Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 15(22)2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38001740

ABSTRACT

There is little argument that the K-RAS onco-protein is the most important single oncoprotein in human cancer [...].

2.
Cancers (Basel) ; 15(16)2023 Aug 16.
Article in English | MEDLINE | ID: mdl-37627161

ABSTRACT

BRCA1 is a tumor suppressor with a complex mode of action. Hereditary mutations in BRCA1 predispose carriers to breast cancer, and spontaneous breast cancers often exhibit defects in BRCA1 expression. However, haploinsufficiency or suppression of BRCA1 expression leads to defects in DNA repair, which can induce DNA damage responses, leading to senescence. Activating mutation or overexpression of the Her2 oncoprotein are also frequent drivers of breast cancer. Yet, over-activation of Her2, working through the RAS oncoprotein, can also induce senescence. It is thought that additional defects in the p53 and Rb tumor suppressor machinery must occur in such tumors to allow an escape from senescence, thus permitting tumor development. Although BRCA1 mutant breast cancers are usually Her2 negative, a significant percentage of Her2 positive tumors also lose their expression of BRCA1. Such Her2+/BRCA1- tumors might be expected to have a particularly high senescence barrier to overcome. An important RAS senescence effector is the protein NORE1A, which can modulate both p53 and Rb. It is an essential senescence effector of the RAS oncoprotein, and it is often downregulated in breast tumors by promotor methylation. Here we show that NORE1A forms a Her2/RAS regulated, endogenous complex with BRCA1 at sites of replication fork arrest. Suppression of NORE1A blocks senescence induction caused by BRCA1 inactivation and Her2 activation. Thus, NORE1A forms a tumor suppressor complex with BRCA1. Its frequent epigenetic inactivation may facilitate the transformation of Her2+/BRCA1- mediated breast cancer by suppressing senescence.

3.
Methods Mol Biol ; 2262: 303-310, 2021.
Article in English | MEDLINE | ID: mdl-33977485

ABSTRACT

RASSF1A is a Ras effector that promotes the anti-proliferative properties of Ras. It acts as a scaffold protein that regulates several pro-apoptotic signaling pathways, thereby linking Ras to their regulation. However, accumulating evidence suggests that RASSF1A functions as a regulator of other additional biological processes, such as DNA repair and transcription, thereby implicating Ras in the modulation of these biological processes. The mechanisms by which RASSF1A modulates these processes is not fully understood but likely involves interacting with other effectors associated with these functions and coordinating their activity. Thus, to fully understand how RASSF1A manifests its activity, it is critical to identify RASSF1A interacting partners.Unfortunately, the reagents available for the detection of RASSF1A are of poor quality and also exhibit low sensitivity. Here we describe an immunoprecipitation protocol, taking into consideration the limitations of currently available reagents, that can reliably detect the endogenous interaction between RASSF1A and its binding partners.


Subject(s)
Carrier Proteins/metabolism , Immunoblotting/methods , Immunoprecipitation/methods , Protein Interaction Domains and Motifs , Tumor Suppressor Proteins/metabolism , Humans
4.
Cancers (Basel) ; 12(12)2020 Dec 17.
Article in English | MEDLINE | ID: mdl-33348649

ABSTRACT

Lung cancer is the leading cause of cancer-related death worldwide. Lung cancer is commonly driven by mutations in the RAS oncogenes, the most frequently activated oncogene family in human disease. RAS-induced tumorigenesis is inhibited by the tumor suppressor RASSF1A, which induces apoptosis in response to hyperactivation of RAS. RASSF1A expression is suppressed in cancer at high rates, primarily owing to promoter hypermethylation. Recent reports have shown that loss of RASSF1A expression uncouples RAS from apoptotic signaling in vivo, thereby enhancing tumor aggressiveness. Moreover, a concomitant upregulation of RAS mitogenic signaling upon RASSF1A loss has been observed, suggesting RASSF1A may directly regulate RAS activation. Here, we present the first mechanistic evidence for control of RAS activation by RASSF1A. We present a novel interaction between RASSF1A and the Ras GTPase Activating Protein (RasGAP) DAB2IP, an important negative regulator of RAS. Using shRNA-mediated knockdown and stable overexpression approaches, we demonstrate that RASSF1A upregulates DAB2IP protein levels in NSCLC cells. Suppression of RASSF1A and subsequent downregulation of DAB2IP enhances GTP loading onto RAS, thus increasing RAS mitogenic signaling in both mutant- and wildtype-RAS cells. Moreover, co-suppression of RASSF1A and DAB2IP significantly enhances in vitro and in vivo growth of wildtype-RAS cells. Tumors expressing wildtype RAS, therefore, may still suffer from hyperactive RAS signaling when RASSF1A is downregulated. This may render them susceptible to the targeted RAS inhibitors currently in development.

5.
J Cell Sci ; 133(3)2020 02 10.
Article in English | MEDLINE | ID: mdl-32041893

ABSTRACT

Mutations that activate the RAS oncoproteins are common in cancer. However, aberrant upregulation of RAS activity often occurs in the absence of activating mutations in the RAS genes due to defects in RAS regulators. It is now clear that loss of function of Ras GTPase-activating proteins (RasGAPs) is common in tumors, and germline mutations in certain RasGAP genes are responsible for some clinical syndromes. Although regulation of RAS is central to their activity, RasGAPs exhibit great diversity in their binding partners and therefore affect signaling by multiple mechanisms that are independent of RAS. The RASSF family of tumor suppressors are essential to RAS-induced apoptosis and senescence, and constitute a barrier to RAS-mediated transformation. Suppression of RASSF protein expression can also promote the development of excessive RAS signaling by uncoupling RAS from growth inhibitory pathways. Here, we will examine how these effectors of RAS contribute to tumor suppression, through both RAS-dependent and RAS-independent mechanisms.


Subject(s)
Neoplasms , ras GTPase-Activating Proteins , Apoptosis , GTPase-Activating Proteins , Humans , Neoplasms/genetics , Signal Transduction
7.
Cancer Res ; 78(10): 2614-2623, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29735543

ABSTRACT

Mutant K-RAS has been shown to have both tumor-promoting and -suppressing functions, and growing evidence suggests that the RASSF family of tumor suppressors can act as RAS apoptosis and senescence effectors. It has been hypothesized that inactivation of the RASSF1A tumor suppressor facilitates K-RAS-mediated transformation by uncoupling it from apoptotic pathways such as the Hippo pathway. In human lung tumors, combined activation of K-RAS and inactivation of RASSF1A is closely associated with the development of the most aggressive and worst prognosis tumors. Here, we describe the first transgenic mouse model for activation of K-RAS in the lung in a RASSF1A-defective background. RASSF1A deficiency profoundly enhanced the development of K-RAS-driven lung tumors in vivo Analysis of these tumors showed loss of RASSF1A-uncoupled RAS from the proapoptotic Hippo pathway as expected. We also observed an upregulation of AKT and RALGEF signaling in the RASSF1A- tumors. Heterozygosity of RASSF1A alone mimicked many of the effects of RAS activation on mitogenic signaling in lung tissue, yet no tumors developed, indicating that nonstandard Ras signaling pathways may be playing a key role in tumor formation in vivo In addition, we observed a marked increase in inflammation and IL6 production in RASSF1A-deficient tumors. Thus, RASSF1A loss profoundly affects RAS-driven lung tumorigenesis and mitogenic signaling in vivo Deregulation of inflammatory pathways due to loss of RASSF1A may be essential for RAS-mediated tumorigenesis. These results may have considerable ramifications for future targeted therapy against RAS+/RASSF1A- tumors.Significance: A transgenic mouse model shows that suppression of RASSF1A dramatically enhances Ras-driven tumorigenesis and alters Ras signaling pathway activity.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/10/2614/F1.large.jpg Cancer Res; 78(10); 2614-23. ©2018 AACR.


Subject(s)
Cell Transformation, Neoplastic/pathology , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , A549 Cells , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , DNA Methylation/genetics , HEK293 Cells , Hippo Signaling Pathway , Humans , Interleukin-6/biosynthesis , Lung Neoplasms/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/genetics
8.
BMC Cancer ; 18(1): 421, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29653561

ABSTRACT

BACKGROUND: Dysregulation of microRNA (miRNA) expression is associated with hallmarks of aggressive tumor phenotypes, e.g., enhanced cell growth, proliferation, invasion, and anchorage independent growth in prostate cancer (PCa). METHODS: Serum-based miRNA profiling involved 15 men diagnosed with non-metastatic (stage I, III) and metastatic (stage IV) PCa and five age-matched disease-free men using miRNA arrays with select targets confirmed by quantitative real-time PCR (qRT-PCR). The effect of miR-186-5p inhibition or ectopic expression on cellular behavior of PCa cells (i.e., PC-3, MDA-PCa-2b, and LNCaP) involved the use bromodeoxyuridine (BrdU) incorporation, invasion, and colony formation assays. Assessment of the impact of miR-186-5p inhibition or overexpression on selected targets entailed microarray analysis, qRT-PCR, and/or western blots. Statistical evaluation used the modified t-test and ANOVA analysis. RESULTS: MiR-186-5p was upregulated in serum from PCa patients and metastatic PCa cell lines (i.e., PC-3, MDA-PCa-2b, LNCaP) compared to serum from disease-free individuals or a normal prostate epithelial cell line (RWPE1), respectively. Inhibition of miR-186-5p reduced cell proliferation, invasion, and anchorage-independent growth of PC-3 and/or MDA-PCa-2b PCa cells. AKAP12, a tumor suppressor target of miR-186-5p, was upregulated in PC-3 and MDA-PCa-2b cells transfected with a miR-186-5p inhibitor. Conversely, ectopic miR-186-5p expression in HEK 293 T cells decreased AKAP12 expression by 30%. Both pAKT and ß-catenin levels were down-regulated in miR-186-5p inhibited PCa cells. CONCLUSIONS: Our findings suggest miR-186-5p plays an oncogenic role in PCa. Inhibition of miR-186-5p reduced PCa cell proliferation and invasion as well as increased AKAP12 expression. Future studies should explore whether miR-186-5p may serve as a candidate prognostic indicator and a therapeutic target for the treatment of aggressive prostate cancer.


Subject(s)
MicroRNAs/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , A Kinase Anchor Proteins/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Circulating MicroRNA , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/blood , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Prostatic Neoplasms/blood , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reproducibility of Results , Transcriptome , beta Catenin/genetics
9.
Cancer Lett ; 400: 30-36, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28455242

ABSTRACT

The Ras genes are the most frequently mutated oncogenes in human cancer. However, Ras biology is quite complex. While Ras promotes tumorigenesis by regulating numerous growth promoting pathways, activated Ras can paradoxically also lead to cell cycle arrest, death, and Oncogene-Induced Senescence (OIS). OIS is thought to be a critical pathway that serves to protect cells against aberrant Ras signaling. Multiple reports have highlighted the importance of the p53 and Rb tumor suppressors in Ras mediated OIS. However, until recently, the molecular mechanisms connecting Ras to these proteins remained unknown. The RASSF family of tumor suppressors has recently been identified as direct effectors of Ras. One of these members, NORE1A (RASSF5), may be the missing link between Ras-induced senescence and the regulation of p53 and Rb. This occurs both quantitatively, by promoting protein stability, as well as qualitatively via promoting critical pro-senescent post-translational modifications. Here we review the mechanisms by which NORE1A can activate OIS as a barrier against Ras-mediated transformation, and how this could lead to improved therapeutic strategies against cancers having lost NORE1A expression.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cellular Senescence , Genes, Tumor Suppressor , Monomeric GTP-Binding Proteins/genetics , Neoplasms/genetics , Oncogenes , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Monomeric GTP-Binding Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Phenotype , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , ras Proteins/genetics , ras Proteins/metabolism
10.
Hepatology ; 65(5): 1462-1477, 2017 05.
Article in English | MEDLINE | ID: mdl-28090674

ABSTRACT

Hepatitis C virus (HCV) infection is a common risk factor for the development of liver cancer. The molecular mechanisms underlying this effect are only partially understood. Here, we show that the HCV protein, nonstructural protein (NS) 5B, directly binds to the tumor suppressor, NORE1A (RASSF5), and promotes its proteosomal degradation. In addition, we show that NORE1A colocalizes to sites of HCV viral replication and suppresses the replication process. Thus, NORE1A has antiviral activity, which is specifically antagonized by NS5B. Moreover, the suppression of NORE1A protein levels correlated almost perfectly with elevation of Ras activity in primary human samples. Therefore, NORE1A inactivation by NS5B may be essential for maximal HCV replication and may make a major contribution to HCV-induced liver cancer by shifting Ras signaling away from prosenescent/proapoptotic signaling pathways. CONCLUSION: HCV uses NS5B to specifically suppress NORE1A, facilitating viral replication and elevated Ras signaling. (Hepatology 2017;65:1462-1477).


Subject(s)
Hepacivirus/physiology , Monomeric GTP-Binding Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Carcinoma, Hepatocellular/virology , Down-Regulation , HEK293 Cells , Humans , Liver/metabolism , Liver/virology , Liver Neoplasms/virology , Proteasome Endopeptidase Complex/metabolism
11.
Mol Cell Oncol ; 3(3): e1055050, 2016 May.
Article in English | MEDLINE | ID: mdl-27314075

ABSTRACT

RAS-induced senescence is a protective mechanism to avoid unrestricted cell growth due to aberrant mitogenic signals; however, the exact mechanism by which RAS induces senescence is not known. We recently identified a novel pathway linking RAS to p53 via NORE1A and HIPK2 that mechanistically explains how Ras induces senescence.

12.
Semin Cell Dev Biol ; 58: 86-95, 2016 10.
Article in English | MEDLINE | ID: mdl-27288568

ABSTRACT

There are six core RASSF family proteins that contain conserved Ras Association domains and may serve as Ras effectors. They lack intrinsic enzymatic activity and appear to function as scaffolding and localization molecules. While initially being associated with pro-apoptotic signaling pathways such as Bax and Hippo, it is now clear that they can also connect Ras to a surprisingly broad range of signaling pathways that control senescence, inflammation, autophagy, DNA repair, ubiquitination and protein acetylation. Moreover, they may be able to impact the activation status of pro-mitogenic Ras effector pathways, such as the Raf pathway. The frequent epigenetic inactivation of RASSF genes in human tumors disconnects Ras from pro-death signaling systems, enhancing Ras driven transformation and metastasis. The best characterized members are RASSF1A and RASSF5 (NORE1A).


Subject(s)
Signal Transduction , Tumor Suppressor Proteins/metabolism , ras Proteins/metabolism , Animals , Humans , Models, Biological
13.
Oncotarget ; 7(23): 33821-31, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27034171

ABSTRACT

The TOR pathway is a vital component of cellular homeostasis that controls the synthesis of proteins, nucleic acids and lipids. Its core is the TOR kinase. Activation of the TOR pathway suppresses autophagy, which plays a vital but complex role in tumorigenesis. The TOR pathway is regulated by activation of the Ras-related protein Rheb, which can bind mTOR. The Hippo pathway is a major growth control module that regulates cell growth, differentiation and apoptosis. Its core consists of an MST/LATS kinase cascade that can be activated by the RASSF1A tumor suppressor. The TOR and Hippo pathways may be coordinately regulated to promote cellular homeostasis. However, the links between the pathways remain only partially understood. We now demonstrate that in addition to mTOR regulation, Rheb also impacts the Hippo pathway by forming a complex with RASSF1A. Using stable clones of two human lung tumor cell lines (NCI-H1792 and NCI-H1299) with shRNA-mediated silencing or ectopic overexpression of RASSF1A, we show that activated Rheb stimulates the Hippo pathway, but is suppressed in its ability to stimulate the TOR pathway. Moreover, by selectively labeling autophagic vacuoles we show that RASSF1A inhibits the ability of Rheb to suppress autophagy and enhance cell growth. Thus, we identify a new connection that impacts coordination of Hippo and TOR signaling. As RASSF1A expression is frequently lost in human tumors, the RASSF1A status of a tumor may impact not just its Hippo pathway status, but also its TOR pathway status.


Subject(s)
Lung Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Apoptosis , Autophagy , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Protein Binding , RNA Interference , Ras Homolog Enriched in Brain Protein/genetics , Time Factors , Transfection , Tumor Suppressor Proteins/genetics
14.
Cancers (Basel) ; 8(4)2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27023610

ABSTRACT

Mouse Double Minute 2 Homolog (MDM2) is a key negative regulator of the master tumor suppressor p53. MDM2 regulates p53 on multiple levels, including acting as an ubiquitin ligase for the protein, thereby promoting its degradation by the proteasome. MDM2 is oncogenic and is frequently found to be over-expressed in human tumors, suggesting its dysregulation plays an important role in human cancers. We have recently found that the Ras effector and RASSF (Ras Association Domain Family) family member RASSF5/NORE1A enhances the levels of nuclear p53. We have also found that NORE1A (Novel Ras Effector 1A) binds the substrate recognition component of the SCF-ubiquitin ligase complex ß-TrCP. Here, we now show that NORE1A regulates MDM2 protein levels by targeting it for ubiquitination by SCF-ß-TrCP. We also show the suppression of NORE1A protein levels enhances MDM2 protein expression. Finally, we show that MDM2 can suppress the potent senescence phenotype induced by NORE1A over-expression. Thus, we identify a mechanism by which Ras/NORE1A can modulate p53 protein levels. As MDM2 has several important targets in addition to p53, this finding has broad implications for cancer biology in tumor cells that have lost expression of NORE1A due to promoter methylation.

15.
Cell Cycle ; 15(17): 2263-4, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26919075

ABSTRACT

Although Ras is a potent oncogene in human tumors it has the paradoxical ability to promote Oncogene Induced Senescence (OIS). This appears to serve as a major barrier to Ras driven transformation in vivo. The signaling pathways used by Ras to promote senescence remain relatively poorly understood, but appear to invoke both the p53 and the Rb master tumor suppressors. Exactly how Ras communicates with p53 and Rb has remained something of a puzzle. NORE1A is a direct Ras effector that is frequently downregulated in human tumors. We have now found that it serves as a powerful Ras senescence effector. Moreover, we have defined signaling mechanisms that allows Ras to control both p53 and Rb post-translational modifications via the NORE1A scaffolding molecule. Indeed, NORE1A can be detected in complex with both p53 and Rb. Thus, by coupling Ras to both tumor suppressors, NORE1A forms a major component of the Ras senescence machinery and serves as the missing link between Ras and p53/Rb.


Subject(s)
Cellular Senescence , Monomeric GTP-Binding Proteins/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , ras Proteins/metabolism , Humans , Models, Biological
16.
J Biol Chem ; 291(6): 3114-23, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26677227

ABSTRACT

Mutations in the Ras oncogene are one of the most frequent events in human cancer. Although Ras regulates numerous growth-promoting pathways to drive transformation, it can paradoxically promote an irreversible cell cycle arrest known as oncogene-induced senescence. Although senescence has clearly been implicated as a major defense mechanism against tumorigenesis, the mechanisms by which Ras can promote such a senescent phenotype remain poorly defined. We have shown recently that the Ras death effector NORE1A plays a critical role in promoting Ras-induced senescence and connects Ras to the regulation of the p53 tumor suppressor. We now show that NORE1A also connects Ras to the regulation of a second major prosenescent tumor suppressor, the retinoblastoma (Rb) protein. We show that Ras induces the formation of a complex between NORE1A and the phosphatase PP1A, promoting the activation of the Rb tumor suppressor by dephosphorylation. Furthermore, suppression of Rb reduces NORE1A senescence activity. These results, together with our previous findings, suggest that NORE1A acts as a critical tumor suppressor node, linking Ras to both the p53 and the Rb pathways to drive senescence.


Subject(s)
Monomeric GTP-Binding Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Retinoblastoma Protein/metabolism , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , COS Cells , Cellular Senescence/genetics , Chlorocebus aethiops , HEK293 Cells , Hep G2 Cells , Humans , Monomeric GTP-Binding Proteins/genetics , Phosphorylation/genetics , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Retinoblastoma Protein/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Front Genet ; 6: 269, 2015.
Article in English | MEDLINE | ID: mdl-26379698

ABSTRACT

BRCA1 is a breast and ovarian tumor suppressor. Hereditary mutations in BRCA1 result in a predisposition to breast cancer, and BRCA1 expression is down-regulated in ~30% of sporadic cases. The function of BRCA1 remains poorly understood, but it appears to play an important role in DNA repair and the maintenance of genetic stability. Mouse models of BRCA1 deficiency have been developed in an attempt to understand the role of the gene in vivo. However, the subtle nature of BRCA1 function and the well-known discrepancies between human and murine breast cancer biology and genetics may limit the utility of mouse systems in defining the function of BRCA1 in cancer and validating the development of novel therapeutics for breast cancer. In contrast to mice, pig biological systems, and cancer genetics appear to more closely resemble their human counterparts. To determine if BRCA1 inactivation in pig cells promotes their transformation and may serve as a model for the human disease, we developed an immortalized porcine breast cell line and stably inactivated BRCA1 using miRNA. The cell line developed characteristics of breast cancer stem cells and exhibited a transformed phenotype. These results validate the concept of using pigs as a model to study BRCA1 defects in breast cancer and establish the first porcine breast tumor cell line.

18.
J Cell Biol ; 208(6): 777-89, 2015 Mar 16.
Article in English | MEDLINE | ID: mdl-25778922

ABSTRACT

The Ras oncoprotein is a key driver of cancer. However, Ras also provokes senescence, which serves as a major barrier to Ras-driven transformation. Ras senescence pathways remain poorly characterized. NORE1A is a novel Ras effector that serves as a tumor suppressor. It is frequently inactivated in tumors. We show that NORE1A is a powerful Ras senescence effector and that down-regulation of NORE1A suppresses senescence induction by Ras and enhances Ras transformation. We show that Ras induces the formation of a complex between NORE1A and the kinase HIPK2, enhancing HIPK2 association with p53. HIPK2 is a tumor suppressor that can induce either proapoptotic or prosenescent posttranslational modifications of p53. NORE1A acts to suppress its proapoptotic phosphorylation of p53 but enhance its prosenescent acetylation of p53. Thus, we identify a major new Ras signaling pathway that links Ras to the control of specific protein acetylation and show how NORE1A allows Ras to qualitatively modify p53 function to promote senescence.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Monomeric GTP-Binding Proteins/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/metabolism , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , COS Cells , Carcinogenesis/metabolism , Cellular Senescence , Chlorocebus aethiops , Enzyme Stability , HEK293 Cells , Hep G2 Cells , Humans , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction
19.
Langmuir ; 31(11): 3326-32, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25714501

ABSTRACT

Magnetic nanoparticle-supported lipid bilayers (SLBs) constructed around core-shell Fe3O4-SiO2 nanoparticles (SNPs) were prepared and evaluated as potential drug carriers. We describe how an oxime ether lipid can be mixed with SNPs to produce lipid-particle assemblies with highly positive ζ potential. To demonstrate the potential of the resultant cationic SLBs, the particles were loaded with either the anticancer drug doxorubicin or an amphiphilic analogue, prepared to facilitate integration into the supported lipid bilayer, and then examined in studies against MCF-7 breast cancer cells. The assemblies were rapidly internalized and exhibited higher toxicity than treatments with doxorubicin alone. The magnetic SLBs were also shown to increase the efficacy of unmodified doxorubicin.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Lipid Bilayers/chemistry , Magnetite Nanoparticles/chemistry , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Humans , MCF-7 Cells
20.
Mol Cell Biol ; 35(1): 277-87, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25368379

ABSTRACT

RASSF1A may be the most frequently inactivated tumor suppressor identified in human cancer so far. It is a proapoptotic Ras effector and plays an important role in the apoptotic DNA damage response (DDR). We now show that in addition to DDR regulation, RASSF1A also plays a key role in the DNA repair process itself. We show that RASSF1A forms a DNA damage-regulated complex with the key DNA repair protein xeroderma pigmentosum A (XPA). XPA requires RASSF1A to exert full repair activity, and RASSF1A-deficient cells exhibit an impaired ability to repair DNA. Moreover, a cancer-associated RASSF1A single-nucleotide polymorphism (SNP) variant exhibits differential XPA binding and inhibits DNA repair. The interaction of XPA with other components of the repair complex, such as replication protein A (RPA), is controlled in part by a dynamic acetylation/deacetylation cycle. We found that RASSF1A and its SNP variant differentially regulate XPA protein acetylation, and the SNP variant hyperstabilizes the XPA-RPA70 complex. Thus, we identify two novel functions for RASSF1A in the control of DNA repair and protein acetylation. As RASSF1A modulates both apoptotic DDR and DNA repair, it may play an important and unanticipated role in coordinating the balance between repair and death after DNA damage.


Subject(s)
DNA Repair , Gene Expression Regulation, Neoplastic , Tumor Suppressor Proteins/metabolism , Xeroderma Pigmentosum Group A Protein/metabolism , Animals , Apoptosis , Cell Line, Tumor , Comet Assay , DNA Damage , HEK293 Cells , Humans , Mice , Mice, Knockout , Polymorphism, Single Nucleotide , Replication Protein A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...