Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Drug Discov Today Technol ; 28: 33-39, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30205879

RESUMEN

The development of genome editing tools capable of modifying specific genomic sequences with unprecedented accuracy has opened up a wide range of new possibilities in targeted gene manipulation. In particular, the CRISPR/Cas9 system, a repurposed prokaryotic adaptive immune system, has been widely adopted because of its unmatched simplicity and flexibility. In this review we discuss achievements and current limitations of CRISPR/Cas9 genome editing in hematopoietic cells with special emphasis on its potential use in ex vivo gene therapy of monogenic blood disorders, HIV and cancer.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Terapia Genética/métodos , Síndrome de Inmunodeficiencia Adquirida/genética , Síndrome de Inmunodeficiencia Adquirida/terapia , Animales , Marcación de Gen/métodos , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/terapia , Humanos , Neoplasias/genética , Neoplasias/terapia
2.
Mol Ther Nucleic Acids ; 10: 1-8, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29499925

RESUMEN

The CRISPR/Cas9 prokaryotic adaptive immune system and its swift repurposing for genome editing enables modification of any prespecified genomic sequence with unprecedented accuracy and efficiency, including targeted gene repair. We used the CRISPR/Cas9 system for targeted repair of patient-specific point mutations in the Cytochrome b-245 heavy chain gene (CYBB), whose inactivation causes chronic granulomatous disease (XCGD)-a life-threatening immunodeficiency disorder characterized by the inability of neutrophils and macrophages to produce microbicidal reactive oxygen species (ROS). We show that frameshift mutations can be effectively repaired in hematopoietic cells by non-integrating lentiviral vectors carrying RNA-guided Cas9 endonucleases (RGNs). Because about 25% of most inherited blood disorders are caused by frameshift mutations, our results suggest that up to a quarter of all patients suffering from monogenic blood disorders could benefit from gene therapy employing personalized, donor template-free RGNs.

3.
Nat Commun ; 9(1): 1281, 2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29599503

RESUMEN

Nitric oxide (NO) synthesis is a late event during differentiation of mouse embryonic stem cells (mESC) and occurs after release from serum and leukemia inhibitory factor (LIF). Here we show that after release from pluripotency, a subpopulation of mESC, kept in the naive state by 2i/LIF, expresses endothelial nitric oxide synthase (eNOS) and endogenously synthesizes NO. This eNOS/NO-positive subpopulation (ESNO+) expresses mesendodermal markers and is more efficient in the generation of cardiovascular precursors than eNOS/NO-negative cells. Mechanistically, production of endogenous NO triggers rapid Hdac2 S-nitrosylation, which reduces association of Hdac2 with the transcriptional repression factor Zeb1, allowing mesendodermal gene expression. In conclusion, our results suggest that the interaction between Zeb1, Hdac2, and eNOS is required for early mesendodermal differentiation of naive mESC.


Asunto(s)
Histona Desacetilasa 2/metabolismo , Células Madre Embrionarias de Ratones/citología , Miocardio/citología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/biosíntesis , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Células HeLa , Humanos , Factor Inhibidor de Leucemia/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Miocardio/metabolismo
4.
Circ Res ; 122(1): 31-46, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29158345

RESUMEN

RATIONALE: Human cardiac mesenchymal cells (CMSCs) are a therapeutically relevant primary cell population. Diabetes mellitus compromises CMSC function as consequence of metabolic alterations and incorporation of stable epigenetic changes. OBJECTIVE: To investigate the role of α-ketoglutarate (αKG) in the epimetabolic control of DNA demethylation in CMSCs. METHODS AND RESULTS: Quantitative global analysis, methylated and hydroxymethylated DNA sequencing, and gene-specific GC methylation detection revealed an accumulation of 5-methylcytosine, 5-hydroxymethylcytosine, and 5-formylcytosine in the genomic DNA of human CMSCs isolated from diabetic donors. Whole heart genomic DNA analysis revealed iterative oxidative cytosine modification accumulation in mice exposed to high-fat diet (HFD), injected with streptozotocin, or both in combination (streptozotocin/HFD). In this context, untargeted and targeted metabolomics indicated an intracellular reduction of αKG synthesis in diabetic CMSCs and in the whole heart of HFD mice. This observation was paralleled by a compromised TDG (thymine DNA glycosylase) and TET1 (ten-eleven translocation protein 1) association and function with TET1 relocating out of the nucleus. Molecular dynamics and mutational analyses showed that αKG binds TDG on Arg275 providing an enzymatic allosteric activation. As a consequence, the enzyme significantly increased its capacity to remove G/T nucleotide mismatches or 5-formylcytosine. Accordingly, an exogenous source of αKG restored the DNA demethylation cycle by promoting TDG function, TET1 nuclear localization, and TET/TDG association. TDG inactivation by CRISPR/Cas9 knockout or TET/TDG siRNA knockdown induced 5-formylcytosine accumulation, thus partially mimicking the diabetic epigenetic landscape in cells of nondiabetic origin. The novel compound (S)-2-[(2,6-dichlorobenzoyl)amino]succinic acid (AA6), identified as an inhibitor of αKG dehydrogenase, increased the αKG level in diabetic CMSCs and in the heart of HFD and streptozotocin mice eliciting, in HFD, DNA demethylation, glucose uptake, and insulin response. CONCLUSIONS: Restoring the epimetabolic control of DNA demethylation cycle promises beneficial effects on cells compromised by environmental metabolic changes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Ácidos Cetoglutáricos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Oxigenasas de Función Mixta/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Timina ADN Glicosilasa/metabolismo , Animales , Células Cultivadas , Citosina/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Inhibidores Enzimáticos/farmacología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ácidos Cetoglutáricos/antagonistas & inhibidores , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos
5.
Matrix Biol ; 59: 109-120, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27645114

RESUMEN

Latent transforming growth factor beta binding protein 4 (LTBP4) belongs to the fibrillin/LTBP family of proteins and plays an important role as a structural component of extracellular matrix (ECM) and local regulator of TGFß signaling. We have previously reported that Ltbp4S knock out mice (Ltbp4S-/-) develop centrilobular emphysema reminiscent of late stage COPD, which could be partially rescued by inactivating the antioxidant protein Sestrin 2 (Sesn2). More recent studies showed that Sesn2 knock out mice upregulate Pdgfrß-controlled alveolar maintenance programs that protect against cigarette smoke induced pulmonary emphysema. Based on this, we hypothesized that the emphysema of Ltbp4S-/- mice is primarily caused by defective Pdgfrß signaling. Here we show that LTBP4 induces Pdgfrß signaling by inhibiting the antioxidant Nrf2/Keap1 pathway in a TGFß-dependent manner. Overall, our data identified Ltbp4 as a major player in lung remodeling and injury repair.


Asunto(s)
Matriz Extracelular/metabolismo , Proteínas de Unión a TGF-beta Latente/genética , Factor 2 Relacionado con NF-E2/genética , Enfisema Pulmonar/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Transformador beta/genética , Animales , Línea Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Matriz Extracelular/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Proteínas de Unión a TGF-beta Latente/deficiencia , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Visón , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Peroxidasas , Plásmidos/química , Plásmidos/metabolismo , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Tropoelastina/deficiencia , Tropoelastina/genética
6.
Cell Rep ; 11(12): 1847-55, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26095368

RESUMEN

The ability of hematopoietic stem cells (HSCs) to self-renew is a prerequisite for the establishment of definitive hematopoiesis and life-long blood regeneration. Here, we report the single-stranded DNA-binding transcriptional regulator far upstream element (FUSE)-binding protein 1 (FUBP1) as an essential factor of HSC self-renewal. Functional inactivation of FUBP1 in two different mouse models resulted in embryonic lethal anemia at around E15.5 caused by severely diminished HSCs. Fetal and adult HSCs lacking FUBP1 revealed an HSC-intrinsic defect in their maintenance, expansion, and long-term blood reconstitution, but could differentiate into all hematopoietic lineages. FUBP1-deficient adult HSCs exhibit significant transcriptional changes, including upregulation of the cell-cycle inhibitor p21 and the pro-apoptotic Noxa molecule. These changes caused an increase in generation time and death of HSCs as determined by video-microscopy-based tracking. Our data establish FUBP1 and its recognition of single-stranded genomic DNA as an important element in the transcriptional regulation of HSC self-renewal.


Asunto(s)
Autorrenovación de las Células/genética , Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Células Madre Fetales/citología , Células Madre Fetales/metabolismo , Regulación de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Ratones , Transducción de Señal/genética
7.
Dis Model Mech ; 6(6): 1378-87, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24046361

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a leading cause of morbidity and mortality worldwide. COPD is caused by chronic exposure to cigarette smoke and/or other environmental pollutants that are believed to induce reactive oxygen species (ROS) that gradually disrupt signalling pathways responsible for maintaining lung integrity. Here we identify the antioxidant protein sestrin-2 (SESN2) as a repressor of PDGFRß signalling, and PDGFRß signalling as an upstream regulator of alveolar maintenance programmes. In mice, the mutational inactivation of Sesn2 prevents the development of cigarette-smoke-induced pulmonary emphysema by upregulating PDGFRß expression via a selective accumulation of intracellular superoxide anions (O2(-)). We also show that SESN2 is overexpressed and PDGFRß downregulated in the emphysematous lungs of individuals with COPD and to a lesser extent in human lungs of habitual smokers without COPD, implicating a negative SESN2-PDGFRß interrelationship in the pathogenesis of COPD. Taken together, our results imply that SESN2 could serve as both a biomarker and as a drug target in the clinical management of COPD.


Asunto(s)
Proteínas Nucleares/fisiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfisema Pulmonar/etiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Transducción de Señal/fisiología , Humo , Regulación hacia Arriba , Animales , Humanos , Pulmón/metabolismo , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Peroxidasas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Superóxidos/metabolismo
8.
Methods ; 53(4): 347-55, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21334922

RESUMEN

Gene trapping is a high-throughput insertional mutagenesis approach that has been primarily used in mouse embryonic stem cells (ESCs). As a high throughput technology, gene trapping helped to generate tenth of thousands of ESC lines harboring mutations in single genes that can be used for making knock-out mice. Ongoing international efforts operating under the umbrella of the International Knockout Mouse Consortium (IKMC; www.knockoutmouse.org) aim to generate conditional alleles for every protein coding gene in the mouse genome by high throughput conditional gene targeting and trapping. Here, we provide protocols for gene trapping in ESCs that can be easily adapted to any other mammalian cell. We further provide protocols for handling and verifying conditional gene trap alleles in ESC lines obtained from the IKMC repositories and describe a highly efficient method for the postinsertional modification of gene trap alleles. More specifically, we describe a protein tagging strategy based on recombinase mediated cassette exchange (RMCE) that enables protein localization and protein-protein interaction studies under physiological conditions.


Asunto(s)
Alelos , Mutagénesis Insercional/métodos , Animales , Técnicas de Cultivo de Célula , Medios de Cultivo , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Componentes del Gen , Técnicas de Inactivación de Genes , Vectores Genéticos , Ratones , Ratones Noqueados , Virus de la Leucemia Murina de Moloney/genética , Reacción en Cadena de la Polimerasa/métodos , Proteómica/métodos , Recombinasas/metabolismo , Análisis de Secuencia de ADN/métodos , Transfección/métodos
9.
Dis Model Mech ; 3(3-4): 246-53, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20106877

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a leading cause of morbidity and mortality worldwide. Cigarette smoking has been identified as one of the major risk factors and several predisposing genetic factors have been implicated in the pathogenesis of COPD, including a single nucleotide polymorphism (SNP) in the latent transforming growth factor (TGF)-beta binding protein 4 (Ltbp4)-encoding gene. Consistent with this finding, mice with a null mutation of the short splice variant of Ltbp4 (Ltbp4S) develop pulmonary emphysema that is reminiscent of COPD. Here, we report that the mutational inactivation of the antioxidant protein sestrin 2 (sesn2) partially rescues the emphysema phenotype of Ltbp4S mice and is associated with activation of the TGF-beta and mammalian target of rapamycin (mTOR) signal transduction pathways. The results suggest that sesn2 could be clinically relevant to patients with COPD who might benefit from antagonists of sestrin function.


Asunto(s)
Silenciador del Gen , Proteínas/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfisema Pulmonar/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Alelos , Animales , Modelos Animales de Enfermedad , Inducción Enzimática , Fibroblastos/metabolismo , Fibroblastos/patología , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Unión a TGF-beta Latente/deficiencia , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Mutación/genética , Proteínas Nucleares , Peroxidasas , Proteínas Serina-Treonina Quinasas/biosíntesis , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfisema Pulmonar/complicaciones , Enfisema Pulmonar/enzimología , Enfisema Pulmonar/patología , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR
10.
J Cell Physiol ; 219(1): 14-22, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19016471

RESUMEN

The latent TGF-beta binding proteins (LTBP) -1, -3, and -4 are extracellular proteins that assist in the secretion and localization of latent TGF-beta. The null mutation of LTBP-4S in mice causes defects in the differentiation of terminal air-sacs, fragmented elastin, and colon carcinomas. We investigated lung development from embryonic day 14.5 (E14.5) to day 7 after birth (P7) in order to determine when the defects in elastin organization initiate and to further examine the relation of TGF-beta signaling levels and air-sac septation in Ltbp4S-/- lungs. We found that defects in elastogenesis are visible as early as E14.5 and are maintained in the alveolar walls, in blood vessel media, and subjacent airway epithelium. The air-sac septation defect was associated with excessive TGF-beta signaling and was reversed by lowering TGF-beta2 levels. Thus, the phenotype is not directly reflective of a change in TGF-beta1, the only TGF-beta isoform known to complex with LTBP-4. Reversal of the air-sac septation defect was not associated with normalization of the elastogenesis indicating two separate functions of LTBP-4 as a regulator of elastic fiber assembly and TGF-beta levels in lungs.


Asunto(s)
Elastina/metabolismo , Proteínas de Unión a TGF-beta Latente/metabolismo , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Isoformas de Proteínas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Benzamidas/metabolismo , Dioxoles/metabolismo , Elastina/genética , Elastina/ultraestructura , Femenino , Proteínas de Unión a TGF-beta Latente/genética , Pulmón/anatomía & histología , Pulmón/metabolismo , Masculino , Ratones , Ratones Noqueados , Fenotipo , Embarazo , Isoformas de Proteínas/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Alveolos Pulmonares/fisiología , Alveolos Pulmonares/ultraestructura , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/genética
11.
J Cell Biol ; 183(3): 409-17, 2008 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-18955553

RESUMEN

The blood-brain barrier (BBB) is confined to the endothelium of brain capillaries and is indispensable for fluid homeostasis and neuronal function. In this study, we show that endothelial Wnt/beta-catenin (beta-cat) signaling regulates induction and maintenance of BBB characteristics during embryonic and postnatal development. Endothelial specific stabilization of beta-cat in vivo enhances barrier maturation, whereas inactivation of beta-cat causes significant down-regulation of claudin3 (Cldn3), up-regulation of plamalemma vesicle-associated protein, and BBB breakdown. Stabilization of beta-cat in primary brain endothelial cells (ECs) in vitro by N-terminal truncation or Wnt3a treatment increases Cldn3 expression, BBB-type tight junction formation, and a BBB characteristic gene signature. Loss of beta-cat or inhibition of its signaling abrogates this effect. Furthermore, stabilization of beta-cat also increased Cldn3 and barrier properties in nonbrain-derived ECs. These findings may open new therapeutic avenues to modulate endothelial barrier function and to limit the devastating effects of BBB breakdown.


Asunto(s)
Barrera Hematoencefálica/fisiología , Sistema Nervioso Central/fisiología , Circulación Cerebrovascular/fisiología , Neovascularización Fisiológica/fisiología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Genes Reporteros , Humanos , Ratones , Modelos Animales , Transducción de Señal , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
12.
Cancer Cell ; 13(5): 407-19, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18455124

RESUMEN

The Drosophila transcription factor Prospero functions as a tumor suppressor, and it has been suggested that the human counterpart of Prospero, PROX1, acts similarly in human cancers. However, we show here that PROX1 promotes dysplasia in colonic adenomas and colorectal cancer progression. PROX1 expression marks the transition from benign colon adenoma to carcinoma in situ, and its loss inhibits growth of human colorectal tumor xenografts and intestinal adenomas in Apc(min/+) mice, while its transgenic overexpression promotes colorectal tumorigenesis. Furthermore, in intestinal tumors PROX1 is a direct and dose-dependent target of the beta-catenin/TCF signaling pathway, responsible for the neoplastic transformation. Our data underscore the complexity of cancer pathogenesis and implicate PROX1 in malignant tumor progression through the regulation of cell polarity and adhesion.


Asunto(s)
Adenoma/genética , Neoplasias del Colon/genética , Proteínas de Homeodominio/genética , Proteínas Supresoras de Tumor/genética , Adenoma/patología , Carcinoma in Situ/genética , Línea Celular Tumoral , Neoplasias del Colon/patología , Neoplasias Colorrectales/genética , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Fenotipo , beta Catenina/fisiología
13.
J Cereb Blood Flow Metab ; 28(6): 1249-60, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18382472

RESUMEN

In the central nervous system, a constant microenvironment required for neuronal cell activity is maintained by the blood-brain barrier (BBB). The BBB is formed by the brain microvascular endothelial cells (BMEC), which are sealed by tight junctions (TJ). To identify genes that are differentially expressed in BMEC compared with peripheral endothelial cells, we constructed a subtractive cDNA library from porcine BMEC (pBMEC) and aortic endothelial cells (AOEC). Screening the library for differentially expressed genes yielded 26 BMEC-specific transcripts, such as solute carrier family 35 member F2 (SLC35F2), ADP-ribosylation factor-like 5B (ARL5B), TSC22 domain family member 1 (TSC22D1), integral membrane protein 2A (ITM2A), and epithelial membrane protein 1 (EMP1). In this study, we show that EMP1 transcript is enriched in pBMEC compared with brain tissue and that EMP1 protein colocalizes with the TJ protein occludin in mouse BMEC by coimmunoprecipitation and in rat brain vessels by immunohistochemistry. Epithelial membrane protein 1 expression was transiently induced in laser-capture microdissected rat brain vessels after a 20-min global cerebral ischemia, in parallel with the loss of occludin immunoreactivity. The study identifies EMP1 as a novel TJ-associated protein of the BBB and suggests its potential role in the regulation of the BBB function in cerebral ischemia.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Uniones Estrechas/metabolismo , Secuencia de Aminoácidos , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Células Cultivadas , Secuencia Conservada , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Alineación de Secuencia , Porcinos
14.
Proc Natl Acad Sci U S A ; 102(20): 7221-6, 2005 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-15870191

RESUMEN

A type of retroviral gene trap vectors has been developed that can induce conditional mutations in most genes expressed in mouse embryonic stem (ES) cells. The vectors rely on directional site-specific recombination systems that can repair and re-induce gene trap mutations when activated in succession. After the gene traps are inserted into the mouse genome, genetic mutations can be produced at a particular time and place in somatic cells. In addition to their conditional features, the vectors create multipurpose alleles amenable to a wide range of post-insertional modifications. Here we have used these directional recombination vectors to assemble the largest library of ES cell lines with conditional mutations in single genes yet assembled, presently totaling 1,000 unique genes. The trapped ES cell lines, which can be ordered from the German Gene Trap Consortium, are freely available to the scientific community.


Asunto(s)
Alelos , Genes/genética , Genoma , Genómica/métodos , Ratones/genética , Células Madre/metabolismo , Animales , Northern Blotting , Western Blotting , Línea Celular , Biología Computacional , Embrión de Mamíferos/citología , Vectores Genéticos/genética , Mutagénesis Sitio-Dirigida , Mutación/genética , Reacción en Cadena de la Polimerasa , Retroviridae , Células Madre/citología
15.
Trends Cell Biol ; 14(12): 657-9, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15564041

RESUMEN

Transforming growth factor (TGF)-betas are powerful cytokines that are secreted as inactive (latent) precursors into the extracellular space. To exert their pleiotropic functions, latent TGF-betas require activation. This requisite restricts TGF-beta signaling to tissues that express TGF-beta-activating proteins such as the adhesion molecule alphavbeta6 integrin. Recent work has uncovered the molecular mechanism by which alphavbeta6 integrin activates latent TGF-beta. Latent-TGF-beta-binding protein 1 has been identified as being the major component of this process, and the integrin-interacting region has been mapped to a poorly conserved sequence stretch called the hinge region.


Asunto(s)
Antígenos de Neoplasias/fisiología , Integrinas/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Antígenos de Neoplasias/metabolismo , Humanos , Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión a TGF-beta Latente , Modelos Biológicos , Isoformas de Proteínas/fisiología , Estrés Mecánico , Factor de Crecimiento Transformador beta/metabolismo
16.
J Cell Biol ; 167(1): 123-33, 2004 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-15466481

RESUMEN

Disruption of latent TGF-beta binding protein (LTBP)-4 expression in the mouse leads to abnormal lung development and colorectal cancer. Lung fibroblasts from these mice produced decreased amounts of active TGF-beta, whereas secretion of latent TGF-beta was significantly increased. Expression and secretion of TGF-beta2 and -beta3 increased considerably. These results suggested that TGF-beta activation but not secretion would be severely impaired in LTBP-4 -/- fibroblasts. Microarrays revealed increased expression of bone morphogenic protein (BMP)-4 and decreased expression of its inhibitor gremlin. This finding was accompanied by enhanced expression of BMP-4 target genes, inhibitors of differentiation 1 and 2, and increased deposition of fibronectin-rich extracellular matrix. Accordingly, increased expression of BMP-4 and decreased expression of gremlin were observed in mouse lung. Transfection of LTBP-4 rescued the -/- fibroblast phenotype, while LTBP-1 was inefficient. Treatment with active TGF-beta1 rescued BMP-4 and gremlin expression to wild-type levels. Our results indicate that the lack of LTBP-4-mediated targeting and activation of TGF-beta1 leads to enhanced BMP-4 signaling in mouse lung.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Northern Blotting , Proteína Morfogenética Ósea 4 , Diferenciación Celular , ADN Complementario/metabolismo , Electroforesis en Gel de Poliacrilamida , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de Unión a TGF-beta Latente , Pulmón/metabolismo , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos/química , Fenotipo , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba
17.
Genes Dev ; 16(17): 2264-73, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12208849

RESUMEN

Transforming growth factor-betas (TGF-betas) are multifunctional growth factors that are secreted as inactive (latent) precursors in large protein complexes. These complexes include the latency-associated propeptide (LAP) and a latent transforming growth factor-beta binding protein (LTBP). Four isoforms of LTBPs (LTBP-1-LTBP-4) have been cloned and are believed to be structural components of connective tissue microfibrils and local regulators of TGF-beta tissue deposition and signaling. By using a gene trap strategy that selects for integrations into genes induced transiently during early mouse development, we have disrupted the mouse homolog of the human LTBP-4 gene. Mice homozygous for the disrupted allele develop severe pulmonary emphysema, cardiomyopathy, and colorectal cancer. These highly tissue-specific abnormalities are associated with profound defects in the elastic fiber structure and with a reduced deposition of TGF-beta in the extracellular space. As a consequence, epithelial cells have reduced levels of phosphorylated Smad2 proteins, overexpress c-myc, and undergo uncontrolled proliferation. This phenotype supports the predicted dual role of LTBP-4 as a structural component of the extracellular matrix and as a local regulator of TGF-beta tissue deposition and signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Cardiomiopatías/genética , Proteínas Portadoras/genética , Neoplasias Colorrectales/genética , Proteínas de Unión a TGF-beta Latente/genética , Pulmón/anomalías , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Proteínas Portadoras/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Tejido Elástico/metabolismo , Tejido Elástico/patología , Matriz Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Humanos , Intrones , Proteínas de Unión a TGF-beta Latente/metabolismo , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Enfisema Pulmonar/genética , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA