Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-37786677

RESUMEN

MITF, a basic-Helix-Loop-Helix Zipper (bHLHZip) transcription factor, plays vital roles in melanocyte development and functions as an oncogene. To explore MITF regulation and its role in melanoma, we conducted a genetic screen for suppressors of the Mitf-associated pigmentation phenotype. An intragenic Mitf mutation was identified, leading to termination of MITF at the K316 SUMOylation site and loss of the C-end intrinsically disordered region (IDR). The resulting protein is more nuclear but less stable than wild-type MITF and retains DNA-binding ability. Interestingly, as a dimer, it can translocate wild-type and mutant MITF partners into the nucleus, improving its own stability and ensuring an active nuclear MITF supply. Interactions between K316 SUMOylation and S409 phosphorylation sites across monomers largely explain the observed effects. Notably, the recurrent melanoma-associated E318K mutation in MITF, which affects K316 SUMOylation, also alters protein regulation in concert with S409, unraveling a novel regulatory mechanism with unexpected disease insights.

2.
Sci Transl Med ; 13(604)2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321320

RESUMEN

Accumulation of the parkin-interacting substrate (PARIS; ZNF746), due to inactivation of parkin, contributes to Parkinson's disease (PD) through repression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α; PPARGC1A) activity. Here, we identify farnesol as an inhibitor of PARIS. Farnesol promoted the farnesylation of PARIS, preventing its repression of PGC-1α via decreasing PARIS occupancy on the PPARGC1A promoter. Farnesol prevented dopaminergic neuronal loss and behavioral deficits via farnesylation of PARIS in PARIS transgenic mice, ventral midbrain transduction of AAV-PARIS, adult conditional parkin KO mice, and the α-synuclein preformed fibril model of sporadic PD. PARIS farnesylation is decreased in the substantia nigra of patients with PD, suggesting that reduced farnesylation of PARIS may play a role in PD. Thus, farnesol may be beneficial in the treatment of PD by enhancing the farnesylation of PARIS and restoring PGC-1α activity.


Asunto(s)
Enfermedad de Parkinson , Animales , Dopamina , Ratones , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Prenilación , Proteínas Represoras/metabolismo , Sustancia Negra/metabolismo
3.
Cancers (Basel) ; 13(2)2021 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-33435458

RESUMEN

A central challenge in cancer genomics is the systematic identification of single and cooperating tumor suppressor gene mutations driving cellular transformation and tumor progression in the absence of oncogenic driver mutation(s). Multiple in vitro and in vivo gene inactivation screens have enhanced our understanding of the tumor suppressor gene landscape in various cancers. However, these studies are limited to single or combination gene effects, specific organs, or require sensitizing mutations. In this study, we developed and utilized a Sleeping Beauty transposon mutagenesis system that functions only as a gene trap to exclusively inactivate tumor suppressor genes. Using whole body transposon mobilization in wild type mice, we observed that cumulative gene inactivation can drive tumorigenesis of solid cancers. We provide a quantitative landscape of the tumor suppressor genes inactivated in these cancers and show that, despite the absence of oncogenic drivers, these genes converge on key biological pathways and processes associated with cancer hallmarks.

4.
Brain ; 142(8): 2380-2401, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31237944

RESUMEN

α-Synuclein misfolding and aggregation plays a major role in the pathogenesis of Parkinson's disease. Although loss of function mutations in the ubiquitin ligase, parkin, cause autosomal recessive Parkinson's disease, there is evidence that parkin is inactivated in sporadic Parkinson's disease. Whether parkin inactivation is a driver of neurodegeneration in sporadic Parkinson's disease or a mere spectator is unknown. Here we show that parkin in inactivated through c-Abelson kinase phosphorylation of parkin in three α-synuclein-induced models of neurodegeneration. This results in the accumulation of parkin interacting substrate protein (zinc finger protein 746) and aminoacyl tRNA synthetase complex interacting multifunctional protein 2 with increased parkin interacting substrate protein levels playing a critical role in α-synuclein-induced neurodegeneration, since knockout of parkin interacting substrate protein attenuates the degenerative process. Thus, accumulation of parkin interacting substrate protein links parkin inactivation and α-synuclein in a common pathogenic neurodegenerative pathway relevant to both sporadic and familial forms Parkinson's disease. Thus, suppression of parkin interacting substrate protein could be a potential therapeutic strategy to halt the progression of Parkinson's disease and related α-synucleinopathies.


Asunto(s)
Enfermedad de Parkinson/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Enfermedad de Parkinson/patología
5.
J Clin Invest ; 128(7): 2927-2943, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29863500

RESUMEN

Although nonmalignant stromal cells facilitate tumor growth and can occupy up to 90% of a solid tumor mass, better strategies to exploit these cells for improved cancer therapy are needed. Here, we describe a potent MMAE-linked antibody-drug conjugate (ADC) targeting tumor endothelial marker 8 (TEM8, also known as ANTXR1), a highly conserved transmembrane receptor broadly overexpressed on cancer-associated fibroblasts, endothelium, and pericytes. Anti-TEM8 ADC elicited potent anticancer activity through an unexpected killing mechanism we term DAaRTS (drug activation and release through stroma), whereby the tumor microenvironment localizes active drug at the tumor site. Following capture of ADC prodrug from the circulation, tumor-associated stromal cells release active MMAE free drug, killing nearby proliferating tumor cells in a target-independent manner. In preclinical studies, ADC treatment was well tolerated and induced regression and often eradication of multiple solid tumor types, blocked metastatic growth, and prolonged overall survival. By exploiting TEM8+ tumor stroma for targeted drug activation, these studies reveal a drug delivery strategy with potential to augment therapies against multiple cancer types.


Asunto(s)
Inmunoconjugados/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/deficiencia , Biomarcadores de Tumor/genética , Brentuximab Vedotina , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Proteínas de Microfilamentos , Neoplasias/metabolismo , Receptores de Péptidos/antagonistas & inhibidores , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Células del Estroma/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Proc Natl Acad Sci U S A ; 115(7): 1635-1640, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29386392

RESUMEN

Mutations in LRRK2 are known to be the most common genetic cause of sporadic and familial Parkinson's disease (PD). Multiple lines of LRRK2 transgenic or knockin mice have been developed, yet none exhibit substantial dopamine (DA)-neuron degeneration. Here we develop human tyrosine hydroxylase (TH) promoter-controlled tetracycline-sensitive LRRK2 G2019S (GS) and LRRK2 G2019S kinase-dead (GS/DA) transgenic mice and show that LRRK2 GS expression leads to an age- and kinase-dependent cell-autonomous neurodegeneration of DA and norepinephrine (NE) neurons. Accompanying the loss of DA neurons are DA-dependent behavioral deficits and α-synuclein pathology that are also LRRK2 GS kinase-dependent. Transmission EM reveals that that there is an LRRK2 GS kinase-dependent significant reduction in synaptic vesicle number and a greater abundance of clathrin-coated vesicles in DA neurons. These transgenic mice indicate that LRRK2-induced DA and NE neurodegeneration is kinase-dependent and can occur in a cell-autonomous manner. Moreover, these mice provide a substantial advance in animal model development for LRRK2-associated PD and an important platform to investigate molecular mechanisms for how DA neurons degenerate as a result of expression of mutant LRRK2.


Asunto(s)
Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/patología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/fisiología , Enfermedades Neurodegenerativas/patología , Norepinefrina/metabolismo , Factores de Edad , Animales , Conducta Animal , Neuronas Dopaminérgicas/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Actividad Motora , Mutación , Enfermedades Neurodegenerativas/metabolismo , alfa-Sinucleína/metabolismo
7.
Genetics ; 207(4): 1335-1345, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29021281

RESUMEN

BRCA2 loss-of-heterozygosity (LOH) is frequently observed in BRCA2-mutated tumors, but its biallelic loss causes embryonic lethality in mice and inhibits proliferation of normal somatic cells. Therefore, it remains unclear how loss of BRCA2 contributes to tumorigenesis. One possibility is that mutation in potential genetic interactors of BRCA2, such as TRP53, is required for cell survival/proliferation in the absence of BRCA2. In this study, using an insertional mutagenesis screen in mouse embryonic stem cells (mESC), we have identified GIPC3 (GAIP-interacting protein C-terminus 3) as a BRCA2 genetic interactor that contributes to survival of Brca2-null mESC. GIPC3 does not compensate for BRCA2 loss in the repair of double-strand breaks. Mass-spectrometric analysis resulted in the identification of G-protein signaling transducers, APPL1 and APPL2, as potential GIPC3-binding proteins. A mutant GIPC3 (His155Ala) that does not bind to APPL1/2 failed to rescue the lethality of Brca2-null mESC, suggesting that the cell viability by GIPC3 is mediated via APPL1/2. Finally, the physiological significance of GIPC3 as a genetic interactor of BRCA2 is supported by the observation that Brca2-null embryos with Gipc3 overexpression are developmentally more advanced than their control littermates. Taken together, we have uncovered a novel role for GIPC3 as a BRCA2 genetic interactor.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteína BRCA2/genética , Neoplasias de la Mama/genética , Carcinogénesis/genética , Animales , Proteína BRCA2/deficiencia , Neoplasias de la Mama/patología , Proteínas Portadoras/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Pérdida de Heterocigocidad/genética , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Mutagénesis Insercional , Mutación
8.
Cancer Cell ; 31(4): 501-515.e8, 2017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28399408

RESUMEN

Targeting the tumor vasculature with antibody-drug conjugates (ADCs) is a promising anti-cancer strategy that in order to be realized must overcome several obstacles, including identification of suitable targets and optimal warheads. Here, we demonstrate that the cell-surface protein CD276/B7-H3 is broadly overexpressed by multiple tumor types on both cancer cells and tumor-infiltrating blood vessels, making it a potentially ideal dual-compartment therapeutic target. In preclinical studies CD276 ADCs armed with a conventional MMAE warhead destroyed CD276-positive cancer cells, but were ineffective against tumor vasculature. In contrast, pyrrolobenzodiazepine-conjugated CD276 ADCs killed both cancer cells and tumor vasculature, eradicating large established tumors and metastases, and improving long-term overall survival. CD276-targeted dual-compartment ablation could aid in the development of highly selective broad-acting anti-cancer therapies.


Asunto(s)
Antígenos B7/genética , Antígenos B7/metabolismo , Inmunoconjugados/farmacología , Neoplasias/irrigación sanguínea , Animales , Antineoplásicos/inmunología , Antineoplásicos/farmacología , Antígenos B7/inmunología , Benzodiazepinas/farmacología , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Línea Celular Tumoral , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Femenino , Humanos , Inmunoconjugados/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Terapia Molecular Dirigida/métodos , Neoplasias/patología , Neoplasias/terapia , Oligopéptidos/farmacología , Pirroles/farmacología , Conejos
9.
Cell Rep ; 16(3): 793-804, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27373150

RESUMEN

The neural network of the temporal lobe is thought to provide a cognitive map of our surroundings. Functional analysis of this network has been hampered by coarse tools that often result in collateral damage to other circuits. We developed a chemogenetic system to temporally control electrical input into the hippocampus. When entorhinal input to the perforant path was acutely silenced, hippocampal firing patterns became destabilized and underwent extensive remapping. We also found that spatial memory acquired prior to neural silencing was impaired by loss of input through the perforant path. Together, our experiments show that manipulation of entorhinal activity destabilizes spatial coding and disrupts spatial memory. Moreover, we introduce a chemogenetic model for non-invasive neuronal silencing that offers multiple advantages over existing strategies in this setting.


Asunto(s)
Hipocampo/fisiología , Red Nerviosa/fisiología , Memoria Espacial/fisiología , Lóbulo Temporal/fisiología , Animales , Corteza Entorrinal/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Modelos Neurológicos , Vía Perforante/fisiología
10.
Neuron ; 90(3): 535-50, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27112497

RESUMEN

Hexanucleotide expansions in C9ORF72 are the most frequent genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Disease mechanisms were evaluated in mice expressing C9ORF72 RNAs with up to 450 GGGGCC repeats or with one or both C9orf72 alleles inactivated. Chronic 50% reduction of C9ORF72 did not provoke disease, while its absence produced splenomegaly, enlarged lymph nodes, and mild social interaction deficits, but not motor dysfunction. Hexanucleotide expansions caused age-, repeat-length-, and expression-level-dependent accumulation of RNA foci and dipeptide-repeat proteins synthesized by AUG-independent translation, accompanied by loss of hippocampal neurons, increased anxiety, and impaired cognitive function. Single-dose injection of antisense oligonucleotides (ASOs) that target repeat-containing RNAs but preserve levels of mRNAs encoding C9ORF72 produced sustained reductions in RNA foci and dipeptide-repeat proteins, and ameliorated behavioral deficits. These efforts identify gain of toxicity as a central disease mechanism caused by repeat-expanded C9ORF72 and establish the feasibility of ASO-mediated therapy.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Demencia Frontotemporal/tratamiento farmacológico , Factores de Intercambio de Guanina Nucleótido/genética , Oligonucleótidos Antisentido/farmacología , ARN/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Proteína C9orf72 , Expansión de las Repeticiones de ADN/genética , Demencia Frontotemporal/genética , Ratones Transgénicos , Neuronas/metabolismo , Oligonucleótidos Antisentido/efectos adversos , Oligonucleótidos Antisentido/genética
11.
Dev Cell ; 35(3): 322-32, 2015 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-26555052

RESUMEN

The mammalian lung forms its elaborate tree-like structure following a largely stereotypical branching sequence. While a number of genes have been identified to play essential roles in lung branching, what coordinates the choice between branch growth and new branch formation has not been elucidated. Here we show that loss of FGF-activated transcription factor genes, Etv4 and Etv5 (collectively Etv), led to prolonged branch tip growth and delayed new branch formation. Unexpectedly, this phenotype is more similar to mutants with increased rather than decreased FGF activity. Indeed, an increased Fgf10 expression is observed, and reducing Fgf10 dosage can attenuate the Etv mutant phenotype. Further evidence indicates that ETV inhibits Fgf10 via directly promoting Shh expression. SHH in turn inhibits local Fgf10 expression and redirects growth, thereby initiating new branches. Together, our findings establish ETV as a key node in the FGF-ETV-SHH inhibitory feedback loop that dictates branching periodicity.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Pulmón/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Factores de Transcripción/metabolismo , Animales , Tipificación del Cuerpo/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Ratones , Ratones Transgénicos , Morfogénesis/genética , Transducción de Señal/genética
12.
Proc Natl Acad Sci U S A ; 112(46): E6321-30, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578792

RESUMEN

Centrosomes are microtubule-organizing centers that facilitate bipolar mitotic spindle assembly and chromosome segregation. Recognizing that centrosome amplification is a common feature of aneuploid cancer cells, we tested whether supernumerary centrosomes are sufficient to drive tumor development. To do this, we constructed and analyzed mice in which centrosome amplification can be induced by a Cre-recombinase-mediated increase in expression of Polo-like kinase 4 (Plk4). Elevated Plk4 in mouse fibroblasts produced supernumerary centrosomes and enhanced the expected mitotic errors, but proliferation continued only after inactivation of the p53 tumor suppressor. Increasing Plk4 levels in mice with functional p53 produced centrosome amplification in liver and skin, but this did not promote spontaneous tumor development in these tissues or enhance the growth of chemically induced skin tumors. In the absence of p53, Plk4 overexpression generated widespread centrosome amplification, but did not drive additional tumors or affect development of the fatal thymic lymphomas that arise in animals lacking p53. We conclude that, independent of p53 status, supernumerary centrosomes are not sufficient to drive tumor formation.


Asunto(s)
División Celular Asimétrica/fisiología , Centrosoma/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Regulación de la Expresión Génica , Linfoma/genética , Linfoma/metabolismo , Linfoma/patología , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Neoplasias del Timo/genética , Neoplasias del Timo/metabolismo , Neoplasias del Timo/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
J Mol Cell Cardiol ; 88: 1-13, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26386426

RESUMEN

Gap junctions (GJ) are intercellular channels composed of connexin subunits that play a critical role in a diverse number of cellular processes in all tissue types. In the heart, GJs mediate electrical coupling between cardiomyocytes and display mislocalization and/or downregulation in cardiac disease (a process known as GJ remodeling), producing an arrhythmogenic substrate. The main constituent of GJs in the ventricular myocardium is Connexin 43 (Cx43), an integral membrane protein that is rapidly turned over and shows decreased expression or function with age. We hypothesized that Wwp1, an ubiquitin ligase whose expression in known to increase in aging-related pathologies, may regulate Cx43 in vivo by targeting it for ubiquitylation and degradation and yield tissue-specific Cx43 loss of function phenotypes. When Wwp1 was globally overexpressed in mice under the control of a ß-actin promoter, the highest induction of Wwp1 expression was observed in the heart which was associated with a 90% reduction in cardiac Cx43 protein levels, left ventricular hypertrophy (LVH), and the development of lethal ventricular arrhythmias around 8weeks of age. This phenotype was completely penetrant in two independent founder lines. Cardiomyocyte-specific overexpression of Wwp1 confirmed that this phenotype was cell autonomous and delineated Cx43-dependent and -independent roles for Wwp1 in arrhythmogenesis and LVH, respectively. Using a cell-based system, it was determined that Wwp1 co-immunoprecipitates with and ubiquitylates Cx43, causing a decrease in the steady state levels of Cx43 protein. These findings offer new mechanistic insights into the regulation of Cx43 which may be exploitable in various gap junctionopathies.


Asunto(s)
Arritmias Cardíacas/genética , Conexina 43/genética , Hipertrofia Ventricular Izquierda/genética , Miocitos Cardíacos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Actinas/genética , Actinas/metabolismo , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Conexina 43/metabolismo , Modelos Animales de Enfermedad , Femenino , Uniones Comunicantes/metabolismo , Uniones Comunicantes/patología , Regulación de la Expresión Génica , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Masculino , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/patología , Fenotipo , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteolisis , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
14.
Cell Rep ; 10(2): 123-30, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25558062

RESUMEN

G protein-coupled receptor 124 (GPR124) is an orphan receptor in the adhesion family of GPCRs, and previous global or endothelial-specific disruption of Gpr124 in mice led to defective CNS angiogenesis and blood-brain barriergenesis. Similar developmental defects were observed following dual deletion of Wnt7a/Wnt7b or deletion of ß-catenin in endothelial cells, suggesting a possible relationship between GPR124 and canonical WNT signaling. Here, we show using in vitro reporter assays, mutation analysis, and genetic interaction studies in vivo that GPR124 functions as a WNT7A/WNT7B-specific costimulator of ß-catenin signaling in brain endothelium. WNT7-stimulated ß-catenin signaling was dependent upon GPR124's intracellular PDZ binding motif and a set of leucine-rich repeats in its extracellular domain. This study reveals a vital role for GPR124 in potentiation of WNT7-induced canonical ß-catenin signaling with important implications for understanding and manipulating CNS-specific angiogenesis and blood-brain barrier-genesis.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Secuencias de Aminoácidos , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Dominios PDZ , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/deficiencia
15.
Sci Transl Med ; 6(242): 242ra84, 2014 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-24964992

RESUMEN

Antiangiogenic agents that block vascular endothelial growth factor (VEGF) signaling are important components of current cancer treatment modalities but are limited by alternative ill-defined angiogenesis mechanisms that allow persistent tumor vascularization in the face of continued VEGF pathway blockade. We identified prostaglandin E2 (PGE2) as a soluble tumor-derived angiogenic factor associated with VEGF-independent angiogenesis. PGE2 production in preclinical breast and colon cancer models was tightly controlled by cyclooxygenase-2 (COX-2) expression, and COX-2 inhibition augmented VEGF pathway blockade to suppress angiogenesis and tumor growth, prevent metastasis, and increase overall survival. These results demonstrate the importance of the COX-2/PGE2 pathway in mediating resistance to VEGF pathway blockade and could aid in the rapid development of more efficacious anticancer therapies.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Neoplasias Hepáticas/prevención & control , Neoplasias Hepáticas/secundario , Neoplasias Mamarias Experimentales/prevención & control , Neoplasias Mamarias Experimentales/secundario , Ensayos Antitumor por Modelo de Xenoinjerto , Inhibidores de la Angiogénesis/farmacología , Animales , Axitinib , Carcinogénesis/patología , Línea Celular Tumoral , Células Clonales , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/metabolismo , Femenino , Humanos , Imidazoles/farmacología , Indazoles/farmacología , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ratones , Terapia Neoadyuvante , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
PLoS One ; 9(1): e85883, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465765

RESUMEN

The LMO2 oncogene is deregulated in the majority of human T-cell leukemia cases and in most gene therapy-induced T-cell leukemias. We made transgenic mice with enforced expression of Lmo2 in T-cells by the CD2 promoter/enhancer. These transgenic mice developed highly penetrant T-ALL by two distinct patterns of gene expression: one in which there was concordant activation of Lyl1, Hhex, and Mycn or alternatively, with Notch1 target gene activation. Most strikingly, this gene expression clustering was conserved in human Early T-cell Precursor ALL (ETP-ALL), where LMO2, HHEX, LYL1, and MYCN were most highly expressed. We discovered that HHEX is a direct transcriptional target of LMO2 consistent with its concordant gene expression. Furthermore, conditional inactivation of Hhex in CD2-Lmo2 transgenic mice markedly attenuated T-ALL development, demonstrating that Hhex is a crucial mediator of Lmo2's oncogenic function. The CD2-Lmo2 transgenic mice offer mechanistic insight into concordant oncogene expression and provide a model for the highly treatment-resistant ETP-ALL subtype.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis/metabolismo , Proteínas con Dominio LIM/metabolismo , Leucemia de Células T/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Antígenos CD2/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Elementos E-Box/genética , Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Leucemia de Células T/genética , Leucemia de Células T/patología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Oncogenes , Penetrancia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , Factores de Transcripción/genética , Transcripción Genética , Regulación hacia Arriba/genética
17.
PLoS One ; 8(4): e62479, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23638095

RESUMEN

The study of axis extension and somitogenesis has been greatly advanced through the use of genetic tools such as the TCre mouse line. In this line, Cre is controlled by a fragment of the T (Brachyury) promoter that is active in progenitor cells that reside within the primitive streak and tail bud and which give rise to lineages emerging from these tissues as the embryonic axis extends. However, because TCre-mediated recombination occurs early in development, gene inactivation can result in an axis truncation that precludes the study of gene function in later or more posterior tissues. To address this limitation, we have generated an inducible TCre transgenic mouse line, called TCreERT2, that provides temporal control, through tamoxifen administration, in all cells emerging from the primitive streak or tail bud throughout development. TCreERT2 activity is mostly silent in the absence of tamoxifen and, in its presence, results in near complete recombination of emerging mesoderm from E7.5 through E13.5. We demonstrate the utility of the TCreERT2 line for determining rate of posterior axis extension and somite formation, thus providing the first in vivo tool for such measurements. To test the usefulness of TCreERT2 for genetic manipulation, we demonstrate that an early deletion of ß-Catenin via TCreERT2 induction phenocopies the TCre-mediated deletion of ß-Catenin defect, whereas a later induction bypasses this early phenotype and produces a similar defect in more caudal tissues. TCreERT2 provides a useful and novel tool for the control of gene expression of emerging embryonic lineages throughout development.


Asunto(s)
Ratones Transgénicos/genética , Línea Primitiva/embriología , Recombinación Genética , Animales , Antagonistas de Estrógenos/administración & dosificación , Femenino , Proteínas Fetales/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Línea Primitiva/citología , Línea Primitiva/metabolismo , Proteínas de Dominio T Box/genética , Tamoxifeno/administración & dosificación
18.
Genome Res ; 22(5): 870-84, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22367191

RESUMEN

Endogenous retrotransposons have caused extensive genomic variation within mammalian species, but the functional implications of such mobilization are mostly unknown. We mapped thousands of endogenous retrovirus (ERV) germline integrants in highly divergent, previously unsequenced mouse lineages, facilitating a comparison of gene expression in the presence or absence of local insertions. Polymorphic ERVs occur relatively infrequently in gene introns and are particularly depleted from genes involved in embryogenesis or that are highly expressed in embryonic stem cells. Their genomic distribution implies ongoing negative selection due to deleterious effects on gene expression and function. A polymorphic, intronic ERV at Slc15a2 triggers up to 49-fold increases in premature transcriptional termination and up to 39-fold reductions in full-length transcripts in adult mouse tissues, thereby disrupting protein expression and functional activity. Prematurely truncated transcripts also occur at Polr1a, Spon1, and up to ∼5% of other genes when intronic ERV polymorphisms are present. Analysis of expression quantitative trait loci (eQTLs) in recombinant BxD mouse strains demonstrated very strong genetic associations between the polymorphic ERV in cis and disrupted transcript levels. Premature polyadenylation is triggered at genomic distances up to >12.5 kb upstream of the ERV, both in cis and between alleles. The parent of origin of the ERV is associated with variable expression of nonterminated transcripts and differential DNA methylation at its 5'-long terminal repeat. This study defines an unexpectedly strong functional impact of ERVs in disrupting gene transcription at a distance and demonstrates that ongoing retrotransposition can contribute significantly to natural phenotypic diversity.


Asunto(s)
Retrovirus Endógenos/genética , Regulación de la Expresión Génica , Transcripción Genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Metilación de ADN , Femenino , Variación Genética , Heterocigoto , Intrones , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Polimorfismo Genético , Biosíntesis de Proteínas/genética , Sitios de Carácter Cuantitativo , Análisis de Secuencia de ADN , Simportadores/genética , Simportadores/metabolismo , Secuencias Repetidas Terminales
19.
Genesis ; 50(2): 112-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21898766

RESUMEN

The Notch1 receptor plays a critical role in cell fate decisions during development. Activation of Notch signaling has been implicated in several types of cancer, particularly T-cell acute lymphoblastic leukemia (T-ALL). Consequently, several transgenic mouse strains have been made to study the role of Notch1 in T-ALL. However, the existing Notch1 transgenic lines mimic a translocation event found in only ∼1% of T-ALL cases. Here we describe three novel NOTCH1 transgenic mouse strains that have Cre-inducible expression of the entire human NOTCH1 locus, each possessing a common mutation found in T-ALL. Unlike existing Notch1 transgenic strains, these NOTCH1 transgenic strains express full-length receptors from an endogenous human promoter that should be susceptible to a number of Notch antagonists that have recently been developed. These strains will allow researchers to modulate Notch signaling to study both normal development and cancer biology.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Ratones Transgénicos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptor Notch1/genética , Animales , Western Blotting , Diferenciación Celular , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Mutación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptor Notch1/metabolismo , Transducción de Señal
20.
PLoS One ; 6(4): e18568, 2011 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-21494637

RESUMEN

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene cause late-onset, autosomal dominant familial Parkinson's disease (PD) and also contribute to idiopathic PD. LRRK2 mutations represent the most common cause of PD with clinical and neurochemical features that are largely indistinguishable from idiopathic disease. Currently, transgenic mice expressing wild-type or disease-causing mutants of LRRK2 have failed to produce overt neurodegeneration, although abnormalities in nigrostriatal dopaminergic neurotransmission have been observed. Here, we describe the development and characterization of transgenic mice expressing human LRRK2 bearing the familial PD mutations, R1441C and G2019S. Our study demonstrates that expression of G2019S mutant LRRK2 induces the degeneration of nigrostriatal pathway dopaminergic neurons in an age-dependent manner. In addition, we observe autophagic and mitochondrial abnormalities in the brains of aged G2019S LRRK2 mice and markedly reduced neurite complexity of cultured dopaminergic neurons. These new LRRK2 transgenic mice will provide important tools for understanding the mechanism(s) through which familial mutations precipitate neuronal degeneration and PD.


Asunto(s)
Sustitución de Aminoácidos/genética , Autofagia , Dopamina/metabolismo , Proteínas Mutantes/metabolismo , Neuritas/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Conducta Animal , Cromatografía Líquida de Alta Presión , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Mesencéfalo/metabolismo , Mesencéfalo/patología , Mesencéfalo/ultraestructura , Ratones , Ratones Transgénicos , Actividad Motora , Neuritas/ultraestructura , Técnicas de Cultivo de Órganos , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...