1
|
Peña-Balderas AM, Martínez-Sánchez M, Olmos-Sánchez I, Calderón-González K, Moctezuma-Dávila M, Rangel-Charqueño M, Hernández-Monge J, Olivares-Illana V. Analysis of pathogenic variants in retinoblastoma reveals a potential gain of function mutation. Genes Cancer 2025; 16:1-15. [PMID: 39845333 PMCID: PMC11748778 DOI: 10.18632/genesandcancer.239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025] Open
Abstract
Retinoblastoma (Rb1) is a gene that codes for a tumour suppressor protein involved in various types of cancer. It was first described in retinoblastoma and is segregated as an autosomal dominant trait with high penetrance. In 1971, Knudson proposed his hypothesis of the two hits, where two mutational events are required to initiate tumour progression. We analysed three different point mutations present in patients' retinoblastoma. We produced three cell lines with retinoblastoma protein (RB) mutated in various regions: the missense pN328H, pD718N, and the nonsense early stop codon pR552*. We studied the effect of these point mutations on levels of mRNA and protein expression, proliferation, viability, localisation, and migration using an RBKO cell line. All three affected their localisation patterns and proliferation. However, the pR552* mutation also increases viability and migration. Moreover, when this mutation is simultaneously expressed with a wild-type RB, the phenotype and proliferation parameters are as with the mutant alone, suggesting that maybe only one mutated allele is needed to trigger the characteristic cancer phenotype. In other words, the pR552* mutant behaves more like a gain-of-function or oncogenic mutant. Indeed, a family carrying this mutation showed complete penetrance and high expressivity.
Collapse
Affiliation(s)
- Ana María Peña-Balderas
- Laboratorio de Interacciones Biomoleculares y Cáncer, Instituto de Física Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| | - Mayra Martínez-Sánchez
- Laboratorio de Interacciones Biomoleculares y Cáncer, Instituto de Física Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| | - Isaí Olmos-Sánchez
- Laboratorio de Interacciones Biomoleculares y Cáncer, Instituto de Física Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| | - Karla Calderón-González
- Laboratorio de Interacciones Biomoleculares y Cáncer, Instituto de Física Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| | - Mariana Moctezuma-Dávila
- Laboratorio de Interacciones Biomoleculares y Cáncer, Instituto de Física Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
- Present address: Houston Methodist Hospital, Department of Pathology and Genomic Medicine, Houston, TX 77030, USA
| | - Martha Rangel-Charqueño
- División de Cirugía, Departamento de Oftalmología, Hospital Central “Ignacio Morones Prieto”, San Luis Potosí, México
| | - Jesús Hernández-Monge
- Investigador por México, Laboratorio de Biomarcadores Moleculares, Instituto de Física, Universidad Autónoma de San Luis Potosí, México City, México
| | - Vanesa Olivares-Illana
- Laboratorio de Interacciones Biomoleculares y Cáncer, Instituto de Física Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| |
Collapse
|
2
|
Curylova L, Staniczkova Zambo I, Neradil J, Kyr M, Jurackova N, Pavlova S, Polaskova K, Mudry P, Sterba J, Veselska R, Skoda J. Dysregulation of the p53 pathway provides a therapeutic target in aggressive pediatric sarcomas with stem-like traits. Cell Oncol (Dordr) 2024; 47:2317-2334. [PMID: 39630408 DOI: 10.1007/s13402-024-01020-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2024] [Indexed: 01/11/2025] Open
Abstract
PURPOSE Pediatric sarcomas are bone and soft tissue tumors that often exhibit high metastatic potential and refractory stem-like phenotypes, resulting in poor outcomes. Aggressive sarcomas frequently harbor a disrupted p53 pathway. However, whether pediatric sarcoma stemness is associated with abrogated p53 function and might be attenuated via p53 reactivation remains unclear. METHODS We utilized a unique panel of pediatric sarcoma models and tumor tissue cohorts to investigate the correlation between the expression of stemness-related transcription factors, p53 pathway dysregulations, tumorigenicity in vivo, and clinicopathological features. TP53 mutation status was assessed by next-generation sequencing. Major findings were validated via shRNA-mediated silencing and functional assays. The p53 pathway-targeting drugs were used to explore the effects and selectivity of p53 reactivation against sarcoma cells with stem-like traits. RESULTS We found that highly tumorigenic stem-like sarcoma cells exhibit dysregulated p53, making them vulnerable to drugs that restore wild-type p53 activity. Immunohistochemistry of mouse xenografts and human tumor tissues revealed that p53 dysregulations, together with enhanced expression of the stemness-related transcription factors SOX2 or KLF4, are crucial features in pediatric osteosarcoma, rhabdomyosarcoma, and Ewing's sarcoma development. p53 dysregulation appears to be an important step for sarcoma cells to acquire a fully stem-like phenotype, and p53-positive pediatric sarcomas exhibit a high frequency of early metastasis. Importantly, reactivating p53 signaling via MDM2/MDMX inhibition selectively induces apoptosis in aggressive, stem-like Ewing's sarcoma cells while sparing healthy fibroblasts. CONCLUSIONS Our results indicate that restoring canonical p53 activity provides a promising strategy for developing improved therapies for pediatric sarcomas with unfavorable stem-like traits.
Collapse
Affiliation(s)
- Lucie Curylova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, 656 91, Czech Republic
| | - Iva Staniczkova Zambo
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, 656 91, Czech Republic
- 1st Department of Pathology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jakub Neradil
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, 656 91, Czech Republic
| | - Michal Kyr
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 613 00, Czech Republic
| | - Nicola Jurackova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- 1st Department of Pathology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sarka Pavlova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, 625 00, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, and Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 625 00, Czech Republic
| | - Kristyna Polaskova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 613 00, Czech Republic
| | - Peter Mudry
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 613 00, Czech Republic
| | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 613 00, Czech Republic
| | - Renata Veselska
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, 656 91, Czech Republic
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 625 00, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, Brno, 656 91, Czech Republic.
| |
Collapse
|
3
|
Synoradzki KJ, Bartnik E, Czarnecka AM, Fiedorowicz M, Firlej W, Brodziak A, Stasinska A, Rutkowski P, Grieb P. TP53 in Biology and Treatment of Osteosarcoma. Cancers (Basel) 2021; 13:4284. [PMID: 34503094 PMCID: PMC8428337 DOI: 10.3390/cancers13174284] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
The TP53 gene is mutated in 50% of human tumors. Oncogenic functions of mutant TP53 maintain tumor cell proliferation and tumor growth also in osteosarcomas. We collected data on TP53 mutations in patients to indicate which are more common and describe their role in in vitro and animal models. We also describe animal models with TP53 dysfunction, which provide a good platform for testing the potential therapeutic approaches. Finally, we have indicated a whole range of pharmacological compounds that modulate the action of p53, stabilize its mutated versions or lead to its degradation, cause silencing or, on the contrary, induce the expression of its functional version in genetic therapy. Although many of the described therapies are at the preclinical testing stage, they offer hope for a change in the approach to osteosarcoma treatment based on TP53 targeting in the future.
Collapse
Affiliation(s)
- Kamil Jozef Synoradzki
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.M.C.); (A.S.); (P.G.)
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, 02-106 Warsaw, Poland;
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Anna M. Czarnecka
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.M.C.); (A.S.); (P.G.)
- Department of Soft Tissue, Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (W.F.); (P.R.)
| | - Michał Fiedorowicz
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Wiktoria Firlej
- Department of Soft Tissue, Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (W.F.); (P.R.)
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Anna Brodziak
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Department of Oncology and Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Agnieszka Stasinska
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.M.C.); (A.S.); (P.G.)
| | - Piotr Rutkowski
- Department of Soft Tissue, Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (W.F.); (P.R.)
| | - Paweł Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.M.C.); (A.S.); (P.G.)
| |
Collapse
|
4
|
Zheng H, Yan B, Wu Q, Zhang J. MicroRNA-9-5p increases the sensitivity of colorectal cancer cells to 5-fluorouracil by downregulating high mobility group A2 expression. Oncol Lett 2021; 21:235. [PMID: 33613724 PMCID: PMC7856691 DOI: 10.3892/ol.2021.12496] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022] Open
Abstract
Chemotherapy drug 5-fluorouracil (5-FU) is the first-line treatment for colorectal cancer (CRC); however, 5-FU resistance decreases CRC therapeutic efficiency. A previous study revealed that microRNA (miR)-9-5p serves an antitumor effect in CRC. However, the effect of miR-9-5p in CRC chemoresistance remains unknown. In the present study, two CRC cell lines, including HT-29 and HCT-116 cells, were used to investigate the impact of miR-9-5p in overcoming 5-FU resistance. The results revealed that treatment with 5-FU decreased CRC cell viability and upregulated miR-9-5p expression in both CRC cells. Knockdown of miR-9-5p decreased HCT-116 cell sensitivity to 5-FU and inhibited apoptosis. By contrast, miR-9-5p overexpression enhanced the sensitivity of HT-29 cells to 5-FU and induced apoptosis. Additionally, it was confirmed that miR-9-5p directly targeted high mobility group A2 (HMGA2). HMGA2 overexpression reversed miR-9-5p-induced HT-29 apoptosis. The present study indicated that miR-9-5p enhanced the sensitivity of CRC cells to 5-FU via downregulating HMGA2 expression.
Collapse
Affiliation(s)
- Huizhe Zheng
- Department of Pathology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China.,Key Laboratory of Tumor Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Bin Yan
- Department of Pathology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China.,Key Laboratory of Tumor Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Qi Wu
- Department of Pathology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China.,Key Laboratory of Tumor Prevention and Treatment of Heilongjiang Province, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Jingli Zhang
- Department of Rheumatology and Immunology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| |
Collapse
|
5
|
MDS and TP53: When One Hit Just Isn't Enough…. Hemasphere 2020; 4:e494. [PMID: 33196012 PMCID: PMC7655086 DOI: 10.1097/hs9.0000000000000494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 11/26/2022] Open
|
6
|
Szpirer C. Rat models of human diseases and related phenotypes: a systematic inventory of the causative genes. J Biomed Sci 2020; 27:84. [PMID: 32741357 PMCID: PMC7395987 DOI: 10.1186/s12929-020-00673-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022] Open
Abstract
The laboratory rat has been used for a long time as the model of choice in several biomedical disciplines. Numerous inbred strains have been isolated, displaying a wide range of phenotypes and providing many models of human traits and diseases. Rat genome mapping and genomics was considerably developed in the last decades. The availability of these resources has stimulated numerous studies aimed at discovering causal disease genes by positional identification. Numerous rat genes have now been identified that underlie monogenic or complex diseases and remarkably, these results have been translated to the human in a significant proportion of cases, leading to the identification of novel human disease susceptibility genes, helping in studying the mechanisms underlying the pathological abnormalities and also suggesting new therapeutic approaches. In addition, reverse genetic tools have been developed. Several genome-editing methods were introduced to generate targeted mutations in genes the function of which could be clarified in this manner [generally these are knockout mutations]. Furthermore, even when the human gene causing a disease had been identified without resorting to a rat model, mutated rat strains (in particular KO strains) were created to analyze the gene function and the disease pathogenesis. Today, over 350 rat genes have been identified as underlying diseases or playing a key role in critical biological processes that are altered in diseases, thereby providing a rich resource of disease models. This article is an update of the progress made in this research and provides the reader with an inventory of these disease genes, a significant number of which have similar effects in rat and humans.
Collapse
Affiliation(s)
- Claude Szpirer
- Université Libre de Bruxelles, B-6041, Gosselies, Belgium.
- , Waterloo, Belgium.
| |
Collapse
|
7
|
Czarnecka AM, Synoradzki K, Firlej W, Bartnik E, Sobczuk P, Fiedorowicz M, Grieb P, Rutkowski P. Molecular Biology of Osteosarcoma. Cancers (Basel) 2020; 12:E2130. [PMID: 32751922 PMCID: PMC7463657 DOI: 10.3390/cancers12082130] [Citation(s) in RCA: 221] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma (OS) is the most frequent primary bone cancer in children and adolescents and the third most frequent in adults. Many inherited germline mutations are responsible for syndromes that predispose to osteosarcomas including Li Fraumeni syndrome, retinoblastoma syndrome, Werner syndrome, Bloom syndrome or Diamond-Blackfan anemia. TP53 is the most frequently altered gene in osteosarcoma. Among other genes mutated in more than 10% of OS cases, c-Myc plays a role in OS development and promotes cell invasion by activating MEK-ERK pathways. Several genomic studies showed frequent alterations in the RB gene in pediatric OS patients. Osteosarcoma driver mutations have been reported in NOTCH1, FOS, NF2, WIF1, BRCA2, APC, PTCH1 and PRKAR1A genes. Some miRNAs such as miR-21, -34a, -143, -148a, -195a, -199a-3p and -382 regulate the pathogenic activity of MAPK and PI3K/Akt-signaling pathways in osteosarcoma. CD133+ osteosarcoma cells have been shown to exhibit stem-like gene expression and can be tumor-initiating cells and play a role in metastasis and development of drug resistance. Although currently osteosarcoma treatment is based on adriamycin chemoregimens and surgery, there are several potential targeted therapies in development. First of all, activity and safety of cabozantinib in osteosarcoma were studied, as well as sorafenib and pazopanib. Finally, novel bifunctional molecules, of potential imaging and osteosarcoma targeting applications may be used in the future.
Collapse
Affiliation(s)
- Anna M Czarnecka
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute-Oncology Centre, 02-781 Warsaw, Poland
| | - Kamil Synoradzki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Wiktoria Firlej
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute-Oncology Centre, 02-781 Warsaw, Poland
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, 02-106 Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Pawel Sobczuk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute-Oncology Centre, 02-781 Warsaw, Poland
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Michal Fiedorowicz
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Interinstitute Laboratory of New Diagnostic Applications of MRI, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Pawel Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute-Oncology Centre, 02-781 Warsaw, Poland
| |
Collapse
|
8
|
Smyczyńska U, Strzemecki D, Czarnecka AM, Fendler W, Fiedorowicz M, Wełniak-Kamińska M, Guzowska M, Synoradzki K, Cheda Ł, Rogulski Z, Grieb P. TP53-Deficient Angiosarcoma Expression Profiling in Rat Model. Cancers (Basel) 2020; 12:cancers12061525. [PMID: 32532104 PMCID: PMC7352674 DOI: 10.3390/cancers12061525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/28/2022] Open
Abstract
Sarcomas are a heterogeneous group of malignant tumors, that develop from mesenchymal cells. Sarcomas are tumors associated with poor prognosis and expected short overall survival. Efforts to improve treatment efficacy and treatment outcomes of advanced and metastatic sarcoma patients have not led to significant improvements in the last decades. In the Tp53C273X/C273X rat model we therefore aimed to characterize specific gene expression pattern of angiosarcomas with a loss of TP53 function. The presence of metabolically active tumors in several locations including the brain, head and neck, extremities and abdomen was confirmed by magnetic resonance imaging (MRI) and positron emission tomography (PET) examinations. Limb angiosarcoma tumors were selected for microarray expression analysis. The most upregulated pathways in angiosarcoma vs all other tissues were related to cell cycle with mitosis and meiosis, chromosome, nucleosome and telomere maintenance as well as DNA replication and recombination. The downregulated genes were responsible for metabolism, including respiratory chain electron transport, tricarboxylic acid (TCA) cycle, fatty acid metabolism and amino-acid catabolism. Our findings demonstrated that the type of developing sarcoma depends on genetic background, underscoring the importance of developing more malignancy susceptibility models in various strains and species to simulate the study of the diverse genetics of human sarcomas.
Collapse
Affiliation(s)
- Urszula Smyczyńska
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (U.S.); (W.F.)
| | - Damian Strzemecki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
| | - Anna M. Czarnecka
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
- Department of Soft Tissue, Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-608-6474
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (U.S.); (W.F.)
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02284-9168, USA
| | - Michał Fiedorowicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Marlena Wełniak-Kamińska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Magdalena Guzowska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| | - Kamil Synoradzki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
| | - Łukasz Cheda
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 02-093 Warsaw, Poland; (Ł.C.); (Z.R.)
| | - Zbigniew Rogulski
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 02-093 Warsaw, Poland; (Ł.C.); (Z.R.)
| | - Paweł Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
| |
Collapse
|
9
|
Gomarteli K, Fleckenstein J, Kirschner S, Bobu V, Brockmann MA, Henzler T, Meyer M, Riffel P, Schönberg SO, Veldwijk MR, Kränzlin B, Hoerner C, Glatting G, Wenz F, Herskind C, Giordano FA. Radiation-induced malignancies after intensity-modulated versus conventional mediastinal radiotherapy in a small animal model. Sci Rep 2019; 9:15489. [PMID: 31664066 PMCID: PMC6820874 DOI: 10.1038/s41598-019-51735-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/07/2019] [Indexed: 12/28/2022] Open
Abstract
A long-standing hypothesis in radiotherapy is that intensity-modulated radiotherapy (IMRT) increases the risk of second cancer due to low-dose exposure of large volumes of normal tissue. Therefore, young patients are still treated with conventional techniques rather than with modern IMRT. We challenged this hypothesis in first-of-its-kind experiments using an animal model. Cancer-prone Tp53+/C273X knockout rats received mediastinal irradiation with 3 × 5 or 3 × 8 Gy using volumetric-modulated arc therapy (VMAT, an advanced IMRT) or conventional anterior-posterior/posterior-anterior (AP/PA) beams using non-irradiated rats as controls (n = 15/group, ntotal = 90). Tumors were assigned to volumes receiving 90–107%, 50–90%, 5–50%, and <5% of the target dose and characterized by histology and loss-of-heterozygosity (LOH). Irradiated rats predominantly developed lymphomas and sarcomas in areas receiving 50–107% (n = 26) rather than 5–50% (n = 7) of the target dose. Latency was significantly shortened only after 3 × 8 Gy vs. controls (p < 0.0001). The frequency (14/28 vs. 19/29; p = 0.29) and latency (218 vs. 189 days; p = 0.17) of radiation-associated tumors were similar after VMAT vs. AP/PA. LOH was strongly associated with sarcoma but not with treatment. The results do not support the hypothesis that IMRT increases the risk of second cancer. Thus the current practice of withholding dose-sparing IMRT from young patients may need to be re-evaluated.
Collapse
Affiliation(s)
- Kaga Gomarteli
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jens Fleckenstein
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Stefanie Kirschner
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Neuroradiology, University Medical Center Mainz, Johannes Gutenberg-University, Mainz, Germany
| | - Vladimir Bobu
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marc A Brockmann
- Department of Neuroradiology, University Medical Center Mainz, Johannes Gutenberg-University, Mainz, Germany
| | - Thomas Henzler
- Department of Clinical Radiology and Nuclear Medicine, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Mathias Meyer
- Department of Clinical Radiology and Nuclear Medicine, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Philipp Riffel
- Department of Clinical Radiology and Nuclear Medicine, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Stefan O Schönberg
- Department of Clinical Radiology and Nuclear Medicine, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Marlon R Veldwijk
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Bettina Kränzlin
- Medical Research Center, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christian Hoerner
- Institute of Pathology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Gerhard Glatting
- Medical Radiation Physics, Department of Nuclear Medicine, Ulm University, Ulm, Germany
| | - Frederik Wenz
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carsten Herskind
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Frank A Giordano
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
10
|
Varghese V, Magnani L, Harada-Shoji N, Mauri F, Szydlo RM, Yao S, Lam EWF, Kenny LM. FOXM1 modulates 5-FU resistance in colorectal cancer through regulating TYMS expression. Sci Rep 2019; 9:1505. [PMID: 30728402 PMCID: PMC6365533 DOI: 10.1038/s41598-018-38017-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 10/07/2018] [Indexed: 12/11/2022] Open
Abstract
Resistance to 5-Fluoruracil (5-FU) has been linked to elevated expression of the main target, thymidylate synthase (TYMS), which catalyses the de novo pathway for production of deoxythymidine monophosphate. The potent oncogenic forkhead box transcription factor, FOXM1 is is regulated by E2F1 which also controls TYMS. This study reveals a significant role of FOXM1 in 5-FU resistance. Overexpression and knock-down studies of FOXM1 in colon cancer cells suggest the importance of FOXM1 in TYMS regulation. ChIP and global ChIP-seq data also confirms that FOXM1 can also potentially regulate other 5-FU targets, such as TYMS, thymidine kinase 1 (TK-1) and thymidine phosphorylase (TYMP). In human colorectal cancer tissue specimens, a strong correlation of FOXM1 and TYMS staining was observed. Elevated FOXM1 and TYMS expression was also observed in acquired 5-FU resistant colon cancer cells (HCT116 5-FU Res). A synergistic effect was observed following treatment of CRC cells with an inhibitor of FOXM1, thiostrepton, in combination with 5-FU. The combination treatment decreased colony formation and migration, and induced cell cycle arrest, DNA damage, and apoptosis in CRC cell lines. In summary, this research demonstrated that FOXM1 plays a pivotal role in 5-FU resistance at least partially through the regulation of TYMS.
Collapse
Affiliation(s)
- Vidhya Varghese
- Department of Surgery and Cancer, Imperial College London, London, USA
| | - Luca Magnani
- Department of Surgery and Cancer, Imperial College London, London, USA
| | | | - Francesco Mauri
- Imperial College Healthcare NHS Trust, Imperial College London, London, USA
| | | | - Shang Yao
- Department of Surgery and Cancer, Imperial College London, London, USA
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, London, USA.
| | - Laura M Kenny
- Department of Surgery and Cancer, Imperial College London, London, USA.
| |
Collapse
|
11
|
Goh WL, Lee MY, Lim TX, Chua JS, Brenner S, Ghadessy FJ, Teo YN. A novel molecular rotor facilitates detection of p53-DNA interactions using the Fluorescent Intercalator Displacement Assay. Sci Rep 2018; 8:12946. [PMID: 30154420 PMCID: PMC6113202 DOI: 10.1038/s41598-018-31197-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 08/13/2018] [Indexed: 01/04/2023] Open
Abstract
We have investigated the use of fluorescent molecular rotors as probes for detection of p53 binding to DNA. These are a class of fluorophores that undergo twisted intramolecular charge transfer (TICT). They are non-fluorescent in a freely rotating conformation and experience a fluorescence increase when restricted in the planar conformation. We hypothesized that intercalation of a molecular rotor between DNA base pairs would result in a fluorescence turn-on signal. Upon displacement by a DNA binding protein, measurable loss of signal would facilitate use of the molecular rotor in the fluorescent intercalator displacement (FID) assay. A panel of probes was interrogated using the well-established p53 model system across various DNA response elements. A novel, readily synthesizable molecular rotor incorporating an acridine orange DNA intercalating group (AO-R) outperformed other conventional dyes in the FID assay. It enabled relative measurement of p53 sequence-specific DNA interactions and study of the dominant-negative effects of cancer-associated p53 mutants. In a further application, AO-R also proved useful for staining apoptotic cells in live zebrafish embryos.
Collapse
Affiliation(s)
- Walter L Goh
- p53 Laboratory, A*STAR, 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore
| | - Min Yen Lee
- Molecular Engineering Lab, Biomedical Sciences Institutes, A*STAR, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Ting Xiang Lim
- p53 Laboratory, A*STAR, 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore
| | - Joy S Chua
- p53 Laboratory, A*STAR, 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore
| | - Sydney Brenner
- Molecular Engineering Lab, Biomedical Sciences Institutes, A*STAR, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Farid J Ghadessy
- p53 Laboratory, A*STAR, 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore.
| | - Yin Nah Teo
- Molecular Engineering Lab, Biomedical Sciences Institutes, A*STAR, 61 Biopolis Drive, Singapore, 138673, Singapore.
| |
Collapse
|
12
|
Dai MS, Hall SJ, Vantangoli Policelli MM, Boekelheide K, Spade DJ. Spontaneous testicular atrophy occurs despite normal spermatogonial proliferation in a Tp53 knockout rat. Andrology 2017; 5:1141-1152. [PMID: 28834365 DOI: 10.1111/andr.12409] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 06/12/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022]
Abstract
The tumor suppressor protein p53 (TP53) has many functions in cell cycle regulation, apoptosis, and DNA damage repair and is also involved in spermatogenesis in the mouse. To evaluate the role of p53 in spermatogenesis in the rat, we characterized testis biology in adult males of a novel p53 knockout rat (SD-Tp53tm1sage ). p53 knockout rats exhibited variable levels of testicular atrophy, including significantly decreased testis weights, atrophic seminiferous tubules, decreased seminiferous tubule diameter, and elevated spermatocyte TUNEL labeling rates, indicating a dysfunction in spermatogenesis. Phosphorylated histone H2AX protein levels and distribution were similar in the non-atrophic seminiferous tubules of both genotypes, showing evidence of pre-synaptic DNA double-strand breaks in leptotene and zygotene spermatocytes, preceding cell death in p53 knockout rat testes. Quantification of the spermatogonial stem cell (SSC) proliferation rate with bromodeoxyuridine (BrdU) labeling, in addition to staining with the undifferentiated type A spermatogonial marker GDNF family receptor alpha-1 (GFRA1), indicated that the undifferentiated spermatogonial population was normal in p53 knockout rats. Following exposure to 0.5 or 5 Gy X-ray, p53 knockout rats exhibited no germ cell apoptotic response beyond their unirradiated phenotype, while germ cell death in wild-type rat testes was elevated to a level similar to the unexposed p53 knockout rats. This study indicates that seminiferous tubule atrophy occurs following spontaneous, elevated levels of spermatocyte death in the p53 knockout rat. This phenomenon is variable across individual rats. These results indicate a critical role for p53 in rat germ cell survival and spermatogenesis.
Collapse
Affiliation(s)
- Matthew S Dai
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Susan J Hall
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | | | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
13
|
Strzemecki D, Guzowska M, Grieb P. Survival rates of homozygotic Tp53 knockout rats as a tool for preclinical assessment of cancer prevention and treatment. Cell Mol Biol Lett 2017; 22:9. [PMID: 28536640 PMCID: PMC5437597 DOI: 10.1186/s11658-017-0039-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/09/2017] [Indexed: 11/24/2022] Open
Abstract
Background The gene that encodes tumor protein p53, Tp53, is mutated or silenced in most human cancers and is recognized as one of the most important cancer drivers. Homozygotic Tp53 knockout mice, which develop lethal cancers early in their lives, are already used in cancer prevention studies, and now Tp53 knockout rats have also been generated. This study assessed feasibility of using homozygous Tp53 knockout rats to evaluate the possible outcome of cancer chemoprevention. Methods A small colony of Tp53 knockout rats with a Wistar strain genetic background was initiated and maintained in the animal house at our institution. Tp53 heterozygotic females were bred with Tp53 homozygous knockout males to obtain a surplus of knockout homozygotes. To evaluate the reproducibility of their lifespan, 4 groups of Tp53 homozygous knockout male rats born during consecutive quarters of the year were kept behind a sanitary barrier in a controlled environment until they reached a moribund state. Their individual lifespan data were used to construct quarterly survival curves. Results The four consecutive quarterly survival curves were highly reproducible. They were combined into a single “master” curve for use as a reference in intervention studies. The average lifespan of untreated male Tp53 homozygous knockout rats was normally distributed, with a median of 133 days. Sample size vs. effect calculations revealed that confirming a 20% and 30% increase in the lifespan would respectively require a sample size of 18 and 9 animals (when assessed using the t-test with a power of 80% and alpha set at 0.05). As an example, the Tp53 homozygous knockout rat model was used to test the chemopreventive properties of carnosine, a dipeptide with suspected anticancer properties possibly involving modulation of the mTOR pathway. The result was negative. Conclusion Further evaluation of the Tp53 homozygous knockout male rat colony is required before it can be confirmed as a viable tool for assessing new methods of cancer prevention or treatment.
Collapse
Affiliation(s)
- Damian Strzemecki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., Warsaw, 02-106 Poland
| | - Magdalena Guzowska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., Warsaw, 02-106 Poland
| | - Paweł Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., Warsaw, 02-106 Poland
| |
Collapse
|
14
|
Hansen SA, Hart ML, Busi S, Parker T, Goerndt A, Jones K, Amos-Landgraf JM, Bryda EC. Fischer-344 Tp53-knockout rats exhibit a high rate of bone and brain neoplasia with frequent metastasis. Dis Model Mech 2016; 9:1139-1146. [PMID: 27528400 PMCID: PMC5087826 DOI: 10.1242/dmm.025767] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/05/2016] [Indexed: 01/31/2023] Open
Abstract
Somatic mutations in the Tp53 tumor suppressor gene are the most commonly seen genetic alterations in cancer, and germline mutations in Tp53 predispose individuals to a variety of early-onset cancers. Development of appropriate translational animal models that carry mutations in Tp53 and recapitulate human disease are important for drug discovery, biomarker development and disease modeling. Current Tp53 mouse and rat models have significant phenotypic and genetic limitations, and often do not recapitulate certain aspects of human disease. We used a marker-assisted speed congenic approach to transfer a well-characterized Tp53-mutant allele from an outbred rat to the genetically inbred Fischer-344 (F344) rat to create the F344-Tp53tm1(EGFP-Pac)Qly/Rrrc (F344-Tp53) strain. On the F344 genetic background, the tumor spectrum shifted, with the primary tumor types being osteosarcomas and meningeal sarcomas, compared to the hepatic hemangiosarcoma and lymphoma identified in the original outbred stock model. The Fischer model is more consistent with the early onset of bone and central nervous system sarcomas found in humans with germline Tp53 mutations. The frequency of osteosarcomas in F344-Tp53 homozygous and heterozygous animals was 57% and 36%, respectively. Tumors were highly representative of human disease radiographically and histologically, with tumors found primarily on long bones with frequent pulmonary metastases. Importantly, the rapid onset of osteosarcomas in this promising new model fills a current void in animal models that recapitulate human pediatric osteosarcomas and could facilitate studies to identify therapeutic targets. Editors' choice: Transferring a Tp53-knockout allele from an outbred rat stock to the F344 inbred rat genetic background alters the spectrum of tumors, providing a model of early-onset brain and bone sarcomas.
Collapse
Affiliation(s)
- Sarah A Hansen
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MI 65211, USA
| | - Marcia L Hart
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MI 65211, USA
| | - Susheel Busi
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MI 65211, USA
| | - Taybor Parker
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MI 65211, USA
| | - Angela Goerndt
- Rat Resource and Research Center, University of Missouri, Columbia, MI 65211, USA
| | - Kevin Jones
- Departments of Orthopaedics and Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - James M Amos-Landgraf
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MI 65211, USA Rat Resource and Research Center, University of Missouri, Columbia, MI 65211, USA
| | - Elizabeth C Bryda
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MI 65211, USA Rat Resource and Research Center, University of Missouri, Columbia, MI 65211, USA
| |
Collapse
|
15
|
Complete reduction of p53 expression by RNA interference following heterozygous knockout in porcine fibroblasts. In Vitro Cell Dev Biol Anim 2016; 52:736-41. [PMID: 27142766 DOI: 10.1007/s11626-016-0026-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/03/2016] [Indexed: 10/21/2022]
Abstract
Tumor suppressor p53 plays a critical role in the regulation of cell cycle and apoptosis in mammals. Mutations of p53 often cause various cancers. Murine models have improved our understanding on tumorigenesis associated with p53 mutations. However, mice and humans are different in many ways. For example, the short lifespans of mice limit the clinical application of the data obtained from this species. Porcine model could be an alternative as pigs share many anatomical and physiological similarities with humans. Here, we modified the expression levels of p53 messenger RNA (mRNA) and protein in porcine fetal fibroblasts using a combination of gene targeting and RNA interference. First, we disrupted the p53 gene to produce p53 knockout (KO) cells. Second, the p53 shRNA expression vector was introduced into fibroblasts to isolate p53 knockdown (KD) cells. We obtained p53 KO, KD, and KO + KD fibroblasts which involve p53 KO and KD either separately or simultaneously. The mRNA expression of p53 in p53 KO fibroblasts was similar to that in the wild-type control. However, the mRNA expression levels of p53 in KD and KO + KD cells were significantly decreased. The p53 protein level significant reduced in p53 KD. Interestingly, no p53 protein was detected in KO + KD, suggesting a complete reduction of the protein by synergistic effect of KO and KD. This study demonstrated that various expression levels of p53 in porcine fibroblasts could be achieved by gene targeting and RNA interference. Moreover, complete abolishment of protein expression is feasible using a combination of gene targeting and RNA interference.
Collapse
|
16
|
Saalfrank A, Janssen KP, Ravon M, Flisikowski K, Eser S, Steiger K, Flisikowska T, Müller-Fliedner P, Schulze É, Brönner C, Gnann A, Kappe E, Böhm B, Schade B, Certa U, Saur D, Esposito I, Kind A, Schnieke A. A porcine model of osteosarcoma. Oncogenesis 2016; 5:e210. [PMID: 26974205 PMCID: PMC4815050 DOI: 10.1038/oncsis.2016.19] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/25/2016] [Accepted: 01/28/2016] [Indexed: 12/15/2022] Open
Abstract
We previously produced pigs with a latent oncogenic TP53 mutation. Humans with TP53 germline mutations are predisposed to a wide spectrum of early-onset cancers, predominantly breast, brain, adrenal gland cancer, soft tissue sarcomas and osteosarcomas. Loss of p53 function has been observed in >50% of human cancers. Here we demonstrate that porcine mesenchymal stem cells (MSCs) convert to a transformed phenotype after activation of latent oncogenic TP53R167H and KRASG12D, and overexpression of MYC promotes tumorigenesis. The process mimics key molecular aspects of human sarcomagenesis. Transformed porcine MSCs exhibit genomic instability, with complex karyotypes, and develop into sarcomas on transplantation into immune-deficient mice. In pigs, heterozygous knockout of TP53 was sufficient for spontaneous osteosarcoma development in older animals, whereas homozygous TP53 knockout resulted in multiple large osteosarcomas in 7–8-month-old animals. This is the first report that engineered mutation of an endogenous tumour-suppressor gene leads to invasive cancer in pigs. Unlike in Trp53 mutant mice, osteosarcoma developed in the long bones and skull, closely recapitulating the human disease. These animals thus promise a model for juvenile osteosarcoma, a relatively uncommon but devastating disease.
Collapse
Affiliation(s)
- A Saalfrank
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - K-P Janssen
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - M Ravon
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - K Flisikowski
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - S Eser
- Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - K Steiger
- Department of Pathology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - T Flisikowska
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - P Müller-Fliedner
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - É Schulze
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - C Brönner
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - A Gnann
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - E Kappe
- Department of Pathology, Bavarian Animal Health Service, Poing, Germany
| | - B Böhm
- Department of Pathology, Bavarian Animal Health Service, Poing, Germany
| | - B Schade
- Department of Pathology, Bavarian Animal Health Service, Poing, Germany
| | - U Certa
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - D Saur
- Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - I Esposito
- Institute of Pathology, Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | - A Kind
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - A Schnieke
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| |
Collapse
|
17
|
Hermsen R, Toonen P, Kuijk E, Youssef SA, Kuiper R, van Heesch S, de Bruin A, Cuppen E, Simonis M. Lack of major genome instability in tumors of p53 null rats. PLoS One 2015; 10:e0122066. [PMID: 25811670 PMCID: PMC4374866 DOI: 10.1371/journal.pone.0122066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/11/2015] [Indexed: 01/20/2023] Open
Abstract
Tumorigenesis is often associated with loss of tumor suppressor genes (such as TP53), genomic instability and telomere lengthening. Previously, we generated and characterized a rat p53 knockout model in which the homozygous rats predominantly develop hemangiosarcomas whereas the heterozygous rats mainly develop osteosarcomas. Using genome-wide analyses, we find that the tumors that arise in the heterozygous and homozygous Tp53C273X mutant animals are also different in their genomic instability profiles. While p53 was fully inactivated in both heterozygous and homozygous knockout rats, tumors from homozygous animals show very limited aneuploidy and low degrees of somatic copy number variation as compared to the tumors from heterozygous animals. In addition, complex structural rearrangements such as chromothripsis and breakage-fusion-bridge cycles were never found in tumors from homozygous animals, while these were readily detectable in tumors from heterozygous animals. Finally, we measured telomere length and telomere lengthening pathway activity and found that tumors of homozygous animals have longer telomeres but do not show clear telomerase or alternative lengthening of telomeres (ALT) activity differences as compared to the tumors from heterozygous animals. Taken together, our results demonstrate that host p53 status in this rat p53 knockout model has a large effect on both tumor type and genomic instability characteristics, where full loss of functional p53 is not the main driver of large-scale structural variations. Our results also suggest that chromothripsis primarily occurs under p53 heterozygous rather than p53 null conditions.
Collapse
Affiliation(s)
- Roel Hermsen
- Hubrecht Institute, KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Pim Toonen
- Hubrecht Institute, KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Ewart Kuijk
- Hubrecht Institute, KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Sameh A. Youssef
- Dutch Molecular Pathology Center. Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Raoul Kuiper
- Dutch Molecular Pathology Center. Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Sebastiaan van Heesch
- Hubrecht Institute, KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Alain de Bruin
- Dutch Molecular Pathology Center. Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Edwin Cuppen
- Hubrecht Institute, KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- * E-mail:
| | - Marieke Simonis
- Hubrecht Institute, KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
18
|
Abstract
ABSTRACT Cancers exhibit differences in metastatic behavior and drug sensitivity that correlate with certain tumor-specific variables such as differentiation grade, growth rate/extent and molecular regulatory aberrations. In practice, patient management is based on the past results of clinical trials adjusted for these biomarkers. Here, it is proposed that treatment strategies could be fine-tuned upfront simply by quantifying tumorigenic spatial (cell growth) and temporal (genetic stability) control losses, as predicted by genetic defects of cell-cycle-regulatory gatekeeper and genome-stabilizing caretaker tumor suppressor genes, respectively. These differential quantifications of tumor dysfunction may in turn be used to create a tumor-specific ‘periodic table’ that guides rational formulation of survival-enhancing anticancer treatment strategies.
Collapse
Affiliation(s)
- Richard J Epstein
- *Clinical Informatics & Research Centre, The Kinghorn Cancer Centre, 370 Victoria St, Darlinghurst 2010, Sydney, Australia
- Laboratory of Genome Evolution, Garvan Institute for Medical Research, 384 Victoria St, Darlinghurst 2010, Sydney, Australia
- Department of Oncology, & UNSW Clinical School, St Vincent's Hospital, 390 Victoria St, Darlinghurst 2010 Sydney, Australia
| |
Collapse
|
19
|
Shetzer Y, Kagan S, Koifman G, Sarig R, Kogan-Sakin I, Charni M, Kaufman T, Zapatka M, Molchadsky A, Rivlin N, Dinowitz N, Levin S, Landan G, Goldstein I, Goldfinger N, Pe'er D, Radlwimmer B, Lichter P, Rotter V, Aloni-Grinstein R. The onset of p53 loss of heterozygosity is differentially induced in various stem cell types and may involve the loss of either allele. Cell Death Differ 2014; 21:1419-31. [PMID: 24832469 DOI: 10.1038/cdd.2014.57] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 02/27/2014] [Accepted: 03/17/2014] [Indexed: 12/12/2022] Open
Abstract
p53 loss of heterozygosity (p53LOH) is frequently observed in Li-Fraumeni syndrome (LFS) patients who carry a mutant (Mut) p53 germ-line mutation. Here, we focused on elucidating the link between p53LOH and tumor development in stem cells (SCs). Although adult mesenchymal stem cells (MSCs) robustly underwent p53LOH, p53LOH in induced embryonic pluripotent stem cells (iPSCs) was significantly attenuated. Only SCs that underwent p53LOH induced malignant tumors in mice. These results may explain why LFS patients develop normally, yet acquire tumors in adulthood. Surprisingly, an analysis of single-cell sub-clones of iPSCs, MSCs and ex vivo bone marrow (BM) progenitors revealed that p53LOH is a bi-directional process, which may result in either the loss of wild-type (WT) or Mut p53 allele. Interestingly, most BM progenitors underwent Mutp53LOH. Our results suggest that the bi-directional p53LOH process may function as a cell-fate checkpoint. The loss of Mutp53 may be regarded as a DNA repair event leading to genome stability. Indeed, gene expression analysis of the p53LOH process revealed upregulation of a specific chromatin remodeler and a burst of DNA repair genes. However, in the case of loss of WTp53, cells are endowed with uncontrolled growth that promotes cancer.
Collapse
Affiliation(s)
- Y Shetzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - S Kagan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - G Koifman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - R Sarig
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - I Kogan-Sakin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - M Charni
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - T Kaufman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - M Zapatka
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - A Molchadsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - N Rivlin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - N Dinowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - S Levin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - G Landan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - I Goldstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - N Goldfinger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - D Pe'er
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - B Radlwimmer
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - P Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - V Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - R Aloni-Grinstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
20
|
Aloni-Grinstein R, Shetzer Y, Kaufman T, Rotter V. p53: the barrier to cancer stem cell formation. FEBS Lett 2014; 588:2580-9. [PMID: 24560790 DOI: 10.1016/j.febslet.2014.02.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 02/07/2014] [Accepted: 02/07/2014] [Indexed: 02/08/2023]
Abstract
The role of p53 as the "guardian of the genome" in differentiated somatic cells, triggering various biological processes, is well established. Recent studies in the stem cell field have highlighted a profound role of p53 in stem cell biology as well. These studies, combined with basic data obtained 20 years ago, provide insight into how p53 governs the quantity and quality of various stem cells, ensuring a sufficient repertoire of normal stem cells to enable proper development, tissue regeneration and a cancer free life. In this review we address the role of p53 in genomically stable embryonic stem cells, a unique predisposed cancer stem cell model and adult stem cells, its role in the generation of induced pluripotent stem cells, as well as its role as the barrier to cancer stem cell formation.
Collapse
Affiliation(s)
- Ronit Aloni-Grinstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yoav Shetzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tom Kaufman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
21
|
Epstein RJ. The unpluggable in pursuit of the undruggable: tackling the dark matter of the cancer therapeutics universe. Front Oncol 2013; 3:304. [PMID: 24377088 PMCID: PMC3859984 DOI: 10.3389/fonc.2013.00304] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/29/2013] [Indexed: 01/19/2023] Open
Abstract
The notion that targeted drugs can unplug gain-of-function tumor pathways has revitalized pharmaceutical research, but the survival benefits of this strategy have so far proven modest. A weakness of oncogene-blocking approaches is that they do not address the problem of cancer progression as selected by the recessive phenotypes of genetic instability and apoptotic resistance which in turn arise from loss-of-function – i.e., undruggable – defects of caretaker (e.g., BRCA, MLH1) or gatekeeper (e.g., TP53, PTEN) suppressor genes. Genetic instability ensures that rapid cell kill is balanced by rapid selection for apoptotic resistance and hence for metastasis, casting doubt on the assumption that cytotoxicity (“response”) remains the best way to identify survival-enhancing drugs. In the absence of gene therapy, it is proposed here that caretaker-defective (high-instability) tumors may be best treated with low-lethality drugs inducing replicative (RAS-RAF-ERK) arrest or dormancy, causing “stable disease” rather than tumorilytic remission. Gatekeeper-defective (death-resistant) tumors, on the other hand, may be best managed by combining survival (PI3K-AKT-mTOR) pathway blockade with metronomic or sequential pro-apoptotic drugs.
Collapse
Affiliation(s)
- Richard J Epstein
- Laboratory of Genome Evolution & Informatics, The Kinghorn Cancer Centre, and Clinical Informatics & Research Centre, Department of Oncology, St Vincent's Hospital, UNSW Clinical School , Sydney, NSW , Australia
| |
Collapse
|
22
|
The new pig on the block: modelling cancer in pigs. Transgenic Res 2013; 22:673-80. [DOI: 10.1007/s11248-013-9720-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/30/2013] [Indexed: 01/09/2023]
|
23
|
Kawamata M, Ochiya T. Two distinct knockout approaches highlight a critical role for p53 in rat development. Sci Rep 2012; 2:945. [PMID: 23230510 PMCID: PMC3517977 DOI: 10.1038/srep00945] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 10/29/2012] [Indexed: 01/06/2023] Open
Abstract
Gene targeting in embryonic stem cells (ESCs) has become the principal technology for generating knockout models. Although numerous studies have predicted that the disruption of p53 leads to increased developmental anomalies and malignancies, most p53 knockout mice develop normally. Therefore, the role of p53 in animal development was examined using rat knockout models. Conventionally generated homozygous KO males developed normally, whereas females rarely survived due to neural tube defects. Mutant chimeras generated via blastocyst injection with p53-null ESCs exhibited high rates of embryonic lethality in both sexes. This phenotype could be observed in one month by the use of zinc-finger nucleases. The p53-null ESCs were resistant to apoptosis and differentiation, and exhibited severe chromosome instabilities in the chimera-contributed cells, suggesting an essential role for p53 in maintaining ESC quality and genomic integrity. These results demonstrate that p53 functions as a guardian of embryogenesis in the rats.
Collapse
Affiliation(s)
- Masaki Kawamata
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute , 1-1, Tsukiji, 5-chome, Chuo-ku, Tokyo 104-0045, Japan
| | | |
Collapse
|
24
|
Leuchs S, Saalfrank A, Merkl C, Flisikowska T, Edlinger M, Durkovic M, Rezaei N, Kurome M, Zakhartchenko V, Kessler B, Flisikowski K, Kind A, Wolf E, Schnieke A. Inactivation and inducible oncogenic mutation of p53 in gene targeted pigs. PLoS One 2012; 7:e43323. [PMID: 23071491 PMCID: PMC3465291 DOI: 10.1371/journal.pone.0043323] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 07/19/2012] [Indexed: 12/24/2022] Open
Abstract
Mutation of the tumor suppressor p53 plays a major role in human carcinogenesis. Here we describe gene-targeted porcine mesenchymal stem cells (MSCs) and live pigs carrying a latent TP53R167H mutant allele, orthologous to oncogenic human mutant TP53R175H and mouse Trp53R172H, that can be activated by Cre recombination. MSCs carrying the latent TP53R167H mutant allele were analyzed in vitro. Homozygous cells were p53 deficient, and on continued culture exhibited more rapid proliferation, anchorage independent growth, and resistance to the apoptosis-inducing chemotherapeutic drug doxorubicin, all characteristic of cellular transformation. Cre mediated recombination activated the latent TP53R167H allele as predicted, and in homozygous cells expressed mutant p53-R167H protein at a level ten-fold greater than wild-type MSCs, consistent with the elevated levels found in human cancer cells. Gene targeted MSCs were used for nuclear transfer and fifteen viable piglets were produced carrying the latent TP53R167H mutant allele in heterozygous form. These animals will allow study of p53 deficiency and expression of mutant p53-R167H to model human germline, or spontaneous somatic p53 mutation. This work represents the first inactivation and mutation of the gatekeeper tumor suppressor gene TP53 in a non-rodent mammal.
Collapse
Affiliation(s)
- Simon Leuchs
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - Anja Saalfrank
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - Claudia Merkl
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - Tatiana Flisikowska
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - Marlene Edlinger
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - Marina Durkovic
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - Nousin Rezaei
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - Mayuko Kurome
- Chair of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, Oberschleissheim, Germany
| | - Valeri Zakhartchenko
- Chair of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, Oberschleissheim, Germany
| | - Barbara Kessler
- Chair of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, Oberschleissheim, Germany
| | | | - Alexander Kind
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - Eckhard Wolf
- Chair of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität München, Oberschleissheim, Germany
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
- * E-mail:
| |
Collapse
|
25
|
Kumar S, Mokhtari RB, Yeger H, Baruchel S. Preclinical models for pediatric solid tumor drug discovery: current trends, challenges and the scopes for improvement. Expert Opin Drug Discov 2012; 7:1093-106. [DOI: 10.1517/17460441.2012.722077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
26
|
McCoy A, Besch-Williford CL, Franklin CL, Weinstein EJ, Cui X. Creation and preliminary characterization of a Tp53 knockout rat. Dis Model Mech 2012; 6:269-78. [PMID: 22917926 PMCID: PMC3529357 DOI: 10.1242/dmm.009704] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The tumor suppressor TP53 plays a crucial role in cancer biology, and the TP53 gene is the most mutated gene in human cancer. Trp53 knockout mouse models have been widely used in cancer etiology studies and in search for a cure of cancer with some limitations that other model organisms might help overcome. Via pronuclear microinjection of zinc finger nucleases (ZFNs), we created a Tp53 knockout rat that contains an 11-bp deletion in exon 3, resulting in a frameshift and premature terminations in the open reading frame. In cohorts of 25 homozygous (Tp53Δ11/Δ11), 37 heterozygous (Tp53Δ11/+) and 30 wild-type rats, the Tp53Δ11/Δ11 rats lived an average of 126 days before death or removal from study because of clinical signs of abnormality or formation of tumors. Half of Tp53Δ11/+ were removed from study by 1 year of age because of tumor formation. Both Tp53Δ11/+ and Tp53Δ11/Δ11 rats developed a wide spectrum of tumors, most commonly sarcomas. Interestingly, there was a strikingly high incidence of brain lesions, especially in Tp53Δ11/Δ11 animals. We believe that this mutant rat line will be useful in studying cancer types rarely observed in mice and in carcinogenicity assays for drug development.
Collapse
Affiliation(s)
- Aaron McCoy
- Sigma Advanced Genetic Engineering Labs, Sigma-Aldrich Corporation, St Louis, MO 63146, USA
| | | | | | | | | |
Collapse
|
27
|
Yan HX, Wu HP, Ashton C, Tong C, Ying QL. Rats deficient for p53 are susceptible to spontaneous and carcinogen-induced tumorigenesis. Carcinogenesis 2012; 33:2001-5. [PMID: 22791818 DOI: 10.1093/carcin/bgs238] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The p53 tumor suppressor gene is highly mutated in human cancers. Individuals who inherit one p53 mutant allele are susceptible to a wide range of tumor types, including breast cancer and sarcoma. We recently generated p53 knockout rats through gene targeting in embryonic stem cells. Here we show that rats homozygous for the null allele are prone to early onset spontaneous sarcomas and lymphoma with high incidence of metastases. Heterozygous rats are also highly predisposed to cancer, but with a delayed onset and a wider spectrum of tumor types compared with homozygotes. Importantly, up to 20% of female heterozygotes developed breast cancer and about 70% of the tumors were positive for estrogen receptor. Exposing p53-deficient rats to a low dose of the carcinogen diethylnitrosamine dramatically decreased the latency for sarcoma development and survival time compared with equivalently treated wild-type rats. These unique features make this knockout line a valuable model for investigating human malignancy and in vivo carcinogenicity of chemicals and therapeutic compounds.
Collapse
Affiliation(s)
- He-Xin Yan
- Department of Cell and Neurobiology, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | |
Collapse
|