Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Oncogene ; 34(39): 5069-79, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-25579177

ABSTRACT

Osteosarcoma (OS) is the most frequent pediatric malignant bone tumor that has a high propensity for metastases. Through osteoblast-specific alteration of p53 status, we developed a genetically engineered mouse model of localized and metastatic OS to gain an understanding into the molecular pathogenesis of OS. Microarray analysis of both localized tumors and metastatic tumors identified the downregulation of the naked cuticle homolog 2 (NKD2) gene, a negative regulator of Wnt signaling. Overexpression of NKD2 in metastatic human and mouse OS cells significantly decreases cell proliferation, migration and invasion ability in vitro and drastically diminishes OS tumor growth and metastasis in vivo, whereas downregulation enhances migratory and invasive potential. Evaluation of NKD2-overexpressing tumors revealed upregulation of tumor-suppressor genes and downregulation of molecules involved in blood vessel formation and cell migration. Furthermore, assessment of primary human OS revealed downregulation of NKD2 in metastatic and recurrent OS. Finally, we provide biological evidence that use of small-molecule inhibitors targeting the Wnt pathway can have therapeutic efficacy in decreasing metastatic properties in OS. Our studies provide compelling evidence that downregulation of NKD2 expression and alterations in associated regulated pathways have a significant role in driving OS tumor growth and metastasis.


Subject(s)
Bone Neoplasms/metabolism , Carrier Proteins/physiology , Cell Proliferation , Neoplasm Metastasis , Osteosarcoma/metabolism , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing , Animals , Bone Neoplasms/pathology , Calcium-Binding Proteins , Cell Line, Tumor , Mice , Neoplasm Invasiveness , Osteosarcoma/pathology
2.
Oncogene ; 31(9): 1155-65, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21765465

ABSTRACT

Wild-type p53-induced phosphatase 1 (WIP1) is a serine/threonine phosphatase that dephosphorylates proteins in the ataxia telangiectasia mutated (ATM)-initiated DNA damage response pathway. WIP1 may have a homeostatic role in ATM signaling by returning the cell to a normal pre-stress state following completion of DNA repair. To better understand the effects of WIP1 on ATM signaling, we crossed Atm-deficient mice to Wip1-deficient mice and characterized phenotypes of the double knockout progeny. We hypothesized that the absence of Wip1 might rescue Atm deficiency phenotypes. Atm null mice, like ATM-deficient humans with the inherited syndrome ataxia telangiectasia, exhibit radiation sensitivity, fertility defects, and are T-cell lymphoma prone. Most double knockout mice were largely protected from lymphoma development and had a greatly extended lifespan compared with Atm null mice. Double knockout mice had increased p53 and H2AX phosphorylation and p21 expression compared with their Atm null counterparts, indicating enhanced p53 and DNA damage responses. Additionally, double knockout splenocytes displayed reduced chromosomal instability compared with Atm null mice. Finally, doubly null mice were partially rescued from gametogenesis defects observed in Atm null mice. These results indicate that inhibition of WIP1 may represent a useful strategy for cancer treatment in general and A-T patients in particular.


Subject(s)
Cell Cycle Proteins/deficiency , DNA-Binding Proteins/deficiency , Phenotype , Phosphoprotein Phosphatases/deficiency , Protein Serine-Threonine Kinases/deficiency , Tumor Suppressor Proteins/deficiency , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromosomal Instability , DNA Damage , DNA-Binding Proteins/genetics , Female , Gene Knockout Techniques , Genetic Association Studies , Germ Cells/metabolism , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/mortality , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2C , Protein Serine-Threonine Kinases/genetics , Radiation Tolerance/genetics , Spleen/cytology , Spleen/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
3.
Oncogene ; 29(35): 4938-46, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20562916

ABSTRACT

Chemoresistance is a major cause of treatment failure and poor outcome in neuroblastoma. In this study, we investigated the expression and function of dual-specificity phosphatase 26 (DUSP26), also known as mitogen-activated protein kinase phophatase-8, in human neuroblastoma. We found that DUSP26 was expressed in a majority of neuroblastoma cell lines and tissue specimens. Importantly, we found that DUSP26 promotes the resistance of human neuroblastoma to doxorubicin-induced apoptosis by acting as a p53 phosphatase to downregulate p53 tumor suppressor function in neuroblastoma cells. Inhibiting DUSP26 expression in the IMR-32 neuroblastoma cell line enhanced doxorubicin-induced p53 phosphorylation at Ser20 and Ser37, p21, Puma, Bax expression as well as apoptosis. In contrast, DUSP26 overexpression in the SK-N-SH cell line inhibited doxorubicin-induced p53 phosphorylation at Ser20 and Ser37, p21, Puma, Bax expression and apoptosis. Using in vitro and in vivo assays, we found that DUSP26 binds to p53 and dephosphorylates p53 at Ser20 and Ser37. In this report, we show that DUSP26 functions as a p53 phosphatase, which suppresses downstream p53 activity in response to genotoxic stress. This suggests that inhibition of this phosphatase may increase neuroblastoma chemosensitivity and DUSP26 is a novel therapeutic target for this aggressive pediatric malignancy.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Neuroblastoma/enzymology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation, Neoplastic , Humans , Mitogen-Activated Protein Kinase Phosphatases/genetics , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phosphorylation , Serine/metabolism , Tumor Suppressor Protein p53/chemistry
4.
Mutat Res ; 540(2): 165-76, 2003 Oct 07.
Article in English | MEDLINE | ID: mdl-14550500

ABSTRACT

Genetically altered mouse models (GAMM) for human cancers have been critical to the investigation and characterization of oncogene and tumor suppressor gene expression and function and the associated cancer phenotype. Similarly, several of the mouse models with defined genetic alterations have shown promise for identification of potential human carcinogens and investigation of mechanisms of carcinogen-gene interactions and tumorigenesis. In particular, both the B6.129N5-Trp53 mouse, heterozygous for a p53 null allele, and the CB6F1-RasH2 mouse, hemizygous for the human H-ras transgene, have been extensively investigated. Using 26-week exposure protocols at or approaching the maximum tolerated dose, the summary results to date indicate the potential for GAMM to identify and, possibly, classify chemicals of potential risk to humans using short-term carcinogenicity experiments. This IWGT session focused on: (1) the development of recommendations for genetic/molecular characterization required in animals, tissues, and tumors before and after treatment for identification of presumptive human carcinogens based on the current state of knowledge, (2) identification of data gaps in our current state of knowledge, and (3) development of recommendations for research strategies for further development of our knowledge base of these particular models. By optimization of protocols and identification of significant outcomes and responses to chemical exposure in appropriate short-term mechanism-based genetically altered rodent models, strategies for prevention and intervention may be developed and employed to the benefit of public health.


Subject(s)
Biological Assay/standards , Genes, Tumor Suppressor , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics , Animals , Carcinogenicity Tests/standards , Disease Models, Animal , Mice , Tumor Suppressor Protein p53/deficiency , ras Proteins/metabolism
5.
Toxicol Pathol ; 29 Suppl: 147-54, 2001.
Article in English | MEDLINE | ID: mdl-11695551

ABSTRACT

The p53 tumor suppressor gene has been shown to be critical in preventing cancer in humans and mice. We have generated and extensively characterized p53-deficient mice lacking one (p53+/-) or both (p53-/-) p53 alleles. The p53-deficient mice are much more susceptible to an array of different tumor types than their wild-type (p53+/+) littermates. The enhanced tumor susceptibility of the p53+/- mice has made them one of several transgenic mouse models that are being considered as substitutes for standard 2-year rodent carcinogenicity assays. In order to fully exploit this model, it will be important to understand some of the basic biological and molecular mechanisms that underlie its enhanced tumor susceptibility. With this in mind, we have explored the fate of the remaining wild-type p53 allele in spontaneously arising p53+/- tumors and have shown that over half of these tumors retain an intact, functional wild-type p53 allele. This suggests that p53 is haploinsufficient for tumor suppression and that mere reduction in p53 dosage is sufficient to promote cancer formation. To support the idea that p53 is indeed a haploinsufficient tumor suppressor, we show here that normal p53+/- cells exhibit reduced parameters of growth control and stress response compared to their p53+/- counterparts. We hypothesize that the reduced p53 dosage in the p53+/- cells provides an environment more conducive to the development of further oncogenic lesions and the initiation of a tumor. Finally, we have assessed p53 loss of heterozygosity (LOH) in carcinogen-induced p53+/- tumors and have found that some agents induce tumors that almost invariably exhibit p53 LOH, whereas other agents induce tumors that often retain the wild-type p53 allele. Our preliminary data suggest that LOH is dependent on both the mechanism of genotoxicity of the agent utilized and the tissue type targeted.


Subject(s)
Carcinogenicity Tests/methods , Carcinogens/toxicity , Genes, p53 , Mutagens/toxicity , Neoplasms, Experimental/chemically induced , Animal Testing Alternatives , Animals , Gene Dosage , Genetic Predisposition to Disease , Genotype , Loss of Heterozygosity , Mice , Mice, Knockout , Mice, Transgenic , Neoplasms, Experimental/genetics
6.
Toxicol Pathol ; 29 Suppl: 24-9, 2001.
Article in English | MEDLINE | ID: mdl-11695559

ABSTRACT

The heterozygous Trp53 null allele C57BL/6 (N5) mouse is susceptible to the rapid development of neoplasia by mutagenic carcinogens relative to control strains. This mouse model of chemical carcinogenesis demonstrates 1) dose-related rapid induction of tumors (26 wks), 2) multiple sites of carcinogen-specific tissue susceptibility, and 3) carcinogen-induced loss of heterozygosity involving the Trp53 wild-type allele or a p53 haploinsufficiency permitting mutation of other critical protooncogenes and/or inactivation of tumor suppressor genes driving tumorigenesis. Demonstration of mutation or loss of heterozygosity involving the Trp53 locus is consistent with a common finding in human cancers and supports extrapolation between rodents and humans. Using diverse experimental protocols, almost all mutagenic rodent carcinogens (including all mutagens that are carcinogenic to humans), but not nonmutagenic rodent carcinogens, induce tumors within 26 weeks of continuous exposure. These characteristics and results indicate that the mouse heterozygous for the Trp53 null allele may be of significant use for the prospective identification of mutagenic carcinogens of potential risk to human health.


Subject(s)
Carcinogenicity Tests/methods , Carcinogens/toxicity , Disease Models, Animal , Genes, p53 , Mutagens/toxicity , Neoplasms, Experimental/genetics , Animals , Female , Heterozygote , Loss of Heterozygosity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/chemically induced , Risk Assessment
7.
Toxicol Pathol ; 29(1): 137-41, 2001.
Article in English | MEDLINE | ID: mdl-11215677

ABSTRACT

Future progress in mechanism-based cancer prevention research may be facilitated by animal models displaying specific genetic susceptibilities for cancer, such as mice deficient in 1 (+/-) or both (-/-) alleles of the p53 tumor suppressor gene. We observed in p53-/- mice that calorie restriction (CR) increased the latency of spontaneous tumor development (mostly lymphomas) by approximately 75%, decreased serum insulin-like growth factor-1 (IGF-1) and leptin levels, slowed thymocyte cell cycle traverse, and induced apoptosis in immature thymocytes. In p53+/- mice, CR and a 1 d/wk fast each delayed spontaneous tumor development (a mix of lymphomas, sarcomas, and epithelial tumors) and decreased serum IGF-1 and leptin levels, even when begun late in life. In p53+/-Wnt-1 transgenic mice, a mammary tumor model, the same interventions increased mammary tumor latency and reduced mean serum IGF-1 and leptin levels to <50% of those of control mice. We capitalized on the susceptibility of p53+/- mice to chronic, low-dose aromatic amine-induced bladder carcinogenesis to develop a useful model for evaluating bladder cancer prevention approaches. These examples clearly indicate that mice with specific (and humanlike) genetic susceptibilities for cancer are powerful models for testing interventions that may inhibit carcinogenesis in humans.


Subject(s)
Neoplasms/prevention & control , Tumor Suppressor Protein p53/deficiency , Animals , Female , Mice , Mice, Transgenic , Pregnancy , Tumor Suppressor Protein p53/genetics
8.
Mol Carcinog ; 29(2): 67-75, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11074603

ABSTRACT

To investigate the role of loss of the p53 tumor suppressor gene in skin carcinogenesis, p53 knockout (p53(-/-)) mice were mated with transgenic mice coexpressing v-Ha-ras, v-fos, or human transforming growth factor alpha (TGFalpha) exclusively in the epidermis by using human keratin 1 (HK1)-based vectors (HK1.ras/fos, HK1.ras/alpha, and HK1.fos/alpha). HK1.ras/fos and HK1.ras/alpha mice displayed epidermal hyperplasia and autonomous benign papillomas to an identical degree between p53(+/+) and p53(+/-) genotypes. However, HK1.ras/fos mice with the p53(-/-) genotype were born with papillomatous skin and died soon after birth. HK1.ras/alpha-p53(-/-) mice also exhibited an increased epidermal hyperplasia, and, similar to HK1.ras/alpha mice with p53(+/+) and p53(+/-) genotypes, these mice rapidly developed spontaneous and 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced papillomas. These results are in contrast to our previous observation that, HK1.ras, HK1.fos, and HK1.TGFalpha transgenic mice with the p53(-/-) genotype display an unexpected delay in both spontaneous and TPA-promoted papilloma formation compared with mice with p53(+/+) and p53(+/-) genotypes. Taken collectively, our mating experiments between HK1 oncogenic transgenic mice and p53 knockout mice may identify a backup system that effectively compensates for p53 loss. Activation of multiple oncogenes not only partly overcomes such compensation but also synergizes with p53 loss. However, HK1.fos/alpha-p53(-/-) mice failed to exhibit either an increased newborn epidermal hyperplasia or an accelerated spontaneous or TPA-induced papillomas, suggesting that certain combinations of oncogenes, such as with activated Ha-ras, are required for this process. Because neither spontaneous nor TPA-elicited papillomas in p53(-/-) mice progressed to malignancy, additional genetic insults appear to be required for malignant progression.


Subject(s)
Genes, fos/physiology , Genes, p53/physiology , Genes, ras/physiology , Papilloma/genetics , Skin Neoplasms/genetics , Transforming Growth Factor alpha/genetics , Animals , Carcinogens , Epidermis/metabolism , Epidermis/physiology , Female , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Keratins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Papilloma/chemically induced , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate
9.
Carcinogenesis ; 21(10): 1891-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11023548

ABSTRACT

Mutations of the p53 tumor suppressor gene constitute one of the most frequent molecular changes in a wide variety of human cancers. Mice deficient in p53 have recently attracted attention for their potential to identify chemical genotoxins. In this study we have investigated the susceptibility of p53 nullizygote (-/-), heterozygote (+/-) and wild-type (+/+) mice to N:-methyl-N:-nitrosourea (MNU) gastric carcinogenesis. p53 knockout mice were treated with 30 p.p.m. MNU in the drinking water 1 week on and 1 week off and killed after 5 weeks. The numbers of pepsinogen-altered pyloric glands (PAPG), putative preneoplastic lesions, were 1.8, 1.7 and 22.6 in p53 (+/+), (+/-) and (-/-) mice, respectively. In a 15 week experiment, adenomas were found in 0 of 19 (+/+) (0%), 2 of 21 (+/-) (9.5%) and 6 of 10 (-/-) (60.0%) animals. Also, one well-differentiated adenocarcinoma was observed in a p53 (-/-) mouse. After 40 weeks treatment with 120 or 30 p.p.m. MNU there was no significant difference in the incidence of gastric tumors between p53 (+/+) and (+/-) mice. However, mortality from carcinogen-induced lymphomas, leukemias and sarcomas was very much greater in the latter group. Homozygous knockout animals could not be maintained long term. PCR-single strand conformation polymorphism analysis of exons 5-8 of the p53 gene of DNA extracts from 68 gastric tumors consisting of 16 and 20 30 p.p.m. MNU-treated p53 (+/+) and (+/-) mice and 14 and 18 120 p.p.m. MNU-treated p53 (+/+) and (+/-) mice demonstrated no mutations. These results suggest that p53 may not be a direct target of MNU but rather play an important role as a gatekeeper in mouse stomach carcinogenesis induced by this direct acting agent.


Subject(s)
Carcinogens/toxicity , Cocarcinogenesis , Genes, p53/genetics , Genetic Predisposition to Disease/genetics , Methylnitrosourea/toxicity , Stomach Neoplasms/genetics , Animals , Female , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gene Dosage , Heterozygote , Male , Mice , Mice, Knockout , Mutation , Pepsinogen A/metabolism , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Precancerous Conditions/chemically induced , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Stomach Neoplasms/chemically induced , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
10.
J Biol Chem ; 275(42): 32649-57, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-10915780

ABSTRACT

The cyclin D1 gene encodes the regulatory subunit of a holoenzyme that phosphorylates and inactivates the pRB tumor suppressor protein. Cyclin D1 is overexpressed in 20-30% of human breast tumors and is induced both by oncogenes including those for Ras, Neu, and Src, and by the beta-catenin/lymphoid enhancer factor (LEF)/T cell factor (TCF) pathway. The ankyrin repeat containing serine-threonine protein kinase, integrin-linked kinase (ILK), binds to the cytoplasmic domain of beta(1) and beta(3) integrin subunits and promotes anchorage-independent growth. We show here that ILK overexpression elevates cyclin D1 protein levels and directly induces the cyclin D1 gene in mammary epithelial cells. ILK activation of the cyclin D1 promoter was abolished by point mutation of a cAMP-responsive element-binding protein (CREB)/ATF-2 binding site at nucleotide -54 in the cyclin D1 promoter, and by overexpression of either glycogen synthase kinase-3beta (GSK-3beta) or dominant negative mutants of CREB or ATF-2. Inhibition of the PI 3-kinase and AKT/protein kinase B, but not of the p38, ERK, or JNK signaling pathways, reduced ILK induction of cyclin D1 expression. ILK induced CREB transactivation and CREB binding to the cyclin D1 promoter CRE. Wnt-1 overexpression in mammary epithelial cells induced cyclin D1 mRNA and targeted overexpression of Wnt-1 in the mammary gland of transgenic mice increased both ILK activity and cyclin D1 levels. We conclude that the cyclin D1 gene is regulated by the Wnt-1 and ILK signaling pathways and that ILK induction of cyclin D1 involves the CREB signaling pathway in mammary epithelial cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclin D1/genetics , Gene Expression Regulation , Protein Serine-Threonine Kinases/metabolism , Zebrafish Proteins , Activating Transcription Factor 2 , Animals , Breast Neoplasms , CD18 Antigens/physiology , Cell Line , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Integrin beta1/physiology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Subunits , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured , Wnt Proteins , Wnt1 Protein
11.
Exp Neurol ; 165(1): 184-90, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10964497

ABSTRACT

Familial amyotrophic lateral sclerosis (ALS) has been linked in some families to dominantly inherited mutations in the gene encoding copper-zinc superoxide dismutase 1 (Cu-Zn SOD1). Transgenic mice expressing a mutant human Cu-Zn SOD1 (G93A) develop a dominantly inherited adult-onset paralytic disorder that replicates many of the clinical and pathological features of familial ALS. Increased p53 immunoreactivity has been reported in the motor cortex and spinal ventral horns of postmortem tissue from ALS patients. The nuclear phosphoprotein p53 is an important regulator of cellular proliferation, and increasing evidence supports the role of p53 in regulating cellular apoptosis. To assess the role of p53-mediated apoptosis in amyotrophic lateral sclerosis, mice deficient in both p53 alleles (p53-/-) were crossed with transgenic mice expressing the G93A mutant (G93A+), creating novel transgenic knockout mice. The animals (p53 +/+G93A+, p53+/-G93A+, p53-/-G93A+) were examined at regular intervals for cage activity, upper and lower extremity strength, and mortality. At 120 days from birth mice from each genotype were sacrificed, and L2-L3 anterior horn motor neurons were counted. There was no significant difference in time to onset of behavioral decline, mortality, or motor neuron degeneration between the different genotypes. Despite evidence that p53 plays an important role after acute neuronal injury, the current study suggests that p53 is not significantly involved in cell death in the G93A+ transgenic mouse model of familial ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Tumor Suppressor Protein p53/deficiency , Amyotrophic Lateral Sclerosis/mortality , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Apoptosis/physiology , Cell Count , Forelimb/physiopathology , Genotype , Humans , Mice , Mice, Transgenic/genetics , Motor Activity , Motor Neurons/pathology , Mutation , Spinal Cord/pathology , Superoxide Dismutase/genetics , Tumor Suppressor Protein p53/physiology
12.
Mol Carcinog ; 28(2): 97-101, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10900466

ABSTRACT

Nullizygous p53 knockout (p53(-/-)) mice are highly susceptible to spontaneous tumorigenesis, in particular malignant lymphomas at an early age. Heterozygous p53 knockout (p53(+/-)) mice develop spontaneous tumors less frequently but may show increased susceptibility to chemical carcinogens. In this study, p53(-/-), p53(+/-), and p53 wild-type (p53(+/+)) mice were treated with N-methylnitrosourea (MNU) by gastric intubation (5 microg/g body weight) three times per week for 5 wk, starting at 5-6 wk of age. The surviving mice were killed when they were 56-57 wk old. All eight p53(-/-) mice treated with MNU developed malignant lymphomas with a shorter latent period (mean age = 16.4+/-0.5 wk) than their spontaneous tumors (61%, at age 23.3+/-1.4 wk). In p53(+/-) mice treated with MNU, malignant lymphomas developed at a higher frequency (eight of 27, 30%) than did spontaneous lymphomas (5%). Development of sarcomas in p53(-/-) and p53(+/-) mice was also significantly enhanced by treatment with MNU. All eight thymic lymphomas and three sarcomas in the p53(+/-) mice showed a loss of the remaining wild-type p53 allele. These results indicate that intragastric MNU treatment significantly enhanced spontaneous development of malignant lymphomas and sarcomas in both p53(-/-) and p53(+/-) mice. In the stomachs of 12 p53(+/-) mice, that were killed at the end of the experiment, two adenomas, one carcinoma in situ, and four adenocarcinomas were observed. In the stomachs of 31 p53(+/+) mice, eight adenomas and one carcinoma in situ were detected. The overall incidence of tumorous changes in the stomachs of p53(+/-) (seven of 12, 58%) and p53(+/+) (nine of 31, 29%) mice were not significantly different (P = 0.090). However, adenocarcinomas invading the submucosa were observed in p53(+/-) mice (four of 12, 33%) but not in p53(+/+) mice (zero of 31; P = 0. 004), suggesting a slightly higher susceptibility to gastric carcinogenesis induced by MNU in p53(+/-) mice. Mol. Carcinog. 28:97-101, 2000.


Subject(s)
Carcinogens/toxicity , Genes, p53 , Methylnitrosourea/toxicity , Stomach Neoplasms/chemically induced , Adenocarcinoma/chemically induced , Alleles , Animals , Carcinogens/administration & dosage , Female , Lymphoma/chemically induced , Male , Methylnitrosourea/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Sarcoma, Experimental/chemically induced , Stomach
13.
Genes Dev ; 14(12): 1448-59, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10859164

ABSTRACT

Chk1, an evolutionarily conserved protein kinase, has been implicated in cell cycle checkpoint control in lower eukaryotes. By gene disruption, we show that CHK1 deficiency results in a severe proliferation defect and death in embryonic stem (ES) cells, and peri-implantation embryonic lethality in mice. Through analysis of a conditional CHK1-deficient cell line, we demonstrate that ES cells lacking Chk1 have a defective G(2)/M DNA damage checkpoint in response to gamma-irradiation (IR). CHK1 heterozygosity modestly enhances the tumorigenesis phenotype of WNT-1 transgenic mice. We show that in human cells, Chk1 is phosphorylated on serine 345 (S345) in response to UV, IR, and hydroxyurea (HU). Overexpression of wild-type Atr enhances, whereas overexpression of the kinase-defective mutant Atr inhibits S345 phosphorylation of Chk1 induced by UV treatment. Taken together, these data indicate that Chk1 plays an essential role in the mammalian DNA damage checkpoint, embryonic development, and tumor suppression, and that Atr regulates Chk1.


Subject(s)
Cell Cycle Proteins , G2 Phase , Mitosis , Protein Kinases/metabolism , Protein Kinases/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Alleles , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Ataxia Telangiectasia Mutated Proteins , Cells, Cultured , Checkpoint Kinase 1 , DNA Damage , DNA Repair , Female , Flow Cytometry , Gamma Rays , Heterozygote , Humans , Hydroxyurea/pharmacology , In Situ Nick-End Labeling , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mitosis/drug effects , Mitosis/radiation effects , Models, Genetic , Mutagenesis , Neoplasm Transplantation , Nocodazole/pharmacology , Phosphorylation , Protein Kinases/genetics , Stem Cells , Time Factors , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
14.
Cancer Res ; 60(11): 2831-5, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10850423

ABSTRACT

The purpose of this study was to examine the role of chromosomal recombination in mediating p53 loss in benzene-induced thymic lymphomas in C57BL/6-Trp53 haploinsufficient (N5) mice (p53+/- mice). We characterized loss of heterozygosity (LOH) on chromosome 11 using seven microsatellite markers in 27 benzene-induced and 6 spontaneous thymic lymphomas. Eleven patterns of LOH were found between the induced and spontaneous tumors, with only one pattern being in common between the tumor groups. Nearly 90% (24 of 27) of benzene-induced tumors exhibited loss of the functional p53 allele locus, and 83% (20 of 24) of these tumors retained two copies of the disrupted p53 allele. The results indicate that benzene induces a high frequency of LOH on chromosome 11 in p53+/- mice, likely mediated by aberrant chromosomal recombination.


Subject(s)
Chromosomes , Gene Deletion , Genes, p53/genetics , Lymphoma/chemically induced , Lymphoma/genetics , Recombination, Genetic , Thymus Neoplasms/chemically induced , Thymus Neoplasms/genetics , Alleles , Animals , Benzene , Blotting, Southern , Gene Dosage , Genotype , Loss of Heterozygosity , Mice , Mice, Transgenic , Microsatellite Repeats , Models, Genetic , Polymerase Chain Reaction
15.
Genomics ; 64(3): 298-306, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10756097

ABSTRACT

The human wildtype p53-induced phosphatase 1 (Wip1; GenBank symbol Ppm1d) gene encodes a type 2C protein phosphatase (PP2C) that is induced by ionizing radiation in a p53-dependent manner. We have cloned and sequenced the mouse Wip1 gene and its encoded mRNA. The mouse Wip1 gene is composed of six exons and spans over 36 kb of DNA. The mouse cDNA sequence predicts a 598-amino-acid protein with a molecular mass of roughly 66 kDa. Comparison of human and mouse Wip1 sequences revealed 83% overall identity at the amino acid level. The 5'-flanking region of exon 1 had promoter elements characteristic of a housekeeping gene. The Wip1 coding sequences share conserved functional regions with other PP2Cs from a diverse array of species. Expression of Wip1 mRNA was detected ubiquitously in adult and embryonic tissues, though expression in the testis was much higher than in other tissues. Wip1 has been mapped near the p53 gene on mouse chromosome 11.


Subject(s)
Neoplasm Proteins , Phosphoprotein Phosphatases/genetics , Saccharomyces cerevisiae Proteins , Tumor Suppressor Protein p53/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Fetus/metabolism , Humans , Mice , Molecular Sequence Data , Organ Specificity , Phosphoprotein Phosphatases/metabolism , Promoter Regions, Genetic , Protein Phosphatase 1 , Protein Phosphatase 2 , Protein Phosphatase 2C , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment
16.
Oncogene ; 19(13): 1635-46, 2000 Mar 23.
Article in English | MEDLINE | ID: mdl-10763820

ABSTRACT

Centrosome hyperamplification and the consequential mitotic defects contribute to chromosome instability in cancers. Loss or mutational inactivation of p53 has been shown to induce chromosome instability through centrosome hyperamplification. It has recently been found that Cdk2-cyclin E is involved in the initiation of centrosome duplication, and that constitutive activation of Cdk2-cyclin E results in the uncoupling of the centrosome duplication cycle and the DNA replication cycle. Cyclin E overexpression and p53 mutations occur frequently in tumors. Here, we show that cyclin E overexpression and loss of p53 synergistically increase the frequency of centrosome hyperamplification in cultured cells as well as in tumors developed in p53-null, heterozygous, and wildtype mice. Through examination of cells derived from Waf1-null mice, we further found that Waf1, a potent inhibitor of Cdk2-cyclin E and a major target of p53's transactivation function, is involved in coordinating the initiation of centrosome duplication and DNA replication, suggesting that Waf1 may act as a molecular link between p53 and Cdk2-cyclin E in the control of the centrosome duplication cycle.


Subject(s)
CDC2-CDC28 Kinases , Centrosome/physiology , Cyclin E/physiology , Mitosis , Nuclear Proteins , Tumor Suppressor Protein p53/physiology , Animals , Cells, Cultured , Cyclin E/biosynthesis , Cyclin E/genetics , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/physiology , Cyclins/deficiency , Cyclins/genetics , Cyclins/physiology , DNA Replication/physiology , Enzyme Activation , Fibroblasts , Genes, p53 , Genotype , Loss of Heterozygosity , Mice , Mice, Knockout , Neoplasm Proteins/analysis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , Tubulin/analysis , Tumor Suppressor Protein p53/deficiency
17.
Proc Natl Acad Sci U S A ; 97(7): 3461-6, 2000 Mar 28.
Article in English | MEDLINE | ID: mdl-10716720

ABSTRACT

The interaction between mutations in the tumor-suppressor genes Apc and p53 was studied in congenic mouse strains to minimize the influence of polymorphic modifiers. The multiplicity and invasiveness of intestinal adenomas of Apc(Min/+) (Min) mice was enhanced by deficiency for p53. In addition, the occurrence of desmoid fibromas was strongly enhanced by p53 deficiency. The genetic modifier Mom1 and the pharmacological agents piroxicam and difluoromethylornithine each reduced intestinal adenoma multiplicity in the absence of p53 function. Mom1 showed no influence on the development of desmoid fibromas, whereas the combination of piroxicam and difluoromethylornithine exerted a moderate effect. The ensemble of tumor suppressors and modifiers of a neoplastic process can be usefully analyzed in respect to tissue specificity and synergy.


Subject(s)
Adenoma/genetics , Intestinal Neoplasms/genetics , Neoplasms, Multiple Primary/genetics , Adenoma/pathology , Animals , Eflornithine/pharmacology , Female , Fibromatosis, Aggressive/genetics , Genes, APC , Genes, p53 , Intestinal Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Neoplasms, Multiple Primary/pathology , Piroxicam/pharmacology
18.
Oncogene ; 19(52): 5988-96, 2000 Dec 07.
Article in English | MEDLINE | ID: mdl-11146550

ABSTRACT

The tumor suppressor p53 transcriptionally regulates a large number of target genes that may affect cell growth and cell death pathways. To better understand the role of p53 loss in tumorigenesis, we have developed a mouse mammary cancer model, the Wnt-1 TG/p53 model. Wnt-1 transgenic females that are p53-/- develop mammary adenocarcinomas that arise sooner, grow faster, appear more anaplastic, and have higher levels of chromosomal instability than their Wnt-1 transgenic p53+/+ counterparts. In this study, we used several assays to determine whether the presence or absence of p53 affects gene expression patterns in the mammary adenocarcinomas. Most of the differentially expressed genes are increased in p53+/+ tumors and many of these represent known target genes of p53 (p21WAF/C1P1, cyclin G1, alpha smooth muscle actin, and cytokeratin 19). Some of these genes (cytokeratin 19, alpha smooth muscle actin, and kappa casein) represent mammary gland differentiation markers which may contribute to the inhibited tumor progression and are consistent with the more differentiated histopathology observed in the p53+/+ tumors. Several differentially expressed genes are growth regulatory in function (p21, c-kit, and cyclin B1) and their altered expression levels correlate well with the differing growth properties of the p53+/+ and p53-/- tumors. Thus, while tumors can arise and progress in the presence of functioning wild type p53, p53 may directly or indirectly regulate expression of an array of genes that facilitate differentiation and inhibit proliferation, contributing to a more differentiated, slow growing, and genomically stable phenotype.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Tumor Suppressor Protein p53/metabolism , Zebrafish Proteins , Actins/genetics , Adenocarcinoma/pathology , Animals , Blotting, Western , Caseins/genetics , Cell Division , Cyclin B/genetics , Cyclin B1 , Cyclin G , Cyclin G1 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Female , Gene Deletion , Immunohistochemistry , Incidence , Keratins/genetics , Mammary Neoplasms, Animal/pathology , Mice , Neoplasm Proteins/genetics , Nuclease Protection Assays , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , RNA, Neoplasm/analysis , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Tumor Suppressor Protein p53/genetics , Wnt Proteins , Wnt1 Protein
19.
Cell Growth Differ ; 10(4): 213-22, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10319991

ABSTRACT

The p21(WAF1/cIP1) cyclin-dependent kinase (cdk) inhibitor is a regulator of the G(1)-S cell cycle checkpoint. Despite the importance of p21 in cell cycle inhibition, its role as a tumor suppressor is uncertain. p21 mutations are infrequent in human tumors, and p21 null mice exhibit no increased tumor incidence. To ascertain whether p21 could influence tumor formation or progression in the context of other oncogenic stimuli, we crossed p21-deficient mice with mammary tumor susceptible Wnt-1 transgenic mice. The p21+/+, p21+/-, and p21-/- Wnt-1 transgenic female offspring were monitored for mammary tumor incidence and growth rates. p21 status had no effect on the age at which mammary tumors formed. However, p21+/- mammary tumors grew significantly faster than p21+/+ and p21-/- mammary tumors. The increased growth rates were confirmed by mitotic index counts and by BrdUrd labelling assays, indicating that a significantly higher percentage of p21+/- tumor cells were in S phase and mitosis than their p21+/+ and p21-/- counterparts. Moreover, cyclin D1-associated phosphorylation of retinoblastoma protein was significantly increased in p21+/- tumor lysates compared with p21+/+ and p21-/- lysates. These results are consistent with data indicating that reduced levels of p21 can facilitate cyclin/cdk complex formation while enhancing cdk activity. Thus, a reduction of p21 dosage may promote tumor progression in the presence of other oncogenic initiators. The dependence of p21 on prior oncogenic stimuli for its tumor-promoting activities suggests that it may behave as a tumor modifier gene rather than as a tumor suppressor gene.


Subject(s)
Cyclin D1/metabolism , Cyclins/genetics , Cyclins/metabolism , Heterozygote , Mammary Neoplasms, Experimental/genetics , Age Factors , Alleles , Animals , Apoptosis , Bromodeoxyuridine/metabolism , Cell Division , Crosses, Genetic , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/pharmacology , Dose-Response Relationship, Drug , Female , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphotransferases/metabolism , Precipitin Tests , Time Factors , Tumor Cells, Cultured
20.
Mol Carcinog ; 24(3): 197-208, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10204804

ABSTRACT

A number of transgenic animal model systems have addressed the mechanistic role of p53 loss in tumor progression. However, many of these tumor models have analyzed p53 function in the context of other transgenes expressing activated oncogenes or defective tumor suppressor genes generated by gene targeting. To examine the role of p53 loss independent of other exogenous oncogenic influences, we analyzed some of the biological aspects of tumor formation and progression in p53-knockout mice containing a null germline p53 allele. We analyzed tumors from p53-/-, p53+/-, and p53+/+ littermates. Some of the p53+/- tumors had lost the remaining p53 allele (p53+/- loss of heterozygosity), whereas others retained the allele (p53+/-). In this report, we show that loss or absence of p53 conferred a tumor growth advantage by increasing the rate of cellular proliferation in a p53 dosage-dependent manner. The apoptotic levels in tumor tissue were found to be modest and not significantly dependent on p53 status. These results contrast with those from some other p53-deficient tumor models, in which p53 loss was associated with more rapid tumor progression through abrogated apoptosis. Finally, as p53 has been shown to regulate certain angiogenic factors, we examined the levels of angiogenesis in p53-containing and p53-deficient tumors. We found no p53-dependent differences in the levels of tumor angiogenesis measured by intratumoral microvessel density.


Subject(s)
Genes, p53 , Neoplasms, Experimental/genetics , Alleles , Animals , Apoptosis/genetics , Cell Division , DNA, Neoplasm/analysis , Genetic Predisposition to Disease , Genotype , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitotic Index , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/genetics , Sequence Deletion , von Willebrand Factor/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...