Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Discov ; 12(2): 562-585, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34561242

RESUMEN

SMARCA4/BRG1 encodes for one of two mutually exclusive ATPases present in mammalian SWI/SNF chromatin remodeling complexes and is frequently mutated in human lung adenocarcinoma. However, the functional consequences of SMARCA4 mutation on tumor initiation, progression, and chromatin regulation in lung cancer remain poorly understood. Here, we demonstrate that loss of Smarca4 sensitizes club cell secretory protein-positive cells within the lung in a cell type-dependent fashion to malignant transformation and tumor progression, resulting in highly advanced dedifferentiated tumors and increased metastatic incidence. Consistent with these phenotypes, Smarca4-deficient primary tumors lack lung lineage transcription factor activities and resemble a metastatic cell state. Mechanistically, we show that Smarca4 loss impairs the function of all three classes of SWI/SNF complexes, resulting in decreased chromatin accessibility at lung lineage motifs and ultimately accelerating tumor progression. Thus, we propose that the SWI/SNF complex via Smarca4 acts as a gatekeeper for lineage-specific cellular transformation and metastasis during lung cancer evolution. SIGNIFICANCE: We demonstrate cell-type specificity in the tumor-suppressive functions of SMARCA4 in the lung, pointing toward a critical role of the cell-of-origin in driving SWI/SNF-mutant lung adenocarcinoma. We further show the direct effects of SMARCA4 loss on SWI/SNF function and chromatin regulation that cause aggressive malignancy during lung cancer evolution.This article is highlighted in the In This Issue feature, p. 275.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Transformación Celular Neoplásica , ADN Helicasas/genética , Neoplasias Pulmonares/genética , Metástasis de la Neoplasia , Proteínas Nucleares/genética , Factores de Transcripción/genética , Adenocarcinoma del Pulmón/secundario , Animales , Modelos Animales de Enfermedad , Humanos , Neoplasias Pulmonares/patología , Ratones
2.
Cancer Res ; 80(18): 3841-3854, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32690724

RESUMEN

Inactivation of SMARCA4/BRG1, the core ATPase subunit of mammalian SWI/SNF complexes, occurs at very high frequencies in non-small cell lung cancers (NSCLC). There are no targeted therapies for this subset of lung cancers, nor is it known how mutations in BRG1 contribute to lung cancer progression. Using a combination of gain- and loss-of-function approaches, we demonstrate that deletion of BRG1 in lung cancer leads to activation of replication stress responses. Single-molecule assessment of replication fork dynamics in BRG1-deficient cells revealed increased origin firing mediated by the prelicensing protein, CDC6. Quantitative mass spectrometry and coimmunoprecipitation assays showed that BRG1-containing SWI/SNF complexes interact with RPA complexes. Finally, BRG1-deficient lung cancers were sensitive to pharmacologic inhibition of ATR. These findings provide novel mechanistic insight into BRG1-mutant lung cancers and suggest that their dependency on ATR can be leveraged therapeutically and potentially expanded to BRG1-mutant cancers in other tissues. SIGNIFICANCE: These findings indicate that inhibition of ATR is a promising therapy for the 10% of non-small cell lung cancer patients harboring mutations in SMARCA4/BRG1. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/18/3841/F1.large.jpg.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/genética , ADN Helicasas/genética , Eliminación de Gen , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona , ADN Helicasas/deficiencia , Progresión de la Enfermedad , Femenino , Factores de Transcripción Forkhead , Edición Génica , Humanos , Inmunoprecipitación , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Análisis de Secuencia de ARN , Factores de Transcripción/deficiencia
3.
Am J Respir Cell Mol Biol ; 59(2): 237-245, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29447458

RESUMEN

Metastatic disease is the primary cause of death of patients with lung cancer, but the mouse models of lung adenocarcinoma do not accurately recapitulate the tumor microenvironment or metastatic disease observed in patients. In this study, we conditionally deleted E-cadherin in an autochthonous lung adenocarcinoma mouse model driven by activated oncogenic Kras and p53 loss. Loss of E-cadherin significantly accelerated lung adenocarcinoma progression and decreased survival of the mice. Kras;p53;E-cadherin mice had a 41% lung tumor burden, invasive grade 4 tumors, and a desmoplastic stroma just 8 weeks after tumor initiation. One hundred percent of the mice developed local metastases to the lymph nodes or chest wall, and 38% developed distant metastases to the liver or kidney. Lung adenocarcinoma cancer cell lines derived from these tumors also had high migratory rates. These studies demonstrate that the Kras;p53;E-cadherin mouse model better emulates the tumor microenvironment and metastases observed in patients with lung adenocarcinoma than previous models and may therefore be useful for studying metastasis and testing new lung cancer treatments in vivo.


Asunto(s)
Adenocarcinoma/patología , Cadherinas/metabolismo , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Adenocarcinoma/genética , Animales , Modelos Animales de Enfermedad , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/genética , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/genética , Proteínas Proto-Oncogénicas p21(ras)/genética
5.
Mol Ther ; 25(9): 1999-2013, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28619647

RESUMEN

Patient-derived induced pluripotent stem cells (iPSCs) hold great promise for autologous cell replacement. However, for many inherited diseases, treatment will likely require genetic repair pre-transplantation. Genome editing technologies are useful for this application. The purpose of this study was to develop CRISPR-Cas9-mediated genome editing strategies to target and correct the three most common types of disease-causing variants in patient-derived iPSCs: (1) exonic, (2) deep intronic, and (3) dominant gain of function. We developed a homology-directed repair strategy targeting a homozygous Alu insertion in exon 9 of male germ cell-associated kinase (MAK) and demonstrated restoration of the retinal transcript and protein in patient cells. We generated a CRISPR-Cas9-mediated non-homologous end joining (NHEJ) approach to excise a major contributor to Leber congenital amaurosis, the IVS26 cryptic-splice mutation in CEP290, and demonstrated correction of the transcript and protein in patient iPSCs. Lastly, we designed allele-specific CRISPR guides that selectively target the mutant Pro23His rhodopsin (RHO) allele, which, following delivery to both patient iPSCs in vitro and pig retina in vivo, created a frameshift and premature stop that would prevent transcription of the disease-causing variant. The strategies developed in this study will prove useful for correcting a wide range of genetic variants in genes that cause inherited retinal degeneration.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Marcación de Gen , Células Madre Pluripotentes Inducidas/metabolismo , Degeneración Retiniana/genética , Trasplante de Células Madre , Alelos , Animales , Línea Celular , Orden Génico , Sitios Genéticos , Terapia Genética , Vectores Genéticos/genética , Recombinación Homóloga , Humanos , Células Madre Pluripotentes Inducidas/citología , Intrones , Mutación , Proteínas Serina-Treonina Quinasas/genética , ARN Guía de Kinetoplastida , Degeneración Retiniana/terapia , Trasplante de Células Madre/métodos , Trasplante Autólogo
6.
Nat Commun ; 8: 14922, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28387316

RESUMEN

Adenosquamous lung tumours, which are extremely poor prognosis, may result from cellular plasticity. Here, we demonstrate lineage switching of KRAS+ lung adenocarcinomas (ADC) to squamous cell carcinoma (SCC) through deletion of Lkb1 (Stk11) in autochthonous and transplant models. Chromatin analysis reveals loss of H3K27me3 and gain of H3K27ac and H3K4me3 at squamous lineage genes, including Sox2, ΔNp63 and Ngfr. SCC lesions have higher levels of the H3K27 methyltransferase EZH2 than the ADC lesions, but there is a clear lack of the essential Polycomb Repressive Complex 2 (PRC2) subunit EED in the SCC lesions. The pattern of high EZH2, but low H3K27me3 mark, is also prevalent in human lung SCC and SCC regions within ADSCC tumours. Using FACS-isolated populations, we demonstrate that bronchioalveolar stem cells and club cells are the likely cells-of-origin for SCC transitioned tumours. These findings shed light on the epigenetics and cellular origins of lineage-specific lung tumours.


Asunto(s)
Adenocarcinoma/genética , Carcinoma de Células Escamosas/genética , Neoplasias Pulmonares/genética , Complejo Represivo Polycomb 2/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Quinasas Activadas por AMP , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Estimación de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Metilación , Ratones de la Cepa 129 , Ratones Noqueados , Complejo Represivo Polycomb 2/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células Tumorales Cultivadas
7.
Sci Transl Med ; 9(376)2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28179501

RESUMEN

Diamond-Blackfan anemia (DBA) is a congenital disorder characterized by the failure of erythroid progenitor differentiation, severely curtailing red blood cell production. Because many DBA patients fail to respond to corticosteroid therapy, there is considerable need for therapeutics for this disorder. Identifying therapeutics for DBA requires circumventing the paucity of primary patient blood stem and progenitor cells. To this end, we adopted a reprogramming strategy to generate expandable hematopoietic progenitor cells from induced pluripotent stem cells (iPSCs) from DBA patients. Reprogrammed DBA progenitors recapitulate defects in erythroid differentiation, which were rescued by gene complementation. Unbiased chemical screens identified SMER28, a small-molecule inducer of autophagy, which enhanced erythropoiesis in a range of in vitro and in vivo models of DBA. SMER28 acted through autophagy factor ATG5 to stimulate erythropoiesis and up-regulate expression of globin genes. These findings present an unbiased drug screen for hematological disease using iPSCs and identify autophagy as a therapeutic pathway in DBA.


Asunto(s)
Anemia de Diamond-Blackfan/tratamiento farmacológico , Descubrimiento de Drogas , Células Madre Hematopoyéticas/metabolismo , Compuestos Alílicos/farmacología , Anemia de Diamond-Blackfan/patología , Antígenos CD34/metabolismo , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/metabolismo , Diferenciación Celular/efectos de los fármacos , Reprogramación Celular , Células Eritroides/efectos de los fármacos , Células Eritroides/patología , Eritropoyesis/efectos de los fármacos , Prueba de Complementación Genética , Globinas/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Quinazolinas/farmacología
8.
Nat Biotechnol ; 33(1): 58-63, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25437882

RESUMEN

Human induced pluripotent stem cells (hiPSCs) are useful in disease modeling and drug discovery, and they promise to provide a new generation of cell-based therapeutics. To date there has been no systematic evaluation of the most widely used techniques for generating integration-free hiPSCs. Here we compare Sendai-viral (SeV), episomal (Epi) and mRNA transfection mRNA methods using a number of criteria. All methods generated high-quality hiPSCs, but significant differences existed in aneuploidy rates, reprogramming efficiency, reliability and workload. We discuss the advantages and shortcomings of each approach, and present and review the results of a survey of a large number of human reprogramming laboratories on their independent experiences and preferences. Our analysis provides a valuable resource to inform the use of specific reprogramming methods for different laboratories and different applications, including clinical translation.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Humanos
9.
Nat Biotechnol ; 32(11): 1151-1157, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25306246

RESUMEN

The ability to differentiate human pluripotent stem cells into endothelial cells with properties of cord-blood endothelial colony-forming cells (CB-ECFCs) may enable the derivation of clinically relevant numbers of highly proliferative blood vessel-forming cells to restore endothelial function in patients with vascular disease. We describe a protocol to convert human induced pluripotent stem cells (hiPSCs) or embryonic stem cells (hESCs) into cells similar to CB-ECFCs at an efficiency of >10(8) ECFCs produced from each starting pluripotent stem cell. The CB-ECFC-like cells display a stable endothelial phenotype with high clonal proliferative potential and the capacity to form human vessels in mice and to repair the ischemic mouse retina and limb, and they lack teratoma formation potential. We identify Neuropilin-1 (NRP-1)-mediated activation of KDR signaling through VEGF165 as a critical mechanism for the emergence and maintenance of CB-ECFC-like cells.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Endoteliales/citología , Células Madre Pluripotentes/citología , Animales , Proliferación Celular/genética , Células Endoteliales/metabolismo , Sangre Fetal/citología , Humanos , Ratones , Neuropilina-1/metabolismo , Células Madre/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
J Am Chem Soc ; 130(46): 15246-7, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-18939837

RESUMEN

A bis-phenanthroline [2]catenane copper complex, consisting of one olefinic macrocycle and one nonolefinic macrocycle, underwent an entropy-driven ring-opening olefin metathesis polymerization (ROMP) to provide a polypseudorotaxane. The polymerization featured an average degree of polymerization of ca. 63 wherein the polymer was effectively saturated with threaded macrocycles. Removal of the copper led to near complete release of the macrocycles from the polymer backbone.


Asunto(s)
Catenanos/química , Polímeros/química , Rotaxanos/química , Estructura Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA