Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Pathol ; 239(2): 206-17, 2016 06.
Article in English | MEDLINE | ID: mdl-27174785

ABSTRACT

The Noonan syndrome (NS) is an autosomal dominant genetic disorder characterized by short stature, craniofacial dysmorphism, and congenital heart defects. A significant proportion of NS patients may also develop myeloproliferative disorders (MPDs), including juvenile myelomonocytic leukaemia (JMML). Surprisingly, scarce information is available in relation to other tumour types in these patients. We have previously developed and characterized a knock-in mouse model that carries one of the most frequent KRAS-NS-related mutations, the K-Ras(V14I) substitution, which recapitulates most of the alterations described in NS patients, including MPDs. The K-Ras(V14I) mutation is a mild activating K-Ras protein; thus, we have used this model to study tumour susceptibility in comparison with mice expressing the classical K-Ras(G12V) oncogene. Interestingly, our studies have shown that these mice display a generalized tumour predisposition and not just MPDs. In fact, we have observed that the K-Ras(V14I) mutation is capable of cooperating with the p16Ink4a/p19Arf and Trp53 tumour suppressors, as well as with other risk factors such as pancreatitis, thereby leading to a higher cancer incidence. In conclusion, our results illustrate that the K-Ras(V14I) activating protein is able to induce cancer, although at a much lower level than the classical K-Ras(G12V) oncogene, and that it can be significantly modulated by both genetic and non-genetic events. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Heart Defects, Congenital/genetics , Lung Neoplasms/genetics , Neoplasms/genetics , Noonan Syndrome/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Alleles , Amino Acid Substitution , Animals , Disease Models, Animal , Disease Susceptibility , Female , Genetic Carrier Screening , Heart Defects, Congenital/pathology , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mutation , Neoplasms/pathology , Noonan Syndrome/pathology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism
2.
Proc Natl Acad Sci U S A ; 111(46): 16395-400, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25359213

ABSTRACT

Noonan syndrome (NS) is an autosomal dominant genetic disorder characterized by short stature, craniofacial dysmorphism, and congenital heart defects. NS also is associated with a risk for developing myeloproliferative disorders (MPD), including juvenile myelomonocytic leukemia (JMML). Mutations responsible for NS occur in at least 11 different loci including KRAS. Here we describe a mouse model for NS induced by K-Ras(V14I), a recurrent KRAS mutation in NS patients. K-Ras(V14I)-mutant mice displayed multiple NS-associated developmental defects such as growth delay, craniofacial dysmorphia, cardiac defects, and hematologic abnormalities including a severe form of MPD that resembles human JMML. Homozygous animals had perinatal lethality whose penetrance varied with genetic background. Exposure of pregnant mothers to a MEK inhibitor rescued perinatal lethality and prevented craniofacial dysmorphia and cardiac defects. However, Mek inhibition was not sufficient to correct these defects when mice were treated after weaning. Interestingly, Mek inhibition did not correct the neoplastic MPD characteristic of these mutant mice, regardless of the timing at which the mice were treated, thus suggesting that MPD is driven by additional signaling pathways. These genetically engineered K-Ras(V14I)-mutant mice offer an experimental tool for studying the molecular mechanisms underlying the clinical manifestations of NS. Perhaps more importantly, they should be useful as a preclinical model to test new therapies aimed at preventing or ameliorating those deficits associated with this syndrome.


Subject(s)
Disease Models, Animal , Genes, ras , Mice, Mutant Strains , Mutation, Missense , Noonan Syndrome/genetics , Point Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Abnormalities, Multiple/prevention & control , Alleles , Amino Acid Substitution , Animals , Body Size/genetics , Cell Lineage , Crosses, Genetic , Dwarfism/genetics , Epistasis, Genetic , Face/abnormalities , Female , Genes, Dominant , Genotype , Heart Defects, Congenital/genetics , Hematopoiesis/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains/genetics , Myeloproliferative Disorders/genetics , Neoplastic Syndromes, Hereditary/embryology , Neoplastic Syndromes, Hereditary/genetics , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/physiology , Radiation Chimera , Signal Transduction/drug effects
3.
Methods Mol Biol ; 1487: 379-408, 2017.
Article in English | MEDLINE | ID: mdl-27924582

ABSTRACT

The RAS/MAPK signaling pathway plays key roles in development, cell survival and proliferation, as well as in cancer pathogenesis. Molecular genetic studies have identified a group of developmental syndromes, the RASopathies, caused by germ line mutations in this pathway. The syndromes included within this classification are neurofibromatosis type 1 (NF1), Noonan syndrome (NS), Noonan syndrome with multiple lentigines (NS-ML, formerly known as LEOPARD syndrome), Costello syndrome (CS), cardio-facio-cutaneous syndrome (CFC), Legius syndrome (LS, NF1-like syndrome), capillary malformation-arteriovenous malformation syndrome (CM-AVM), and hereditary gingival fibromatosis (HGF) type 1. Although these syndromes present specific molecular alterations, they are characterized by a large spectrum of functional and morphological abnormalities, which include heart defects, short stature, neurocognitive impairment, craniofacial malformations, and, in some cases, cancer predisposition. The development of genetically modified animals, such as mice (Mus musculus), flies (Drosophila melanogaster), and zebrafish (Danio rerio), has been instrumental in elucidating the molecular and cellular bases of these syndromes. Moreover, these models can also be used to determine tumor predisposition, the impact of different genetic backgrounds on the variable phenotypes found among the patients and to evaluate preventative and therapeutic strategies. Here, we review a wide range of genetically modified mouse models used in the study of RASopathies and the potential application of novel technologies, which hopefully will help us resolve open questions in the field.


Subject(s)
Disease Models, Animal , Genetic Association Studies , Genetic Predisposition to Disease , ras Proteins/genetics , ras Proteins/metabolism , Animals , Gene Targeting , Genetic Loci , Humans , MAP Kinase Signaling System , Mice , Mice, Transgenic , Signal Transduction
4.
Expert Rev Endocrinol Metab ; 12(5): 367-378, 2017 09.
Article in English | MEDLINE | ID: mdl-30058892

ABSTRACT

INTRODUCTION: Noonan syndrome is a RASopathy that results from activating mutations in different members of the RAS/MAPK signaling pathway. At least eleven members of this pathway have been found mutated, PTPN11 being the most frequently mutated gene affecting about 50% of the patients, followed by SOS1 (10%), RAF1 (10%) and KRAS (5%). Recently, even more infrequent mutations have been newly identified by next generation sequencing. This spectrum of mutations leads to a broad variety of clinical symptoms such as cardiopathies, short stature, facial dysmorphia and neurocognitive impairment. The genetic variability of this syndrome makes it difficult to establish a genotype-phenotype correlation, which will greatly help in the clinical management of the patients. Areas covered: Studies performed with different genetically engineered mouse models (GEMMs) developed up to date. Expert commentary: GEMMs have helped us understand the role of some genes and the effect of the different mutations in the development of the syndrome. However, few models have been developed and more characterization of the existing ones should be performed to learn about the impact of the different modifiers in the phenotypes, the potential cancer risk in patients, as well as preventative and therapeutic strategies.

5.
Cancer Res ; 77(3): 707-718, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27872088

ABSTRACT

Genetic studies in mice have provided evidence that H-Ras and K-Ras proteins are bioequivalent. However, human tumors display marked differences in the association of RAS oncogenes with tumor type. Thus, to further assess the bioequivalence of oncogenic H-Ras and K-Ras, we replaced the coding region of the murine K-Ras locus with H-RasG12V oncogene sequences. Germline expression of H-RasG12V or K-RasG12V from the K-Ras locus resulted in embryonic lethality. However, expression of these genes in adult mice led to different tumor phenotypes. Whereas H-RasG12V elicited papillomas and hematopoietic tumors, K-RasG12V induced lung tumors and gastric lesions. Pulmonary expression of H-RasG12V created a senescence-like state caused by excessive MAPK signaling. Likewise, H-RasG12V but not K-RasG12V induced senescence in mouse embryonic fibroblasts. Label-free quantitative analysis revealed that minor differences in H-RasG12V expression levels led to drastically different biological outputs, suggesting that subtle differences in MAPK signaling confer nonequivalent functions that influence tumor spectra induced by RAS oncoproteins. Cancer Res; 77(3); 707-18. ©2016 AACR.


Subject(s)
Genes, ras/genetics , Neoplasms/genetics , ras Proteins/genetics , ras Proteins/metabolism , Animals , Disease Models, Animal , Immunoblotting , Immunohistochemistry , Mice , Mice, Transgenic , Regulatory Sequences, Nucleic Acid
6.
Rare Dis ; 3(1): e1045169, 2015.
Article in English | MEDLINE | ID: mdl-26458870

ABSTRACT

Noonan syndrome (NS) is an autosomal dominant genetic disorder characterized by short stature, craniofacial dysmorphism, and congenital heart defects. A significant fraction of NS-patients also develop myeloproliferative disorders. The penetrance of these defects varies considerably among patients. In this study, we have examined the effect of 2 genetic backgrounds (C57BL/6J.OlaHsd and 129S2/SvPasCrl) on the phenotypes displayed by a mouse model of NS induced by germline expression of the mutated K-Ras (V14I) allele, one of the most frequent NS-KRAS mutations. Our results suggest the presence of genetic modifiers associated to the genetic background that are essential for heart development and function at early stages of postnatal life as well as in the severity of the haematopoietic alterations.

7.
PLoS One ; 8(12): e83028, 2013.
Article in English | MEDLINE | ID: mdl-24391734

ABSTRACT

BACKGROUND: Insulin-like Growth Factor 1 (IGF1) is a multifunctional regulator of somatic growth and development throughout evolution. IGF1 signaling through IGF type 1 receptor (IGF1R) controls cell proliferation, survival and differentiation in multiple cell types. IGF1 deficiency in mice disrupts lung morphogenesis, causing altered prenatal pulmonary alveologenesis. Nevertheless, little is known about the cellular and molecular basis of IGF1 activity during lung development. METHODS/PRINCIPAL FINDINGS: Prenatal Igf1(-/-) mutant mice with a C57Bl/6J genetic background displayed severe disproportional lung hypoplasia, leading to lethal neonatal respiratory distress. Immuno-histological analysis of their lungs showed a thickened mesenchyme, alterations in extracellular matrix deposition, thinner smooth muscles and dilated blood vessels, which indicated immature and delayed distal pulmonary organogenesis. Transcriptomic analysis of Igf1(-/-) E18.5 lungs using RNA microarrays identified deregulated genes related to vascularization, morphogenesis and cellular growth, and to MAP-kinase, Wnt and cell-adhesion pathways. Up-regulation of immunity-related genes was verified by an increase in inflammatory markers. Increased expression of Nfib and reduced expression of Klf2, Egr1 and Ctgf regulatory proteins as well as activation of ERK2 MAP-kinase were corroborated by Western blot. Among IGF-system genes only IGFBP2 revealed a reduction in mRNA expression in mutant lungs. Immuno-staining patterns for IGF1R and IGF2, similar in both genotypes, correlated to alterations found in specific cell compartments of Igf1(-/-) lungs. IGF1 addition to Igf1(-/-) embryonic lungs cultured ex vivo increased airway septa remodeling and distal epithelium maturation, processes accompanied by up-regulation of Nfib and Klf2 transcription factors and Cyr61 matricellular protein. CONCLUSIONS/SIGNIFICANCE: We demonstrated the functional tissue specific implication of IGF1 on fetal lung development in mice. Results revealed novel target genes and gene networks mediators of IGF1 action on pulmonary cellular proliferation, differentiation, adhesion and immunity, and on vascular and distal epithelium maturation during prenatal lung development.


Subject(s)
Insulin-Like Growth Factor I/deficiency , Lung/embryology , Lung/metabolism , Animals , Connective Tissue Growth Factor/genetics , Cysteine-Rich Protein 61/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Inflammation Mediators/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Kruppel-Like Transcription Factors/genetics , Lung/abnormalities , Mice , Mice, Inbred C57BL , Mice, Knockout , NFI Transcription Factors/genetics , Organogenesis/genetics , Organogenesis/physiology , Pregnancy , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Signal Transduction
8.
Cancer Cell ; 22(3): 318-30, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22975375

ABSTRACT

Clinical evidence indicates that mutation/activation of EGF receptors (EGFRs) is mutually exclusive with the presence of K-RAS oncogenes in lung and colon tumors. We have validated these observations using genetically engineered mouse models. However, development of pancreatic ductal adenocarcinomas driven by K-Ras oncogenes are totally dependent on EGFR signaling. Similar results were obtained using human pancreatic tumor cell lines. EGFRs were also essential even in the context of pancreatic injury and absence of p16Ink4a/p19Arf. Only loss of p53 made pancreatic tumors independent of EGFR signaling. Additional inhibition of PI3K and STAT3 effectively prevented proliferation of explants derived from these p53-defective pancreatic tumors. These findings may provide the bases for more rational approaches to treat pancreatic tumors in the clinic.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , ErbB Receptors/metabolism , Genes, ras , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction , Adenocarcinoma , Animals , Carcinoma, Pancreatic Ductal/genetics , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Epithelial Cells , ErbB Receptors/genetics , Erlotinib Hydrochloride , Humans , Mice , Mice, Transgenic , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/genetics , Quinazolines/pharmacology , Quinazolines/therapeutic use , STAT3 Transcription Factor/antagonists & inhibitors , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology , ras Proteins/genetics
9.
Cancer Cell ; 19(6): 728-39, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21665147

ABSTRACT

Pancreatic acinar cells of adult mice (≥P60) are resistant to transformation by some of the most robust oncogenic insults including expression of K-Ras oncogenes and loss of p16Ink4a/p19Arf or Trp53 tumor suppressors. Yet, these acinar cells yield pancreatic intraepithelial neoplasias (mPanIN) and ductal adenocarcinomas (mPDAC) if exposed to limited bouts of non-acute pancreatitis, providing they harbor K-Ras oncogenes. Pancreatitis contributes to tumor progression by abrogating the senescence barrier characteristic of low-grade mPanINs. Attenuation of pancreatitis-induced inflammation also accelerates tissue repair and thwarts mPanIN expansion. Patients with chronic pancreatitis display senescent PanINs, providing they have received antiinflammatory drugs. These results support the concept that antiinflammatory treatment of people diagnosed with pancreatitis may reduce their risk of developing PDAC.


Subject(s)
Cellular Senescence , Genes, ras , Pancreatic Neoplasms/etiology , Pancreatitis/complications , Adenocarcinoma/etiology , Animals , Anti-Inflammatory Agents/therapeutic use , Carcinoma, Pancreatic Ductal/etiology , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p16/physiology , Genes, p53/physiology , Humans , Mice , Pancreas, Exocrine/pathology , Pancreatic Neoplasms/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL