151
|
Dance-Barnes ST, Kock ND, Floyd HS, Moore JE, Mosley LJ, D'Agostino RB, Pettenati MJ, Miller MS. Effects of mutant human Ki-ras(G12C) gene dosage on murine lung tumorigenesis and signaling to its downstream effectors. Toxicol Appl Pharmacol 2008; 231:77-84. [PMID: 18565564 PMCID: PMC2596665 DOI: 10.1016/j.taap.2008.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 04/08/2008] [Accepted: 04/18/2008] [Indexed: 11/16/2022]
Abstract
Studies in cell culture have suggested that the level of RAS expression can influence the transformation of cells and the signaling pathways stimulated by mutant RAS expression. However, the levels of RAS expression in vivo appear to be subject to feedback regulation, limiting the total amount of RAS protein that can be expressed. We utilized a bitransgenic mouse lung tumor model that expressed the human Ki-ras(G12C) allele in a tetracycline-inducible, lung-specific manner. Treatment for 12 months with 500 microg/ml of doxycycline (DOX) allowed for maximal expression of the human Ki-ras(G12C) allele in the lung, and resulted in the development of focal hyperplasia and adenomas. We determined if different levels of mutant RAS expression would influence the phenotype of the lung lesions. Treatment with 25, 100 and 500 microg/ml of DOX resulted in dose-dependent increases in transgene expression and tumor multiplicity. Microscopic analysis of the lungs of mice treated with the 25 microg/ml dose of DOX revealed infrequent foci of hyperplasia, whereas mice treated with the 100 and 500 microg/ml doses exhibited numerous hyperplastic foci and also adenomas. Immunohistochemical and RNA analysis of the downstream effector pathways demonstrated that different levels of mutant RAS transgene expression resulted in differences in the expression and/or phosphorylation of specific signaling molecules. Our results suggest that the molecular alterations driving tumorigenesis may differ at different levels of mutant Ki-ras(G12C) expression, and this should be taken into consideration when inducible transgene systems are utilized to promote tumorigenesis in mouse models.
Collapse
Affiliation(s)
- Stephanie T Dance-Barnes
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
152
|
Hong HHL, Ton TVT, Kim Y, Wakamatsu N, Clayton NP, Chan PC, Sills RC, Lahousse SA. Genetic alterations in K-ras and p53 cancer genes in lung neoplasms from B6C3F1 mice exposed to cumene. Toxicol Pathol 2008; 36:720-6. [PMID: 18648094 PMCID: PMC2803761 DOI: 10.1177/0192623308320280] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The incidences of alveolar/bronchiolar adenomas and carcinomas in cumene-treated B6C3F1 mice were significantly greater than those of the control animals. We evaluated these lung neoplasms for point mutations in the K-ras and p53 genes that are often mutated in humans. K-ras and p53 mutations were detected by cycle sequencing of PCR-amplified DNA isolated from paraffin-embedded neoplasms. K-ras mutations were detected in 87% of cumene-induced lung neoplasms, and the predominant mutations were exon 1 codon 12 G to T transversions and exon 2 codon 61 A to G transitions. P53 protein expression was detected by immunohistochemistry in 56% of cumene-induced neoplasms, and mutations were detected in 52% of neoplasms. The predominant mutations were exon 5, codon 155 G to A transitions, and codon 133 C to T transitions. No p53 mutations and one of seven (14%) K-ras mutations were detected in spontaneous neoplasms. Cumene-induced lung carcinomas showed loss of heterozygosity (LOH) on chromosome 4 near the p16 gene (13%) and on chromosome 6 near the K-ras gene (12%). No LOH was observed in spontaneous carcinomas or normal lung tissues examined. The pattern of mutations identified in the lung tumors suggests that DNA damage and genomic instability may be contributing factors to the mutation profile and development of lung cancer in mice exposed to cumene.
Collapse
Affiliation(s)
- Hue-Hua L Hong
- 1 Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | | | | | | | | | |
Collapse
|
153
|
Patek CE, Arends MJ, Rose L, Luo F, Walker M, Devenney PS, Berry RL, Lawrence NJ, Ridgway RA, Sansom OJ, Hooper ML. The pro-apoptotic K-Ras 4A proto-oncoprotein does not affect tumorigenesis in the ApcMin/+ mouse small intestine. BMC Gastroenterol 2008; 8:24. [PMID: 18554389 PMCID: PMC2442095 DOI: 10.1186/1471-230x-8-24] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Accepted: 06/13/2008] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alterations in gene splicing occur in human sporadic colorectal cancer (CRC) and may contribute to tumour progression. The K-ras proto-oncogene encodes two splice variants, K-ras 4A and 4B, and K-ras activating mutations which jointly affect both isoforms are prevalent in CRC. Past studies have established that splicing of both the K-ras oncogene and proto-oncogene is altered in CRC in favour of K-ras 4B. The present study addressed whether the K-Ras 4A proto-oncoprotein can suppress tumour development in the absence of its oncogenic allele, utilising the ApcMin/+ (Min) mouse that spontaneously develops intestinal tumours that do not harbour K-ras activating mutations, and the K-rastmDelta4A/tmDelta4A mouse that can express the K-ras 4B splice variant only. By this means tumorigenesis in the small intestine was compared between ApcMin/+, K-ras+/+ and ApcMin/+, K-rastmDelta4A/tmDelta4A mice that can, and cannot, express the K-ras 4A proto-oncoprotein respectively. METHODS The relative levels of expression of the K-ras splice variants in normal small intestine and small intestinal tumours were quantified by real-time RT-qPCR analysis. Inbred (C57BL/6) ApcMin/+, K-ras+/+ and ApcMin/+, K-rastmDelta4A/tmDelta4A mice were generated and the genotypes confirmed by PCR analysis. Survival of stocks was compared by the Mantel-Haenszel test, and tumour number and area compared by Student's t-test in outwardly healthy mice at approximately 106 and 152 days of age. DNA sequencing of codons 12, 13 and 61 was performed to confirm the intestinal tumours did not harbour a K-ras activating mutation. RESULTS The K-ras 4A transcript accounted for about 50% of K-ras expressed in the small intestine of both wild-type and Min mice. Tumours in the small intestine of Min mice showed increased levels of K-ras 4B transcript expression, but no appreciable change in K-ras 4A transcript levels. No K-ras activating mutations were detected in 27 intestinal tumours derived from Min and compound mutant Min mice. K-Ras 4A deficiency did not affect mouse survival, or tumour number, size or histopathology. CONCLUSION The K-Ras 4A proto-oncoprotein does not exhibit tumour suppressor activity in the small intestine, even though the K-ras 4A/4B ratio is reduced in adenomas lacking K-ras activating mutations.
Collapse
Affiliation(s)
- Charles E Patek
- Sir Alastair Currie Cancer Research UK Laboratories, Molecular Medicine Centre, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Mark J Arends
- Department of Pathology, The University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 2QQ, UK
| | - Lorraine Rose
- Sir Alastair Currie Cancer Research UK Laboratories, Molecular Medicine Centre, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
- Rheumatic Diseases Unit, Molecular Medicine Centre, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Feijun Luo
- Department of Pathology, The University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 2QQ, UK
| | - Marion Walker
- Sir Alastair Currie Cancer Research UK Laboratories, Molecular Medicine Centre, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
- Division of Oncology, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, ,EH4 2XU, UK
| | - Paul S Devenney
- Sir Alastair Currie Cancer Research UK Laboratories, Molecular Medicine Centre, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
- Comparative and Developmental Genetics, MRC Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Rachel L Berry
- Sir Alastair Currie Cancer Research UK Laboratories, Molecular Medicine Centre, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
- Comparative and Developmental Genetics, MRC Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Nicola J Lawrence
- Sir Alastair Currie Cancer Research UK Laboratories, Molecular Medicine Centre, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
- Department of Genetics, Erasmus University Medical Centre, Dr. Molewaterplein, Rotterdam, 3015 GE, The Netherlands
| | - Rachel A Ridgway
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Owen J Sansom
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Martin L Hooper
- Sir Alastair Currie Cancer Research UK Laboratories, Molecular Medicine Centre, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| |
Collapse
|
154
|
Patek CE, Arends MJ, Wallace WAH, Luo F, Hagan S, Brownstein DG, Rose L, Devenney PS, Walker M, Plowman SJ, Berry RL, Kolch W, Sansom OJ, Harrison DJ, Hooper ML. Mutationally activated K-ras 4A and 4B both mediate lung carcinogenesis. Exp Cell Res 2008; 314:1105-1114. [PMID: 18062963 DOI: 10.1016/j.yexcr.2007.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 11/08/2007] [Accepted: 11/08/2007] [Indexed: 12/30/2022]
Abstract
To examine the roles of endogenous K-ras 4A and K-ras 4B splice variants in tumorigenesis, murine lung carcinogenesis was induced by N-methyl-N-nitrosourea (MNU), which causes a K-ras mutation (G12D) that jointly affects both isoforms. Compared with age-matched K-ras(tmDelta4A/-) mice (where tumours can express mutationally activated K-ras 4B only), tumour number and size were significantly higher in K-ras(+/-) mice (where tumours can also express mutationally activated K-ras 4A), and significantly lower in K-ras(tmDelta4A/tmDelta4A) mice (where tumours can express both wild-type and activated K-ras 4B). MNU induced significantly more, and larger, tumours in wild-type than K-ras(tmDelta4A/tmDelta4A) mice which differ in that only tumours in wild-type mice can express wild-type and activated K-ras 4A. Lung tumours in all genotypes were predominantly papillary adenomas, and tumours from K-ras(+/-) and K-ras(tmDelta4A/-) mice exhibited phospho-Erk1/2 and phospho-Akt staining. Hence (1) mutationally activated K-ras 4B is sufficient to activate the Raf/MEK/ERK(MAPK) and PI3-K/Akt pathways, and initiate lung tumorigenesis, (2) when expressed with activated K-ras 4B, mutationally activated K-ras 4A further promotes lung tumour formation and growth (both in the presence and absence of its wild-type isoform) but does not affect either tumour pathology or progression, and (3) wild-type K-ras 4B, either directly or indirectly, reduces tumour number and size.
Collapse
Affiliation(s)
- Charles E Patek
- Sir Alastair Currie Cancer Research UK Laboratories, Molecular Medicine Centre, The University of Edinburgh, Western General Hospital, Edinburgh, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Abstract
The retinoblastoma tumor suppressor gene, Rb, and the ras proto-oncogenes regulate various cellular processes, including differentiation and proliferation. Rb and ras genetically interact to positively influence differentiation in the mouse. This genetic interaction between Rb and ras also affects tumor development, either positively or negatively depending on cell type. Loss of one or two N-ras alleles allows medullary thyroid (C cell) adenomas occurring in Rb heterozygous mice to progress to metastatic carcinomas, an event associated with C cells displaying a less-differentiated phenotype. Here, we discuss the genetic interaction between Rb and ras and the development of a mouse model of medullary thyroid carcinoma.
Collapse
Affiliation(s)
- Chiaki Takahashi
- The 21st Century Center of Excellence Program, Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | |
Collapse
|
156
|
Agudo-Ibáñez L, Núñez F, Calvo F, Berenjeno IM, Bustelo XR, Crespo P. Transcriptomal profiling of site-specific Ras signals. Cell Signal 2007; 19:2264-76. [PMID: 17714917 PMCID: PMC2085357 DOI: 10.1016/j.cellsig.2007.06.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Accepted: 06/19/2007] [Indexed: 11/30/2022]
Abstract
Ras proteins are distributed in distinct plasma-membrane microdomains and endomembranes. The biochemical signals generated by Ras therein differ qualitatively and quantitatively, but the extent to which this spatial variability impacts on the genetic program switched-on by Ras is unknown. We have used microarray technology to identify the transcriptional targets of localization-specific Ras subsignals in NIH3T3 cells expressing H-RasV12 selectively tethered to distinct cellular microenvironments. We report that the transcriptomes resulting from site-specific Ras activation show a significant overlap. However, distinct genetic signatures can also be found for each of the Ras subsignals. Our analyses unveil 121 genes uniquely regulated by Ras signals emanating from plasma-membrane microdomains. Interestingly, not a single gene is specifically controlled by lipid raft-anchored Ras. Furthermore, only 9 genes are exclusive for Ras signals from endomembranes. Also, we have identified 31 genes common to the site-specific Ras subsignals capable of inducing cellular transformation. Among these are the genes coding for Vitamin D receptor and for p120-GAP and we have assessed their impact in Ras-induced transformation. Overall, this report reveals the complexity and variability of the different genetic programs orchestrated by Ras from its main sublocalizations.
Collapse
Affiliation(s)
- Lorena Agudo-Ibáñez
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC), Departamento de Biología Molecular, Unidad de Biomedicina, CSIC-Universidad de Cantabria, Santander, E-39011, Spain
| | - Fátima Núñez
- Centro de Investigación del Cancer, CSIC-Universidad de Salamanca, Salamanca E-37007, Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), CSIC-Universidad de Salamanca, Salamanca E-37007, Spain
| | - Fernando Calvo
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC), Departamento de Biología Molecular, Unidad de Biomedicina, CSIC-Universidad de Cantabria, Santander, E-39011, Spain
| | - Inmaculada M. Berenjeno
- Centro de Investigación del Cancer, CSIC-Universidad de Salamanca, Salamanca E-37007, Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), CSIC-Universidad de Salamanca, Salamanca E-37007, Spain
| | - Xosé R. Bustelo
- Centro de Investigación del Cancer, CSIC-Universidad de Salamanca, Salamanca E-37007, Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), CSIC-Universidad de Salamanca, Salamanca E-37007, Spain
| | - Piero Crespo
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC), Departamento de Biología Molecular, Unidad de Biomedicina, CSIC-Universidad de Cantabria, Santander, E-39011, Spain
| |
Collapse
|
157
|
Noutsopoulos D, Markopoulos G, Koliou M, Dova L, Vartholomatos G, Kolettas E, Tzavaras T. Vanadium Induces VL30 Retrotransposition at an Unusually High Level: A Possible Carcinogenesis Mechanism. J Mol Biol 2007; 374:80-90. [DOI: 10.1016/j.jmb.2007.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Revised: 08/14/2007] [Accepted: 09/04/2007] [Indexed: 10/22/2022]
|
158
|
Affiliation(s)
- Ernesto Diaz-Flores
- Department of Pediatrics, University of California, San Francisco, California 94143, USA
| | | |
Collapse
|
159
|
Fotiadou PP, Takahashi C, Rajabi HN, Ewen ME. Wild-type NRas and KRas perform distinct functions during transformation. Mol Cell Biol 2007; 27:6742-55. [PMID: 17636015 PMCID: PMC2099215 DOI: 10.1128/mcb.00234-07] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 03/16/2007] [Accepted: 07/09/2007] [Indexed: 12/30/2022] Open
Abstract
The ras proto-oncogenes, of which there are four isoforms, are molecular switches that function in signal transduction pathways to control cell differentiation, proliferation, and survival. How the Ras isoforms orchestrate cellular processes that affect behavior is poorly understood. Further, why cells express two or more Ras isoforms is unknown. Here, using a genetically defined system, we show that the presence of both wild-type KRas and NRas isoforms is required for transformation because they perform distinct nonoverlapping functions: wild-type NRas regulates adhesion, and KRas coordinates motility. Remarkably, we find that Ras isoforms achieve functional specificity by engaging different signaling pathways to affect the same cellular processes, thereby coordinating cellular outcome. Although we find that signaling from both isoforms intersects in actin and microtubule cytoskeletons, our results suggest that KRas signals through Akt and Cdc42 while NRas signals through Raf and RhoA. Our analyses suggest a previously unappreciated convergence of different Ras isoforms on the dynamics of the processes involved in transformation.
Collapse
Affiliation(s)
- Poppy P Fotiadou
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
160
|
Li H, Cao HF, Li Y, Zhu ML, Wan J. Changes in gene-expression profiles of colon carcinoma cells induced by wild type K-ras2. World J Gastroenterol 2007; 13:4620-5. [PMID: 17729418 PMCID: PMC4611839 DOI: 10.3748/wjg.v13.i34.4620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To further elucidate the possible molecular biological activity of wild type K-ras2 gene by detecting changes in wild type K-ras2 gene-induced gene-expression profiles of colon carcinoma cells using cDNA microarray techniques.
METHODS: Total RNA was isolated from peripheral blood of health volunteers. Reverse transcription of RNA and polymerase chain reaction were used to synthesize wild type K-ras2 cDNA. K-ras2 cDNA fragment was cloned into a T easy vector and sequenced. A eukaryotic expression vector pCI-neo-K-ras2 was constructed and transfected to Caco2 cell line using the liposome method. Finally, mRNA was isolated, reverse-transcribed to cDNA from pCI-neo-K-ras2 or pCI-neo blank vector-transfected Caco cells, and analyzed by cDNA microarray assay.
RESULTS: Restriction enzyme analysis and DNA sequencing verified that the constructed expression vector was accurate. High-quality RNA was extracted and reverse transcribed to cDNA for microarray assay. Among the 135 genes, the expression was up-regulated in 24 and down-regulated in 121. All these differentially expressed genes were related to cell proliferation, differentiation, apoptosis and signal transduction.
CONCLUSION: Differentially expressed genes can be successfully screened from wild type K-ras2-transfected colon carcinoma cells using microarray techniques. The results of our study suggest that wild type K-ras2 is related to the negative regulation of cell proliferation, metabolism and transcriptional control, and provide new clues to the further elucidation of its possible biological activity.
Collapse
Affiliation(s)
- Hong Li
- Inpatient Department of Special Need Treatment, General Hospital of Chinese PLA, Beijing, China
| | | | | | | | | |
Collapse
|
161
|
Vimalachandran D, Ghaneh P, Costello E, Neoptolemos JP. Genetics and prevention of pancreatic cancer. Cancer Control 2007; 11:6-14. [PMID: 14749618 DOI: 10.1177/107327480401100102] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Pancreatic cancer is an aggressive disease with a poor prognosis. Hereditary factors have been reported in up to 10% of cases of pancreatic cancer. The clinical characteristics and genetic abnormalities have been identified for a proportion of this high-risk group, and the development of preventive strategies for these individuals is now a primary goal of cancer clinicians. METHODS A review of the current literature regarding the genetics, screening, and prevention of pancreatic cancer and its precursor lesions was undertaken. RESULTS Risk factors for pancreatic cancer include smoking, chronic pancreatitis, and a genetic predisposition. The role of diabetes or a diet high in fat or meat remains unclear. The genetic mutations that accompany pancreatic cancer appear to occur in a temporal sequence, beginning in the earliest of precursor lesions. These mutations are detectable in pancreatic juice and, in conjunction with imaging, form the basis of screening programs for high-risk individuals. Not all precursor lesions will undergo malignant transformation, and testing is currently limited in its ability to determine which lesions will undergo transformation. CONCLUSIONS Avoiding tobacco smoking and minimizing risk factors associated with chronic pancreatitis are recommended to reduce the risk of pancreatic cancer. Individuals with a high-risk genetic background require counseling, genetic testing if appropriate (BRCA2 mutation or p16INK4A inactivity) and secondary screening for pancreatic cancer in specialist centers. Risk stratification will improve as more genetic abnormalities causing pancreatic cancer are defined.
Collapse
Affiliation(s)
- Dale Vimalachandran
- Department of Surgery, University of Liverpool, Royal Liverpool University Hospital, United Kingdom
| | | | | | | |
Collapse
|
162
|
Abstract
Animal models of cancer provide an alternative means to determine the causes of and treatments for malignancy, thus representing a resource of immense potential for cancer medicine. The sophistication of modelling cancer in mice has increased to the extent that investigators can both observe and manipulate a complex disease process in a manner impossible to perform in patients. However, owing to limitations in model design and technology development, and the surprising underuse of existing models, only now are we realising the full potential of mouse models of cancer and what new approaches are needed to derive the maximum value for cancer patients from this investment.
Collapse
|
163
|
Vantaggiato C, Formentini I, Bondanza A, Bonini C, Naldini L, Brambilla R. ERK1 and ERK2 mitogen-activated protein kinases affect Ras-dependent cell signaling differentially. J Biol 2007; 5:14. [PMID: 16805921 PMCID: PMC1781522 DOI: 10.1186/jbiol38] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2005] [Revised: 02/17/2006] [Accepted: 04/06/2006] [Indexed: 12/30/2022] Open
Abstract
Background The mitogen-activated protein (MAP) kinases p44ERK1 and p42ERK2 are crucial components of the regulatory machinery underlying normal and malignant cell proliferation. A currently accepted model maintains that ERK1 and ERK2 are regulated similarly and contribute to intracellular signaling by phosphorylating a largely common subset of substrates, both in the cytosol and in the nucleus. Results Here, we show that ablation of ERK1 in mouse embryo fibroblasts and NIH 3T3 cells by gene targeting and RNA interference results in an enhancement of ERK2-dependent signaling and in a significant growth advantage. By contrast, knockdown of ERK2 almost completely abolishes normal and Ras-dependent cell proliferation. Ectopic expression of ERK1 but not of ERK2 in NIH 3T3 cells inhibits oncogenic Ras-mediated proliferation and colony formation. These phenotypes are independent of the kinase activity of ERK1, as expression of a catalytically inactive form of ERK1 is equally effective. Finally, ectopic expression of ERK1 but not ERK2 is sufficient to attenuate Ras-dependent tumor formation in nude mice. Conclusion These results reveal an unexpected interplay between ERK1 and ERK2 in transducing Ras-dependent cell signaling and proliferation. Whereas ERK2 seems to have a positive role in controlling normal and Ras-dependent cell proliferation, ERK1 probably affects the overall signaling output of the cell by antagonizing ERK2 activity.
Collapse
Affiliation(s)
- Chiara Vantaggiato
- Istituto Scientifico San Raffaele and Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy
- Current address: Istituto Scientifico E. Medea, 23848 Bosisio Parini, Italy
| | - Ivan Formentini
- Istituto Scientifico San Raffaele and Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | - Attilio Bondanza
- Istituto Scientifico San Raffaele and Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | - Chiara Bonini
- Istituto Scientifico San Raffaele and Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | - Luigi Naldini
- Istituto Scientifico San Raffaele and Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | - Riccardo Brambilla
- Istituto Scientifico San Raffaele and Università Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| |
Collapse
|
164
|
Karow A, Steinemann D, Göhring G, Hasle H, Greiner J, Harila-Saari A, Flotho C, Zenker M, Schlegelberger B, Niemeyer CM, Kratz CP. Clonal duplication of a germline PTPN11 mutation due to acquired uniparental disomy in acute lymphoblastic leukemia blasts from a patient with Noonan syndrome. Leukemia 2007; 21:1303-5. [PMID: 17361219 DOI: 10.1038/sj.leu.2404651] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
165
|
Izeradjene K, Combs C, Best M, Gopinathan A, Wagner A, Grady WM, Deng CX, Hruban RH, Adsay NV, Tuveson DA, Hingorani SR. Kras(G12D) and Smad4/Dpc4 haploinsufficiency cooperate to induce mucinous cystic neoplasms and invasive adenocarcinoma of the pancreas. Cancer Cell 2007; 11:229-43. [PMID: 17349581 DOI: 10.1016/j.ccr.2007.01.017] [Citation(s) in RCA: 277] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2006] [Revised: 11/01/2006] [Accepted: 01/19/2007] [Indexed: 01/08/2023]
Abstract
Oncogenic Kras initiates pancreatic tumorigenesis, while subsequent genetic events shape the resultant disease. We show here that concomitant expression of Kras(G12D) and haploinsufficiency of the Smad4/Dpc4 tumor suppressor gene engenders a distinct class of pancreatic tumors, mucinous cystic neoplasms (MCNs), which culminate in invasive ductal adenocarcinomas. Disease evolves along a progression scheme analogous to, but distinct from, the classical PanIN-to-ductal adenocarcinoma sequence, and also portends a markedly different prognosis. Progression of MCNs is accompanied by LOH of Dpc4 and mutation of either p53 or p16. Thus, these distinct phenotypic routes to invasive adenocarcinoma nevertheless share the same overall mutational spectra. Our findings suggest that the sequence, as well as the context, in which these critical mutations are acquired helps determine the ensuing pathology.
Collapse
Affiliation(s)
- Kamel Izeradjene
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Li H, Cao HF, Wan J, Li Y, Zhu ML, Zhao P. Growth inhibitory effect of wild-type Kras2 gene on a colonic adenocarcinoma cell line. World J Gastroenterol 2007; 13:934-8. [PMID: 17352027 PMCID: PMC4065933 DOI: 10.3748/wjg.v13.i6.934] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the growth inhibitory effect of wild-type Kras2 gene on a colonic adenocarcinoma cell line Caco-2.
METHODS: Recombinant plasmid pCI-neo-Kras2 with wild type Kras2 open reading frame was constructed. The Caco-2 cells were transfected with either pCI-neo or pCI-neo-Kras2 using Lipofectamine 2000. The expression of wild type Kras2 was examined by Northern blot analysis. And the expression of wild type Kras2 protein was examined by Western blot analysis. The effects of wild-type Kras2 on cell proliferation were analyzed by monotetrazolium (MTT) assay, meanwhile analyses of cell cycle and spontaneous apoptosis rate were carried out by flow cytometry (FCM).
RESULTS: The plasmid of pCI-neo-Kras2 was successfully established. The growth rate of cells transfected with pCI-neo-Kras2 was significantly lower than the control cells transfected with the empty pCI-neo vector (P < 0.05). Cell cycle analysis revealed arrest of the pCI-neo-Kras2 transfected cells in G0/G1 phases, decreased DNA synthesis and decreased fractions of cells in S phase. The proliferative index of cells transfected with pCI-neo-Kras2 was decreased compared with the control cells (49.78% vs 64.21%),while the apoptotic rate of Caco-2 cells with stable Kras2 expression increased (0.30% vs 0.02%).
CONCLUSION: The wild-type Kras2 gene effectively inhibits the growth of the colonic adenocarcinoma cell line Caco-2.
Collapse
Affiliation(s)
- Hong Li
- Inpatient Department of Medical Health Center, Chinese PLA General Hospital, Beijing 100853, China
| | | | | | | | | | | |
Collapse
|
167
|
Yan Y, Tan Q, Wang Y, Wang D, Jin M, Gordon T, Lubet RA, You M. Enhanced lung tumor development in tobacco smoke-exposed p53 transgenic and Kras2 heterozygous deficient mice. Inhal Toxicol 2007; 19 Suppl 1:183-7. [PMID: 17886066 DOI: 10.1080/08958370701496160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
A/J mice bearing either a mutation in the p53 gene or a Kras2 heterozygous deficiency were investigated for their susceptibility to tobacco smoke-induced lung tumorigenesis. Transgenic mice and their wild-type littermates were exposed to mainstream tobacco smoke (MS) for 5 mo, followed by 4 mo of recovery in filtered air. In sham (filtered air) groups, p53 transgenic mice did not exhibit a higher tumor multiplicity but did exhibit larger tumors, with tumor load increased 3.6-fold, when compared with wild-type mice. With exposure to MS, tumor multiplicity was increased 60% but there was a strikingly increased tumor load (15.9-fold) in p53 transgenic mice. Increased tumor load (5.3-fold) but not tumor multiplicity was seen in MS-exposed Kras2 heterozygous deficient mice. Interestingly, MS exposure did not increase benzo[a]pyrene-induced lung tumorigenesis when MS exposure was initiated after BaP treatment. These results indicate that a p53 mutation or loss of a Kras2 allele increases susceptibility to MS-induced lung tumor development.
Collapse
Affiliation(s)
- Ying Yan
- Department of Surgery, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
168
|
Wakamatsu N, Devereux TR, Hong HHL, Sills RC. Overview of the molecular carcinogenesis of mouse lung tumor models of human lung cancer. Toxicol Pathol 2007; 35:75-80. [PMID: 17325975 PMCID: PMC2094362 DOI: 10.1080/01926230601059993] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Lung cancer is the leading cause of cancer death worldwide, and the need to develop better diagnostic techniques and therapies is urgent. Mouse models have been utilized for studying carcinogenesis of human lung cancers, and many of the major genetic alterations detected in human lung cancers have also been identified in mouse lung tumors. The importance of mouse models for understanding human lung carcinogenic processes and in developing early diagnostic techniques, preventive measures and therapies cannot be overstated. In this report, the major known molecular alterations in lung tumorigenesis of mice are reviewed and compared to those in humans.
Collapse
Affiliation(s)
- Nobuko Wakamatsu
- Laboratory of Experimental Pathology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | | | | | |
Collapse
|
169
|
Calvert RJ, Tepper S, Kammouni W, Anderson LM, Kritchevsky D. Elevated K-ras activity with cholestyramine and lovastatin, but not konjac mannan or niacin in lung--importance of mouse strain. Biochem Pharmacol 2006; 72:1749-55. [PMID: 17005160 PMCID: PMC1849957 DOI: 10.1016/j.bcp.2006.08.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Revised: 08/28/2006] [Accepted: 08/28/2006] [Indexed: 10/24/2022]
Abstract
Our previous work established that hypocholesterolemic agents altered K-ras intracellular localization in lung. Here, we examined K-ras activity to define further its potential importance in lung carcinogenesis. K-ras activity in lungs from male A/J, Swiss and C57BL/6 mice was examined. For 3 weeks, mice consumed either 2 or 4% cholestyramine (CS), 1% niacin, 5% konjac mannan (KM), or were injected with lovastatin 25mg/kg three or five times weekly (Lov-3X and Lov-5X). A pair-fed (PF) group was fed the same quantity of diet consumed by the Lov-5X mice to control for lower body weights in Lov-5X mice. After 3 weeks, serum cholesterol was assayed with a commercial kit. Activated K-ras protein from lung was affinity precipitated with a Raf-1 ras binding domain-glutathione-S-transferase fusion protein bound to glutathione-agarose beads, followed by Western blotting, K-ras antibody treatment, and chemiluminescent detection. Only KM reduced serum cholesterol (in two of three mouse strains). In C56BL/6 mice treated with Lov-3X, lung K-ras activity increased 1.8-fold versus control (p=0.009). In normal lung with wild-type K-ras, this would be expected to be associated with maintenance of differentiation. In A/J mice fed 4% CS, K-ras activity increased 2.1-fold (p=0.02), which might be responsible for the reported enhancement of carcinogenesis in carcinogen-treated rats fed CS. KM feeding and PF treatment had no significant effects on K-ras activity. These data are consistent with the concept that K-ras in lung has an oncogenic function when mutated, but may act as a tumor suppressor when wild-type.
Collapse
Affiliation(s)
- Richard J Calvert
- Division of Research and Applied Technology, Office of Nutritional Products, Labeling, and Dietary Supplements, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, College Park, MD 20740, USA.
| | | | | | | | | |
Collapse
|
170
|
BAC Consensus Conference, November 4???6, 2004: Epidemiology, Pathogenesis, and Preclinical Models. J Thorac Oncol 2006. [DOI: 10.1097/01243894-200611001-00002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
171
|
Christiani DC, Pao W, DeMartini JC, Linnoila RI, Malkinson AM, Onn A, Politi KA, Sharp M, Kim K. BAC Consensus Conference, November 4–6, 2004: Epidemiology, Pathogenesis, and Preclinical Models. J Thorac Oncol 2006. [DOI: 10.1016/s1556-0864(15)30002-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
172
|
|
173
|
Jang JW, Boxer RB, Chodosh LA. Isoform-specific ras activation and oncogene dependence during MYC- and Wnt-induced mammary tumorigenesis. Mol Cell Biol 2006; 26:8109-21. [PMID: 16908535 PMCID: PMC1636749 DOI: 10.1128/mcb.00404-06] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We have previously shown that c-MYC-induced mammary tumorigenesis in mice proceeds via a preferred secondary pathway involving spontaneous activating mutations in Kras2 (C. M. D'Cruz, E. J. Gunther, R. B. Boxer, J. L. Hartman, L. Sintasath, S. E. Moody, J. D. Cox, S. I. Ha, G. K. Belka, A. Golant, R. D. Cardiff, and L. A. Chodosh, Nat. Med. 7:235-239, 2001). In contrast, we now demonstrate that Wnt1-induced mammary tumorigenesis proceeds via a pathway that preferentially activates Hras1. In addition, we find that expression of oncogenic forms of Kras2 and Hras1 from their endogenous promoters has markedly different consequences for the progression of tumors to oncogene independence. Spontaneous activating Kras2 mutations occurring in either MYC- or Wnt1-induced tumors were strongly associated with oncogene-independent tumor growth following MYC or Wnt1 downregulation. In contrast, Hras1-mutant Wnt1-induced tumors consistently remained oncogene dependent. Additionally, Kras2-mutant tumors exhibited substantially higher levels of ras-GTP, phospho-Erk1/2, and phospho-Mek1/2 compared to Hras1-mutant tumors, suggesting the involvement of the ras/mitogen-activated protein kinase (MAPK) pathway in the acquisition of oncogene independence. Consistent with this, by use of carcinogen-induced ras mutations as well as knock-in mice harboring a latent activated Kras2 allele, we demonstrate that Kras2 activation strongly synergizes with both c-MYC and Wnt1 in mammary tumorigenesis and promotes the progression of tumors to oncogene independence. Together, our findings support a model for tumorigenesis in which c-MYC and Wnt1 select for the outgrowth of cells harboring mutations in specific ras isoforms and that these secondary mutations, in turn, determine the extent of ras/MAPK pathway activation and the potential for oncogene-independent growth.
Collapse
Affiliation(s)
- Joanne W Jang
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6160, USA
| | | | | |
Collapse
|
174
|
To MD, Perez-Losada J, Mao JH, Hsu J, Jacks T, Balmain A. A functional switch from lung cancer resistance to susceptibility at the Pas1 locus in Kras2LA2 mice. Nat Genet 2006; 38:926-30. [PMID: 16823377 PMCID: PMC4461000 DOI: 10.1038/ng1836] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Accepted: 06/06/2006] [Indexed: 11/09/2022]
Abstract
Pulmonary adenoma susceptibility 1 (Pas1) is the major mouse lung cancer susceptibility locus on chromosome 6 (ref. 1). Kras2 is a common target of somatic mutation in chemically induced mouse lung tumors and is a candidate Pas1 gene. M. spretus mice (SPRET/Ei) carry a Pas1 resistance haplotype for chemically induced lung tumors. We demonstrate that the SPRET/Ei Pas1 allele is switched from resistance to susceptibility by fixation of the parental origin of the mutant Kras2 allele. This switch correlates with low expression of endogenous Kras2 in SPRET/Ei. We propose that the Pas1 modifier effect is due to Kras2, and that a sensitive balance between the expression levels of wild-type and mutant alleles determines lung tumor susceptibility. These data demonstrate that cancer predisposition should also be considered in the context of somatic events and could have major implications for the design of human association studies to identify cancer susceptibility genes.
Collapse
Affiliation(s)
- Minh D To
- University of California San Francisco (UCSF) Comprehensive Cancer Center, San Francisco, California 94115, USA
| | | | | | | | | | | |
Collapse
|
175
|
Li H, Cao HF, Wan J, Li Y, Zhu ML, Han WD. Screening of genes differentially expressed in Caco2 cells transfected with wild type K-ras2 by cDNA microarray. Shijie Huaren Xiaohua Zazhi 2006; 14:1970-1976. [DOI: 10.11569/wcjd.v14.i20.1970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the genes differentially expressed in human colon carcinoma cell line Caco2 transfected with wild type K-ras2-expressing plasmid and further elucidate the potential molecular biological function of wild type K-ras2.
METHODS: Sequence specific primers were designed and synthesized, and the wild type K-ras2 DNA fragment was amplified with polymerase chain reaction (PCR) technique. The expressive vector of pCI-neo-K-ras2 was constructed by routine molecular biological methods. cDNA microarray technique was employed to detect the mRNA expression in Caco2 cells transfected with pCI-neo-K-ras2 and pCI-neo, respectively, using lipofectamine.
RESULTS: The expressive vector was constructed and confirmed by restriction enzyme digestion and DNA sequencing analysis. High quality RNA and cDNA were prepared and successful microarray screening was conducted. The scanning results indicated that among 8568 genes which were obtained from gene expression profile analysis, there were 135 different ones of which 121 were down-regulated and 24 were up-regulated in the wild type K-ras2-expressing Caco2 cells. These genes differentially regulated by wild type K-ras2 included human genes encoding proteins involved in cell signal transduction, cell apoptosis, cell proliferation and differentiation.
CONCLUSION: cDNA microarray is successfully used to screen the genes differentially expressed in wild type K-ras2-expressing Caco2 cells, and the alteration of expression profile induced by the wide-type K-ras2 suggested the negatively regulatory function of wild type K-ras2 for cell signal transduction, apoptosis, cell proliferation and differentiation.
Collapse
|
176
|
Albertson DG. Gene amplification in cancer. Trends Genet 2006; 22:447-55. [PMID: 16787682 DOI: 10.1016/j.tig.2006.06.007] [Citation(s) in RCA: 376] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Revised: 04/26/2006] [Accepted: 06/08/2006] [Indexed: 02/07/2023]
Abstract
Gene amplification is a copy number increase of a restricted region of a chromosome arm. It is prevalent in some tumors and is associated with overexpression of the amplified gene(s). Amplified DNA can be organized as extrachromosomal elements, as repeated units at a single locus or scattered throughout the genome. Common chromosomal fragile sites, defects in DNA replication or telomere dysfunction might promote amplification. Some regions of amplification are complex, yet elements of the pattern are reproduced in different tumor types. A genetic basis for amplification is suggested by its relative frequency in some tumor subtypes, and its occurrence in "early" preneoplastic lesions. Clinically, amplification has prognostic and diagnostic usefulness, and is a mechanism of acquired drug resistance.
Collapse
Affiliation(s)
- Donna G Albertson
- Cancer Research Institute and Comprehensive Cancer Center, University of California-San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
177
|
Ton TV, Hong HH, Devereux TR, Melnick RL, Sills RC, Kim Y. Evaluation of genetic alterations in cancer-related genes in lung and brain tumors from B6C3F1 mice exposed to 1,3-butadiene or chloroprene. Chem Biol Interact 2006; 166:112-20. [PMID: 16860786 DOI: 10.1016/j.cbi.2006.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2005] [Revised: 04/12/2006] [Accepted: 04/19/2006] [Indexed: 10/24/2022]
Abstract
1,3-Butadiene and chloroprene are multisite carcinogens in B6C3F1 mice with the strongest tumor response being the induction of lung neoplasms in females. Incidence of brain tumors in mice exposed to 1,3-butadiene was equivocal. This article reviews the efforts of our laboratory and others to uncover the mechanisms of butadiene and chloroprene induced lung and brain tumor responses in the B6C3F1 mouse. The formation of lung tumors by these chemicals involved mutations in the K-ras cancer gene and loss of heterozygosity in the region of K-ras on distal chromosome 6, while alterations in p53 and p16 were implicated in brain tumorigenesis.
Collapse
Affiliation(s)
- Thai-Vu Ton
- Environmental Toxicology Program and Environmental Carcinogenesis Program, National Institute of Environmental Health Sciences, MD B3-08, 111 Alexander Drive, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | |
Collapse
|
178
|
Wang Y, Zhang Z, Yao R, Jia D, Wang D, Lubet RA, You M. Prevention of lung cancer progression by bexarotene in mouse models. Oncogene 2006; 25:1320-9. [PMID: 16247446 DOI: 10.1038/sj.onc.1209180] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bexarotene (Targretin), is a synthetic high-affinity RXR receptor agonist with limited affinity for RAR receptors. Bexarotene has shown efficacy in a phase I/II trial of non-small-cell lung cancers. However, the chemopreventive efficacy of bexarotene has not been determined in mouse lung cancer models. In this study, we have investigated the ability of bexarotene to inhibit lung tumor progression in the mutant A/J mouse models with genetic alterations in p53 or K-ras, two of the most commonly altered genes in human lung tumorigenesis. Mice were administered vinyl carbamate (VC), a carcinogen, by a single intraperitoneal injection (i.p.) at 6 weeks of age. Bexarotene was given by gavage starting at 16 weeks after VC and was continued for 12 weeks. Although all mice developed lung tumors, only 7% of lung tumors were adenocarcinomas in wild-type mice, whereas 22 and 26% of lung tumors were adenocarcinomas in p53 transgenic or K-ras heterozygous deficient mice. Bexarotene inhibited both tumor multiplicity and tumor volume in mice of all three genotypes. Furthermore, bexarotene reduced the progression of adenoma to adenocarcinoma by approximately 50% in both p53(wt/wt)K-ras(ko/wt) and p53(wt/wt)K-ras(wt/wt) mice. Thus, bexarotene appears to be an effective preventive agent against lung tumor growth and progression.
Collapse
Affiliation(s)
- Y Wang
- Department of Surgery and the Siteman Cancer Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
179
|
Wang Y, Zhang Z, Lubet RA, You M. A mouse model for tumor progression of lung cancer in ras and p53 transgenic mice. Oncogene 2006; 25:1277-80. [PMID: 16247444 DOI: 10.1038/sj.onc.1209182] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although ras and p53 are the most commonly found oncogene and tumor suppressor gene, respectively, in human cancers, their collective roles in tumor progression have yet to be defined in animal models. Here, we demonstrated the synergistic effect between ras and p53 in promoting tumor progression during lung tumorigenesis using bitransgenic mice. Mice with a heterozygous knockout of K-ras (K-ras(wt/ko)) were mated to p53 transgenic mice (p53(val135/wt)) in lung tumorigenesis (K-ras(wt/ko) x p53(val135/wt)). F(1) mice exhibited a significant increase in lung tumor load (tumor multiplicity x tumor volume) when compared to those seen in either K-ras(wt/ko) mice or p53(val135/wt) mice alone. Furthermore, over 50% of the lung tumors were lung adenocarcinomas in bitransgenic mice compared to only 3% in wild-type mice. Alterations of ras and p53 appear to promote the development of lung adenocarcinomas. These results provide the in vivo experimental evidence of synergistic interactions of ras and p53 in lung tumor progression.
Collapse
Affiliation(s)
- Y Wang
- Department of Surgery and The Alvin J Siteman Cancer Center, Washington University School of Medicine, St Louis, MI 63110, USA
| | | | | | | |
Collapse
|
180
|
Allen T, van Tuyl M, Iyengar P, Jothy S, Post M, Tsao MS, Lobe CG. Grg1 acts as a lung-specific oncogene in a transgenic mouse model. Cancer Res 2006; 66:1294-301. [PMID: 16452182 DOI: 10.1158/0008-5472.can-05-1634] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Groucho proteins are transcriptional corepressors that are recruited to gene regulatory regions by numerous transcription factors. Long isoforms, such as Grg1, have all the domains of the prototype Drosophila Groucho. Short Groucho proteins, such as Grg5, have only the amino-terminal Q and G/P domains. We generated Grg1 and Grg5 transgenic mice and found that Grg1 overexpression induces lung adenocarcinoma, whereas Grg5 overexpression does not. Coexpression of Grg5 with Grg1 reduces tumor burden. Grg1 and Grg5 both diminish p53 protein levels; however, only Grg1 overexpression induces elevated levels of ErbB1 and ErbB2 receptor tyrosine kinases. The molecular and biological changes that accompany tumor progression in Grg1 transgenic mice closely reiterate events seen in human lung cancer. We also found that within a human lung tumor tissue array, a significant number of carcinomas overexpress Grg1/TLE1. Our data suggest that Grg1 overexpression contributes to malignancy in human lung cancers.
Collapse
Affiliation(s)
- Thaddeus Allen
- Molecular and Cellular Biology Division, Sunnybrook and Women's College Health Science Centre S-236, 2075 Bayview Avenue, Toronto, Ontario, Canada M4N 3M5
| | | | | | | | | | | | | |
Collapse
|
181
|
Wan J, Li H, Li Y, Zhu ML, Zhao P. Loss of heterozygosity of Kras2 gene on 12p12-13 in Chinese colon carcinoma patients. World J Gastroenterol 2006; 12:1033-7. [PMID: 16534842 PMCID: PMC4087893 DOI: 10.3748/wjg.v12.i7.1037] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the loss of heterozygosity (LOH) on 12p12-13 in Chinese colon carcinoma patients.
METHODS: DNA was extracted from 10 specimens of cancer tissue, 10 specimens of adjacent tissue and 10 specimens of normal tissue, respectively. LOH of Kras2 gene was analyzed by polymerase chain reaction (PCR) and denaturing polyacrylamide gel electrophoresis using 11 microsatellite markers on 12p-12-13.
RESULTS: LOH of Kras gene was detected at least on one marker of 12p-12-13 in 30% (3/10) of adjacent tissue specimens. The highest frequency of LOH was identified on D12S1034 in 28.57% (2/7) of adjacent tissue specimens. LOH was detected at least on one marker of 12p12-13 in 60% (6/10) of carcinoma tissue specimens, the most frequent LOH was found on D12S1034 and D12S1591 in 42.86% (3/7) of carcinoma tissue specimens. LOH was detected in 30% (3/10) of carcinoma tissue specimens, 30% (3/10) of adjacent tissue specimens, and no signal in 1% (1/0) carcinoma tissue specimen. The occurrence of LOH did not correlate with sex, age, tumor size and lymph node metastasis.
CONCLUSION: Genomic instability may occur on 12p-12-13 of Kras2 gene in the development and progression of colon carcinoma. The high LOH of Kras2 gene may directly influence the transcription and translation of wild type Kras2 gene.
Collapse
Affiliation(s)
- Jun Wan
- Department of Grastroenterology, General Hospital of the Chinese PLA, Beijing, China
| | | | | | | | | |
Collapse
|
182
|
Abstract
Krüppel-like factors are transcriptional regulators that influence several cellular functions, including proliferation. Recent studies have shown that one family member, KLF4, can function both as a tumour suppressor and an oncogene. The ability of KLF4 to affect the levels of expression of the cell-cycle regulator p21 seems to be involved, in that this protein might function as a switch that determines the outcome of KLF4 signalling. Is this role of p21 restricted to KLF4, or does p21 represent a nodal point for signals from multiple other factors with opposing functions in cancer?
Collapse
Affiliation(s)
- Benjamin D Rowland
- Division of Molecular Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | |
Collapse
|
183
|
Takahashi C, Contreras B, Iwanaga T, Takegami Y, Bakker A, Bronson RT, Noda M, Loda M, Hunt JL, Ewen ME. Nras loss induces metastatic conversion of Rb1-deficient neuroendocrine thyroid tumor. Nat Genet 2006; 38:118-23. [PMID: 16369533 DOI: 10.1038/ng1703] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2005] [Accepted: 10/13/2005] [Indexed: 12/28/2022]
Abstract
Mutations in the gene encoding the retinoblastoma tumor suppressor predispose humans and mice to tumor development. Here we have assessed the effect of Nras loss on tumor development in Rb1 heterozygous mice. Loss of one or two Nras alleles is shown to significantly reduce the severity of pituitary tumors arising in Rb1(+/-) animals by enhancing their differentiation. By contrast, C-cell thyroid adenomas occurring in Rb1(+/-) mice progress to metastatic medullary carcinomas after loss of Nras. In Rb1(+/-)Nras(+/-) animals, distant medullary thyroid carcinoma metastases are associated with loss of the remaining wild-type Nras allele. Loss of Nras in Rb1-deficient C cells results in elevated Ras homolog family A (RhoA) activity, and this is causally linked to the invasiveness and metastatic behavior of these cells. These findings suggest that the loss of the proto-oncogene Nras in certain cellular contexts can promote malignant tumor progression.
Collapse
Affiliation(s)
- Chiaki Takahashi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Calvert RJ, Kammouni W, Kikawa KD. Optimization of a nonradioactive method for consistent and sensitive determination of activated K-ras protein. Anal Biochem 2005; 343:283-92. [PMID: 16018961 DOI: 10.1016/j.ab.2005.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2005] [Revised: 05/25/2005] [Accepted: 06/03/2005] [Indexed: 11/17/2022]
Abstract
Accurate measurement of activity of wild-type K-ras protein is important due to its tumor suppressor action in tissues such as lung. A published method by Taylor and co-workers uses plasmid-containing Escherichia coli cells to produce a glutathione-S-transferase/raf-1 ras binding domain (GST-RBD) fusion protein attached to glutathione beads to isolate activated ras protein. We systematically optimized the method before use on lung tissues. Changing the GST-RBD protein induction temperature from the original 37 to 30 degrees C produced a consistently greater yield of fusion protein. To improve stability of the GST-RBD beads so as to perform large-scale experiments, 0.1% NaN(3) was added. NaN(3)-treated beads retained full affinity for at least 24 days. Sensitivity was improved by using a polyvinylidene difluoride membrane rather than nitrocellulose for immunoblotting. We also compared our GST-RBD beads with two commercial assay kits and found that our beads had both superior sensitivity and reduced variability. In summary, our modification of the GST-RBD affinity method to recover activated K-ras greatly increased the yield of fusion protein, prolonged the useful life of GST-RBD beads to at least 24 days, and enhanced detection sensitivity.
Collapse
Affiliation(s)
- Richard J Calvert
- U.S. Food and Drug Administration, Office of Nutritional Products, Labeling, and Dietary Supplements, Division of Research and Applied Technology, College Park, MD 20740, USA.
| | | | | |
Collapse
|
185
|
Zhang Z, Yao R, Li J, Wang Y, Boone CW, Lubet RA, You M. Induction of Invasive Mouse Skin Carcinomas in Transgenic Mice with Mutations in Both H-ras and p53. Mol Cancer Res 2005; 3:563-74. [PMID: 16254190 DOI: 10.1158/1541-7786.mcr-05-0144] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Synergistic interaction between H-ras and p53 were systematically examined during skin tumorigenesis. Concurrent expression of an activated H-ras gene and a mutant p53 gene was accomplished by crossing p53(Val135/wt) mice with TG.AC mice. Topical application to wild-type mice with benzo(a)pyrene (BaP) alone produced approximately 26% skin tumor incidence, whereas BaP treatment of p53(wt/wt)Hras(TG.AC/wt), p53(Val135/wt)Hras(wt/wt), and p53(Val135/wt)Hras(TG.AC/wt) mice produced a 75%, 77%, and 100% incidence of skin tumors, respectively. An average of 0.33 tumor per mouse was observed in wild-type (p53(wt/wt)Hras(wt/wt)) mice, whereas approximately 1.54, 1.96, and 3.08 tumors per mouse were seen in BaP-treated p53(wt/wt)Hras(TG.AC/wt), p53(Val135/wt)Hras(wt/wt), and p53(Val135/wt)Hras(TG.AC/wt) mice, respectively. The effects on total tumor volume were even more striking with 7-, 48-, and 588-fold increases in tumor volume compared with wild-type (p53(wt/wt)Hras(wt/wt)) in p53(wt/wt)Hras(TG.AC/wt), p53(Val135/wt)Hras(wt/wt), and p53(Val135/wt)Hras(TG.AC/wt) mice, respectively. Histopathologically, all tumors from p53(wt/wt)Hras(wt/wt) mice were either papillomas or well-differentiated squamous cell carcinomas, whereas the tumors in p53(wt/wt)Hras(TG.AC/wt), p53(Val135/wt)Hras(wt/wt), and p53(Val135/wt)Hras(TG.AC/wt) mice were principally squamous cell carcinomas with varying degree of invasiveness. Particularly, tumors in p53(Val135/wt)Hras(TG.AC/wt) mice exhibited the most rapid growth and the extreme form of tumor invasion. Microarray analysis revealed that dominant-negative p53 (Val135) and activated H-ras affected several cellular processes involved in tumorigenesis possibly through its effects on apoptosis, cell cycle arrest, and Ras-mitogen-activated protein kinase pathways. The present study provides the first in vivo evidence that a germ line p53 mutation and activated H-ras act synergistically to profoundly enhance tumor progression.
Collapse
Affiliation(s)
- Zhongqiu Zhang
- Department of Surgery and The Alvin J. Siteman Cancer Center, Washington University School of Medicine, 660 Euclid Avenue, Box 8109, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | |
Collapse
|
186
|
Affiliation(s)
- Onno Kranenburg
- Department of Surgery, University Medical Center Utrecht, Utrecht.
| |
Collapse
|
187
|
Abstract
The cellular Ras is known to play an important role in cellular proliferation mediated by growth factor receptor. Evidence also points to its role in growth arrest. Substantiated proof for growth-suppressive activity of wild-type Ras comes from studies that showed 1) loss of wild-type ras allele in tumors, 2) suppression of growth in cells transformed by oncogenic ras upon overexpression of wild-type Ras, and 3) up-regulation of Ras expression during postnatal development and following growth arrest of untransformed cells in culture. To understand the mechanism by which the wild-type Ras brings about these diverse actions, we evaluated its well-known role in actively proliferating cells and its less understood role in growth arrest. This led to the proposal that wild-type Ras in either GDP or GTP-bound state can antagonize the function of oncogenic Ras.-Singh, A., Sowjanya, A. P., Ramakrishna, G. The wild-type Ras: road ahead.
Collapse
Affiliation(s)
- Arvind Singh
- Centre for DNA Fingerprinting and Diagnostics, Nacharam, Hyderabad, Andhra Pradesh, India
| | | | | |
Collapse
|
188
|
Tanuma JI, Hirano M, Hirayama Y, Semba I, Ogawa K, Shisa H, Hiai H, Kitano M. Genetic predisposition to 4NQO-induced tongue carcinogenesis in the rat. Med Princ Pract 2005; 14:297-305. [PMID: 16103694 DOI: 10.1159/000086926] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2004] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE This study aims to elucidate the genetic basis of predisposition to 4-nitroquinoline 1-oxide (4NQO)-induced tongue cancers (TCs). MATERIALS AND METHODS We have reported that inbred Dark-Agouti (DA) strain rats were highly susceptible to 4NQO-induced TCs, whereas Wistar/Furth (WF) rats were resistant to tongue squamous cell carcinomas induced by oral administration of 4NQO. Using size and number of the tumours as quantitative parameters, responsible host loci were analysed by an interval mapping of F2 intercross of DA and WF given carcinogenic regimen. Also, loss of heterozygosity (LOH) at these loci was analysed in tongue cancers in (DA x WF) F1. RESULTS We identified and mapped 5 significant quantitative trait loci (QTL), the Tongue squamous cell carcinoma 1-5 (Tscc1-5), and several other suggestive QTL that determine susceptibility to 4NQO-induced TC. Study of TCs induced in (DA x WF)F1 rats revealed a high frequency of LOH in the chromosomal regions of Tscc2, 3, and 4 and also of suggestive QTL on chromosomes 5 and 6. The fact that LOH was found only in larger TCs indicates that LOH occurred in the process of tumour progression. In most LOH, the allele of the resistant WF strain was lost, suggesting that these loci may encode tumour suppressor genes. In larger TCs, in addition to LOH, point mutations and the methylation of possible candidate genes were accumulated. CONCLUSION These observations indicate that the 4NQO-induced TC in the rat is a multifactorial disease of a polygenic trait. This model will be useful to understand the complicated genetic basis of predisposition to oral cancers.
Collapse
Affiliation(s)
- Jun-ichi Tanuma
- Department of Oral Pathology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
189
|
Abstract
The Ras and Notch signaling pathways are used over and over again during development to control many different biological processes. Frequently, these two signaling pathways intersect to influence common processes, but sometimes they cooperate and sometimes they antagonize each other. The Caenorhabditis elegans vulva and the Drosophila eye are two classic paradigms for understanding how Ras and Notch affect cell fates, and how the two pathways work together to control biological pattern. Recent advances in these systems reveal some of the mechanisms by which Ras and Notch can interact. Similar types of interactions in mammals may be important for determining whether and how alterations in Ras or Notch lead to cancer.
Collapse
Affiliation(s)
- Meera V Sundaram
- Department of Genetics, University of Pennsylvania, Philadelphia, 19104, USA
| |
Collapse
|
190
|
Klampfer L, Swaby LA, Huang J, Sasazuki T, Shirasawa S, Augenlicht L. Oncogenic Ras increases sensitivity of colon cancer cells to 5-FU-induced apoptosis. Oncogene 2005; 24:3932-41. [PMID: 15856030 DOI: 10.1038/sj.onc.1208552] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Despite the fact that objective response rates to 5-FU are as low as 20%, 5-FU remains the most commonly used drug for the treatment of colorectal cancer. The lack of understanding of resistance to 5-FU, therefore, remains a significant impediment in maximizing its efficacy. We used intestinal epithelial cells with an inducible K-RasV12 to demonstrate that expression of oncogenic Ras promotes cell death upon 5-FU treatment. Accordingly, transient expression of the mutant RasV12, but not the WT Ras, enhanced 5-FU-induced apoptosis in 293T cells. Consistent with these data, we showed that targeted deletion of the mutant Ras allele in the HCT116 colon cancer cell line protected cells from 5-FU-induced apoptosis. Using isogenic colon cancer cell lines that differ only by the presence of the mutant Ras allele, HCT116 and Hke-3 cells, we demonstrated that signaling by oncogenic Ras promotes both accumulation of p53 and its phosphorylation on serine15 in response to 5-FU, a situation that favors apoptosis over growth arrest. However, despite the differential induction of p53 in HCT116 and Hke-3 cells, the expression of Puma, a gene with an important role in p53-dependent apoptosis, was not affected by Ras signaling. In contrast, we showed that Ras interferes with 5-FU-induced expression of gelsolin, a protein with known antiapoptotic activity. We ascertained the role of gelsolin in 5-FU-induced apoptosis by demonstrating that silencing of gelsolin expression through RNAi sensitized cells to 5-FU-induced apoptosis and that re-expression of gelsolin in cells harboring mutant Ras protected cells from 5-FU-induced apoptosis. These data therefore demonstrate that Ras mutations increase sensitivity to 5-FU-induced apoptosis at least in part through the negative regulation of gelsolin expression. Our data indicate that Ras mutations promote apoptosis in response to 5-FU treatment and imply that tumors with Ras mutations and/or reduced expression of gelsolin may show enhanced apoptosis in response to 5-FU also in vivo.
Collapse
Affiliation(s)
- Lidija Klampfer
- Albert Einstein Cancer Center, Montefiore Medical Center, Department of Oncology, Bronx, NY 10467, USA.
| | | | | | | | | | | |
Collapse
|
191
|
Germeshausen M, Schulze H, Kratz C, Wilkens L, Repp R, Shannon K, Welte K, Ballmaier M. An acquired G-CSF receptor mutation results in increased proliferation of CMML cells from a patient with severe congenital neutropenia. Leukemia 2005; 19:611-7. [PMID: 15729385 DOI: 10.1038/sj.leu.2403663] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Severe congenital neutropenia (CN) is characterized by a maturation arrest of myelopoiesis at the promyelocyte stage. Treatment with pharmacological doses of recombinant human granulocyte colony-stimulating factor (rh-G-CSF) stimulates neutrophil production and decreases the risk of major infectious complications. However, approximately 15% of CN patients develop myeloid malignancies that have been associated with somatic mutations in the G-CSF receptor (G-CSFR) and RAS genes as well as with acquired monosomy 7. We report a CN patient with chronic myelomonocytic leukemia (CMML) who never received rh-G-CSF. Molecular analysis demonstrated a somatic G-CSFR mutation (C2390T), which led to expression of a truncated G-CSFR protein in the CMML. Normal G-CSFR expression was unexpectedly absent in primary and cultured CMML. In addition, CMML cells showed monosomy 7 and an oncogenic NRAS mutation. In vitro culture revealed a G-CSF-dependent proliferation of CMML cells, which subsequently differentiated along the monocytic/macrophage lineage. Our results provide direct evidence for the in vivo expression of a truncated G-CSFR in leukemic cells, which emerged in the absence of rh-G-CSF treatment and transduces proliferative signals.
Collapse
Affiliation(s)
- M Germeshausen
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
192
|
Wang Y, Zhang Z, Lubet R, You M. Tobacco smoke-induced lung tumorigenesis in mutant A/J mice with alterations in K-ras, p53, or Ink4a/Arf. Oncogene 2005; 24:3042-9. [PMID: 15846305 DOI: 10.1038/sj.onc.1208390] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A/J mice with genetic alterations in K-ras, p53, or Ink4a/Arf were employed to investigate whether mice carrying these germline mutations would be susceptible to tobacco smoke-induced lung tumorigenesis. Transgenic mice of both genders and their wild-type littermates were exposed to environmental cigarette smoke for 6 months, followed by recovery in air for 5 months. A significant increase of lung tumor multiplicity was observed in K-ras, p53, or Ink4a/Arf mutant mice when compared with wild-type mice. Furthermore, an additive effect was observed between the mice with a mutant p53 transgene and an Ink4A/Arf deletion during tobacco smoke-induced lung tumorigenesis. Sequence analysis of the K-ras gene indicated that the mutations had occurred at either codon 12/13 or 61 in both spontaneously occurring (air control) and tobacco smoke-induced lung tumors. K-ras mutations were found in 62% of the tumors from air-control animals and 83% in those exposed to tobacco smoke. The mutation spectrum found in tumors from mice exposed to tobacco smoke is somewhat similar to that in tumors from air-control mice. In addition, we identified three novel mutations at codon 12: GGT (Gly) --> TTT (Phe), ATT (Ile), and CTT (Leu). These findings provide evidence that K-ras, p53, and Ink4a/Arf mutations play a role in tobacco smoke-related lung carcinogenesis. The similarity of the mutation spectra in the K-ras oncogene observed in tobacco smoke-induced tumors, as compared to spontaneous tumors, suggests that tobacco smoke enhances lung tumorigenesis primarily through promoting spontaneously occurring K-ras mutations.
Collapse
Affiliation(s)
- Yian Wang
- Department of Surgery, School of Medicine, Siteman Cancer Center, The Washington University in St Louis, 660 S Euclid Avenue, St Louis, MO, USA
| | | | | | | |
Collapse
|
193
|
Kaye FJ. A Curious Link Between Epidermal Growth Factor Receptor Amplification and Survival: Effect of “Allele Dilution” on Gefitinib Sensitivity? ACTA ACUST UNITED AC 2005; 97:621-3. [PMID: 15870427 DOI: 10.1093/jnci/dji127] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
194
|
Abstract
In recent years several new mouse models for lung cancer have been described. These include models for both non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC). Tumorigenesis in these conditional mouse tumor models can be initiated in adult mice through Cre-recombinase-induced activation of oncogenic mutations in a subset of the cells. They present a marked improvement over mouse models that depend on carcinogen induction of tumors. These models permit us to study the consecutive steps involved in initiation and progression and allow us to address questions like the cell of origin, and the role of cancer stem cells in the maintenance of these tumors. They now need to be validated as suitable preclinical models for intervention studies in which questions with respect to therapy response and resistance can be addressed.
Collapse
Affiliation(s)
- Ralph Meuwissen
- Division of Molecular Genetics and Center of Biomedical Genetics, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | | |
Collapse
|
195
|
Yao R, Wang Y, D'Agostini F, Izzotti A, Lubet RA, You M, De Flora S. K-ras mutations in lung tumors from p53 mutant mice exposed to cigarette smoke. Exp Lung Res 2005; 31:271-81. [PMID: 15824025 DOI: 10.1080/0190214059090386] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In this study, we used p53 transgenic mice to investigate whether mice carrying this germline mutation would be susceptible to tobacco smoke-induced lung tumorigenesis. We subjected male transgenic mice and their wild-type littermates to whole-body exposure to environmental cigarette smoke (ECS) for up to 9.5 months. K-ras gene expression was significantly increased, 28 days after ECS exposure, in the apparently healthy lung of p53 mutant mice. An increase of lung tumor incidence and multiplicity was observed in p53 transgenic mice after exposure to ECS for either 5 months, followed by recovery in air for 4.5 months, or 9.5 continuative months of exposure. Conversely, no tumorigenic effect was observed in their wild-type littermates. Sequence analysis of the K-ras gene indicated that mutations had occurred at codon 12, 13 or codon 61 in tumors both from the air control group and tobacco smoke treatment groups. K-ras mutations were found in 100 %, 100 % and 77 % of tumors from animals exposed to air, ECS for 5 months, followed by recovery in air for 4.5 months, and ECS for 9.5 continuative months, respectively. The K-ras mutations were seemingly not related to the p53 genotype of the animals or to ECS exposure. The mutation spectrum was similar in tumors from the different groups. An apparently higher incidence of K-ras codon 12 mutations in the 9.5 months ECS group was not statistically significant. These findings provide evidence that mice carrying a mutant p53 transgene appear to be more sensitive to ECS-induced lung tumors than the corresponding wild-type littermates. K-ras mutations seem to be independent of the p53 status but the early overexpression of this oncogene is related to the p53 status in ECS-exposed mice. These results suggest that tobacco smoke enhances lung tumorigenesis primarily through promoting spontaneously occurring K-ras mutations.
Collapse
Affiliation(s)
- Ruisheng Yao
- Department of Surgery and The Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | | | |
Collapse
|
196
|
Koch CA. Molecular pathogenesis of MEN2-associated tumors. Fam Cancer 2005; 4:3-7. [PMID: 15883703 DOI: 10.1007/s10689-004-7022-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2004] [Accepted: 11/30/2004] [Indexed: 10/25/2022]
Abstract
Although the gene responsible for multiple endocrine neoplasia type 2 (MEN2) was discovered many years ago, the exact mechanisms of tumor development in patients affected with RET germline mutations remain unknown. In vitro studies have certain pitfalls, one of which is the use of cell culture systems such as the NIH3T3 cells, in which RET usually is not expressed in contrast to the in vivo situation. Recent data suggest that an overrepresentation of mutant RET as a 'second hit' event might trigger tumorigenesis. However, alterations in other genes might contribute to this overrepresentation of RET or impact on MEN 2-related tumor development through completely different mechanisms and pathways. The final goal of further elucidating the natural history and pathogenesis of MEN2-related tumors should be the chance to offer patients with RET germline mutations an optimal cancer prevention (e.g. codon specific recommendations for prophylactic thyroidectomy) and treatment program, especially for metastatic medullary thyroid carcinoma for which presently no effective therapy other than surgery exists.
Collapse
Affiliation(s)
- Christian A Koch
- University of Leipzig, Philipp-Rosenthalstr. 27, 04103, Leipzig, Germany.
| |
Collapse
|
197
|
Diaz R, Lue J, Mathews J, Yoon A, Ahn D, Garcia-España A, Leonardi P, Vargas MP, Pellicer A. Inhibition of Ras oncogenic activity by Ras protooncogenes. Int J Cancer 2005; 113:241-8. [PMID: 15386411 DOI: 10.1002/ijc.20563] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Point mutations in ras genes have been found in a large number and wide variety of human tumors. These oncogenic Ras mutants are locked in an active GTP-bound state that leads to a constitutive and deregulated activation of Ras function. The dogma that ras oncogenes are dominant, whereby the mutation of a single allele in a cell will predispose the host cell to transformation regardless of the presence of the normal allele, is being challenged. We have seen that increasing amounts of Ras protooncogenes are able to inhibit the activity of the N-Ras oncogene in the activation of Elk in NIH 3T3 cells and in the formation of foci. We have been able to determine that the inhibitory effect is by competition between Ras protooncogenes and the N-Ras oncogene that occurs first at the effector level at the membranes, then at the processing level and lastly at the effector level in the cytosol. In addition, coexpression of the N-Ras protooncogene in thymic lymphomas induced by the N-Ras oncogene is associated with increased levels of p107, p130 and cyclin A and decreased levels of Rb. In the present report, we have shown that the N-Ras oncogene is not truly dominant over Ras protooncogenes and their competing activities might be depending on cellular context.
Collapse
Affiliation(s)
- Roberto Diaz
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Tommasi S, Dammann R, Zhang Z, Wang Y, Liu L, Tsark WM, Wilczynski SP, Li J, You M, Pfeifer GP. Tumor Susceptibility of Rassf1a Knockout Mice. Cancer Res 2005. [DOI: 10.1158/0008-5472.92.65.1] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Abstract
The human Ras association domain family 1 (RASSF1) gene is located at 3p21.3 in an area that is believed to harbor at least one important tumor suppressor gene. The two major isoforms of RASSF1, RASSF1A and RASSF1C, are distinguished by alternative NH2-terminal exons and the two transcripts initiate in two separate CpG islands. RASSF1A is one of the most frequently inactivated genes described thus far in human solid tumors. Inactivation of RASSF1A most commonly involves methylation of the promoter and CpG island associated with the RASSF1A isoform. In contrast, RASSF1C is almost never inactivated in tumors. Here, we have derived Rassf1a knockout mice in which exon 1-α of the Rassf1 gene was deleted, leading to specific loss of Rassf1a but not Rassf1c transcripts. Rassf1a-targeted mice were viable and fertile. Rassf1a−/− mice were prone to spontaneous tumorigenesis in advanced age (18–20 months). Whereas only two tumors developed in 48 wild-type mice, six tumors were found in 35 Rassf1a+/− mice (P < 0.05) and thirteen tumors were found in 41 Rassf1a−/− mice (P < 0.001). The tumors in Rassf1a-targeted mice included lung adenomas, lymphomas, and one breast adenocarcinoma. Rassf1a−/− and wild-type mice were treated with two chemical carcinogens, benzo(a)pyrene and urethane, to induce skin tumors and lung tumors, respectively. Rassf1a−/− and Rassf1a+/− mice showed increased tumor multiplicity and tumor size relative to control animals. The data are consistent with the tumor-suppressive role of Rassf1a, which may explain its frequent epigenetic inactivation in human tumors.
Collapse
Affiliation(s)
| | - Reinhard Dammann
- 3Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany; and
| | - Zhongqiu Zhang
- 4Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Yian Wang
- 4Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | | | | | - Sharon P. Wilczynski
- 2Department of Anatomic Pathology, City of Hope National Medical Center, Duarte, California
| | - Jie Li
- 4Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Ming You
- 4Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | | |
Collapse
|
199
|
De Vita G, Bauer L, da Costa VMC, De Felice M, Baratta MG, De Menna M, Di Lauro R. Dose-Dependent Inhibition of Thyroid Differentiation by RAS Oncogenes. Mol Endocrinol 2005; 19:76-89. [PMID: 15388794 DOI: 10.1210/me.2004-0172] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Activating mutations in RAS protooncogenes are associated with several different histotypes of thyroid cancer, including anaplastic thyroid carcinoma. The latter is the most aggressive cancer of the thyroid gland, showing little or no expression of the differentiated phenotype. Likewise, expression of viral RAS oncogenes in FRTL-5 rat thyroid cells mimics such loss of differentiation. We established FRTL-5 cell lines stably expressing constitutively active forms of RAS, either of viral (v-Ha-RAS or v-Ki-RAS) or cellular (H-RAS(V12)) origin and generated a tamoxifen-inducible RAS oncoprotein to analyze the timing of RAS effects on thyroid differentiation. In RAS-transformed FRTL-5 cells, we measured the expression of many thyroid-specific genes by real-time PCR and observed that a clear loss of differentiation was only obtained in the presence of high RAS oncogene expression. In contrast, TSH-independent growth appeared to be induced in the presence of both low and high levels of oncogenic RAS expression. We also showed that inhibition of differentiation is an early RAS-induced phenomenon. Finally, we demonstrated that only high doses of RAS oncogenes are able to inhibit the activity of Titf1 and Pax8, two transcription factors essential for the maintenance of thyroid differentiation, and that the homeodomain of Titf1 is a target of the inhibitory action of RAS. Our results represent the first evidence of a dose-dependent effect of RAS oncogenes on thyroid epithelial differentiation.
Collapse
Affiliation(s)
- Gabriella De Vita
- Stazione Zoologica Anton Dohrn, Villa Comunale 1, 80121 Napoli, Italy
| | | | | | | | | | | | | |
Collapse
|
200
|
Abstract
The revolution in cancer research can be summed up in a single sentence: cancer is, in essence, a genetic disease. In the last decade, many important genes responsible for the genesis of various cancers have been discovered, their mutations precisely identified, and the pathways through which they act characterized. The purposes of this review are to highlight examples of progress in these areas, indicate where knowledge is scarce and point out fertile grounds for future investigation.
Collapse
Affiliation(s)
- Bert Vogelstein
- Howard Hughes Medical Institute and The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University Medical Institutions, Baltimore, Maryland 21231, USA.
| | | |
Collapse
|