Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cancer Cell ; 40(10): 1111-1127.e9, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36113478

RESUMEN

Glioblastoma (GBM) is poorly responsive to therapy and invariably lethal. One conceivable strategy to circumvent this intractability is to co-target distinctive mechanistic components of the disease, aiming to concomitantly disrupt multiple capabilities required for tumor progression and therapeutic resistance. We assessed this concept by combining vascular endothelial growth factor (VEGF) pathway inhibitors that remodel the tumor vasculature with the tricyclic antidepressant imipramine, which enhances autophagy in GBM cancer cells and unexpectedly reprograms immunosuppressive tumor-associated macrophages via inhibition of histamine receptor signaling to become immunostimulatory. While neither drug is efficacious as monotherapy, the combination of imipramine with VEGF pathway inhibitors orchestrates the infiltration and activation of CD8 and CD4 T cells, producing significant therapeutic benefit in several GBM mouse models. Inclusion up front of immune-checkpoint blockade with anti-programmed death-ligand 1 (PD-L1) in eventually relapsing tumors markedly extends survival benefit. The results illustrate the potential of mechanism-guided therapeutic co-targeting of disparate biological vulnerabilities in the tumor microenvironment.


Asunto(s)
Glioblastoma , Animales , Antidepresivos Tricíclicos/metabolismo , Antidepresivos Tricíclicos/uso terapéutico , Autofagia , Antígeno B7-H1/metabolismo , Glioblastoma/patología , Imipramina/metabolismo , Imipramina/uso terapéutico , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Macrófagos/metabolismo , Ratones , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptor de Muerte Celular Programada 1 , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Cell Rep ; 28(8): 2064-2079.e11, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31433983

RESUMEN

Identifying cellular programs that drive cancers to be stem-like and treatment resistant is critical to improving outcomes in patients. Here, we demonstrate that constitutive extracellular signal-regulated kinase 1/2 (ERK1/2) activation sustains a stem-like state in glioblastoma (GBM), the most common primary malignant brain tumor. Pharmacological inhibition of ERK1/2 activation restores neurogenesis during murine astrocytoma formation, inducing neuronal differentiation in tumorspheres. Constitutive ERK1/2 activation globally regulates miRNA expression in murine and human GBMs, while neuronal differentiation of GBM tumorspheres following the inhibition of ERK1/2 activation requires the functional expression of miR-124 and the depletion of its target gene SOX9. Overexpression of miR124 depletes SOX9 in vivo and promotes a stem-like-to-neuronal transition, with reduced tumorigenicity and increased radiation sensitivity. Providing a rationale for reports demonstrating miR-124-induced abrogation of GBM aggressiveness, we conclude that reversal of an ERK1/2-miR-124-SOX9 axis induces a neuronal phenotype and that enforcing neuronal differentiation represents a therapeutic strategy to improve outcomes in GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Diferenciación Celular , Glioblastoma/patología , Sistema de Señalización de MAP Quinasas , MicroARNs/metabolismo , Neuronas/patología , Factor de Transcripción SOX9/metabolismo , Animales , Astrocitoma/genética , Astrocitoma/patología , Benzamidas/farmacología , Neoplasias Encefálicas/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Difenilamina/análogos & derivados , Difenilamina/farmacología , Progresión de la Enfermedad , Femenino , Glioblastoma/genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones Desnudos , MicroARNs/genética , Invasividad Neoplásica , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Tolerancia a Radiación/efectos de los fármacos
3.
Cancer Res ; 76(24): 7181-7193, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27754848

RESUMEN

Pancreatic cancer is a highly aggressive tumor, mostly resistant to the standard treatments. Nucleolin is overexpressed in cancers and its inhibition impairs tumor growth. Herein, we showed that nucleolin was overexpressed in human specimens of pancreatic ductal adenocarcinoma (PDAC) and that the overall survival significantly increased in patients with low levels of nucleolin. The nucleolin antagonist N6L strongly impaired the growth of primary tumors and liver metastasis in an orthotopic mouse model of PDAC (mPDAC). Similar antitumor effect of N6L has been observed in a highly angiogenic mouse model of pancreatic neuroendocrine tumor RIP-Tag2. N6L significantly inhibited both human and mouse pancreatic cell proliferation and invasion. Notably, the analysis of tumor vasculature revealed a strong increase of pericyte coverage and vessel perfusion both in mPDAC and RIP-Tag2 tumors, in parallel to an inhibition of tumor hypoxia. Nucleolin inhibition directly affected endothelial cell (EC) activation and changed a proangiogenic signature. Among the vascular activators, nucleolin inhibition significantly decreased angiopoietin-2 (Ang-2) secretion and expression in ECs, in the tumor and in the plasma of mPDAC mice. As a consequence of the observed N6L-induced tumor vessel normalization, pre-treatment with N6L efficiently improved chemotherapeutic drug delivery and increased the antitumor properties of gemcitabine in PDAC mice. In conclusion, nucleolin inhibition is a new anti-pancreatic cancer therapeutic strategy that dually blocks tumor progression and normalizes tumor vasculature, improving the delivery and efficacy of chemotherapeutic drugs. Moreover, we unveiled Ang-2 as a potential target and suitable response biomarker for N6L treatment in pancreatic cancer. Cancer Res; 76(24); 7181-93. ©2016 AACR.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Neovascularización Patológica/patología , Neoplasias Pancreáticas/patología , Péptidos/farmacología , Fosfoproteínas/antagonistas & inhibidores , Proteínas de Unión al ARN/antagonistas & inhibidores , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones , Análisis de Matrices Tisulares , Nucleolina
4.
Cancer Cell ; 28(4): 456-471, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26412325

RESUMEN

The associations of tricyclic antidepressants (TCAs) with reduced incidence of gliomas and elevated autophagy in glioma cells motivated investigation in mouse models of gliomagenesis. First, we established that imipramine, a TCA, increased autophagy and conveyed modest therapeutic benefit in tumor-bearing animals. Then we screened clinically approved agents suggested to affect autophagy for their ability to enhance imipramine-induced autophagy-associated cell death. The anticoagulant ticlopidine, which inhibits the purinergic receptor P2Y12, potentiated imipramine, elevating cAMP, a modulator of autophagy, reducing cell viability in culture, and increasing survival in glioma-bearing mice. Efficacy of the combination was obviated by knockdown of the autophagic regulatory gene ATG7, implicating cell-lethal autophagy. This seemingly innocuous combination of TCAs and P2Y12 inhibitors may have applicability for treating glioma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Imipramina/administración & dosificación , Ticlopidina/administración & dosificación , Animales , Anticoagulantes/administración & dosificación , Anticoagulantes/farmacología , Antidepresivos Tricíclicos/administración & dosificación , Antidepresivos Tricíclicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Autofagia/efectos de los fármacos , Proteína 7 Relacionada con la Autofagia , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Supervivencia Celular/efectos de los fármacos , Reposicionamiento de Medicamentos , Sinergismo Farmacológico , Técnicas de Silenciamiento del Gen , Glioma/mortalidad , Glioma/patología , Imipramina/farmacología , Ratones , Proteínas Asociadas a Microtúbulos/genética , Análisis de Supervivencia , Ticlopidina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
PLoS One ; 10(4): e0120348, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25927437

RESUMEN

Motivated by the recent implication of cysteine protease cathepsin L as a potential target for anti-cancer drug development, we used a conditional MycERTAM;Bcl-xL model of pancreatic neuroendocrine tumorigenesis (PNET) to assess the role of cathepsin L in Myc-induced tumor progression. By employing a cysteine cathepsin activity probe in vivo and in vitro, we first established that cathepsin activity increases during the initial stages of MycERTAM;Bcl-xL tumor development. Among the cathepsin family members investigated, only cathepsin L was predominately produced by beta-tumor cells in neoplastic pancreata and, consistent with this, cathepsin L mRNA expression was rapidly upregulated following Myc activation in the beta cell compartment. By contrast, cathepsins B, S and C were highly enriched in tumor-infiltrating leukocytes. Genetic deletion of cathepsin L had no discernible effect on the initiation of neoplastic growth or concordant angiogenesis. However, the tumors that developed in the cathepsin L-deficient background were markedly reduced in size relative to their typical wild-type counterparts, indicative of a role for cathepsin L in enabling expansive tumor growth. Thus, genetic blockade of cathepsin L activity is inferred to retard Myc-driven tumor growth, encouraging the potential utility of pharmacological inhibitors of cysteine cathepsins in treating late stage tumors.


Asunto(s)
Catepsina L/deficiencia , Transformación Celular Neoplásica/metabolismo , Neoplasias Experimentales/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Leucocitos/metabolismo , Leucocitos/patología , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(16): E1480-9, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23542378

RESUMEN

Based on clinical presentation, glioblastoma (GBM) is stratified into primary and secondary types. The protein 53 (p53) pathway is functionally incapacitated in most GBMs by distinctive type-specific mechanisms. To model human gliomagenesis, we used a GFAP-HRas(V12) mouse model crossed into the p53ER(TAM) background, such that either one or both copies of endogenous p53 is replaced by a conditional p53ER(TAM) allele. The p53ER(TAM) protein can be toggled reversibly in vivo between wild-type and inactive conformations by administration or withdrawal of 4-hydroxytamoxifen (4-OHT), respectively. Surprisingly, gliomas that develop in GFAP-HRas(V12);p53(+/KI) mice abrogate the p53 pathway by mutating p19(ARF)/MDM2 while retaining wild-type p53 allele. Consequently, such tumors are unaffected by restoration of their p53ER(TAM) allele. By contrast, gliomas arising in GFAP-HRas(V12);p53(KI/KI) mice develop in the absence of functional p53. Such tumors retain a functional p19(ARF)/MDM2-signaling pathway, and restoration of p53ER(TAM) allele triggers p53-tumor-suppressor activity. Congruently, growth inhibition upon normalization of mutant p53 by a small molecule, Prima-1, in human GBM cultures also requires p14(ARF)/MDM2 functionality. Notably, the antitumoral efficacy of p53 restoration in tumor-bearing GFAP-HRas(V12);p53(KI/KI) animals depends on the duration and frequency of p53 restoration. Thus, intermittent exposure to p53ER(TAM) activity mitigated the selective pressure to inactivate the p19(ARF)/MDM2/p53 pathway as a means of resistance, extending progression-free survival. Our results suggest that intermittent dosing regimes of drugs that restore wild-type tumor-suppressor function onto mutant, inactive p53 proteins will prove to be more efficacious than traditional chronic dosing by similarly reducing adaptive resistance.


Asunto(s)
Modelos Animales de Enfermedad , Glioblastoma/tratamiento farmacológico , Glioblastoma/fisiopatología , Transducción de Señal/fisiología , Tamoxifeno/análogos & derivados , Proteína p53 Supresora de Tumor/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Glioblastoma/metabolismo , Técnicas Histológicas , Humanos , Immunoblotting , Estimación de Kaplan-Meier , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Análisis de Secuencia de ADN , Transducción de Señal/genética , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico
7.
Nature ; 466(7303): 253-7, 2010 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-20613842

RESUMEN

Although it is known that the methylation of DNA in 5' promoters suppresses gene expression, the role of DNA methylation in gene bodies is unclear. In mammals, tissue- and cell type-specific methylation is present in a small percentage of 5' CpG island (CGI) promoters, whereas a far greater proportion occurs across gene bodies, coinciding with highly conserved sequences. Tissue-specific intragenic methylation might reduce, or, paradoxically, enhance transcription elongation efficiency. Capped analysis of gene expression (CAGE) experiments also indicate that transcription commonly initiates within and between genes. To investigate the role of intragenic methylation, we generated a map of DNA methylation from the human brain encompassing 24.7 million of the 28 million CpG sites. From the dense, high-resolution coverage of CpG islands, the majority of methylated CpG islands were shown to be in intragenic and intergenic regions, whereas less than 3% of CpG islands in 5' promoters were methylated. The CpG islands in all three locations overlapped with RNA markers of transcription initiation, and unmethylated CpG islands also overlapped significantly with trimethylation of H3K4, a histone modification enriched at promoters. The general and CpG-island-specific patterns of methylation are conserved in mouse tissues. An in-depth investigation of the human SHANK3 locus and its mouse homologue demonstrated that this tissue-specific DNA methylation regulates intragenic promoter activity in vitro and in vivo. These methylation-regulated, alternative transcripts are expressed in a tissue- and cell type-specific manner, and are expressed differentially within a single cell type from distinct brain regions. These results support a major role for intragenic methylation in regulating cell context-specific alternative promoters in gene bodies.


Asunto(s)
Encéfalo/metabolismo , Secuencia Conservada/genética , Metilación de ADN , Regiones Promotoras Genéticas/genética , Animales , Encéfalo/anatomía & histología , Encéfalo/citología , Proteínas Portadoras/genética , Línea Celular , Islas de CpG/genética , ADN Intergénico/genética , ADN Intergénico/metabolismo , Lóbulo Frontal/metabolismo , Regulación de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos , Persona de Mediana Edad , Proteínas del Tejido Nervioso , Especificidad de Órganos , Transcripción Genética/genética
8.
Cancer Cell ; 14(6): 447-57, 2008 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-19061836

RESUMEN

Deregulated Myc triggers a variety of intrinsic tumor suppressor programs that serve to restrain Myc's oncogenic potential. Since Myc activity is also required for normal cell proliferation, activation of intrinsic tumor suppression must be triggered only when Myc signaling is oncogenic. However, how cells discriminate between normal and oncogenic Myc is unknown. Here we show that distinct threshold levels of Myc govern its output in vivo: low levels of deregulated Myc are competent to drive ectopic proliferation of somatic cells and oncogenesis, but activation of the apoptotic and ARF/p53 intrinsic tumor surveillance pathways requires Myc overexpression. The requirement to keep activated oncogenes at a low level to avoid engaging tumor suppression is likely an important selective pressure governing the early stages of tumor microevolution.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Apoptosis , Núcleo Celular/metabolismo , Proliferación Celular , Fibroblastos/metabolismo , Genotipo , Humanos , Ratones , Ratones Transgénicos , Neoplasias/genética , Reacción en Cadena de la Polimerasa , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
9.
Nat Med ; 13(10): 1211-8, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17906636

RESUMEN

An association between inflammation and cancer has long been recognized, but the cause and effect relationship linking the two remains unclear. Myc is a pleiotropic transcription factor that is overexpressed in many human cancers and instructs many extracellular aspects of the tumor tissue phenotype, including remodeling of tumor stroma and angiogenesis. Here we show in a beta-cell tumor model that activation of Myc in vivo triggers rapid recruitment of mast cells to the tumor site-a recruitment that is absolutely required for macroscopic tumor expansion. In addition, treatment of established beta-cell tumors with a mast cell inhibitor rapidly triggers hypoxia and cell death of tumor and endothelial cells. Inhibitors of mast cell function may therefore prove therapeutically useful in restraining expansion and survival of pancreatic and other cancers.


Asunto(s)
Transformación Celular Neoplásica/genética , Mastocitos/metabolismo , Neovascularización Patológica/etiología , Neoplasias Pancreáticas/irrigación sanguínea , Proteínas Proto-Oncogénicas c-myc/fisiología , Animales , Células de la Médula Ósea/citología , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CCL5/metabolismo , Fémur/citología , Regulación Neoplásica de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica/genética , Neovascularización Patológica/fisiopatología , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/fisiopatología , Proteínas Proto-Oncogénicas c-myc/genética
10.
Cancer Res ; 67(15): 7059-61, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17671171

RESUMEN

Both tumors and normal tissues need a blood supply for oxygen, nutrients, and waste removal. However, whereas normal vasculature is hierarchically assembled into efficient networks of arteries, capillaries, and veins, the blood vessels of tumors are a mess-chaotic, leaky, inefficient, and barely making do. Why the difference? Do tumor vessels lack the signals to mature or, instead, is their maturation actively suppressed? What triggers and maintains tumor vasculature? In a recent study using a switchable Myc-driven mouse tumor model, we addressed these fundamental questions. We identified the inflammatory cytokine interleukin-1beta as an essential initiating trigger of vascular endothelial growth factor-dependent angiogenesis. Here, we consider how kinetic studies using regulatable forms of Myc or other oncogenes can shed new light on the way tumors initiate and maintain their aberrant blood supplies.


Asunto(s)
Inductores de la Angiogénesis/uso terapéutico , Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias/irrigación sanguínea , Neovascularización Patológica/tratamiento farmacológico , Animales , Humanos
11.
Genes Dev ; 20(18): 2527-38, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16980582

RESUMEN

Although induction of blood vessel growth is acknowledged as a pivotal requirement for the evolution of macroscopic tumors, the events that trigger onset of tumor angiogenesis remain largely obscure. The pervasive Myc oncoprotein is itself a potent inducer of angiogenesis in a wide range of tissues. We have used a reversibly switchable mouse transgenic model of Myc-dependent beta-cell carcinogenesis to delineate the kinetics and causal sequence of angiogenic processes following acute Myc activation. We show that onset of endothelial cell proliferation is induced shortly after Myc-induced cell cycle entry of beta cells. Endothelial cell proliferation is not indirectly induced by local tissue hypoxia but instead via a diffusible angiogenic signal produced by Myc-expressing beta cells. This signal triggers the release of pre-existing, sequestered VEGF from the islet extracellular matrix, that then homes to the endothelial compartment where it induces endothelial cell proliferation. Myc activation in beta cells rapidly induces expression and release of the proinflammatory cytokine interleukin 1beta (IL-1beta). We show that IL-1beta is the principal effector downstream of Myc responsible for triggering rapid onset of islet angiogenesis. Together, our data delineate a complete pathway in vivo by which the highly pleiotropic Myc oncoproteins elicits coexpansion of the vascular compartment during tumorigenic progression.


Asunto(s)
Insulinoma/irrigación sanguínea , Interleucina-1/fisiología , Neovascularización Patológica/etiología , Neoplasias Pancreáticas/irrigación sanguínea , Proteínas Proto-Oncogénicas c-myc/fisiología , Animales , Proliferación Celular , Células Endoteliales/patología , Regulación Neoplásica de la Expresión Génica , Genes myc , Técnicas In Vitro , Insulinoma/etiología , Insulinoma/genética , Insulinoma/fisiopatología , Islotes Pancreáticos/irrigación sanguínea , Ratones , Ratones Transgénicos , Neovascularización Patológica/genética , Neovascularización Patológica/fisiopatología , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/fisiopatología , Proteínas Proto-Oncogénicas c-myc/genética , Factor A de Crecimiento Endotelial Vascular/fisiología , Proteína bcl-X/genética
12.
Cancer Cell ; 10(2): 113-20, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16904610

RESUMEN

Overexpression of Bcl-xL, loss of p19 ARF, and loss of p53 all accelerate Myc oncogenesis. All three lesions are implicated in suppressing Myc-induced apoptosis, suggesting that this is a common mechanism by which they synergize with Myc. However, using an acutely switchable model of Myc-induced tumorigenesis, we demonstrate that each lesion cooperates with Myc in vivo by a distinct mechanism. While Bcl-xL blocks Myc-induced apoptosis, inactivation of p19 ARF enhances it. However, this increase in apoptosis is matched by increased Myc-induced proliferation. p53 inactivation shares features of both lesions, partially suppressing apoptosis while augmenting proliferation. Bcl-xL and p19 ARF loss together synergize to further accelerate Myc oncogenesis. Thus, differing lesions cooperate oncogenically with Myc by discrete mechanisms that can themselves synergize with each other.


Asunto(s)
Apoptosis , Transformación Celular Neoplásica , Genes myc/fisiología , Proteína p14ARF Supresora de Tumor/fisiología , Proteína bcl-X/fisiología , Animales , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Secretoras de Insulina/patología , Ratones , Ratones Transgénicos , Proteína p53 Supresora de Tumor/fisiología
13.
Cancer Res ; 66(9): 4591-601, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16651409

RESUMEN

Deregulated expression of the Myc transcription factor is a frequent causal mutation in human cancer. Thousands of putative Myc target genes have been identified in in vitro studies, indicating that Myc exerts highly pleiotropic effects within cells and tissues. However, the complexity and diversity of Myc gene targets has confounded attempts at identifying which of these genes are the critical targets mediating Myc-driven tumorigenesis in vivo. Acute activation of Myc in a reversibly switchable transgenic model of Myc-mediated beta cell tumorigenesis induces rapid tumor onset, whereas subsequent Myc deactivation triggers equally rapid tumor regression. Thus, sustained Myc activity is required for tumor maintenance. We have used this reversibly switchable kinetic tumor model in combination with high-density oligonucleotide microarrays to develop an unbiased strategy for identifying candidate Myc-regulated genes responsible for maintenance of Myc-dependent tumors. Consistent with known Myc functions, some Myc-regulated genes are involved in cell growth, cycle, and proliferation. In addition, however, many Myc-regulated genes are specific to beta cells, indicating that a significant component of Myc action is cell type specific. Finally, we identify a very restricted cadre of genes with expression that is inversely regulated upon Myc activation-induced tumor progression and deactivation-induced tumor regression. By definition, such genes are candidates for tumor maintenance functions. Combining reversibly switchable, transgenic models of tumor formation and regression with genomic profiling offers a novel strategy with which to deconvolute the complexities of oncogenic signaling pathways in vivo.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas c-myc/fisiología , Animales , Ciclo Celular/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Marcación de Gen , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/fisiología , Ratones , Ratones Transgénicos , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-myc/genética , Activación Transcripcional
14.
Nat Genet ; 37(6): 645-51, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15895082

RESUMEN

CpG islands are present in one-half of all human and mouse genes and typically overlap with promoters or exons. We developed a method for high-resolution analysis of the methylation status of CpG islands genome-wide, using arrays of BAC clones and the methylation-sensitive restriction enzyme NotI. Here we demonstrate the accuracy and specificity of the method. By computationally mapping all NotI sites, methylation events can be defined with single-nucleotide precision throughout the genome. We also demonstrate the unique expandability of the array method using a different methylation-sensitive restriction enzyme, BssHII. We identified and validated new CpG island loci that are methylated in a tissue-specific manner in normal human tissues. The methylation status of the CpG islands is associated with gene expression for several genes, including SHANK3, which encodes a structural protein in neuronal postsynaptic densities. Defects in SHANK3 seem to underlie human 22q13 deletion syndrome. Furthermore, these patterns for SHANK3 are conserved in mice and rats.


Asunto(s)
Proteínas Portadoras/metabolismo , Cromosomas Artificiales Bacterianos , Islas de CpG , Metilación de ADN , Animales , Secuencia Conservada , Desoxirribonucleasas de Localización Especificada Tipo II , Humanos , Ratones , Proteínas del Tejido Nervioso , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Secuencias Reguladoras de Ácidos Nucleicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Cell Cycle ; 3(12): 1606-11, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15611657

RESUMEN

We have previously demonstrated that overexpression of Cell Death Inhibiting RNA (CDIR), a portion of the 3'untranslated region (UTR) of KIAA0425, inhibits Interferon-gamma (IFN-gamma) induced apoptosis in HeLa cells (Shchors et al., J Biol Chem 2002; 277:47061-72). IFN-gamma is known to sensitize cells to killing induced by the death receptor ligands such as Fas/APO-1/CD95 and TNF-related apoptosis-inducing ligand (TRAIL/Apo-2L). Here we report that while CDIR does not alter the response of cells to Fas or TRAIL, it significantly modulates IFN-gamma-induced sensitization of HeLa cells to these death-inducing ligands. Interestingly, while CDIR abrogates the IFN-gamma-modulated sensitization to Fas, it enhances the sensitization to TRAIL. Expression of CDIR did not alter initial steps of IFN-gamma signaling including induction of Signal Transducer and Activator-1 (Stat1), caspase-1 or Interferon Regulatory Factor-1 (IRF1) transcription. In contrast, although expression of CDIR does not affect the protein level of caspase-1 or STAT1, it does significantly reduce the level of IRF1 protein. Thus, CDIR mediates IFN-gamma-induced apoptosis, at least in part, by reducing the level of the pro-apoptotic tumor suppressor gene IRF1 via a post-transcriptional mechanism. Since tumor cells are often less sensitive to Fas and more sensitive to TRAIL than normal cells, we suggest that CDIR or CDIR-like activity could contribute to such a phenotype of tumor cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Interferón gamma/metabolismo , Glicoproteínas de Membrana/metabolismo , ARN/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Receptor fas/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Caspasa 1/metabolismo , Células HeLa , Humanos , Inmunización , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Interferón gamma/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF , Transfección , Factor de Necrosis Tumoral alfa/genética
16.
Exp Cell Res ; 283(1): 17-21, 2003 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-12565816

RESUMEN

Constitutive expression of the proto-oncogene c-myc results in oncogenic activation and contributes to progression of a wide range of human and animal tumors. Myc executes its multiple activities mostly through transcriptional regulation of the target genes. The special interest of this review is the mechanism of transcriptional repression of cell cycle inhibitors by Myc. Myc suppresses expression of cell cycle/growth arrest genes gas1, p15, p21, p27, and gadd34, -45, and -153. It appears that Myc represses growth arrest gene transcription by at least two distinct mechanisms. One mechanism is limited to the binding of Myc-Max heterodimers to the Inr element in their promoters and inhibition of Miz-1 or other transcriptional activators via the C-terminal domain of c-Myc. This mechanism requires DNA binding of the Myc-Max complex to Inr sequences. The other mechanism is dependent on c-Myc binding to the Sp1 transcription factor via the c-Myc central region and inhibition of Sp1 transcriptional activity. At this time it is not entirely clear which Sp1-containing promoters will be repressed by c-Myc and what other modes of c-Myc transcriptional repression may exist. The ability of c-Myc to repress transcription of growth arrest genes may contribute to its potential to promote proliferation and oncogenesis.


Asunto(s)
Proteínas de Ciclo Celular/genética , Células Eucariotas/metabolismo , Genes Reguladores/genética , Genes cdc/fisiología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Represoras/genética , Animales , Proteínas de Ciclo Celular/metabolismo , División Celular/genética , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-myc/metabolismo
17.
Oncogene ; 22(6): 797-806, 2003 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-12584558

RESUMEN

Here we describe the Achilles' Heel Method (AHM), a new function-based approach for identification of inhibitors of signaling pathways, optimized for human cells. The principle of AHM is the identification of 'sensitizing' cDNAs based on their decreased abundance following selection. As a proof of principle, we have employed AHM for the identification of Fas/CD95/APO-1 pathway inhibitors. HeLa cells were transfected with an antisense cDNA expression library in an episomal vector followed by selection with a suboptimal dose of the apoptotic inducer. Antisense inactivation of Fas inhibitors rendered the cells more sensitive to apoptosis resulting in their preferential death and consequent loss of their sensitizing episomes that were identified by subtraction. We show that the resulting products were enriched for sensitizing cDNAs as seven out of eight candidates tested were confirmed as inhibitors of Fas-induced killing either by transfection or by pharmacological inhibition. Furthermore, we demonstrate by multiple approaches that one candidate, NF-E2 related factor 2 (Nrf2), is an inhibitor of Fas-induced apoptosis. Inactivation of Nrf2 by antisense or by a membrane permeable dominant-negative polypeptide sensitized cells while overexpression of Nrf2 protected cells from Fas-induced apoptosis. In addition, dicumarol, an inhibitor of the phase II detoxifying enzyme NQO1, a downstream target of Nrf2, sensitized cells. Nrf2 induces the production of Glutathione (GSH) and we demonstrated that N-acetyl L-cysteine (NAC), a precursor to GSH, protected cells from Fas-mediated killing. Taken together, AHM is a powerful approach for the identification of inhibitors of a signaling pathway with a low rate of false positives that opens new avenues for function profiling of human genes and discovery of new drug targets.


Asunto(s)
Apoptosis/fisiología , Proteínas de Unión al ADN/metabolismo , Perfilación de la Expresión Génica/métodos , Transducción de Señal/genética , Transactivadores/metabolismo , Receptor fas/metabolismo , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Factor 2 Relacionado con NF-E2 , Transactivadores/biosíntesis , Transactivadores/genética
18.
J Biol Chem ; 277(49): 47061-72, 2002 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-12356764

RESUMEN

Regulators of programmed cell death were previously identified using a technical knockout genetic screen. Among the elements that inhibited interferon-gamma-induced apoptosis of HeLa cells was a 441-nucleotide fragment derived from the 3'-untranslated region (UTR) of KIAA0425, a gene of unknown function. This fragment was termed cell death inhibiting RNA (CDIR). Deletion and mutation analyses of CDIR were employed to identify the features required for its anti-apoptotic activity. Single nucleotide alterations within either copy of the duplicated U-rich motif found in the CDIR sequence abolished the anti-apoptotic activity of CDIR and altered its in vitro association with a protein complex. Further analysis of the CDIR-binding complex indicated that it contained heat shock protein 27 (Hsp27) and the regulator of mRNA turnover AUF1 (heterogeneous nuclear ribonucleoprotein D). In addition, recombinant AUF1 bound directly to CDIR. Furthermore, expression of another AUF1-binding RNA element, derived from the 3'-UTR of c-myc, inhibited apoptosis. We also demonstrate that the level and the stability of p21(waf1/Cip1/sdi1) mRNA, a target of AUF1 with anti-apoptotic activity, were increased in CDIR-transfected cells. The level of mRNA and protein of Bcl-2, another anti-apoptotic gene, containing an AUF1 binding site in its 3'-UTR was also increased in CDIR-transfected cells. Our data suggest that AUF1 regulates apoptosis by altering mRNA turnover. We propose that CDIR inhibits apoptosis by acting as a competitive inhibitor of AUF1, preventing AUF1 from binding to its targets.


Asunto(s)
Proteínas de Choque Térmico , Ribonucleoproteína Heterogénea-Nuclear Grupo D/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Mensajero/metabolismo , ARN Mensajero/fisiología , Regiones no Traducidas 3' , Secuencias de Aminoácidos , Apoptosis , Secuencia de Bases , Sitios de Unión , Unión Competitiva , Northern Blotting , Western Blotting , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Citometría de Flujo , Eliminación de Gen , Proteínas de Choque Térmico HSP27 , Células HeLa , Ribonucleoproteína Nuclear Heterogénea D0 , Humanos , Interferón gamma/metabolismo , Modelos Genéticos , Chaperonas Moleculares , Datos de Secuencia Molecular , Mutación , Conformación de Ácido Nucleico , Sistemas de Lectura Abierta , Plásmidos/metabolismo , Unión Proteica , Isoformas de Proteínas , Estructura Secundaria de Proteína , ARN Mensajero/química , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA