Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Genes Dev ; 26(10): 1055-69, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22549727

RESUMEN

Human melanocytic nevi (moles) are benign lesions harboring activated oncogenes, including BRAF. Although this oncogene initially acts mitogenically, eventually, oncogene-induced senescence (OIS) ensues. Nevi can infrequently progress to melanomas, but the mechanistic relationship with OIS is unclear. We show here that PTEN depletion abrogates BRAF(V600E)-induced senescence in human fibroblasts and melanocytes. Correspondingly, in established murine BRAF(V600E)-driven nevi, acute shRNA-mediated depletion of PTEN prompted tumor progression. Furthermore, genetic analysis of laser-guided microdissected human contiguous nevus-melanoma specimens recurrently revealed identical mutations in BRAF or NRAS in adjacent benign and malignant melanocytes. The PI3K pathway was often activated through either decreased PTEN or increased AKT3 expression in melanomas relative to their adjacent nevi. Pharmacologic PI3K inhibition in melanoma cells suppressed proliferation and induced the senescence-associated tumor suppressor p15(INK4B). This treatment also eliminated subpopulations resistant to targeted BRAF(V600E) inhibition. Our findings suggest that a significant proportion of melanomas arise from nevi. Furthermore, these results demonstrate that PI3K pathway activation serves as a rate-limiting event in this setting, acting at least in part by abrogating OIS. The reactivation of senescence features and elimination of cells refractory to BRAF(V600E) inhibition by PI3K inhibition warrants further investigation into the therapeutic potential of simultaneously targeting these pathways in melanoma.


Asunto(s)
Senescencia Celular , Melanoma/patología , Nevo/patología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/patología , Sustitución de Aminoácidos , Proliferación Celular , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Activación Enzimática , Fibroblastos/metabolismo , Fibroblastos/patología , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Humanos , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/metabolismo , Nevo/metabolismo , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Cutáneas/metabolismo , Valina/genética , Valina/metabolismo
2.
PLoS Biol ; 12(3): e1001807, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24618719

RESUMEN

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.


Asunto(s)
Daño del ADN , Reparación del ADN/fisiología , Genes ras/fisiología , Proteínas de Homeodominio/fisiología , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Células Cultivadas , Senescencia Celular/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Neoplasias Mamarias Experimentales/genética , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estrés Oxidativo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción
3.
J Pathol ; 229(1): 132-40, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22951975

RESUMEN

Undifferentiated pleomorphic sarcoma (UPS) is one of the most common soft tissue malignancies. Patients with large, high-grade sarcomas often develop fatal lung metastases. Understanding the mechanisms underlying sarcoma metastasis is needed to improve treatment of these patients. Micro-RNAs (miRNAs) are a class of small RNAs that post-transcriptionally regulate gene expression. Global alterations in miRNAs are frequently observed in a number of disease states including cancer. The signalling pathways that regulate miRNA biogenesis are beginning to emerge. To test the relevance of specific oncogenic mutations in miRNA biogenesis in sarcoma, we used primary soft tissue sarcomas expressing either Braf(V600E) or Kras(G12D). We found that Braf(V600E) mutant tumours, which have increased MAPK signalling, have higher levels of mature miRNAs and enhanced miRNA processing. To investigate the relevance of oncogene-dependent alterations in miRNA biogenesis, we introduced conditional mutations in Dicer and showed that Dicer haploinsufficiency promotes the development of distant metastases in an oncogene-dependent manner. These results demonstrate that a specific oncogenic mutation can cooperate with mutation in Dicer to promote tumour progression in vivo.


Asunto(s)
Diferenciación Celular , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Sarcoma/genética , Neoplasias de los Tejidos Blandos/genética , Animales , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Haploinsuficiencia , Sistema de Señalización de MAP Quinasas , Ratones , Mutación , Invasividad Neoplásica , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Sarcoma/metabolismo , Sarcoma/secundario , Neoplasias de los Tejidos Blandos/metabolismo , Neoplasias de los Tejidos Blandos/patología , Factores de Tiempo
4.
Proc Natl Acad Sci U S A ; 108(21): 8663-7, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21555580

RESUMEN

The phosphorylation of H2Ax on its S139 site, γH2Ax, is important during DNA double-strand repair and is considered necessary for assembly of repair complexes, but its functional role after other kinds of DNA damage is less clear. We have measured the survival of isogenic mouse cell lines with the H2Ax gene knocked out, and replaced with wild-type or mutant (S139A) H2Ax genes, exposed to a range of agents with varied mechanisms of DNA damage. Knockout and mutant cells were sensitive to γ-rays, etoposide, temozolamide, and endogenously generated reactive oxygen species, each of which can include double-strand breaks among their spectra of DNA lesions. The absence or mutation of H2Ax had no influence on sensitivity to cisplatin or mitomycin C. Although UV light induced the highest levels of γH2Ax, mutation of S139 had no influence on UV sensitivity or the UV DNA damage response. Complete loss of H2Ax reduced the survival of cells exposed to UV light and reduced pChk1 induction, suggesting that sites other than S139 may impact the ATR-pChk1 pathway. The relative intensity of γH2Ax measured in Western blots in wild-type cells did not correlate with the functional importance of γH2Ax. The use of γH2Ax as a general biomarker of DNA damage is therefore potentially misleading because it is not an unambiguous indicator of double-strand breaks, and a significant fraction of DNA repair, especially involving nucleotide excision or crosslink repair, can occur without functional involvement of γH2Ax.


Asunto(s)
Daño del ADN , Reparación del ADN/genética , Histonas/fisiología , Animales , Línea Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Técnicas de Inactivación de Genes , Histonas/genética , Ratones , Mutación , Fosforilación , Proteínas Quinasas/metabolismo , Transgenes
5.
Cancer Res ; 84(8): 1333-1351, 2024 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-38277141

RESUMEN

Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors are approved for breast cancer treatment and show activity against other malignancies, including KRAS-mutant non-small cell lung cancer (NSCLC). However, the clinical efficacy of CDK4/6 inhibitors is limited due to frequent drug resistance and their largely cytostatic effects. Through a genome-wide cDNA screen, we identified that bromodomain-containing protein 4 (BRD4) overexpression conferred resistance to the CDK4/6 inhibitor palbociclib in KRAS-mutant NSCLC cells. Inhibition of BRD4, either by RNA interference or small-molecule inhibitors, synergized with palbociclib to induce senescence in NSCLC cells and tumors, and the combination prolonged survival in a KRAS-mutant NSCLC mouse model. Mechanistically, BRD4-inhibition enhanced cell-cycle arrest and reactive oxygen species (ROS) accumulation, both of which are necessary for senescence induction; this in turn elevated GPX4, a peroxidase that suppresses ROS-triggered ferroptosis. Consequently, GPX4 inhibitor treatment selectively induced ferroptotic cell death in the senescent cancer cells, resulting in tumor regression. Cotargeting CDK4/6 and BRD4 also promoted senescence and ferroptosis vulnerability in pancreatic and breast cancer cells. Together, these findings reveal therapeutic vulnerabilities and effective combinations to enhance the clinical utility of CDK4/6 inhibitors. SIGNIFICANCE: The combination of cytostatic CDK4/6 and BRD4 inhibitors induces senescent cancer cells that are primed for activation of ferroptotic cell death by targeting GPX4, providing an effective strategy for treating cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Citostáticos , Ferroptosis , Neoplasias Pulmonares , Animales , Ratones , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Quinasa 4 Dependiente de la Ciclina , Proteínas Nucleares/metabolismo , Citostáticos/uso terapéutico , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Pulmonares/genética , Línea Celular Tumoral , Factores de Transcripción/metabolismo , Quinasa 6 Dependiente de la Ciclina , Inhibidores de Proteínas Quinasas/farmacología
6.
Mol Cancer Res ; 20(5): 770-781, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35046109

RESUMEN

Oncogenic activation of the RTK-RAS-RAF-MEK-ERK pathway occurs in approximately 25% of all human cancers, yet activated RAS, BRAF, or MEK expression in primary cells leads to a prolonged and predominantly irreversible cell-cycle arrest termed oncogene-induced senescence (OIS). OIS acts as an intrinsic tumor suppressor mechanism, serving as a barrier to tumor progression. Screening a library of activated kinases and kinase-regulatory proteins we identified MOB3A, a Mps-one binder coactivator (MOB) protein family member, whose constitutive expression permits proliferation and suppresses senescence in response to oncogenic RAS and BRAF signals. MOB3A is one of seven human MOB genes, which are highly conserved from yeast to human and that function to activate the Hippo pathway kinases (MST/LATS) or NDR kinases through direct association. Here we show that within the MOB family of genes MOB3A and C are unique in their ability to allow primary cell proliferation in the face of sustained oncogene signaling. Unlike the canonical MOB1A/B proteins, MOB3A inhibits Hippo/MST/LATS signaling and constitutive MOB3A membrane localization phenocopies OIS bypass seen with elevated YAP expression. Moreover, inhibition of MOB3 family member expression results in decreased proliferation and tumor growth of cancer cell lines. Together these data identify MOB3A's role in bypass of oncogene induced senescence and its role as a Hippo pathway inhibitor. IMPLICATIONS: These results suggest that MOB3 targeting to re-engage the Hippo pathway, or direct targeting of YAP/TAZ, may be viable therapeutic strategies potential for RAS-pathway driven tumours.


Asunto(s)
Genes ras , Proteínas Asociadas a Microtúbulos , Proteínas Proto-Oncogénicas B-raf , Humanos , Senescencia Celular , Vía de Señalización Hippo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal
7.
Nat Cell Biol ; 6(6): 515-22, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15156151

RESUMEN

Clinical studies have revealed that cancer patients whose tumours have increased ErbB2 expression tend to have more aggressive, metastatic disease, which is associated with parameters predicting a poor outcome. The molecular basis underlying ErbB2-dependent cell motility and metastases formation, however, still remains poorly understood. In this study, we show that activation of a set of signalling molecules, including MAPK, phosphatidylinositol-3-OH kinase (PI(3)K) and Src, is required for Neu/ErbB2-dependent lamellipodia formation and for motility of breast carcinoma cells. Stimulation of these molecules, however, failed to induce efficient cell migration in the absence of Neu/ErbB2 phosphorylation at Tyr 1201 or Tyr 1227. We describe a novel molecule, Memo (mediator of ErbB2-driven cell motility), that interacts with a phospho-Tyr 1227-containing peptide, most probably through the Shc adaptor protein. After Neu/ErbB2 activation, Memo-defective cells form actin fibres and grow lamellipodia, but fail to extend microtubules towards the cell cortex. Our data suggest that Memo controls cell migration by relaying extracellular chemotactic signals to the microtubule cytoskeleton.


Asunto(s)
Proteínas Portadoras/metabolismo , Movimiento Celular/fisiología , Proteínas de la Membrana/metabolismo , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Receptor ErbB-2/genética , Transducción de Señal/genética , Citoesqueleto de Actina/metabolismo , Humanos , Proteínas de la Membrana/aislamiento & purificación , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Seudópodos/metabolismo , Receptor ErbB-2/metabolismo , Células Tumorales Cultivadas , Familia-src Quinasas/metabolismo
8.
Mol Cancer Res ; 19(5): 874-885, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33500359

RESUMEN

Cellular senescence is characterized by a prolonged and predominantly irreversible cell-cycle arrest state, which is linked to loss of tissue function and aging in mammals. Moreover, in response to aberrant oncogenic signals such as those from oncogenic RAS or BRAF, senescence functions as an intrinsic tumor suppressor mechanism restraining tumor progression. In addition to this durable proliferative block, senescent cells adopt altered morphologies, transcriptional profiles, and metabolism, while often possessing unusual heterochromatin formation termed senescence-associated heterochromatic foci. To uncover genes that are required to permit proliferation in the face of sustained oncogene signaling, we conducted an shRNA-based genetic screen in primary cells expressing inducible BRAF. Here we show that depletion of a known glycolysis regulator, islet amylin polypeptide (IAPP also known as amylin), prevents RAS and BRAF oncogene-induced senescence (OIS) in human cells. Importantly, depletion of IAPP resulted in changes of the cells' metabolome and this metabolic reprogramming was associated with widespread alterations in chromatin modifications compared with senescent cells. Conversely, exogenous treatment of IAPP-depleted cells with amylin restored OIS. Together, our results demonstrate that the metabolic regulator IAPP is important regulator of OIS. Moreover, they suggest that IAPP analog treatment or activation of IAPP signaling in RAS/BRAF mutant tumors may have therapeutic potential through senescence induction. IMPLICATIONS: These findings demonstrate that IAPP is a novel metabolic regulator of oncogene-induced senescence and use of IAPP analogs may be therapeutically effective to restore growth arrest to BRAF and/or RAS mutant cancers.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas ras/genética , Proliferación Celular/fisiología , Senescencia Celular/fisiología , Células HEK293 , Humanos , Mutación , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas ras/metabolismo
9.
J Clin Invest ; 131(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33690225

RESUMEN

Melanomas commonly undergo a phenotype switch, from a proliferative to an invasive state. Such tumor cell plasticity contributes to immunotherapy resistance; however, the mechanisms are not completely understood and thus are therapeutically unexploited. Using melanoma mouse models, we demonstrated that blocking the MNK1/2-eIF4E axis inhibited melanoma phenotype switching and sensitized melanoma to anti-PD-1 immunotherapy. We showed that phospho-eIF4E-deficient murine melanomas expressed high levels of melanocytic antigens, with similar results verified in patient melanomas. Mechanistically, we identified phospho-eIF4E-mediated translational control of NGFR, a critical effector of phenotype switching. Genetic ablation of phospho-eIF4E reprogrammed the immunosuppressive microenvironment, exemplified by lowered production of inflammatory factors, decreased PD-L1 expression on dendritic cells and myeloid-derived suppressor cells, and increased CD8+ T cell infiltrates. Finally, dual blockade of the MNK1/2-eIF4E axis and the PD-1/PD-L1 immune checkpoint demonstrated efficacy in multiple melanoma models regardless of their genomic classification. An increase in the presence of intratumoral stem-like TCF1+PD-1+CD8+ T cells, a characteristic essential for durable antitumor immunity, was detected in mice given a MNK1/2 inhibitor and anti-PD-1 therapy. Using MNK1/2 inhibitors to repress phospho-eIF4E thus offers a strategy to inhibit melanoma plasticity and improve response to anti-PD-1 immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Factor 4E Eucariótico de Iniciación/inmunología , Inmunidad Celular , Sistema de Señalización de MAP Quinasas/inmunología , Melanoma Experimental/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Línea Celular Tumoral , Factor 4E Eucariótico de Iniciación/genética , Inmunoterapia , Sistema de Señalización de MAP Quinasas/genética , Melanoma Experimental/genética , Melanoma Experimental/terapia , Ratones , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Proteínas Serina-Treonina Quinasas/genética , Receptor de Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/inmunología
10.
J Clin Invest ; 134(9)2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690739
11.
Mol Biol Cell ; 16(2): 550-61, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15548598

RESUMEN

Activation of the hepatocyte growth factor receptor Met induces a morphogenic response and stimulates the formation of branching tubules by Madin-Darby canine kidney (MDCK) epithelial cells in three-dimensional cultures. A constitutively activated ErbB2/Neu receptor, NeuNT, promotes a similar invasive morphogenic program in MDCK cells. Because both receptors are expressed in breast epithelia, are associated with poor prognosis, and hepatocyte growth factor (HGF) is expressed in stroma, we examined the consequence of cooperation between these signals. We show that HGF disrupts NeuNT-induced epithelial morphogenesis, stimulating the breakdown of cell-cell junctions, dispersal, and invasion of single cells. This correlates with a decrease in junctional proteins claudin-1 and E-cadherin, in addition to the internalization of the tight junction protein ZO-1. HGF-induced invasion of NT-expressing cells is abrogated by pretreatment with a pharmacological inhibitor of the mitogen-activated protein kinase kinase (MEK) pathway, which restores E-cadherin and ZO-1 at cell-cell junctions, establishing the involvement of MEK-dependent pathways in this process. These results demonstrate that physiological signals downstream from the HGF/Met receptor synergize with ErbB2/Neu to enhance the malignant phenotype, promoting the breakdown of cell-cell junctions and enhanced cell invasion. This is particularly important for cancers where ErbB2/Neu is overexpressed and HGF is a physiological growth factor found in the stroma.


Asunto(s)
Movimiento Celular , Células Epiteliales/citología , Genes erbB-2 , Factor de Crecimiento de Hepatocito/metabolismo , Morfogénesis , Animales , Anoicis , Western Blotting , Cadherinas/metabolismo , Línea Celular , Claudina-1 , Colágeno/metabolismo , Perros , Combinación de Medicamentos , Matriz Extracelular/metabolismo , Laminina/metabolismo , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Modelos Biológicos , Fosfoproteínas/metabolismo , Pruebas de Precipitina , Proteoglicanos/metabolismo , Receptor ErbB-2/metabolismo , Uniones Estrechas , Proteína de la Zonula Occludens-1
12.
Oncogene ; 24(51): 7599-607, 2005 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-16170374

RESUMEN

c-Src associates with and is activated by the ErbB-2 receptor tyrosine kinase, but is unable to bind the EGFR. Although c-Src has been found to interact directly and specifically with the ErbB-2 receptor, the significance of this interaction is unclear. Using both chimeric receptor and site-directed mutagenesis approaches, the region of interaction of c-Src on ErbB-2 was identified. Significantly, EGFR could be converted into a receptor capable of binding c-Src by replacement of a catalytic domain of ErbB-2. We further demonstrated that MDCK cells that express mutant EGFR that are competent in c-Src recruitment lose epithelial polarity in organoid cultures, whereas cells overexpressing the wild-type EGFR retain a polarized phenotype. ErbB-2-dependent activation of c-Src results in disruption of epithelial cell-cell contacts leading to cell dispersal that correlates with the re-localization of phospho-MAPK to focal adhesions. Taken together, these observations suggest that recruitment of c-Src to these closely related EGFR family members plays a critical role in modulating cell polarity.


Asunto(s)
Polaridad Celular/fisiología , Transformación Celular Neoplásica , Receptores ErbB/genética , Proteínas Tirosina Quinasas/metabolismo , Receptor ErbB-2/fisiología , Animales , Sitios de Unión , Proteína Tirosina Quinasa CSK , Dominio Catalítico , Diferenciación Celular , Línea Celular , Perros , Receptores ErbB/fisiología , Adhesiones Focales , Humanos , Riñón/citología , Mutagénesis Sitio-Dirigida , Mutación , Ratas , Proteínas Recombinantes de Fusión , Transducción de Señal , Familia-src Quinasas
13.
Methods Mol Biol ; 1448: 3-21, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27317169

RESUMEN

Elucidating gene function is heavily reliant on the ability to modulate gene expression in biological model systems. Although transient expression systems can provide useful information about the biological outcome resulting from short-term gene overexpression or silencing, methods providing stable integration of desired expression constructs (cDNA or RNA interference) are often preferred for functional studies. To this end, lentiviral vectors offer the ability to deliver long-term and regulated gene expression to mammalian cells, including the expression of gene targeting small hairpin RNAs (shRNAmirs). Unfortunately, constructing vectors containing the desired combination of cDNAs, markers, and shRNAmirs can be cumbersome and time-consuming if using traditional sequence based restriction enzyme and ligation-dependent methods. Here we describe the use of a recombination based Gateway cloning strategy to rapidly and efficiently produce recombinant lentiviral vectors for the expression of one or more cDNAs with or without simultaneous shRNAmir expression. Additionally, we describe a luciferase-based approach to rapidly triage shRNAs for knockdown efficacy and specificity without the need to create stable shRNAmir expressing cells.


Asunto(s)
ADN Complementario/genética , Técnicas de Silenciamiento del Gen/métodos , Vectores Genéticos , Lentivirus/genética , Expresión Génica/genética , Humanos , Interferencia de ARN , ARN Interferente Pequeño/genética
14.
Oncogene ; 22(13): 1916-26, 2003 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-12673197

RESUMEN

The ErbB-2/Neu receptor tyrosine kinase plays a causal role in tumorigenesis in mammals. Neu's carboxyl terminus contains five phosphorylated tyrosines that mediate transformation through interaction with cytoplasmic SH2 or PTB containing adaptor proteins. We show that Drosophila adaptors signal from individual phosphotyrosine sites of rat Neu. Activated Neu expression in the midline glia suppressed apoptosis, similar to that seen with activated Drosophila EGF-R expression. Expression in eye and wing tissues generated graded phenotypes suitable for dosage-sensitive modifier genetics. Suppression of ErbB-2/Neu-induced phenotypes in tissues haplosufficient for genes encoding adaptor protein or second messengers suggests that pTyr 1227(YD) signals require Shc, and that pTyr 1253 (YE) signalling does not employ Ras, but does require Raf function. Signalling from pTyr (YB) was affected by a haplosufficiency in drk (Grb-2), and in genes thought to function downstream of Grb-2: dab, sos, csw (Shp-2), and dos (Gab-1). These data demonstrate the power of Drosophila genetics to unmask the molecules that signal from oncogenic ErbB-2/Neu.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular , Proteínas de Drosophila/fisiología , Fosfotirosina/química , Proteínas Quinasas , Procesamiento Proteico-Postraduccional , Receptor ErbB-2/química , Transducción de Señal/fisiología , Animales , Animales Modificados Genéticamente , Apoptosis/genética , Apoptosis/fisiología , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Receptores ErbB/fisiología , Ojo/crecimiento & desarrollo , Proteínas del Ojo/genética , Proteínas del Ojo/fisiología , Dosificación de Gen , Regulación del Desarrollo de la Expresión Génica , Morfogénesis/genética , Morfogénesis/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Fenotipo , Fosforilación , Estructura Terciaria de Proteína , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/fisiología , Proteínas Tirosina Fosfatasas no Receptoras , Proteínas/genética , Proteínas/fisiología , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/fisiología , Ratas , Receptor ErbB-2/fisiología , Receptores de Péptidos de Invertebrados/fisiología , Proteínas Recombinantes de Fusión/fisiología , Proteínas Adaptadoras de la Señalización Shc , Transducción de Señal/genética , Proteína Son Of Sevenless Drosofila/genética , Proteína Son Of Sevenless Drosofila/fisiología , Relación Estructura-Actividad , Alas de Animales/crecimiento & desarrollo
15.
Cancer Res ; 75(15): 3167-80, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26001956

RESUMEN

Lung carcinogenesis is a multistep process in which normal lung epithelial cells are converted to cancer cells through the sequential acquisition of multiple genetic or epigenetic events. Despite the utility of current genetically engineered mouse (GEM) models of lung cancer, most do not allow temporal dissociation of the cardinal events involved in lung tumor initiation and cancer progression. Here we describe a novel two-switch GEM model for BRAF(V600E)-induced lung carcinogenesis allowing temporal dissociation of these processes. In mice carrying a Flp recombinase-activated allele of Braf (Braf(FA)) in conjunction with Cre-regulated alleles of Trp53, Cdkn2a, or c-MYC, we demonstrate that secondary genetic events can promote bypass of the senescence-like proliferative arrest displayed by BRAF(V600E)-induced lung adenomas, leading to malignant progression. Moreover, restoring or activating TP53 in cultured BRAF(V600E)/TP53(Null) or BRAF(V600E)/INK4A-ARF(Null) lung cancer cells triggered a G1 cell-cycle arrest regardless of p19(ARF) status. Perhaps surprisingly, neither senescence nor apoptosis was observed upon TP53 restoration. Our results establish a central function for the TP53 pathway in restricting lung cancer development, highlighting the mechanisms that limit malignant progression of BRAF(V600E)-initiated tumors.


Asunto(s)
Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas B-raf/genética , Proteína p53 Supresora de Tumor/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenoma/genética , Adenoma/patología , Animales , Benzamidas/farmacología , Puntos de Control del Ciclo Celular/genética , Proliferación Celular , Supervivencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Difenilamina/análogos & derivados , Difenilamina/farmacología , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Neoplasias Pulmonares/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Ratones Transgénicos , Proteína p53 Supresora de Tumor/metabolismo
16.
Cancer Cell ; 27(1): 41-56, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25584893

RESUMEN

Braf(V600E) induces benign, growth-arrested melanocytic nevus development, but also drives melanoma formation. Cdkn2a loss in Braf(V600E) melanocytes in mice results in rare progression to melanoma, but only after stable growth arrest as nevi. Immediate progression to melanoma is prevented by upregulation of miR-99/100, which downregulates mTOR and IGF1R signaling. mTORC1 activation through Stk11 (Lkb1) loss abrogates growth arrest of Braf(V600E) melanocytic nevi, but is insufficient for complete progression to melanoma. Cdkn2a loss is associated with mTORC2 and Akt activation in human and murine melanocytic neoplasms. Simultaneous Cdkn2a and Lkb1 inactivation in Braf(V600E) melanocytes results in activation of both mTORC1 and mTORC2/Akt, inducing rapid melanoma formation in mice. In this model, activation of both mTORC1/2 is required for Braf-induced melanomagenesis.


Asunto(s)
Melanoma Experimental/patología , Complejos Multiproteicos/metabolismo , Nevo/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP , Animales , Línea Celular Tumoral , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Melanocitos/metabolismo , Melanoma Experimental/metabolismo , Ratones , MicroARNs/metabolismo , Datos de Secuencia Molecular , Mutación , Nevo/patología , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Neoplasias Cutáneas/patología
17.
PLoS One ; 9(1): e84745, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24489653

RESUMEN

Lung cancer is currently the most deadly malignancy in industrialized countries and accounts for 18% of all cancer-related deaths worldwide. Over 70% of patients with non-small cell lung cancer (NSCLC) are diagnosed at a late stage, with a 5-year survival below 10%. KRAS and the EGFR are frequently mutated in NSCLC and while targeted therapies for patients with EGFR mutations exist, oncogenic KRAS is thus far not druggable. KRAS activates multiple signalling pathways, including the PI3K/Akt pathway, the Raf-Mek-Erk pathway and the RalGDS/Ral pathway. Lung-specific expression of BrafV600E, the most prevalent BRAF mutation found in human tumors, results in Raf-Mek-Erk pathway activation and in the formation of benign adenomas that undergo widespread senescence in a Cre-activated Braf mouse model (Braf(CA)). However, oncogenic KRAS expression in mice induces adenocarcinomas, suggesting additional KRAS-activated pathways cooperate with sustained RAF-MEK-ERK signalling to bypass the oncogene-induced senescence proliferation arrest. To determine which KRAS effectors were responsible for tumor progression, we created four effector domain mutants (S35, G37, E38 and C40) in G12V-activated KRAS and expressed these alone or with BrafV600E in mouse lungs... The S35 and E38 mutants bind to Raf proteins but not PI3K or RalGDS; the G37 mutant binds to RalGDS and not Raf or PI3K and the C40 mutant is specific to PI3K. We designed lentiviral vectors to code for Cre recombinase along with KRAS mutants (V12, V12/S35, V12/G37, V12/E38 or V12/C40) or EGFP as a negative control.. These lentiviruses were used to infect Braf(CA) and wild-type mice. Surprisingly there was a significant decrease in tumor number and penetrance with each KRAS effector domain mutant relative to controls, suggesting that KRAS directly activates effectors with tumor suppressive functions.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas ras/metabolismo , Animales , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Neoplasias Pulmonares/genética , Ratones , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas ras/genética
18.
Mol Cancer Res ; 11(12): 1530-41, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24152792

RESUMEN

UNLABELLED: Mutational activation of BRAF leading to expression of the BRAF(V600E) oncoprotein was recently identified in a high percentage of specific hematopoietic neoplasms in monocyte/histiocyte and mature B-cell lineages. Although BRAF(V600E) is a driver oncoprotein and pharmacologic target in solid tumors such as melanoma, lung, and thyroid cancer, it remains unknown whether BRAF(V600E) is an appropriate therapeutic target in hematopoietic neoplasms. To address this critical question, we generated a mouse model expressing inducible BRAF(V600E) in the hematopoietic system, and evaluated the efficacy of pathway-targeted therapeutics against primary hematopoietic cells. In this model, BRAF(V600E) expression conferred cytokine-independent growth to monocyte/macrophage-lineage progenitors leading to aberrant in vivo and in vitro monocyte/macrophage expansion. Furthermore, transplantation of BRAF(V600E)-expressing bone marrow cells promoted an in vivo pathology most notable for monocytosis in hematopoietic tissues and visceral organs. In vitro analysis revealed that MAP-ERK kinase inhibition, but not RAF inhibition, effectively suppressed cytokine-independent clonal growth of monocyte/macrophage-lineage progenitors. However, combined RAF and phosphoinositide 3-kinase (PI3K) inhibition effectively inhibited cytokine-independent colony formation, suggesting autocrine PI3K pathway activation. Taken together, these results provide evidence that constitutively activated BRAF(V600E) drives aberrant proliferation of monocyte-lineage cells. IMPLICATIONS: This study supports the development of pathway-targeted therapeutics in the treatment of BRAF(V600E)-expressing hematopoietic neoplasms in the monocyte/histiocyte lineage.


Asunto(s)
Resistencia a Antineoplásicos , Indoles/farmacología , Células Precursoras de Monocitos y Macrófagos/fisiología , Monocitos/fisiología , Proteínas Proto-Oncogénicas B-raf/metabolismo , Sulfonamidas/farmacología , Quinasas raf/antagonistas & inhibidores , Animales , Benzamidas/farmacología , Trasplante de Médula Ósea , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Difenilamina/análogos & derivados , Difenilamina/farmacología , Eritropoyesis , Furanos/farmacología , Regulación Neoplásica de la Expresión Génica , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Precursoras de Monocitos y Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Mielopoyesis , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Piridinas/farmacología , Pirimidinas/farmacología , Transducción de Señal
19.
PLoS One ; 8(10): e76279, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24146852

RESUMEN

The ability to express exogenous cDNAs while suppressing endogenous genes via RNAi represents an extremely powerful research tool with the most efficient non-transient approach being accomplished through stable viral vector integration. Unfortunately, since traditional restriction enzyme based methods for constructing such vectors are sequence dependent, their construction is often difficult and not amenable to mass production. Here we describe a non-sequence dependent Gateway recombination cloning system for the rapid production of novel lentiviral (pLEG) and retroviral (pREG) vectors. Using this system to recombine 3 or 4 modular plasmid components it is possible to generate viral vectors expressing cDNAs with or without inhibitory RNAs (shRNAmirs). In addition, we demonstrate a method to rapidly produce and triage novel shRNAmirs for use with this system. Once strong candidate shRNAmirs have been identified they may be linked together in tandem to knockdown expression of multiple targets simultaneously or to improve the knockdown of a single target. Here we demonstrate that these recombinant vectors are able to express cDNA and effectively knockdown protein expression using both cell culture and animal model systems.


Asunto(s)
Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Vectores Genéticos/genética , Lentivirus/genética , Retroviridae/genética , Animales , Biomarcadores/metabolismo , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Células 3T3 NIH , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción Genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Cancer Res ; 72(18): 4765-76, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22836754

RESUMEN

Both the PI3K → Akt → mTOR and mitogen-activated protein kinase (MAPK) signaling pathways are often deregulated in prostate tumors with poor prognosis. Here we describe a new genetically engineered mouse model of prostate cancer in which PI3K-Akt-mTOR signaling is activated by inducible disruption of PTEN, and extracellular signal-regulated kinase 1/2 (ERK1/2) MAPK signaling is activated by inducible expression of a BRAF(V600E) oncogene. These tissue-specific compound mutant mice develop lethal prostate tumors that are inherently resistant to castration. These tumors bypass cellular senescence and disseminate to lymph nodes, bone marrow, and lungs where they form overt metastases in approximately 30% of the cases. Activation of PI3K → Akt → mTOR and MAPK signaling pathways in these prostate tumors cooperate to upregulate c-Myc. Accordingly, therapeutic treatments with rapamycin and PD0325901 to target these pathways, respectively, attenuate c-Myc levels and reduce tumor and metastatic burden. Together, our findings suggest a generalized therapeutic approach to target c-Myc activation in prostate cancer by combinatorial targeting of the PI3K → Akt → mTOR and ERK1/2 MAPK signaling pathways.


Asunto(s)
Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal/fisiología , Animales , Western Blotting , Modelos Animales de Enfermedad , Activación Enzimática/fisiología , Perfilación de la Expresión Génica , Inmunoprecipitación , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA