1
|
van Oers JMM, Edwards Y, Chahwan R, Zhang W, Smith C, Pechuan X, Schaetzlein S, Jin B, Wang Y, Bergman A, Scharff MD, Edelmann W. The MutSβ complex is a modulator of p53-driven tumorigenesis through its functions in both DNA double-strand break repair and mismatch repair. Oncogene 2013; 33:3939-46. [PMID: 24013230 DOI: 10.1038/onc.2013.365] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/19/2013] [Accepted: 07/30/2013] [Indexed: 01/10/2023]
Abstract
Loss of the DNA mismatch repair (MMR) protein MSH3 leads to the development of a variety of tumors in mice without significantly affecting survival rates, suggesting a modulating role for the MutSβ (MSH2-MSH3) complex in late-onset tumorigenesis. To better study the role of MSH3 in tumor progression, we crossed Msh3(-/-) mice onto a tumor predisposing p53-deficient background. Survival of Msh3/p53 mice was not reduced compared with p53 single mutant mice; however, the tumor spectrum changed significantly from lymphoma to sarcoma, indicating MSH3 as a potent modulator of p53-driven tumorigenesis. Interestingly, Msh3(-/-) mouse embryonic fibroblasts displayed increased chromatid breaks and persistence of γH2AX foci following ionizing radiation, indicating a defect in DNA double-strand break repair (DSBR). Msh3/p53 tumors showed increased loss of heterozygosity, elevated genome-wide copy-number variation and a moderate microsatellite instability phenotype compared with Msh2/p53 tumors, revealing that MSH2-MSH3 suppresses tumorigenesis by maintaining chromosomal stability. Our results show that the MSH2-MSH3 complex is important for the suppression of late-onset tumors due to its roles in DNA DSBR as well as in DNA MMR. Further, they demonstrate that MSH2-MSH3 suppresses chromosomal instability and modulates the tumor spectrum in p53-deficient tumorigenesis and possibly has a role in other chromosomally unstable tumors as well.
Collapse
Affiliation(s)
- J M M van Oers
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Y Edwards
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - R Chahwan
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - W Zhang
- Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - C Smith
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - X Pechuan
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - S Schaetzlein
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - B Jin
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Y Wang
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - A Bergman
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - M D Scharff
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - W Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
2
|
Bartesaghi S, Salomoni P. Tumor suppressive pathways in the control of neurogenesis. Cell Mol Life Sci 2013; 70:581-97. [PMID: 22802124 PMCID: PMC11113109 DOI: 10.1007/s00018-012-1063-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 06/16/2012] [Accepted: 06/18/2012] [Indexed: 12/16/2022]
Abstract
The generation of specialized neural cells in the developing and postnatal central nervous system is a highly regulated process, whereby neural stem cells divide to generate committed neuronal progenitors, which then withdraw from the cell cycle and start to differentiate. Cell cycle checkpoints play a major role in regulating the balance between neural stem cell expansion and differentiation. Loss of tumor suppressors involved in checkpoint control can lead to dramatic alterations of neurogenesis, thus contributing to neoplastic transformation. Here we summarize and critically discuss the existing literature on the role of tumor suppressive pathways and their regulatory networks in the control of neurogenesis and transformation.
Collapse
Affiliation(s)
- Stefano Bartesaghi
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6DD UK
| | - Paolo Salomoni
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6DD UK
| |
Collapse
|
3
|
Saba I, Kosan C, Vassen L, Klein-Hitpass L, Möröy T. Miz-1 is required to coordinate the expression of TCRbeta and p53 effector genes at the pre-TCR "beta-selection" checkpoint. THE JOURNAL OF IMMUNOLOGY 2011; 187:2982-92. [PMID: 21841135 DOI: 10.4049/jimmunol.1101451] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Miz-1 is a Broad-complex, Tramtrack and Bric-à-brac/pox virus zinc finger domain (BTB/POZ)-containing protein expressed in lymphoid precursors that can activate or repress transcription. We report in this article that mice expressing a nonfunctional Miz-1 protein lacking the BTB/POZ domain (Miz-1(ΔPOZ)) have a severe differentiation block at the pre-T cell "β-selection" checkpoint, evident by a drastic reduction of CD4(-)CD8(-) double-negative-3 (DN3) and DN4 cell numbers. T cell-specific genes including Rag-1, Rag-2, CD3ε, pTα, and TCRβ are expressed in Miz-1-deficient cells and V(D)J recombination is intact, but few DN3/DN4 cells express a surface pre-TCR. Miz-1-deficient DN3 cells are highly apoptotic and do not divide, which is consistent with enhanced expression of p53 target genes such as Cdkn1a, PUMA, and Noxa. However, neither coexpression of the antiapoptotic protein Bcl2 nor the deletion of p21(CIP1) nor the combination of both relieved Miz-1-deficient DN3/DN4 cells from their differentiation block. Only the coexpression of rearranged TCRαβ and Bcl2 fully rescued Miz-1-deficient DN3/DN4 cell numbers and enabled them to differentiate into DN4TCRβ(+) and double-positive cells. We propose that Miz-1 is a critical factor for the β-selection checkpoint and is required for both the regulation of p53 target genes and proper expression of the pre-TCR to support the proliferative burst of DN3 cells during T cell development.
Collapse
Affiliation(s)
- Ingrid Saba
- Institut de Recherches Cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
| | | | | | | | | |
Collapse
|
4
|
Gramling S, Reisman D. Discovery of BRM Targeted Therapies: Novel Reactivation of an Anti-cancer Gene. LETT DRUG DES DISCOV 2011; 8:93-99. [PMID: 23565070 PMCID: PMC3615482 DOI: 10.2174/157018011793663840] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Drug discovery in the field of oncology has been advanced mainly through the targeting of receptor tyrosine kinases. Both antibodies and small molecule inhibitors have been found to have successful applications in blocking the proliferative functions of these cell surface receptors. Based on these early successes, additional kinases within the cytoplasm have been found to promote cancer and, as such, have been recognized as feasible targets for additional modes of therapies. Unlike these oncogene targets, most tumor suppressors are irreversibly altered during cancer progression and therefore are not feasible targets for therapy. However, a subset of these genes is reversibly epigenetically suppressed. One such gene is BRM, and when it is re-expressed in cancer cells, this gene halts their growth. Moreover, as the key catalytic subunit of the SWI/SNF complex, BRM is centrally important to a host of anticancer pathways and cellular mechanisms, and its status may serve as a biomarker. Restoring its expression will both reconnect a number of growth-controlling pathways and affect cellular adhesion, DNA repair, and immune functions. For these reasons, restoring BRM expression is not only feasible, but potentially a potent form of anticancer therapy. To identify BRM-restoring compounds, we developed a cell-based luciferase assay. In this review, we discuss some of the challenges we encountered, issues related to this type of drug discovery, and our future ambitions. We hope this review will provide insight to this type of endeavor and lead to more investigations pursuing this type of drug research.
Collapse
Affiliation(s)
- Sarah Gramling
- Department of Medicine, Division of Hematology/Oncology, University of Florida, Gainesville Florida 32611, USA
| | - David Reisman
- Department of Medicine, Division of Hematology/Oncology, University of Florida, Gainesville Florida 32611, USA
| |
Collapse
|
5
|
Mutant alleles of HD improve the life span of p53(-/-) mice. Mech Ageing Dev 2008; 129:238-41. [PMID: 18242663 DOI: 10.1016/j.mad.2007.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Revised: 12/03/2007] [Accepted: 12/04/2007] [Indexed: 11/20/2022]
Abstract
Loss of function mutations in the p53 tumor suppressor gene predispose mice and humans to cancer, resulting in abbreviated life spans. A dominant mutation in the murine HD gene, similar to mutations that cause Huntington's disease in humans, reverses some of the effects of p53 mutations on longevity. We attribute this to the enhanced apoptotic effect of the expanded polyglutamine region in the HD protein on proliferating cells lacking p53.
Collapse
|
6
|
Amatruda JF, Patton EE. Chapter 1 Genetic Models of Cancer in Zebrafish. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2008; 271:1-34. [DOI: 10.1016/s1937-6448(08)01201-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
7
|
Merk HF, Baron JM, Heise R, Fritsche E, Schroeder P, Abel J, Krutmann J. Concepts in molecular dermatotoxicology. Exp Dermatol 2006; 15:692-704. [PMID: 16881966 DOI: 10.1111/j.1600-0625.2006.00475.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In the recent years, molecular research has successfully elucidated some of the major mechanisms through which environmental noxae damage human skin. From this knowledge, novel concepts for skin protection have been developed. Here, we provide a brief overview of some of the most exciting and intriguing concepts in molecular dermatotoxicology.
Collapse
Affiliation(s)
- Hans F Merk
- Department of Dermatology and Allergology, RWTH Aachen, Aachen, Germany.
| | | | | | | | | | | | | |
Collapse
|
8
|
Maddalena AS, Hainfellner JA, Hegi ME, Glatzel M, Aguzzi A. No complementation between TP53 or RB-1 and v-src in astrocytomas of GFAP-v-src transgenic mice. Brain Pathol 2006; 9:627-37. [PMID: 10517501 PMCID: PMC8098330 DOI: 10.1111/j.1750-3639.1999.tb00544.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human low-grade astrocytomas frequently recur and progress to states of higher malignancy. During tumor progression TP53 alterations are among the first genetic changes, while derangement of the p16/p14ARF/RB-1 system occurs later. To probe the pathogenetic significance of TP53 and RB-1 alterations, we introduced a v-src transgene driven by glial fibrillary acidic protein (GFAP) regulatory elements (which causes preneoplastic astrocytic lesions and stochastically astrocytomas of varying degrees of malignancy) into TP53+/- or RB-1+/- mice. Hemizygosity for TP53 or RB-1 did not increase the incidence or shorten the latency of astrocytic tumors in GFAP-v-src mice over a period of up to 76 weeks. Single strand conformation analysis of exons 5 to 8 of non-ablated TP53 alleles revealed altered migration patterns in only 3/16 tumors analyzed. Wild-type RB-1 alleles were retained in all RB-1+/-GFAP-v-src mice-derived astrocytic tumors analyzed, and pRb immunostaining revealed protein expression in all tumors. Conversely, the GFAP-v-src transgene did not influence the development of extraneural tumors related to TP53 or RB-1 hemizygosity. Therefore, the present study indicates that neither loss of RB-1 nor of TP53 confer a growth advantage in vivo to preneoplastic astrocytes expressing v-src, and suggests that RB-1 and TP53 belong to one single complementation group along with v-src in this transgenic model of astrocytoma development. The stochastic development of astrocytic tumors in GFAP-v-src, TP53+/- GFAP-v-src, and RB-1+/- GFAP-v-src transgenic mice indicates that additional hitherto unknown genetic lesions of astrocytes contribute to tumorigenesis, whose elucidation may prove important for our understanding of astrocytoma initiation and progression.
Collapse
Affiliation(s)
| | - Johannes A. Hainfellner
- Institute of Neuropathology, University Hospital of Zurich, Switzerland
- Institute of Neurology, University of Vienna, Austria
| | - Monika E. Hegi
- Institute of Neuropathology, University Hospital of Zurich, Switzerland
- Laboratory for Tumor Biology and Genetics, Department of Neurosurgery, University Hospital, Lausanne, Switzerland
| | - Markus Glatzel
- Institute of Neuropathology, University Hospital of Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, Switzerland
| |
Collapse
|
9
|
Merk HF, Abel J, Baron JM, Krutmann J. Molecular pathways in dermatotoxicology. Toxicol Appl Pharmacol 2004; 195:267-77. [PMID: 15020189 DOI: 10.1016/j.taap.2003.09.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2003] [Accepted: 09/24/2003] [Indexed: 01/09/2023]
Affiliation(s)
- H F Merk
- Institut für Umweltmedizinische Forschung (IUF) at the Heinrich Heine University Düsseldorf gGmbH, Düsseldorf, Germany
| | | | | | | |
Collapse
|
10
|
Abstract
The INK4a/ARF locus encodes two proteins whose expression limits cellular proliferation. Whilst the biochemical activities of the two proteins appear very different, they both converge on regulating the retinoblastoma and p53 tumour suppressor pathways. Neither protein is required for normal development, but lack of either predisposes to the development of malignancy. Both proteins have also been implicated in the establishment of senescence states in response to a variety of stresses, signalling imbalances and telomere shortening. The INK4a/Arf regulatory circuits appear to be partially redundant and show evidence of rapid evolution. Especially intriguing are the large number of biological differences documented between mice and man. We review here the brief history of INK4a/Arf and explore possible links with organismal aging and the evolution of longevity.
Collapse
Affiliation(s)
- Carol J Collins
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
| | | |
Collapse
|
11
|
Wei W, Jobling WA, Chen W, Hahn WC, Sedivy JM. Abolition of cyclin-dependent kinase inhibitor p16Ink4a and p21Cip1/Waf1 functions permits Ras-induced anchorage-independent growth in telomerase-immortalized human fibroblasts. Mol Cell Biol 2003; 23:2859-70. [PMID: 12665584 PMCID: PMC152557 DOI: 10.1128/mcb.23.8.2859-2870.2003] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2002] [Revised: 10/29/2002] [Accepted: 01/28/2003] [Indexed: 11/20/2022] Open
Abstract
Human cells are more resistant to both immortalization and malignant transformation than rodent cells. Recent studies have established the basic genetic requirements for the transformation of human cells, but much of this work relied on the expression of transforming proteins derived from DNA tumor viruses. We constructed an isogenic panel of human fibroblast cell lines using a combination of gene targeting and ectopic expression of dominantly acting mutants of cellular genes. Abolition of p21(Cip1/Waf1) and p16(Ink4a) functions prevented oncogenically activated Ras from inducing growth arrest and was sufficient for limited anchorage-independent growth but not tumorigenesis. Deletion of the tumor suppressor p53 combined with abolition of p16(Ink4a) function failed to mimic the introduction of simian virus 40 large T antigen, indicating that large T antigen may target additional cellular functions. Ha-Ras and Myc cooperated only to a limited extent, but in the absence of Ras, Myc cooperated strongly with the simian virus 40 small t antigen to elicit aggressive anchorage-independent growth. The experiments reported here further define specific components of human transformation pathways.
Collapse
Affiliation(s)
- Wenyi Wei
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island 02912, USA
| | | | | | | | | |
Collapse
|
12
|
Wei W, Hemmer RM, Sedivy JM. Role of p14(ARF) in replicative and induced senescence of human fibroblasts. Mol Cell Biol 2001; 21:6748-57. [PMID: 11564860 PMCID: PMC99853 DOI: 10.1128/mcb.21.20.6748-6757.2001] [Citation(s) in RCA: 174] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Following a proliferative phase of variable duration, most normal somatic cells enter a growth arrest state known as replicative senescence. In addition to telomere shortening, a variety of environmental insults and signaling imbalances can elicit phenotypes closely resembling senescence. We used p53(-/-) and p21(-/-) human fibroblast cell strains constructed by gene targeting to investigate the involvement of the Arf-Mdm2-p53-p21 pathway in natural as well as premature senescence states. We propose that in cell types that upregulate p21 during replicative exhaustion, such as normal human fibroblasts, p53, p21, and Rb act sequentially and constitute the major pathway for establishing growth arrest and that the telomere-initiated signal enters this pathway at the level of p53. Our results also revealed a number of significant differences between human and rodent fibroblasts in the regulation of senescence pathways.
Collapse
Affiliation(s)
- W Wei
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 02912, USA
| | | | | |
Collapse
|
13
|
Jacobs H. TCR-independent T cell development mediated by gain-of-oncogene function or loss-of-tumor-suppressor gene function. Semin Immunol 2000; 12:487-502. [PMID: 11085181 DOI: 10.1006/smim.2000.0262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The mechanisms that govern differentiation of T cell precursors during intrathymic development bridge an interdisciplinary research field of immunology, oncology and developmental biology. Critical checkpoints controlling early thymic T cell development and homeostasis are set by the proper signaling function of the IL-7 receptor, c-Kit receptor, and the pre-T cell antigen receptor (pre-TCR). Given the intimate link between cell cycle control and differentiation in T cell development, proto-oncogenes and tumor suppressors participate as physiological effectors downstream of these receptors not only to influence the cell cycle but also to determine differentiation and survival. Gain- or loss-of-function mutations of these downstream effectors uncouples partially or completely T cell precursors from these checkpoints, providing a selective advantage and enabling aberrant development. These effectors can be identified by provirus tagging in normal mice and more readily by complementation tagging in mice with a predefined block in T cell differentiation.
Collapse
Affiliation(s)
- H Jacobs
- Basel Institute for Immunology, Switzerland
| |
Collapse
|
14
|
Hollstein M, Hergenhahn M, Yang Q, Bartsch H, Wang ZQ, Hainaut P. New approaches to understanding p53 gene tumor mutation spectra. Mutat Res 1999; 431:199-209. [PMID: 10635987 DOI: 10.1016/s0027-5107(99)00162-1] [Citation(s) in RCA: 159] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The first p53 gene mutation arising in a human tumor was described a decade ago by Baker et al. [S.J. Baker, E.R. Fearon, J.M. Nigro, S.R. Hamilton, A.C. Preisinger, J.M. Jessup, P. van Tuinen, D.H. Ledbetter, D.F. Barker, Y. Nakamura, R. White, B. Vogelstein, Chromosome 17 deletions and p53 gene mutations in colorectal carcinomas, Science 244 (1989) 217-221]. There are now over 10,000 mutations extracted from the published literature in the IARC database of human p53 tumor mutations [P. Hainaut, T. Hernandez, A. Robinson, P. Rodriguez-Tome, T. Flores, M. Hollstein, C.C. Harris, R. Montesano, IARC database of p53 gene mutations in human tumors and cell lines: updated compilation, revised formats and new visualization tools, Nucleic Acids Res. 26 (1998) 205-213; Version R3, January 1999]. A large and diverse collection of tumor mutations in cancer patients provides important information on the nature of environmental factors or biological processes that are important causes of human gene mutation, since xenobiotic mutagens as well as endogenous mechanisms of genetic change produce characteristic types of patterns in target DNA [J.H. Miller, Mutational specificity in bacteria, Annu. Rev. Genet. 17 (1983) 215-238; T. Lindahl, Instability and decay of the primary structure of DNA, Nature 362 (1993) 709-715; S.P. Hussain, C.C. Harris, Molecular epidemiology of human cancer: contribution of mutation spectra studies of tumor suppressor genes, Cancer Res. 58 (1998) 4023-4037; P. Hainaut, M. Hollstein, p53 and human cancer: the first ten thousand mutations, Adv. Cancer Res. 2000]. P53 gene mutations in cancers can be compared to point mutation spectra at the HPRT locus of human lymphocytes from patients or healthy individuals with known exposure histories, and accumulated data indicate that mutation patterns at the two loci share certain general features. Hypotheses regarding specific cancer risk factors can be tested by comparing p53 tumor mutations typical of a defined patient group against mutations generated experimentally in rodents or in prokaryotic and eukaryotic cells in vitro. Refinements of this approach to hypothesis testing are being explored that employ human p53 sequences introduced artificially into experimental organisms used in laboratory mutagenesis assays. P53-specific laboratory models, combined with DNA microchips designed for high through-put mutation screening promise to unmask information currently hidden in the compilation of human tumor p53 mutations.
Collapse
Affiliation(s)
- M Hollstein
- Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
15
|
Aubrecht J, Secretan MB, Bishop AJ, Schiestl RH. Involvement of p53 in X-ray induced intrachromosomal recombination in mice. Carcinogenesis 1999; 20:2229-36. [PMID: 10590213 DOI: 10.1093/carcin/20.12.2229] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The tumor suppressor gene Trp53 (also known as p53) is the most frequently mutated gene in human cancers. p53 is induced in response to DNA damage and effects a G(1) cell cycle arrest. It is believed that p53 plays a key role in maintaining genomic integrity following exposure to DNA-damaging agents. We determined the frequency of spontaneous and DNA damage-induced homologous intrachromosomal recombination in p53-deficient mouse embryos. Homologous intrachromosomal recombination events resulting in deletions at the pink eyed unstable (p(un)) locus result in reversion to the p gene. Reversions occurring in embryonic premelanocytes give rise to black spots on the gray fur of the offspring. Pregnant C57BL/6J p(un)/p(un) p53(+/-) mice were exposed to X-rays (1 Gy) or administered benzo¿apyrene (B¿aP; 30 or 150 mg/kg i.p.) 10 days after conception. Frequencies of spontaneous p(un) reversions in p53(-/-) and p53(+/-) animals were not significantly different compared with their wild-type littermates. X-ray treatment increased the recombination frequency in wild-type and p53(+/-), but surprisingly not in p53(-/-) offspring. In contrast, B¿aP treatment caused a dose-dependent increase in p(un) reversion frequencies in all three genotypes. Western blot analysis of embryos indicated that p53 protein levels increased approximately 3-fold following X-ray treatment, while B¿aP had no effect on p53 expression. These results are in agreement with the proposal that p53 is involved in the DNA damage response following X-ray exposure and suggest that X-ray-induced double-strand breaks are processed differently in p53(-/-) animals.
Collapse
Affiliation(s)
- J Aubrecht
- Department of Cancer Cell Biology, Harvard School of Public Health, 665 Huntington Avenue, Boston, MA 2115, USA
| | | | | | | |
Collapse
|
16
|
Dass SB, Bucci TJ, Heflich RH, Casciano DA. Evaluation of the transgenic p53+/- mouse for detecting genotoxic liver carcinogens in a short-term bioassay. Cancer Lett 1999; 143:81-5. [PMID: 10465341 DOI: 10.1016/s0304-3835(99)00196-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The transgenic p53-deficient heterozygous (p53+/-) mouse is prone to both spontaneous and induced tumors and has been proposed for use in a sensitive, short-term (6 months) assay for identifying genotoxic, multispecies carcinogens. It is not clear, however, if a short-term assay with p53+/- mice detects agents that target certain organs, in particular, the liver. In this study, we treated neonatal male p53+/- and p53+/+ mice with the genotoxic carcinogens dimethylnitrosamine (DMN), 2-amino-1-methyl-6-phenylimidazo [4,5-b]pyridine (PhIP), and 6-nitrochrysene (6-NC). In keeping with the methodology of the proposed short-term assay, the p53+/- mice were evaluated for tumors 7 months after treatment. Wild-type neonatal mice treated with genotoxic carcinogens are known to develop tumors within 1 year; hence, the p53+/+ animals used as controls were subjected to pathological examination at 1 year of age. Our results showed that PhIP was not tumorigenic in either group of mice. Liver tumor incidence increased significantly in the p53+/+ mice treated with DMN and 6-NC, indicating that the conditions of the bioassay were conducive to the promotion of liver tumorigenesis. On the other hand, these two chemicals failed to induce a significant increase in liver tumors in the p53+/- mice by seven months. This result suggests that a deficiency in the amount of p53 protein does not lead to accelerated liver tumorigenesis in mice, and contrasts with previous reports that show a decreased latency of tumors in non-liver targets.
Collapse
Affiliation(s)
- S B Dass
- Division of Genetic and Reproductive Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | | | | | | |
Collapse
|
17
|
Haks MC, Krimpenfort P, van den Brakel JH, Kruisbeek AM. Pre-TCR signaling and inactivation of p53 induces crucial cell survival pathways in pre-T cells. Immunity 1999; 11:91-101. [PMID: 10435582 DOI: 10.1016/s1074-7613(00)80084-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Signaling through the pre-TCR is essential for early T cell development and is severely impaired in mice lacking the CD3 gamma chain of the pre-TCR. We here address the molecular mechanisms underlying this defect. Impaired pre-TCR signaling is shown to be associated with a profound increase in the number of apoptotic CD4- CD8- (DN) thymocytes. Introduction of p53 deficiency into CD3 gamma-deficient mice completely reverses the cell survival defect in CD3 gamma-deficient DN thymocytes and rescues the block in pre-T cell differentiation. In addition, the CD4+ CD8+ (DP) compartment is expanded to its normal size. These findings suggest that the pre-TCR regulates progression through the DNA-damage checkpoint of the DN to DP transition by inactivating p53.
Collapse
Affiliation(s)
- M C Haks
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam
| | | | | | | |
Collapse
|
18
|
Nozawa H, Oda E, Nakao K, Ishihara M, Ueda S, Yokochi T, Ogasawara K, Nakatsuru Y, Shimizu S, Ohira Y, Hioki K, Aizawa S, Ishikawa T, Katsuki M, Muto T, Taniguchi T, Tanaka N. Loss of transcription factor IRF-1 affects tumor susceptibility in mice carrying the Ha-ras transgene or nullizygosity for p53. Genes Dev 1999; 13:1240-5. [PMID: 10346812 PMCID: PMC316726 DOI: 10.1101/gad.13.10.1240] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The transcription factor IRF-1 has been implicated in tumor suppression: IRF-1 suppresses cell transformation and mediates apoptosis in vitro. Here we show that the loss of IRF-1 alleles per se has no effect on spontaneous tumor development in the mouse but dramatically exacerbates previous tumor predispositions caused by the c-Ha-ras transgene or by nullizygosity for p53. Grossly altered tumor spectrum, as compared to p53-null mice, was also observed in mice lacking both IRF-1 and p53, and cells from these mice show significantly higher mutation rate. Our results suggest that IRF-1 is a new member of the tumor susceptibility genes.
Collapse
Affiliation(s)
- H Nozawa
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Sedivy JM. Can ends justify the means?: telomeres and the mechanisms of replicative senescence and immortalization in mammalian cells. Proc Natl Acad Sci U S A 1998; 95:9078-81. [PMID: 9689036 PMCID: PMC33878 DOI: 10.1073/pnas.95.16.9078] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Finite replicative lifespan, or senescence, of mammalian cells in culture is a phenomenon that has generated much curiosity since its description. The obvious significance of senescence to organismal aging and the development of cancer has engendered a long-lasting and lively debate about its mechanisms. Recent discoveries concerning the phenotypes of telomerase knockout mice, the consequences of telomerase reexpression in somatic cells, and genes that regulate senescence have provided striking molecular insights but also have uncovered important new questions. The objective of this review is to reconcile old observations with new molecular details and to focus attention on the key remaining puzzles.
Collapse
Affiliation(s)
- J M Sedivy
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
20
|
Hüsler MR, Kotopoulis KA, Sundberg JP, Tennent BJ, Kunig SV, Knowles BB. Lactation-induced WAP-SV40 Tag transgene expression in C57BL/6J mice leads to mammary carcinoma. Transgenic Res 1998; 7:253-63. [PMID: 9859214 DOI: 10.1023/a:1008865911660] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Two transgenic lineages were generated by directing the expression of SV40 T antigen to the mammary gland of inbred C57BL/6J mice using the whey acidic protein (WAP) promoter. In one lineage, WAPTag 1, multiparous female mice developed mammary adenocarcinoma with an average latency period of 13 months. The histopathological phenotype was heterogeneous, tumours occurred in a stochastic fashion, normal tissue was located next to neoplastic tissue, the mammary tumours usually developed and were remarkably similar to that observed in human cases. In addition, male and virgin females developed a poorly differentiated SV40 T antigen-positive soft tissue sarcoma, also at 13 months of age. In the other lineage, WAPTag 3, some parous females developed mammary tumours, but most mice succumbed to osteosarcomas arising from the os petrosum at 5.5 to 6 months of age and on necropsy, renal adenocarcinomas were also found. Appearance of these unexpected tumour types demonstrates the non-specific expression of SV40 Tag under the control of the WAP promoter. The expression of SV40 Tag in mammary glands at different stages of development was also examined, and only actively lactating glands were positive. This suggests that the abundant cyclic synthesis of SV40 Tag associated with pregnancy is required for mammary tumorigenesis in these lineages.
Collapse
MESH Headings
- Adenocarcinoma, Papillary/genetics
- Adenocarcinoma, Papillary/pathology
- Animals
- Antigens, Polyomavirus Transforming/genetics
- Base Sequence
- DNA Primers/genetics
- Female
- Gene Expression
- Humans
- Lactation/genetics
- Male
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Milk Proteins/genetics
- Osteosarcoma/genetics
- Osteosarcoma/pathology
- Parity
- Pregnancy
- Promoter Regions, Genetic
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/pathology
- Soft Tissue Neoplasms/genetics
- Soft Tissue Neoplasms/pathology
Collapse
Affiliation(s)
- M R Hüsler
- Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | | | | | | | | |
Collapse
|
21
|
Roberts RA. Transgenic rodent mutation/cancer bioassays: cell cycle control, cell proliferation and apoptosis as modifiers of outcome. Mutat Res 1998; 398:189-95. [PMID: 9626979 DOI: 10.1016/s0027-5107(97)00239-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- R A Roberts
- Zeneca Central Toxicology Laboratory, Alderley Park, Macclesfield, UK.
| |
Collapse
|
22
|
Mayr B, Reifinger M, Loupal G. Polymorphisms in feline tumour suppressor gene p53. Mutations in an osteosarcoma and a mammary carcinoma. Vet J 1998; 155:103-6. [PMID: 9455165 DOI: 10.1016/s1090-0233(98)80044-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- B Mayr
- Institute for Animal Breeding and Genetics, Veterinary University, Vienna, Austria
| | | | | |
Collapse
|
23
|
Opalka B, Kasimir-Bauer S. Looking at p53: theoretical implications and methodological aspects. ARCHIVES OF TOXICOLOGY. SUPPLEMENT. = ARCHIV FUR TOXIKOLOGIE. SUPPLEMENT 1997; 19:17-28. [PMID: 9079191 DOI: 10.1007/978-3-642-60682-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- B Opalka
- Klinikum der Universität (GHS) Essen
| | | |
Collapse
|
24
|
Bedell MA, Largaespada DA, Jenkins NA, Copeland NG. Mouse models of human disease. Part II: recent progress and future directions. Genes Dev 1997; 11:11-43. [PMID: 9000048 DOI: 10.1101/gad.11.1.11] [Citation(s) in RCA: 141] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- M A Bedell
- Mammalian Genetics Laboratory, ABL-Basic Research Program, NCI-Frederick Cancer Research and Development Center, Frederick, Maryland 21702-1201, USA
| | | | | | | |
Collapse
|