factors , E2F1 Transcription Factor , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Mice , Mutation , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Transcription Factors/genetics , Transfection , Tumor Suppressor Protein p14ARF/deficiency , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
16.
Cancer Res ; 65(6): 2065-9, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15781613

ABSTRACT

Homozygous deletion of the INK4a-ARF locus is one of the most frequent mutations found in human glioblastoma. We have previously shown that combined Ink4a-Arf loss can increase tumor incidence in both glial progenitor cells and astrocytes during mouse gliomagenesis. Here we have investigated the separate contribution of loss of each of the tumor suppressor genes in glial progenitor cells and astrocytes in Akt + Kras-induced gliomagenesis. We show that Arf is the major tumor suppressor gene in both cell types. Arf loss generated glioblastomas from both nestin-expressing glial progenitor cells and glial fibrillary acidic protein-expressing astrocytes, with a significantly higher incidence in astrocytes. Ink4a loss, on the other hand, could only significantly contribute to gliomagenesis from glial progenitor cells and the induced tumors were of lower malignancy than those seen in Arf-deficient mice. Thus, Ink4a and Arf have independent and differential tumor suppressor functions in vivo in the glial cell compartment.


Subject(s)
Brain Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Genes, ras/physiology , Glioma/genetics , Neuroglia/physiology , Stem Cells/physiology , Tumor Suppressor Protein p14ARF/genetics , Animals , Astrocytes/pathology , Astrocytes/physiology , Brain Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Genes, Tumor Suppressor , Glioma/pathology , Mice , Mice, Transgenic , Neuroglia/cytology , Stem Cells/cytology , Tumor Suppressor Protein p14ARF/deficiency
17.
Biochim Biophys Acta ; 1744(1): 76-87, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15878400

ABSTRACT

In the healthy adult liver, quiescent hepatic stellate cells (HSCs) present the major site for vitamin A storage in cytoplasmic lipid droplets. During liver injury due to viral infection or alcohol intoxication, HSCs get activated and produce high amounts of extracellular matrix components for tissue repair and fibrogenesis. Employing p19 ARF deficiency, we established a non-transformed murine HSC model to investigate their plasticity and the dynamics of HSC activation. Primary HSCs isolated from livers of adult p19 ARF null mice underwent spontaneous activation through long-term passaging without an obvious replicative limit. The immortalized cell line, referred to as M1-4HSC, showed stellate cell characteristics including the expression of desmin, glial fibrillary acidic protein, alpha-smooth muscle actin and pro-collagen I. Treatment of these non-tumorigenic M1-4HSC with pro-fibrogenic TGF-beta1 provoked a morphological transition to a myofibroblastoid cell type which was accompanied by enhanced cellular turnover and impaired migration. In addition, M1-4HSCs expressed constituents of cell adhesion complexes such as p120(ctn) and beta-catenin at cell borders, which dislocalized in the cytoplasm during stimulation to myofibroblasts, pointing to the epitheloid characteristics of HSCs. By virtue of its non-transformed phenotype and unlimited availability of cells, the p19(ARF) deficient model of activated HSCs and corresponding myofibroblasts render this system a highly valuable tool for studying the cellular and molecular basis of hepatic fibrogenesis.


Subject(s)
Cell Line , Liver Cirrhosis, Experimental/etiology , Liver/cytology , Tumor Suppressor Protein p14ARF/deficiency , Actins/metabolism , Animals , Apoptosis , Cell Communication , Cell Differentiation/drug effects , Cell Division , Cell Movement , Cyclin-Dependent Kinase Inhibitor p16 , Desmin/metabolism , Extracellular Matrix/metabolism , Glial Fibrillary Acidic Protein/metabolism , Liver/drug effects , Liver/metabolism , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Mice , Procollagen/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Protein p14ARF/genetics
18.
Mech Ageing Dev ; 127(7): 633-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16620915

ABSTRACT

The murine INK4a locus encodes the critical tumor suppressor proteins, p16(INK4a) and p19(ARF). Mice lacking both p16(INK4a) and p19(ARF) (INK4a-/-) in their FVB/NJ genetic backgrounds developed cataracts and microophthalmia. Histopathologically, INK4a-/- mice showed defects in the developmental regression of the hyaloid vascular system (HVS), retinal dysplasia, and cataracts with numerous vacuolations, closely resembling human persistent hyperplastic primary vitreous (PHPV). Ocular defects, such as retinal fold and abnormal migration of lens fiber cells, were observed as early as embryonic day (E) 15.5, thereby resulting in the abnormal differentiation of the lens. We also found that ectopic expression of p16(INK4a) resulted in the induction of gammaF-crystallin, suggesting an important role of INK4a locus during mouse eye development, and also providing insights into the potential genetic basis of human cataract genesis.


Subject(s)
Cataract/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Eye/embryology , Quantitative Trait Loci/genetics , Tumor Suppressor Protein p14ARF/metabolism , Animals , Cataract/genetics , Cataract/pathology , Cell Movement/genetics , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Eye/blood supply , Eye/pathology , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Mice, Knockout , Retinal Dysplasia/genetics , Retinal Dysplasia/metabolism , Tumor Suppressor Protein p14ARF/deficiency , gamma-Crystallins/biosynthesis
19.
Cancer Res ; 62(7): 2184-91, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11929842

ABSTRACT

p19(ARF) is a key regulator of the p53-mediated apoptotic and tumor suppressor pathway. The proapoptotic Bax gene is a transcription target of p53, yet genetic studies in some animal models have suggested that Bax and p53 loss may cooperate in tumorigenesis. ARF-deficient mice are tumor prone, and to determine whether Bax loss could cooperate in the development of these tumors, we generated mice null for both ARF and Bax. The tumor latency of Bax+/+ARF-/-, Bax+/-ARF-/- and Bax-/-ARF-/- mice was similar with a mean survival of 48.9, 48.1, and 47.6 weeks, respectively. In Bax+/+ARF-/- mice, the predominant tumor type was B- and T-cell lymphoma followed by sarcomas and a lack of carcinomas. However, the frequency of lymphoma development dramatically decreased, whereas that of sarcomas and carcinomas increased, in a gene dosage-dependent manner in Bax+/-ARF-/- and Bax-/-ARF-/- mice. Furthermore, uncommon tumors of ARF-/- mice (osteosarcoma and hemangiosarcoma) were observed in Bax/ARF-double null mice, and tumor types not described previously in ARF-null mice (mixed germ cell tumor, Triton tumor, and histiocytic sarcoma) also developed in Bax-/-ARF-/- animals. Importantly, multiple primary malignant tumors of different lineage arose in 25% of the Bax-/-ARF-/- mice, whereas only one tumor type per animal was observed in Bax+/+ARF-null littermates. Finally, the wild-type Bax allele was retained in tumors arising in Bax+/-ARF-/- mice. Thus, Bax appears to function as a tumor modifier rather than as a classic tumor suppressor, and the combined loss of Bax and the ARF allows for the emergence of multiple malignant tumor types, an alteration of the tumor spectrum, and tumors not observed previously in ARF-null mice.


Subject(s)
Neoplasms, Experimental/genetics , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p14ARF/deficiency , Animals , Cyclin-Dependent Kinase Inhibitor p16 , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/physiology , bcl-2-Associated X Protein
20.
Oncogene ; 23(2): 379-85, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14724566

ABSTRACT

Mounting genetic evidence suggests that each product of the Ink4a/Arf locus, p16(INK4a) and p19(ARF), possesses tumor-suppressor activity (Kamijo et al., 1997; Krimpenfort et al., 2001; Sharpless et al., 2001a). We report the generation and characterization of a p19(ARF)-specific knockout allele (p19(ARF)-/-) and direct comparison with mice and derivative cells deficient for p16(INK4a), both p16(INK4a) and p19(ARF), and p53. Like Ink4a/Arf-/- murine embryo fibroblasts (MEFs), p19(ARF)-/- MEFs were highly susceptible to oncogenic transformation, exhibited enhanced subcloning efficiency at low density, and resisted both RAS- and culture-induced growth arrest. In contrast, the biological profile of p16(INK4a)-/- MEFs in these assays more closely resembled that of wild-type cells. In vivo, however, both p19(ARF)-/- and p16(INK4a)-/- animals were significantly more tumor prone than wild-type animals, but each less so than p53-/- or Ink4a/Arf-/- animals, and with differing tumor spectra. These data confirm the predominant role of p19(ARF) over p16(INK4a) in cell culture-based assays of MEFs, yet also underscore the importance of the analysis of tumor suppressors across many cell types within the organism. The cancer-prone conditions of mice singly deficient for either p16(INK4a) or p19(ARF) agree with data derived from human cancer genetics, and reinforce the view that both gene products play significant and nonredundant roles in suppressing malignant transformation in vivo.


Subject(s)
Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Neoplasms/metabolism , Neoplasms/pathology , Tumor Suppressor Protein p14ARF/deficiency , Alleles , Animals , Cell Division , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Embryo, Mammalian/cytology , Fibroblasts , Gene Deletion , Genotype , Mice , Neoplasms/genetics , Phenotype , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL