Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
1.
Vet Pathol ; 53(2): 468-76, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26604215

ABSTRACT

Genetic and environmental factors both play a role in the occurrence of age-related disease. To examine the genetic contribution to the development of spontaneous lesions in aging animals, a complete range of tissues was comprehensively analyzed by histopathology from 180 individually housed ad libitum-fed or 40% calorically restricted 24-month-old male and female mice of 2 parental strains-DBA/2NNia (D2) and C57BL/6NNia (B6)-and the F1 cross B6D2F1/NNia. Several strain- and diet-dependent patterns of lesions were identified. Many lesions were genotype dependent and exhibited recessive phenotypic expression, defined as being common in 1 parental strain but infrequently observed in the F1 cross (eg, glomerulonephritis in B6 mice), while others were maintained from 1 parental strain to the F1 with similar frequencies (eg, reproductive tract leiomyoma in D2 mice). Other lesions were common regardless of genotype (osteoarthritis, periodontitis). Only rare lesions were more common in the F1 but underrepresented in the 2 parental strains. Furthermore, F1 mice had a lower number of overall total lesions and a lower number of tumors than either parental strain. Caloric restriction reduced the total number of lesions and neoplasms regardless of genotype but differentially affected genotype-dependent lesions in B6 and D2 mice, with B6 mice more sensitive to the effects of caloric restriction than D2 mice. In summary, genetics and environmental factors (eg, dietary restriction) both substantially contribute to the pattern of lesions that develop as animals age.


Subject(s)
Aging/pathology , Caloric Restriction , Gene-Environment Interaction , Neoplasms/genetics , Animals , Diet , Disease Models, Animal , Female , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasms/pathology , Phenotype , Species Specificity
2.
Curr Mol Med ; 15(7): 634-41, 2015.
Article in English | MEDLINE | ID: mdl-26299770

ABSTRACT

BACKGROUND: Seven isoforms of histone deacetylase Class III have been reported - Sirtuin (SIRT) 1-7. We recently demonstrated that EX-527, an inhibitor of SIRT1, reduces mortality in a mouse model of lethal-cecal-ligationand- puncture (CLP)-induced septic shock. Our present study was aimed at determining whether selective inhibition of SIRT2, with AGK2, would decrease animal death and attenuate the inflammatory response in a septic model. METHODS: Experiment I: C57BL/6J mice were intraperitoneally given either AGK2 (82 mg/kg) in dimethyl sulfoxide (DMSO) or DMSO alone, and 2 h later subjected to CLP. Survival was monitored for 240 hours. Experiment II: mice treated the same way as Experiment I, were grouped into (i) DMSO vehicle, and (ii) AGK2, with sham mice (operating but without any treatment) serving as controls. Peritoneal fluid and peripheral blood were examined at 24 and 48 hours for cytokine production. Samples of blood at 48 h were also allocated to assess coagulability using Thrombelastography (TEG). Morphological changes of bone marrow were evaluated from long bones (femurs and tibias) with hematoxylin and eosin (H&E) staining. Bone marrow atrophy was quantified by a blinded pathologist. Experiment III: cytokines in supernatant of the cultured normal primary splenocytes were measured after the cells were stimulated by lipopolysaccharide and treated with or without AGK2 (10 µM) for 6 hours. RESULTS: AGK2 significantly reduced mortality and decreased levels of cytokines in blood (TNF-α: 298.3±24.6 vs 26.8±2.8 pg/ml, p=0.0034; IL-6: 633.4±82.8 vs 232.6±133.0 pg/ml, p=0.0344) and peritoneal fluid (IL-6: 704.8±67.7 vs 391.4±98.5 pg/ml, p=0.033) compared to vehicle control. Also, AGK2 suppressed the TNF-α and IL-6 production in the cultured splenocytes (TNF-α: 68.1±6.4 vs 23.9±2.8 pg/ml, p=0.0009; IL-6: 73.1±4.2 vs 49.6±3.0 pg/ml; p=0.0051). The TEG data showed that the mice subjected to CLP displayed prolonged fibrin formation and fibrin cross-linkage time, slower clot formation, decreased platelet function, and clot rigidity. AGK2 treatment was associated with dramatic improvements in fibrin cross-linkage and clot formation times, without a significant impact on the clot initiation parameters or platelet function. Additionally, AGK2 significantly attenuated the bone marrow atrophy (58.3±6.5 vs 30.0±8.2%, p=0.0262). CONCLUSION: Selective inhibition of SIRT2 significantly improves survival, and attenuates sepsis-associated "cytokine storm", coagulopathy, and bone marrow atrophy in a mouse model of lethal septic shock.


Subject(s)
Furans/administration & dosage , Histone Deacetylase Inhibitors/administration & dosage , Quinolines/administration & dosage , Shock, Septic/drug therapy , Sirtuin 2/antagonists & inhibitors , Animals , Atrophy/prevention & control , Bone Marrow/drug effects , Bone Marrow/pathology , Cells, Cultured , Cytokines/blood , Drug Evaluation, Preclinical , Injections, Intraperitoneal , Lipopolysaccharides/pharmacology , Male , Mice, Inbred C57BL , Shock, Septic/blood , Shock, Septic/enzymology , Shock, Septic/immunology , Sirtuin 2/metabolism
3.
Oncogene ; 34(48): 5890-9, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-25745996

ABSTRACT

The retinoblastoma gene (Rb) is mutated at significant frequency in various human epithelial tumors, including colorectal cancer, and is strongly associated with metastatic disease. However, sole inactivation of Rb in the mouse has so far failed to yield epithelial cancers. Here, we specifically inactivate Rb and/or p53 in the urogenital epithelium and the intestine. We find that the loss of both tumor suppressors is unable to yield tumors in the transitional epithelium lining the bladder, kidneys and ureters. Instead, these mice develop highly metastatic tumors of neuroendocrine, not epithelial, origin within the urogenital tract to give prostate cancer in the males and vaginal tumors in the females. Additionally, we discovered that the sole inactivation of Rb in the intestine was sufficient to induce formation of metastatic colorectal adenocarcinomas. These tumors closely mirror the human disease in regard to the age of onset, histological appearance, invasiveness and metastatic potential. Like most human colorectal carcinomas, our murine Rb-deficient tumors demonstrate genomic instability and they show activation of ß-catenin. Deregulation of the Wnt/ß-catenin pathway is specific to the intestinal tumors, as genomic instability but not activation of ß-catenin was observed in the neuroendocrine tumors. To date, attempts to generate genetically engineered mouse models of colorectal cancer tumors have yielded mostly cancer of the small intestine, which rarely occurs in humans. Our system provides the opportunity to accurately model and study colorectal cancer in the mouse via a single gene mutation.


Subject(s)
Adenocarcinoma/etiology , Colorectal Neoplasms/etiology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Retinoblastoma Protein/physiology , Tumor Suppressor Protein p53/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Intestinal Neoplasms/etiology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/secondary , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Lung Neoplasms/etiology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Mice , Mice, Knockout , Mutation/genetics , Neuroendocrine Tumors/etiology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/secondary , Prostatic Neoplasms/etiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/secondary , Tumor Cells, Cultured , Wnt Signaling Pathway
4.
Oncogene ; 34(40): 5187-97, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25659581

ABSTRACT

The mucin 1 (MUC1) oncoprotein has been linked to the inflammatory response by promoting cytokine-mediated activation of the NF-κB pathway. The TGF-ß-activated kinase 1 (TAK1) is an essential effector of proinflammatory NF-κB signaling that also regulates cancer cell survival. The present studies demonstrate that the MUC1-C transmembrane subunit induces TAK1 expression in colon cancer cells. MUC1 also induces TAK1 in a MUC1(+/-)/IL-10(-/-) mouse model of colitis and colon tumorigenesis. We show that MUC1-C promotes NF-κB-mediated activation of TAK1 transcription and, in a positive regulatory loop, MUC1-C contributes to TAK1-induced NF-κB signaling. In this way, MUC1-C binds directly to TAK1 and confers the association of TAK1 with TRAF6, which is necessary for TAK1-mediated activation of NF-κB. Targeting MUC1-C thus suppresses the TAK1NF-κB pathway, downregulates BCL-XL and in turn sensitizes colon cancer cells to MEK inhibition. Analysis of colon cancer databases further indicates that MUC1, TAK1 and TRAF6 are upregulated in tumors associated with decreased survival and that MUC1-C-induced gene expression patterns predict poor outcomes in patients. These results support a model in which MUC1-C-induced TAK1NF-κB signaling contributes to intestinal inflammation and colon cancer progression.


Subject(s)
Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/physiology , MAP Kinase Kinase Kinases/metabolism , Mucin-1/metabolism , Signal Transduction/physiology , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Disease Progression , Humans , Immunoblotting , Immunoprecipitation , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Polymerase Chain Reaction , Proportional Hazards Models
5.
Vet Pathol ; 51(4): 846-57, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24009271

ABSTRACT

Detailed histopathological diagnoses of inbred mouse strains are important for interpreting research results and defining novel models of human diseases. The aim of this study was to histologically detect lesions affecting the KK/HlJ inbred strain. Mice were examined at 6, 12, and 20 months of age and near natural death (ie, moribund mice). Histopathological lesions were quantified by percentage of affected mice per age group and sex. Predominant lesions were mineralization, hyperplasia, and fibro-osseous lesions. Mineralization was most frequently found in the connective tissue dermal sheath of vibrissae, the heart, and the lung. Mineralization was also found in many other organs but to a lesser degree. Hyperplasia was found most commonly in the pancreatic islets, and fibro-osseous lesions were observed in several bones. The percentage of lesions increased with age until 20 months. This study shows that KK/HlJ mice demonstrate systemic aberrant mineralization, with greatest frequency in aged mice. The detailed information about histopathological lesions in the inbred strain KK/HlJ can help investigators to choose the right model and correctly interpret the experimental results.


Subject(s)
Calcinosis/pathology , Mice, Inbred Strains/abnormalities , Models, Animal , Phenotype , Vibrissae/pathology , Age Factors , Animals , Mice , Sex Factors
6.
Oncogene ; 32(35): 4078-85, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-22986529

ABSTRACT

The proapoptotic BCL-2 family proteins BAX and BAK serve as essential gatekeepers of the intrinsic apoptotic pathway and, when activated, transform into pore-forming homo-oligomers that permeabilize the mitochondrial outer membrane. Deletion of Bax and Bak causes marked resistance to death stimuli in a variety of cell types. Bax(-/-)Bak(-/-) mice are predominantly non-viable and survivors exhibit multiple developmental abnormalities characterized by cellular excess, including accumulation of neural progenitor cells in the periventricular, hippocampal, cerebellar and olfactory bulb regions of the brain. To explore the long-term pathophysiological consequences of BAX/BAK deficiency in a stem cell niche, we generated Bak(-/-) mice with conditional deletion of Bax in Nestin-positive cells. Aged Nestin(Cre)Bax(fl/fl)Bak(-/-) mice manifest progressive brain enlargement with a profound accumulation of NeuN- and Sox2-positive neural progenitor cells within the subventricular zone (SVZ). One-third of the mice develop frank masses comprised of neural progenitors, and in 20% of these cases, more aggressive, hypercellular tumors emerged. Unexpectedly, 60% of Nestin(Cre)Bax(fl/fl)Bak(-/-) mice harbored high-grade tumors within the testis, a peripheral site of Nestin expression. This in vivo model of severe apoptotic blockade highlights the constitutive role of BAX/BAK in long-term regulation of Nestin-positive progenitor cell pools, with loss of function predisposing to adult-onset tumorigenesis.


Subject(s)
Brain Neoplasms/etiology , Neural Stem Cells/physiology , Testicular Neoplasms/etiology , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology , Animals , Hyperplasia , Intermediate Filament Proteins/analysis , Male , Megalencephaly/etiology , Mice , Nerve Tissue Proteins/analysis , Nestin , Neural Stem Cells/chemistry , Neurons/pathology , Transcriptome , Tumor Suppressor Protein p53/physiology , bcl-2 Homologous Antagonist-Killer Protein/analysis , bcl-2-Associated X Protein/analysis
7.
Oncogene ; 31(25): 3039-50, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22020333

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive brain tumor for which there is no cure. Overexpression of wild-type epidermal growth factor receptor (EGFR) and loss of the tumor suppressor genes Ink4a/Arf and PTEN are salient features of this deadly cancer. Surprisingly, targeted inhibition of EGFR has been clinically disappointing, demonstrating an innate ability for GBM to develop resistance. Efforts at modeling GBM in mice using wild-type EGFR have proven unsuccessful to date, hampering endeavors at understanding molecular mechanisms of therapeutic resistance. Here, we describe a unique genetically engineered mouse model of EGFR-driven gliomagenesis that uses a somatic conditional overexpression and chronic activation of wild-type EGFR in cooperation with deletions in the Ink4a/Arf and PTEN genes in adult brains. Using this model, we establish that chronic activation of wild-type EGFR with a ligand is necessary for generating tumors with histopathological and molecular characteristics of GBMs. We show that these GBMs are resistant to EGFR kinase inhibition and we define this resistance molecularly. Inhibition of EGFR kinase activity using tyrosine kinase inhibitors in GBM tumor cells generates a cytostatic response characterized by a cell cycle arrest, which is accompanied by a substantial change in global gene expression levels. We demonstrate that an important component of this pattern is the transcriptional activation of the MET receptor tyrosine kinase and that pharmacological inhibition of MET overcomes the resistance to EGFR inhibition in these cells. These findings provide important new insights into mechanisms of resistance to EGFR inhibition and suggest that inhibition of multiple targets will be necessary to provide therapeutic benefit for GBM patients.


Subject(s)
Disease Models, Animal , ErbB Receptors/genetics , Glioblastoma/genetics , Mice , Proto-Oncogene Proteins c-met/genetics , Animals , ErbB Receptors/antagonists & inhibitors , Genes, Tumor Suppressor , Glioblastoma/physiopathology , Humans , Mice, Transgenic
8.
Oncogene ; 29(12): 1857-64, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-20010873

ABSTRACT

The adenomatous polyposis coli (APC) gene product is mutated in the vast majority of human colorectal cancers. APC negatively regulates the WNT pathway by aiding in the degradation of beta-catenin, which is the transcription factor activated downstream of WNT signaling. APC mutations result in beta-catenin stabilization and constitutive WNT pathway activation, leading to aberrant cellular proliferation. APC mutations associated with colorectal cancer commonly fall in a region of the gene termed the mutation cluster region and result in expression of an N-terminal fragment of the APC protein. Biochemical and molecular studies have revealed localization of APC/Apc to different sub-cellular compartments and various proteins outside of the WNT pathway that associate with truncated APC/Apc. These observations and genotype-phenotype correlations have led to the suggestion that truncated APC bears neomorphic and/or dominant-negative function that support tumor development. To analyze this possibility, we have generated a novel allele of Apc in the mouse that yields complete loss of Apc protein. Our studies reveal that whole-gene deletion of Apc results in more rapid tumor development than the APC multiple intestinal neoplasia (Apc(Min)) truncation. Furthermore, we found that adenomas bearing truncated Apc had increased beta-catenin activity when compared with tumors lacking Apc protein, which could lead to context-dependent inhibition of tumorigenesis.


Subject(s)
Adenomatous Polyposis Coli/genetics , Gene Deletion , Genes, APC , Adenomatous Polyposis Coli/prevention & control , Animals , Codon/genetics , Codon, Nonsense , Disease Models, Animal , Genetic Carrier Screening , Genotype , Humans , Intestinal Neoplasms/genetics , Mice , Mice, Inbred C57BL/genetics , Multigene Family/genetics , Mutation , Phenotype , beta Catenin/metabolism
9.
Oncogene ; 28(17): 1928-38, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19330024

ABSTRACT

Coexistence of pulmonary tuberculosis (TB) and lung cancer in clinic poses significant challenges for the diagnostic and treatment of both diseases. Although association of chronic inflammation and cancer is well-documented, causal relationship between TB infection and lung cancer are not understood. We present experimental evidence that chronic TB infection induces cell dysplasia and squamous cell carcinoma (SCC) in a lung-specific manner. First, squamous cell aggregates consistently appeared within the lung tissue associated with chronic TB lesions, and in some cases resembled SCCs. A transplantable tumor was established after the transfer of cells isolated from TB lung lesions into syngeneic recipients. Second, the (Mycobacterium tuberculosis) MTB-infected macrophages play a pivotal role in TB-induced carcinogenesis by inducing DNA damage in their vicinity and by the production of a potent epidermal growth factor epiregulin, which may serve as a paracrine survival and growth factor responsible for squamous metaplasia and tumorigenesis. Third, lung carcinogenesis during the course of chronic TB infection was more pronounced in animals with severe lung tissue damage mediated by TB-susceptibility locus sst1. Together, our experimental findings showed a causal link between pulmonary TB and lung tumorigenesis and established a genetic model for further analysis of carcinogenic mechanisms activated by TB infection.


Subject(s)
Carcinoma, Squamous Cell/genetics , Lung Neoplasms/genetics , Tuberculosis, Pulmonary/genetics , Animals , Antitubercular Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/etiology , Cell Transformation, Neoplastic/genetics , Chronic Disease , Disease Models, Animal , Epidermal Growth Factor/genetics , Epiregulin , Female , Gene Expression , Genetic Predisposition to Disease/genetics , Host-Pathogen Interactions , Isoniazid/therapeutic use , Lung/metabolism , Lung/microbiology , Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/etiology , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Congenic , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/physiology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tuberculosis, Pulmonary/complications , Tuberculosis, Pulmonary/drug therapy
10.
Oncogene ; 28(4): 500-8, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-18997819

ABSTRACT

The retinoblastoma protein pRB suppresses tumorigenesis largely through regulation of the E2F transcription factors. E2F4, the most abundant E2F protein, is thought to act in cooperation with pRB to restrain cell proliferation. In this study, we analyse how loss of E2f4 affects the tumorigenicity of pRB-deficient tissues. As Rb(-/-);E2f4(-/-) germline mice die in utero, we generated Rb(-/-);E2f4(-/-) chimeric animals to allow examination of adult tumor phenotypes. We found that loss of E2f4 had a differential effect on known Rb-associated neuroendocrine tumors. It did not affect thyroid and adrenal glands tumors but partially suppressed lung neuroendocrine hyperplasia. The most striking effect was in the pituitary where E2F4 loss delayed the development, and reduced the incidence, of Rb mutant tumors. This tumor suppression increased the longevity of the Rb(-/-);E2f4(-/-) chimeric animals allowing us to identify novel tumor types. We observed ganglionic neuroendocrine neoplasms, lesions not associated earlier with mutation of either Rb or E2f4. Moreover, a subset of the Rb(-/-);E2f4(-/-) chimeras developed either low- or high-grade carcinomas in the urothelium transitional epithelium supporting a key role for Rb in bladder cancer.


Subject(s)
E2F4 Transcription Factor/genetics , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Retinoblastoma Protein/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , E2F4 Transcription Factor/metabolism , Longevity/genetics , Mice , Mice, Knockout , Pituitary Neoplasms/pathology , Retinoblastoma Protein/metabolism , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology , Urothelium/metabolism , Urothelium/pathology
11.
Oncogene ; 27(51): 6561-70, 2008 Nov 20.
Article in English | MEDLINE | ID: mdl-18663357

ABSTRACT

The E2f transcription factors are key downstream targets of the retinoblastoma protein tumor suppressor that control cell proliferation. E2F3 has garnered particular attention because it is amplified in various human tumors. E2f3 mutant mice typically die around birth and E2f3-deficient cells have a proliferation defect that correlates with impaired E2f target gene activation and also induction of p19(Arf) and p53. The E2f3 locus encodes two isoforms, E2f3a and E2f3b, which differ in their N-termini. However, it is unclear how E2f3a versus E2f3b contributes to E2f3's requirement in either proliferation or development. To address this, we use E2f3a- and E2f3b-specific knockouts. We show that inactivation of E2f3a results in a low penetrance proliferation defect in vitro whereas loss of E2f3b has no effect. This proliferation defect appears insufficient to disrupt normal development as E2f3a and E2f3b mutant mice are both fully viable and have no detectable defects. However, when combined with E2f1 mutation, inactivation of E2f3a, but not E2f3b, causes significant proliferation defects in vitro, neonatal lethality and also a striking cartilage defect. Thus, we conclude that E2f3a and E2f3b have largely overlapping functions in vivo and that E2f3a can fully substitute for E2f1 and E2f3 in most murine tissues.


Subject(s)
E2F3 Transcription Factor/metabolism , E2F3 Transcription Factor/physiology , Signal Transduction/physiology , ADP-Ribosylation Factors/genetics , Animals , Cell Cycle/genetics , Cell Proliferation , Cell Survival/genetics , Down-Regulation , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/physiology , E2F3 Transcription Factor/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Signal Transduction/genetics
12.
Oncogene ; 26(41): 6010-20, 2007 Sep 06.
Article in English | MEDLINE | ID: mdl-17384673

ABSTRACT

Nonhomologous end joining (NHEJ) is a critical DNA repair pathway, with proposed tumor suppression functions in many tissues. Mutations in the NHEJ factor ARTEMIS cause radiation-sensitive severe combined immunodeficiency in humans and may increase susceptibility to lymphoma in some settings. We now report that deficiency for Artemis (encoded by Dclre1c/Art in mouse) accelerates tumorigenesis in several tissues in a Trp53 heterozygous setting, revealing tumor suppression roles for NHEJ in lymphoid and non-lymphoid cells. We also show that B-lineage lymphomas in these mice undergo loss of Trp53 heterozygosity by allele replacement, but arise by mechanisms distinct from those in Art Trp53 double null mice. These findings demonstrate a general tumor suppression function for NHEJ, and reveal that interplay between NHEJ and Trp53 loss of heterozygosity influences the sequence of multi-hit oncogenesis. We present a model where p53 status at the time of tumor initiation is a key determinant of subsequent oncogenic mechanisms. Because Art deficient mice represent a model for radiation-sensitive severe combined immunodeficiency, our findings suggest that these patients may be at risk for both lymphoid and non-lymphoid cancers.


Subject(s)
DNA Repair , Genes, p53 , Loss of Heterozygosity , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Animals , Endonucleases , Humans , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Sarcoma, Experimental/genetics , Sarcoma, Experimental/pathology , Severe Combined Immunodeficiency/genetics , Tumor Suppressor Protein p53/deficiency
13.
Oncogene ; 26(20): 2815-21, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17072335

ABSTRACT

An adequate and appropriate response to physiological and pathophysiological stresses is critical for long-term homeostasis and viability of the aging organism. Previous work has pointed to the immune system, telomeres and DNA repair pathways as important and distinct determinants of a normal healthy lifespan. In this study, we explored the genetic interactions of telomeres and DNA-PKcs, a protein involved in non-homologous end-joining (NHEJ) and immune responses, in the context of a key aspect of aging and lifespan--the capacity to mount an acute and appropriate immune-mediated stress response. We observed that the combination of DNA-PKcs deficiency and telomere dysfunction resulted in a shortened lifespan that was reduced further following viral infection or experimental activation of the innate immune response. Analysis of the innate immune response in the DNA-PKcs-deficient mice with short dysfunctional telomeres revealed high basal serum levels of tumor necrosis factor alpha (TNFalpha) and hyper-active cytokine responses upon challenge with polyinosinic-polycytidylic acid (poly-IC). We further show that serum cytokine levels become elevated in telomere dysfunctional mice as a function of age. These results raise speculation that these genetic factors may contribute to misdirected immune responses of the aged under conditions of acute and chronic stress.


Subject(s)
DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/genetics , Longevity/genetics , Nuclear Proteins/genetics , Stress, Physiological/genetics , Stress, Physiological/mortality , Telomere/metabolism , Animals , Crosses, Genetic , Hepatitis, Animal/blood , Hepatitis, Animal/genetics , Hepatitis, Animal/immunology , Interleukin-1beta/blood , Interleukin-6/blood , Liver/pathology , Mice , Mice, Transgenic , Murine hepatitis virus/immunology , RNA/genetics , Stress, Physiological/pathology , Telomerase/genetics , Telomere/physiology , Tumor Necrosis Factor-alpha/blood
15.
Oncogene ; 25(14): 2105-12, 2006 Mar 30.
Article in English | MEDLINE | ID: mdl-16288213

ABSTRACT

Activating mutations in K-ras are one of the most common genetic alterations in human lung cancer. To dissect the role of K-ras activation in bronchial epithelial cells during lung tumorigenesis, we created a model of lung adenocarcinoma by generating a conditional mutant mouse with both Clara cell secretory protein (CC10)-Cre recombinase and the Lox-Stop-Lox K-ras(G12D) alleles. The activation of K-ras mutant allele in CC10 positive cells resulted in a progressive phenotype characterized by cellular atypia, adenoma and ultimately adenocarcinoma. Surprisingly, K-ras activation in the bronchiolar epithelium is associated with a robust inflammatory response characterized by an abundant infiltration of alveolar macrophages and neutrophils. These mice displayed early mortality in the setting of this pulmonary inflammatory response with a median survival of 8 weeks. Bronchoalveolar lavage fluid from these mutant mice contained the MIP-2, KC, MCP-1 and LIX chemokines that increased significantly with age. Cell lines derived from these tumors directly produced MIP-2, LIX and KC. This model demonstrates that K-ras activation in the lung induces the elaboration of inflammatory chemokines and provides an excellent means to further study the complex interactions between inflammatory cells, chemokines and tumor progression.


Subject(s)
Genes, ras , Lung Neoplasms/genetics , Pneumonia/genetics , Animals , Base Sequence , Bronchoalveolar Lavage Fluid , Cell Line, Tumor , DNA Primers , Humans , Immunohistochemistry , Lung Neoplasms/complications , Lung Neoplasms/physiopathology , Macrophages, Alveolar/pathology , Mice , Mice, Mutant Strains , Pneumonia/complications , Reverse Transcriptase Polymerase Chain Reaction
16.
Mol Cell Biol ; 23(3): 1044-53, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12529408

ABSTRACT

Targeted disruption of the retinoblastoma gene in mice leads to embryonic lethality in midgestation accompanied by defective erythropoiesis. Rb(-/-) embryos also exhibit inappropriate cell cycle activity and apoptosis in the central nervous system (CNS), peripheral nervous system (PNS), and ocular lens. Loss of p53 can prevent the apoptosis in the CNS and lens; however, the specific signals leading to p53 activation have not been determined. Here we test the hypothesis that hypoxia caused by defective erythropoiesis in Rb-null embryos contributes to p53-dependent apoptosis. We show evidence of hypoxia in CNS tissue from Rb(-/-) embryos. The Cre-loxP system was then used to generate embryos in which Rb was deleted in the CNS, PNS and lens, in the presence of normal erythropoiesis. In contrast to the massive CNS apoptosis in Rb-null embryos at embryonic day 13.5 (E13.5), conditional mutants did not have elevated apoptosis in this tissue. There was still significant apoptosis in the PNS and lens, however. Rb(-/-) cells in the CNS, PNS, and lens underwent inappropriate S-phase entry in the conditional mutants at E13.5. By E18.5, conditional mutants had increased brain size and weight as well as defects in skeletal muscle development. These data support a model in which hypoxia is a necessary cofactor in the death of CNS neurons in the developing Rb mutant embryo.


Subject(s)
Cell Cycle/genetics , Central Nervous System/cytology , Central Nervous System/embryology , Genes, Retinoblastoma , Mutation , Animals , Apoptosis/genetics , Erythropoiesis/genetics , Female , Hypoxia/genetics , Lens, Crystalline/cytology , Lens, Crystalline/embryology , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Models, Biological , Muscle, Skeletal/abnormalities , Muscle, Skeletal/embryology , Peripheral Nerves/cytology , Peripheral Nerves/embryology , Pregnancy , S Phase/genetics
17.
Genes Dev ; 15(24): 3243-8, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751630

ABSTRACT

Adenocarcinoma of the lung is the most common form of lung cancer, but the cell of origin and the stages of progression of this tumor type are not well understood. We have developed a new model of lung adenocarcinoma in mice harboring a conditionally activatable allele of oncogenic K-ras. Here we show that the use of a recombinant adenovirus expressing Cre recombinase (AdenoCre) to induce K-ras G12D expression in the lungs of mice allows control of the timing and multiplicity of tumor initiation. Through the ability to synchronize tumor initiation in these mice, we have been able to characterize the stages of tumor progression. Of particular significance, this system has led to the identification of a new cell type contributing to the development of pulmonary adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Genes, ras/genetics , Lung Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Codon , Disease Progression , Gene Expression , Immunoenzyme Techniques , Integrases/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Knockout , Models, Animal , Mutation , Proto-Oncogene Proteins p21(ras)/metabolism , Time Factors , Viral Proteins/metabolism
18.
Am J Pathol ; 159(5): 1949-56, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11696456

ABSTRACT

In vitro and in vivo data indicate that thrombospondin-1 (TSP1) inhibits tumor progression in several ways including direct effects on cellular growth and apoptosis in the stromal compartment. To evaluate the importance of TSP1 for the progression of naturally arising tumors in vivo, we have crossed TSP1-deficient mice with p53-deficient mice. In p53-null mice, the absence of TSP1 decreases survival from 160 +/- 52 days to 149 +/- 42 days. A log-rank test comparing survival curves for these two populations yields a two-sided P value of 0.0272. For mice that are heterozygous for the p53-null allele, survival is 500 +/- 103 days in the presence of TSP1 expression, and 426 +/- 125 days in its absence (P = 0.0058). Whereas TSP1 expression did not cause a measurable change in the incidence of the majority of tumor types, a statistically significant (P < or = 0.05) decrease in the incidence of osteosarcomas is observed in the absence of TSP1. To determine more directly if host TSP1 inhibits tumor growth, B16F10 melanoma and F9 testicular teratocarcinoma cells have been implanted in C57BL/6J and 129Sv TSP1-null mice, respectively. The B16F10 tumors grow approximately twice as fast in the TSP1-null background and exhibit an increase in vascular density, a decrease in the rate of tumor cell apoptosis, and an increase in the rate of tumor cell proliferation. Increased tumor growth is also observed in the absence of TSP1 on the 129Sv genetic background. These data indicate that endogenous host TSP1 functions as a modifier or landscaper gene to suppress tumor growth.


Subject(s)
Gene Expression/physiology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/mortality , Thrombospondin 1/genetics , Tumor Suppressor Protein p53/deficiency , Animals , Female , Genotype , Loss of Heterozygosity , Male , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Teratocarcinoma/genetics , Teratocarcinoma/pathology , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
19.
J Neurosci ; 21(17): 6758-71, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11517264

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) plays a pivotal role in brain development and neuronal migration. Cdk5 is abundant in postmitotic, terminally differentiated neurons. The ability of Cdk5 to phosphorylate substrates is dependent on activation by its neuronal-specific activators p35 and p39. There exist striking differences in the phenotypic severity of Cdk5-deficient mice and p35-deficient mice. Cdk5-null mutants show a more severe disruption of lamination in the cerebral cortex, hippocampus, and cerebellum. In addition, Cdk5-null mice display perinatal lethality, whereas p35-null mice are viable. These discrepancies have been attributed to the function of other Cdk5 activators, such as p39. To understand the roles of p39 and p35, we created p39-null mice and p35/p39 compound-mutant mice. Interestingly, p39-null mice show no obvious detectable abnormalities, whereas p35(-/-)p39(-/-) double-null mutants are perinatal lethal. We show here that the p35(-/-)p39(-/-) mutants exhibit phenotypes identical to those of the Cdk5-null mutant mice. Other compound-mutant mice with intermediate phenotypes allow us to determine the distinct and redundant functions between p35 and p39. Our data strongly suggest that p35 and p39 are essential for Cdk5 activity during the development of the nervous system. Thus, p35 and p39 are likely to be the principal, if not the only, activators of Cdk5.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Nerve Tissue Proteins/metabolism , Animals , Animals, Newborn , Brain/abnormalities , Brain/enzymology , Brain/pathology , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/genetics , Enzyme Activation/genetics , Fetal Viability/genetics , Gene Dosage , Gene Targeting , Genes, Lethal , Homozygote , Macromolecular Substances , Mice , Mice, Neurologic Mutants , Motor Neurons/pathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nervous System Malformations/enzymology , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Phenotype , Phosphorylation , Transplantation Chimera
20.
Cytogenet Cell Genet ; 93(1-2): 77-82, 2001.
Article in English | MEDLINE | ID: mdl-11474184

ABSTRACT

The paracentric inversion In(3)55Rk on mouse Chromosome 3 (Chr 3) was induced by cesium irradiation. Genetic crosses indicate the proximal breakpoint cosegregates with D3Mit324 and D3Mit92; the distal breakpoint cosegregates with D3Mit127, D3Mit160, and D3Mit200. Giemsa-banded chromosomes show the inversion spans approximately 80% of Chr 3. The proximal breakpoint occurs within band 3A2, not 3B as reported previously; the distal breakpoint occurs within band 3H3. Mice homozygous for the inversion exhibit nephropathy indicative of uricase deficiency. Southern blot analyses of urate oxidase, Uox, show two RFLPs of genomic mutant DNA: an EcoRI site between exons 4-8 and a BamHI site 3' to exon 6. Mutant cDNA fails to amplify downstream of base 844 at the 3' end of exon 7. FISH analysis of chromosomes from inversion heterozygotes, using a cosmid clone containing genomic wild-type DNA for Uox exons 2-4, shows that a 5' segment of the mutated Uox allele on the inverted chromosome has been transposed from the distal breakpoint region to the proximal breakpoint region. Clinical, histopathological, and Northern analyses indicate that our radiation-induced mutation, uox(In), is a putative null.


Subject(s)
Chromosome Inversion , Kidney Diseases/genetics , Mutation/genetics , Urate Oxidase/genetics , Alleles , Animals , Blotting, Southern , Chromosome Banding , Chromosome Mapping , Crosses, Genetic , DNA Mutational Analysis , Exons/genetics , Female , In Situ Hybridization, Fluorescence , Kidney Diseases/enzymology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , Mice, Mutant Strains , Polymorphism, Restriction Fragment Length , RNA, Messenger/genetics , RNA, Messenger/metabolism , Uric Acid/blood , Uric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...