Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 835
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Genet ; 17(8): e1009738, 2021 08.
Article in English | MEDLINE | ID: mdl-34411095

ABSTRACT

Activation of Ras signaling occurs in ~30% of human cancers. However, activated Ras alone is insufficient to produce malignancy. Thus, it is imperative to identify those genes cooperating with activated Ras in driving tumoral growth. In this work, we have identified a novel EGFR inhibitor, which we have named EGFRAP, for EGFR adaptor protein. Elimination of EGFRAP potentiates activated Ras-induced overgrowth in the Drosophila wing imaginal disc. We show that EGFRAP interacts physically with the phosphorylated form of EGFR via its SH2 domain. EGFRAP is expressed at high levels in regions of maximal EGFR/Ras pathway activity, such as at the presumptive wing margin. In addition, EGFRAP expression is up-regulated in conditions of oncogenic EGFR/Ras activation. Normal and oncogenic EGFR/Ras-mediated upregulation of EGRAP levels depend on the Notch pathway. We also find that elimination of EGFRAP does not affect overall organogenesis or viability. However, simultaneous downregulation of EGFRAP and its ortholog PVRAP results in defects associated with increased EGFR function. Based on these results, we propose that EGFRAP is a new negative regulator of the EGFR/Ras pathway, which, while being required redundantly for normal morphogenesis, behaves as an important modulator of EGFR/Ras-driven tissue hyperplasia. We suggest that the ability of EGFRAP to functionally inhibit the EGFR pathway in oncogenic cells results from the activation of a feedback loop leading to increase EGFRAP expression. This could act as a surveillance mechanism to prevent excessive EGFR activity and uncontrolled cell growth.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Genes, ras/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle , Cell Proliferation/genetics , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, ras/physiology , Imaginal Discs/metabolism , Morphogenesis , Phosphorylation , Receptors, Invertebrate Peptide/antagonists & inhibitors , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Signal Transduction/genetics , ras Proteins/genetics
2.
Genes Dev ; 27(13): 1447-61, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23824538

ABSTRACT

Macroautophagy (autophagy hereafter) degrades and recycles proteins and organelles to support metabolism and survival in starvation. Oncogenic Ras up-regulates autophagy, and Ras-transformed cell lines require autophagy for mitochondrial function, stress survival, and engrafted tumor growth. Here, the essential autophagy gene autophagy-related-7 (atg7) was deleted concurrently with K-ras(G12D) activation in mouse models for non-small-cell lung cancer (NSCLC). atg7-deficient tumors accumulated dysfunctional mitochondria and prematurely induced p53 and proliferative arrest, which reduced tumor burden that was partly relieved by p53 deletion. atg7 loss altered tumor fate from adenomas and carcinomas to oncocytomas-rare, predominantly benign tumors characterized by the accumulation of defective mitochondria. Surprisingly, lipid accumulation occurred in atg7-deficient tumors only when p53 was deleted. atg7- and p53-deficient tumor-derived cell lines (TDCLs) had compromised starvation survival and formed lipidic cysts instead of tumors, suggesting defective utilization of lipid stores. atg7 deficiency reduced fatty acid oxidation (FAO) and increased sensitivity to FAO inhibition, indicating that with p53 loss, Ras-driven tumors require autophagy for mitochondrial function and lipid catabolism. Thus, autophagy is required for carcinoma fate, and autophagy defects may be a molecular basis for the occurrence of oncocytomas. Moreover, cancers require autophagy for distinct roles in metabolism that are oncogene- and tumor suppressor gene-specific.


Subject(s)
Adenoma, Oxyphilic/physiopathology , Autophagy , Carcinoma, Non-Small-Cell Lung/physiopathology , Genes, ras/physiology , Lipid Metabolism , Lung Neoplasms/physiopathology , Animals , Cell Line, Tumor , Gene Deletion , Gene Expression Regulation, Neoplastic , Genes, p53/genetics , Homeostasis , Longevity/genetics , Mice , Mitochondria/pathology , Tumor Cells, Cultured
3.
Toxicol Appl Pharmacol ; 402: 115121, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32621833

ABSTRACT

Aberrant activation of H-Ras is often associated with tumor aggressiveness in breast cancer. Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1  (Pin1) is a unique enzyme that interacts with phosphorylated serine or threonine of a target protein and isomerizes the adjacent proline residue. Pin1 is prevalently overexpressed in human cancers, and its overexpression correlates with poor prognosis. Nuclear factor E2-related factor 2 (Nrf2) is a master regulator of cellular redox homeostasis. The sustained activation/accumulation of Nrf2 has been observed in many different types of human malignancies, conferring an advantage for growth and survival of cancer cells. The activated form of H-Ras (GTP-H-Ras) is highly overexpressed in human breast cancer tissues. In our present study, silencing of H-Ras decreased the invasiveness of MDA-MB-231 human breast cancer cells and abrogated the interaction between Pin1 and Nrf2 in these cells. Pin1 knockdown blocked the accumulation of Nrf2, thereby suppressing proliferation and clonogenicity of MCF10A-Ras human mammary epithelial cells. We found that Pin1 binds to Nrf2 which stabilizes this transcription factor by hampering proteasomal degradation. In conclusion, H-Ras activation in cooperation with the Pin1-Nrf2 complex represents a novel mechanism underlying breast cancer progression and constitutive activation of Nrf2 and can be exploited as a therapeutic target.


Subject(s)
Breast Neoplasms/metabolism , Genes, ras/physiology , NF-E2-Related Factor 2/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Silencing , Genes, ras/genetics , HEK293 Cells , Humans , NF-E2-Related Factor 2/genetics , NIMA-Interacting Peptidylprolyl Isomerase/genetics
4.
Curr Opin Pediatr ; 32(1): 48-56, 2020 02.
Article in English | MEDLINE | ID: mdl-31815779

ABSTRACT

PURPOSE OF REVIEW: The current review aims to highlight the frequency of RAS mutations in pediatric leukemias and solid tumors and to propose strategies for targeting oncogenic RAS in pediatric cancers. RECENT FINDINGS: The three RAS genes (HRAS, NRAS, and KRAS) comprise the most frequently mutated oncogene family in human cancer. RAS mutations are commonly observed in three of the leading causes of cancer death in the United States, namely lung cancer, pancreatic cancer, and colorectal cancer. The association of RAS mutations with these aggressive malignancies inspired the creation of the National Cancer Institute RAS initiative and spurred intense efforts to develop strategies to inhibit oncogenic RAS, with much recent success. RAS mutations are frequently observed in pediatric cancers; however, recent advances in anti-RAS drug development have yet to translate into pediatric clinical trials. SUMMARY: We find that RAS is mutated in common and rare pediatric malignancies and that oncogenic RAS confers a functional dependency in these cancers. Many strategies for targeting RAS are being pursued for malignancies that primarily affect adults and there is a clear need for inclusion of pediatric patients in clinical trials of these agents.


Subject(s)
Genes, ras/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Child , GTP Phosphohydrolases/genetics , Genes, ras/physiology , Humans , Membrane Proteins/genetics , Molecular Targeted Therapy , Mutation , Proto-Oncogene Proteins p21(ras)/genetics
5.
Mol Cancer ; 17(1): 33, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29455666

ABSTRACT

Lung neoplasms are the leading cause of death by cancer worldwide. Non-small cell lung cancer (NSCLC) constitutes more than 80% of all lung malignancies and the majority of patients present advanced disease at onset. However, in the last decade, multiple oncogenic driver alterations have been discovered and each of them represents a potential therapeutic target. Although KRAS mutations are the most frequently oncogene aberrations in lung adenocarcinoma patients, effective therapies targeting KRAS have yet to be developed. Moreover, the role of KRAS oncogene in NSCLC remains unclear and its predictive and prognostic impact remains controversial. The study of the underlying biology of KRAS in NSCLC patients could help to determine potential candidates to evaluate novel targeted agents and combinations that may allow a tailored treatment for these patients. The aim of this review is to update the current knowledge about KRAS-mutated lung adenocarcinoma, including a historical overview, the biology of the molecular pathways involved, the clinical relevance of KRAS mutations as a prognostic and predictive marker and the potential therapeutic approaches for a personalized treatment of KRAS-mutated NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Genes, ras/genetics , Lung Neoplasms/genetics , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Genes, ras/physiology , Humans , Lung Neoplasms/drug therapy , Mutation/genetics
6.
Am J Pathol ; 187(5): 973-979, 2017 May.
Article in English | MEDLINE | ID: mdl-28279655

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a risk factor for lung cancer. COPD is characterized by chronic airway inflammation and lung infections. The airways of patients with COPD are frequently colonized with bacteria [eg, nontypeable Haemophilus influenzae (NTHi)] that cause pulmonary inflammation and exacerbations. Pulmonary adenocarcinomas are frequently associated with an activating mutation in the KRAS gene. We determined the function of Toll-like receptor (TLR) signaling on the progression of Kras-induced early adenomatous lesions in the lung. Wild-type (WT) mice and mice doubly deficient in Tlr-2 and -4 (Tlr2/4-/-), both with an oncogenic Kras allele in lung epithelium, were exposed to NTHi for 4 weeks. Exposure to NTHi resulted in increased tumor proliferation and growth in WT mice, but not in Tlr2/4-/- mice. Alveolar adenomatous hyperplasia and adenocarcinoma were significantly increased in WT mice compared with Tlr2/4-/- mice. The average size of tumors was significantly larger in WT mice, whereas there was no difference in the number of alveolar lesions between WT and Tlr2/4-/- mice. NTHi-induced pulmonary neutrophilic inflammation and tumor-associated neutrophils were reduced in Tlr2/4-/- mice. Thus, subsequent to a driver mutation, NTHi-induced inflammation promotes proliferation of early adenomatous lesions in a TLR-dependent manner.


Subject(s)
Genes, ras/physiology , Pulmonary Disease, Chronic Obstructive/physiopathology , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology , Adenocarcinoma/physiopathology , Animals , Cell Proliferation/physiology , Haemophilus Infections/physiopathology , Haemophilus influenzae/physiology , Lung Neoplasms/physiopathology , Mice , Neutrophils/physiology , Pulmonary Disease, Chronic Obstructive/virology , Signal Transduction/physiology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 4/deficiency , ras Proteins/metabolism
7.
BMC Cancer ; 18(1): 1201, 2018 Dec 03.
Article in English | MEDLINE | ID: mdl-30509235

ABSTRACT

BACKGROUND: Peritoneal dissemination is a critical prognostic factor in ovarian cancer. Although stabilized spheroid formation promotes cancer cell peritoneal dissemination in ovarian cancer, the associated oncogenes are unknown. In this study, we assessed the role of the KRAS oncogene in ovarian cancer cell dissemination, focusing on the stability of cells in spheroid condition, as well as the modulation of intracellular signaling following spheroid transformation. METHODS: We used ID8, a murine ovarian cancer cell line, and ID8-KRAS, an oncogenic KRAS (G12 V)-transduced ID8 cell line in this study. Spheroid-forming (3D) culture and cell proliferation assays were performed to evaluate the growth characteristics of these cells. cDNA microarray analysis was performed to identify genes involved in KRAS-associated signal transduction in floating condition. A MEK inhibitor was used to evaluate the effect on cancer peritoneal dissemination. RESULTS: Cell viability and proliferation in monolayer (2D) cultures did not differ between ID8 and ID8-KRAS cells. However, the proportions of viable and proliferating ID8-KRAS cells in 3D culture were approximately 2-fold and 5-fold higher than that of ID8, respectively. Spheroid-formation was increased in ID8-KRAS cells. Analysis of peritoneal floating cells obtained from mice intra-peritoneally injected with cancer cells revealed that the proportion of proliferating cancer cells was approximately 2-fold higher with ID8-KRAS than with ID8 cells. Comprehensive cDNA microarray analysis revealed that pathways related to cell proliferation, and cell cycle checkpoint and regulation were upregulated specifically in ID8-KRAS cells in 3D culture, and that some genes partially regulated by the MEK-ERK pathway were upregulated only in ID8-KRAS cells in 3D culture. Furthermore, a MEK inhibitor, trametinib, suppressed spheroid formation in 3D culture of ID8-KRAS cells, although trametinib did not affect 2D-culture cell proliferation. Finally, we demonstrated that trametinib dramatically improved the prognosis for mice with ID8-KRAS tumors in an in vivo mouse model. CONCLUSIONS: Our data indicated that KRAS promoted ovarian cancer dissemination by stabilizing spheroid formation and that the MEK pathway is important for stabilized spheroid formation. Disruption of spheroid formation by a MEK inhibitor could be a therapeutic target for cancer peritoneal dissemination.


Subject(s)
Cell Proliferation/physiology , Genes, ras/physiology , MAP Kinase Signaling System/physiology , Ovarian Neoplasms/metabolism , Spheroids, Cellular/metabolism , Animals , Cell Culture Techniques/methods , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/pathology
8.
Biochim Biophys Acta ; 1863(4): 770-84, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26844774

ABSTRACT

Melanoma has markedly increased worldwide during the past several decades in the Caucasian population and is responsible for 80% of skin cancer deaths. Considering that metastatic melanoma is almost completely resistant to most current therapies and is linked with a poor patient prognosis, it is crucial to further investigate potential molecular targets. Major cell-autonomous drivers in the pathogenesis of this disease include the classical MAPK (i.e., RAS-RAF-MEK-ERK), WNT, and PI3K signaling pathways. These pathways play a major role in defining the progression of melanoma, and some have been the subject of recent pharmacological strategies to treat this belligerent disease. This review describes the latest advances in the understanding of melanoma progression and the major molecular pathways involved. In addition, we discuss the roles of emerging molecular players that are involved in melanoma pathogenesis, including the functional role of the melanoma tumor antigen, p97/MFI2 (melanotransferrin).


Subject(s)
Melanoma/genetics , Melanoma/pathology , Oncogenes/physiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Animals , Disease Progression , Genes, ras/physiology , Humans , MAP Kinase Signaling System/physiology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Wnt Signaling Pathway/physiology , raf Kinases/physiology
9.
PLoS Biol ; 12(3): e1001807, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24618719

ABSTRACT

The Cut homeobox 1 (CUX1) gene is a target of loss-of-heterozygosity in many cancers, yet elevated CUX1 expression is frequently observed and is associated with shorter disease-free survival. The dual role of CUX1 in cancer is illustrated by the fact that most cell lines with CUX1 LOH display amplification of the remaining allele, suggesting that decreased CUX1 expression facilitates tumor development while increased CUX1 expression is needed in tumorigenic cells. Indeed, CUX1 was found in a genome-wide RNAi screen to identify synthetic lethal interactions with oncogenic RAS. Here we show that CUX1 functions in base excision repair as an ancillary factor for the 8-oxoG-DNA glycosylase, OGG1. Single cell gel electrophoresis (comet assay) reveals that Cux1⁺/⁻ MEFs are haploinsufficient for the repair of oxidative DNA damage, whereas elevated CUX1 levels accelerate DNA repair. In vitro base excision repair assays with purified components demonstrate that CUX1 directly stimulates OGG1's enzymatic activity. Elevated reactive oxygen species (ROS) levels in cells with sustained RAS pathway activation can cause cellular senescence. We show that elevated expression of either CUX1 or OGG1 prevents RAS-induced senescence in primary cells, and that CUX1 knockdown is synthetic lethal with oncogenic RAS in human cancer cells. Elevated CUX1 expression in a transgenic mouse model enables the emergence of mammary tumors with spontaneous activating Kras mutations. We confirmed cooperation between Kras(G12V) and CUX1 in a lung tumor model. Cancer cells can overcome the antiproliferative effects of excessive DNA damage by inactivating a DNA damage response pathway such as ATM or p53 signaling. Our findings reveal an alternate mechanism to allow sustained proliferation in RAS-transformed cells through increased DNA base excision repair capability. The heightened dependency of RAS-transformed cells on base excision repair may provide a therapeutic window that could be exploited with drugs that specifically target this pathway.


Subject(s)
DNA Damage , DNA Repair/physiology , Genes, ras/physiology , Homeodomain Proteins/physiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cellular Senescence/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mammary Neoplasms, Experimental/genetics , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidative Stress , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors
10.
Clin Exp Dermatol ; 42(6): 645-647, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28691211

ABSTRACT

The MS1/SVR system, in which MS1 represents immortalized endothelial cells and SVR represents MS1 cells transformed with oncogenic human-rat sarcoma protein (H-Ras), has been used for around 20 years as a valuable tool to study angiogenesis and carcinogenesis. Despite the use of these cells in numerous studies, a comprehensive profile of the signalling differences due to oncogenic H-Ras transformation has not been performed previously. In this study, we profiled the well-known MS1 and SVR cell lines using a combination of both Western blot and gene chip assays.


Subject(s)
Genes, ras/physiology , Hemangiosarcoma/metabolism , Signal Transduction , Animals , Blotting, Western , Cell Line, Tumor , Hemangiosarcoma/genetics , Mice , Neovascularization, Pathologic , Oligonucleotide Array Sequence Analysis , Rabbits
11.
Genes Dev ; 23(7): 877-89, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19339691

ABSTRACT

The genetic heterogeneity of cancer influences the trajectory of tumor progression and may underlie clinical variation in therapy response. To model such heterogeneity, we produced genetically and pathologically accurate mouse models of common forms of human acute myeloid leukemia (AML) and developed methods to mimic standard induction chemotherapy and efficiently monitor therapy response. We see that murine AMLs harboring two common human AML genotypes show remarkably diverse responses to conventional therapy that mirror clinical experience. Specifically, murine leukemias expressing the AML1/ETO fusion oncoprotein, associated with a favorable prognosis in patients, show a dramatic response to induction chemotherapy owing to robust activation of the p53 tumor suppressor network. Conversely, murine leukemias expressing MLL fusion proteins, associated with a dismal prognosis in patients, are drug-resistant due to an attenuated p53 response. Our studies highlight the importance of genetic information in guiding the treatment of human AML, functionally establish the p53 network as a central determinant of chemotherapy response in AML, and demonstrate that genetically engineered mouse models of human cancer can accurately predict therapy response in patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Disease Models, Animal , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mice, Inbred C57BL , Animals , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, ras/genetics , Genes, ras/physiology , Genotype , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/physiopathology , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Prognosis , RUNX1 Translocation Partner 1 Protein , Tumor Suppressor Protein p53/genetics
12.
Circulation ; 131(4): 390-400; discussion 400, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25369805

ABSTRACT

BACKGROUND: Cardiac hypertrophy is an early hallmark during the clinical course of heart failure and is regulated by various signaling pathways. However, the molecular mechanisms that negatively regulate these signal transduction pathways remain poorly understood. METHODS AND RESULTS: Here, we characterized Carabin, a protein expressed in cardiomyocytes that was downregulated in cardiac hypertrophy and human heart failure. Four weeks after transverse aortic constriction, Carabin-deficient (Carabin(-/-)) mice developed exaggerated cardiac hypertrophy and displayed a strong decrease in fractional shortening (14.6±1.6% versus 27.6±1.4% in wild type plus transverse aortic constriction mice; P<0.0001). Conversely, compensation of Carabin loss through a cardiotropic adeno-associated viral vector encoding Carabin prevented transverse aortic constriction-induced cardiac hypertrophy with preserved fractional shortening (39.9±1.2% versus 25.9±2.6% in control plus transverse aortic constriction mice; P<0.0001). Carabin also conferred protection against adrenergic receptor-induced hypertrophy in isolated cardiomyocytes. Mechanistically, Carabin carries out a tripartite suppressive function. Indeed, Carabin, through its calcineurin-interacting site and Ras/Rab GTPase-activating protein domain, functions as an endogenous inhibitor of calcineurin and Ras/extracellular signal-regulated kinase prohypertrophic signaling. Moreover, Carabin reduced Ca(2+)/calmodulin-dependent protein kinase II activation and prevented nuclear export of histone deacetylase 4 after adrenergic stimulation or myocardial pressure overload. Finally, we showed that Carabin Ras-GTPase-activating protein domain and calcineurin-interacting domain were both involved in the antihypertrophic action of Carabin. CONCLUSIONS: Our study identifies Carabin as a negative regulator of key prohypertrophic signaling molecules, calcineurin, Ras, and Ca(2+)/calmodulin-dependent protein kinase II and implicates Carabin in the development of cardiac hypertrophy and failure.


Subject(s)
Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , GTPase-Activating Proteins/biosynthesis , Genes, ras/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Rats , Signal Transduction/physiology
13.
Proc Natl Acad Sci U S A ; 110(17): 6853-8, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23569271

ABSTRACT

The transformation of normal cells to malignant, metastatic tumor cells is a multistep process caused by the sequential acquirement of genetic changes. To identify these changes, we compared the transcriptomes and levels and distribution of proteins in a four-stage cell model of isogenically matched normal, immortalized, transformed, and metastatic human cells, using deep transcriptome sequencing and immunofluorescence microscopy. The data show that ∼6% (n = 1,357) of the human protein-coding genes are differentially expressed across the stages in the model. Interestingly, the majority of these genes are down-regulated, linking malignant transformation to dedifferentiation. The up-regulated genes are mainly components that control cellular proliferation, whereas the down-regulated genes consist of proteins exposed on or secreted from the cell surface. As many of the identified gene products control basic cellular functions that are defective in cancers, the data provide candidates for follow-up studies to investigate their functional roles in tumor formation. When we further compared the expression levels of four of the identified proteins in clinical cancer cohorts, similar differences were observed between benign and cancer cells, as in the cell model. This shows that this comprehensive demonstration of the molecular changes underlying malignant transformation is a relevant model to study the process of tumor formation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic/physiology , Models, Biological , Proteins/metabolism , Antigens, Polyomavirus Transforming/metabolism , Cell Line, Tumor , Fibroblasts/metabolism , Gene Expression Profiling , Genes, ras/physiology , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Microscopy, Fluorescence , Principal Component Analysis , Telomerase/metabolism
14.
Nat Genet ; 39(6): 750-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17468755

ABSTRACT

Stem cell function is central for the maintenance of normal tissue homeostasis. Here we show that deletion of p38alpha mitogen-activated protein (MAP) kinase in adult mice results in increased proliferation and defective differentiation of lung stem and progenitor cells both in vivo and in vitro. We found that p38alpha positively regulates factors such as CCAAT/enhancer-binding protein that are required for lung cell differentiation. In addition, p38alpha controls self-renewal of the lung stem and progenitor cell population by inhibiting proliferation-inducing signals, most notably epidermal growth factor receptor. As a consequence, the inactivation of p38alpha leads to an immature and hyperproliferative lung epithelium that is highly sensitized to K-Ras(G12V)-induced tumorigenesis. Our results indicate that by coordinating proliferation and differentiation signals in lung stem and progenitor cells, p38alpha has a key role in the regulation of lung cell renewal and tumorigenesis.


Subject(s)
Cell Differentiation , Cell Proliferation , Lung/cytology , Mitogen-Activated Protein Kinase 14/physiology , Stem Cells/cytology , Animals , Cells, Cultured , Female , Genes, ras/physiology , Immunoenzyme Techniques , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 14/genetics , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
15.
J Neurosci ; 34(3): 776-83, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24431436

ABSTRACT

In dendritic spines, Ras plays a critical role in synaptic plasticity but its regulation mechanism is not fully understood. Here, using a fluorescence resonance energy transfer/fluorescence lifetime imaging microscopy-based Ras imaging technique in combination with 2-photon glutamate uncaging, we show that neurofibromin, in which loss-of-function mutations cause Neurofibromatosis Type 1 (NF1), contributes to the majority (∼90%) of Ras inactivation in dendritic spines of pyramidal neurons in the CA1 region of the rat hippocampus. Loss of neurofibromin causes sustained Ras activation in spines, which leads to impairment of spine structural plasticity and loss of spines in an activity-dependent manner. Therefore, deregulation of postsynaptic Ras signaling may explain, at least in part, learning disabilities associated with NF1.


Subject(s)
Dendritic Spines/physiology , Genes, ras/physiology , Neurofibromin 1/physiology , Animals , Cells, Cultured , Dendritic Spines/genetics , Dendritic Spines/ultrastructure , Excitatory Postsynaptic Potentials/genetics , Female , Hippocampus/physiology , Hippocampus/ultrastructure , Long-Term Potentiation/physiology , Male , Neurofibromin 1/deficiency , Neurofibromin 1/genetics , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
16.
J Neurosci ; 34(6): 2169-90, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24501358

ABSTRACT

Neural cell fate specification is well understood in the embryonic cerebral cortex, where the proneural genes Neurog2 and Ascl1 are key cell fate determinants. What is less well understood is how cellular diversity is generated in brain tumors. Gliomas and glioneuronal tumors, which are often localized in the cerebrum, are both characterized by a neoplastic glial component, but glioneuronal tumors also have an intermixed neuronal component. A core abnormality in both tumor groups is overactive RAS/ERK signaling, a pro-proliferative signal whose contributions to cell differentiation in oncogenesis are largely unexplored. We found that RAS/ERK activation levels differ in two distinct human tumors associated with constitutively active BRAF. Pilocytic astrocytomas, which contain abnormal glial cells, have higher ERK activation levels than gangliogliomas, which contain abnormal neuronal and glial cells. Using in vivo gain of function and loss of function in the mouse embryonic neocortex, we found that RAS/ERK signals control a proneural genetic switch, inhibiting Neurog2 expression while inducing Ascl1, a competing lineage determinant. Furthermore, we found that RAS/ERK levels control Ascl1's fate specification properties in murine cortical progenitors--at higher RAS/ERK levels, Ascl1(+) progenitors are biased toward proliferative glial programs, initiating astrocytomas, while at moderate RAS/ERK levels, Ascl1 promotes GABAergic neuronal and less glial differentiation, generating glioneuronal tumors. Mechanistically, Ascl1 is phosphorylated by ERK, and ERK phosphoacceptor sites are necessary for Ascl1's GABAergic neuronal and gliogenic potential. RAS/ERK signaling thus acts as a rheostat to influence neural cell fate selection in both normal cortical development and gliomagenesis, controlling Neurog2-Ascl1 expression and Ascl1 function.


Subject(s)
Brain Neoplasms/metabolism , Cerebral Cortex/metabolism , Genes, ras/physiology , Glioma/metabolism , MAP Kinase Signaling System/physiology , Neurons/metabolism , Animals , Brain Neoplasms/pathology , Cerebral Cortex/embryology , Cerebral Cortex/pathology , Female , Glioma/pathology , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Pregnancy
17.
Int J Cancer ; 136(6): E508-20, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25155634

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) process allows carcinoma cells to dissociate from the primary tumor thereby facilitating tumor cell invasion and metastasis. Ras-dependent hyperactive signaling is commonly associated with tumorigenesis, invasion, EMT, and metastasis. However, the downstream effectors by which Ras regulates EMT remain ill defined. In this study, we show that the H-Ras pathway leads to mesenchymal-like phenotypic changes in human breast epithelial cells by controlling the ZEB1/microRNA-200c axis. Moreover, H-Ras suppresses the expression of the discoidin domain receptor 1 (DDR1), a collagen receptor tyrosine kinase, via ZEB1, thus identifying ZEB1 as a novel transcriptional repressor of DDR1. Mutation studies on the putative promoter of the DDR1 gene revealed that bipartite Z- and E-box elements play a key role in transcriptional repression of DDR1 in Hs578T and MDA-MB-231 breast carcinoma cell lines by ZEB1. Furthermore, we found an inverse correlation between ZEB1 and DDR1 expression in various cancer cell lines and in human breast carcinoma tissues. Consistently, overexpression of DDR1 reduced the invasive phenotype of mesenchymal-like triple-negative breast cancer cells in 3D cultures and in vivo. Thus, ZEB1's role in maintenance of EMT in breast carcinoma cells is mediated in part by its ability to suppress DDR1 expression and consequently contribute to the activation of the invasive phenotype. Taken together, our results unveil a novel H-Ras/ZEB1/DDR1 network that contributes to breast cancer progression in triple-negative breast cancers.


Subject(s)
Breast/pathology , Epithelial-Mesenchymal Transition , Genes, ras/physiology , Homeodomain Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Mitogen/physiology , Transcription Factors/physiology , Cell Line, Tumor , Cytoskeleton/physiology , Discoidin Domain Receptors , Epithelial Cells/pathology , Female , Humans , MicroRNAs/physiology , Morphogenesis , Zinc Finger E-box-Binding Homeobox 1
18.
BMC Cancer ; 15: 172, 2015 Mar 25.
Article in English | MEDLINE | ID: mdl-25885317

ABSTRACT

BACKGROUND: Mutant Ras plays multiple functions in tumorigenesis including tumor formation and metastasis. Reversion-inducing cysteine-rich protein with Kazal motifs (RECK), a metastasis inhibitor gene, suppresses matrix metalloproteinase (MMP) activity in the metastatic cascade. Clarifying the relationship between Ras and RECK and understanding the underlying molecular mechanism may lead to the development of better treatment for Ras-related tumors. METHODS: Suppression subtractive hybridization PCR (SSH PCR) was conducted to identify Ha-ras (val12) up-regulated genes in bladder cancer cells. Stable cell lines of human breast cancer (MCF-7-ras) and mouse NIH3T3 fibroblasts (7-4) harboring the inducible Ha-ras (val12) oncogene, which could be induced by isopropylthio-ß-D-galactoside (IPTG), were used to clarify the relationship between Ras and the up-regulated genes. Chromatin immunoprecipitation (ChIP) assay, DNA affinity precipitation assay (DAPA) and RECK reporter gene assay were utilized to confirm the complex formation and binding with promoters. RESULTS: Retinoblastoma binding protein-7 (RbAp46) was identified and confirmed as a Ha-ras (val12) up-regulated gene. RbAp46 could bind with histone deacetylase (HDAC1) and Sp1, followed by binding to RECK promoter at the Sp1 site resulting in repression of RECK expression. High expression of Ras protein accompanied with high RbAp46 and low RECK expression were detected in 75% (3/4) of the clinical bladder cancer tumor tissues compared to the adjacent normal parts. Ras induced RbAp46 expression increases invasion of the bladder cancer T24 cells and MMP-9 activity was increased, which was confirmed by specific lentiviral shRNAs inhibitors against Ras and RbAp46. Similarly, knockdown of RbAp46 expression in the stable NIH3T3 cells "7-4" by shRNA decreased Ras-related lung metastasis using a xenograft nude mice model. CONCLUSIONS: We confirmed that RbAp46 is a Ha-ras (val12) up-regulated gene and binds with HDAC1 and Sp1. Furthermore, RbAp46 binds to the RECK promoter at the Sp1 site via recruitment by Sp1. RECK is subsequently activated, leading to increased MMP9 activity, which may lead to increased metastasis in vivo. Our findings of Ras upregulation of RbAp46 may lead to revealing a novel mechanism of Ras-related tumor cell metastasis.


Subject(s)
GPI-Linked Proteins/metabolism , Genes, ras , Lung Neoplasms/metabolism , Promoter Regions, Genetic , Retinoblastoma-Binding Protein 7/biosynthesis , Up-Regulation , Animals , Female , GPI-Linked Proteins/antagonists & inhibitors , Genes, ras/physiology , Humans , Lung Neoplasms/pathology , MCF-7 Cells , Mice , Mice, Nude , NIH 3T3 Cells , Promoter Regions, Genetic/physiology , Up-Regulation/physiology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/secondary
19.
J Pathol ; 234(2): 214-27, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24889936

ABSTRACT

Pancreatic cancer has a dismal prognosis and is currently the fourth leading cause of cancer-related death in developed countries. The inhibition of poly(ADP-ribose) polymerase-1 (Parp-1), the major protein responsible for poly(ADP-ribosy)lation in response to DNA damage, has emerged as a promising treatment for several tumour types. Here we aimed to elucidate the involvement of Parp-1 in pancreatic tumour progression. We assessed Parp-1 protein expression in normal, preneoplastic and pancreatic tumour samples from humans and from K-Ras- and c-myc-driven mouse models of pancreatic cancer. Parp-1 was highly expressed in acinar cells in normal and cancer tissues. In contrast, ductal cells expressed very low or undetectable levels of this protein, both in a normal and in a tumour context. The Parp-1 expression pattern was similar in human and mouse samples, thereby validating the use of animal models for further studies. To determine the in vivo effects of Parp-1 depletion on pancreatic cancer progression, Ela-myc-driven pancreatic tumour development was analysed in a Parp-1 knock-out background. Loss of Parp-1 resulted in increased tumour necrosis and decreased proliferation, apoptosis and angiogenesis. Interestingly, Ela-myc:Parp-1(-/-) mice displayed fewer ductal tumours than their Ela-myc:Parp-1(+/+) counterparts, suggesting that Parp-1 participates in promoting acinar-to-ductal metaplasia, a key event in pancreatic cancer initiation. Moreover, impaired macrophage recruitment can be responsible for the ADM blockade found in the Ela-myc:Parp-1(-/-) mice. Finally, molecular analysis revealed that Parp-1 modulates ADM downstream of the Stat3-MMP7 axis and is also involved in transcriptional up-regulation of the MDM2, VEGFR1 and MMP28 cancer-related genes. In conclusion, the expression pattern of Parp-1 in normal and cancer tissue and the in vivo functional effects of Parp-1 depletion point to a novel role for this protein in pancreatic carcinogenesis and shed light into the clinical use of Parp-1 inhibitors.


Subject(s)
Pancreatic Neoplasms/genetics , Poly(ADP-ribose) Polymerases/genetics , Proto-Oncogene Proteins c-myc/metabolism , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Genes, ras/physiology , Humans , Male , Mice , Mice, Knockout , Middle Aged , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1
20.
Cancer Cell ; 12(2): 160-70, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17692807

ABSTRACT

We investigated the influence of normal cell phenotype on the neoplastic phenotype by comparing tumors derived from two different normal human mammary epithelial cell populations, one of which was isolated using a new culture medium. Transformation of these two cell populations with the same set of genetic elements yielded cells that formed tumor xenografts exhibiting major differences in histopathology, tumorigenicity, and metastatic behavior. While one cell type (HMECs) yielded squamous cell carcinomas, the other cell type (BPECs) yielded tumors closely resembling human breast adenocarcinomas. Transformed BPECs gave rise to lung metastases and were up to 10(4)-fold more tumorigenic than transformed HMECs, which are nonmetastatic. Hence, the pre-existing differences between BPECs and HMECs strongly influence the phenotypes of their transformed derivatives.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast/cytology , Cell Transformation, Neoplastic , Epithelial Cells/cytology , Adenocarcinoma/etiology , Adenocarcinoma/pathology , Adult , Animals , Antigens, Polyomavirus Transforming/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , Cell Division , Cells, Cultured , Female , Gene Expression Profiling , Genes, ras/physiology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Middle Aged , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL