Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Cell ; 174(1): 172-186.e21, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29958106

RESUMEN

The fusion oncoprotein CBFß-SMMHC, expressed in leukemia cases with chromosome 16 inversion, drives leukemia development and maintenance by altering the activity of the transcription factor RUNX1. Here, we demonstrate that CBFß-SMMHC maintains cell viability by neutralizing RUNX1-mediated repression of MYC expression. Upon pharmacologic inhibition of the CBFß-SMMHC/RUNX1 interaction, RUNX1 shows increased binding at three MYC distal enhancers, where it represses MYC expression by mediating the replacement of the SWI/SNF complex component BRG1 with the polycomb-repressive complex component RING1B, leading to apoptosis. Combining the CBFß-SMMHC inhibitor with the BET inhibitor JQ1 eliminates inv(16) leukemia in human cells and a mouse model. Enhancer-interaction analysis indicated that the three enhancers are physically connected with the MYC promoter, and genome-editing analysis demonstrated that they are functionally implicated in deregulation of MYC expression. This study reveals a mechanism whereby CBFß-SMMHC drives leukemia maintenance and suggests that inhibitors targeting chromatin activity may prove effective in inv(16) leukemia therapy.


Asunto(s)
Apoptosis , Cromatina/metabolismo , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Apoptosis/efectos de los fármacos , Azepinas/farmacología , Azepinas/uso terapéutico , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Inversión Cromosómica/efectos de los fármacos , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , ADN/química , ADN/metabolismo , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Triazoles/farmacología , Triazoles/uso terapéutico
2.
Mol Cell ; 82(6): 1225-1238.e6, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35196517

RESUMEN

The long-range interactions of cis-regulatory elements (cREs) play a central role in gene regulation. cREs can be characterized as accessible chromatin sequences. However, it remains technically challenging to comprehensively identify their spatial interactions. Here, we report a new method HiCAR (Hi-C on accessible regulatory DNA), which utilizes Tn5 transposase and chromatin proximity ligation, for the analysis of open-chromatin-anchored interactions with low-input cells. By applying HiCAR in human embryonic stem cells and lymphoblastoid cells, we demonstrate that HiCAR identifies high-resolution chromatin contacts with an efficiency comparable with that of in situ Hi-C over all distance ranges. Interestingly, we found that the "poised" gene promoters exhibit silencer-like function to repress the expression of distal genes via promoter-promoter interactions. Lastly, we applied HiCAR to 30,000 primary human muscle stem cells and demonstrated that HiCAR is capable of analyzing chromatin accessibility and looping using low-input primary cells and clinical samples.


Asunto(s)
Cromatina , Secuencias Reguladoras de Ácidos Nucleicos , Cromatina/genética , ADN , Regulación de la Expresión Génica , Humanos , Regiones Promotoras Genéticas
4.
Nature ; 590(7844): 129-133, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33408418

RESUMEN

Regeneration is a complex chain of events that restores a tissue to its original size and shape. The tissue-wide coordination of cellular dynamics that is needed for proper morphogenesis is challenged by the large dimensions of regenerating body parts. Feedback mechanisms in biochemical pathways can provide effective communication across great distances1-5, but how they might regulate growth during tissue regeneration is unresolved6,7. Here we report that rhythmic travelling waves of Erk activity control the growth of bone in time and space in regenerating zebrafish scales, millimetre-sized discs of protective body armour. We find that waves of Erk activity travel across the osteoblast population as expanding concentric rings that are broadcast from a central source, inducing ring-like patterns of tissue growth. Using a combination of theoretical and experimental analyses, we show that Erk activity propagates as excitable trigger waves that are able to traverse the entire scale in approximately two days and that the frequency of wave generation controls the rate of scale regeneration. Furthermore, the periodic induction of synchronous, tissue-wide activation of Erk in place of travelling waves impairs tissue growth, which indicates that wave-distributed Erk activation is key to regeneration. Our findings reveal trigger waves as a regulatory strategy to coordinate cell behaviour and instruct tissue form during regeneration.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Osteoblastos/citología , Osteoblastos/metabolismo , Regeneración , Pez Cebra/fisiología , Escamas de Animales/citología , Escamas de Animales/enzimología , Escamas de Animales/crecimiento & desarrollo , Escamas de Animales/fisiología , Animales , Difusión , Femenino , Masculino , Pez Cebra/crecimiento & desarrollo
5.
Development ; 149(4)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35179181

RESUMEN

The epicardium is a mesothelial tissue layer that envelops the heart. Cardiac injury activates dynamic gene expression programs in epicardial tissue, which in zebrafish enables subsequent regeneration through paracrine and vascularizing effects. To identify tissue regeneration enhancer elements (TREEs) that control injury-induced epicardial gene expression during heart regeneration, we profiled transcriptomes and chromatin accessibility in epicardial cells purified from regenerating zebrafish hearts. We identified hundreds of candidate TREEs, which are defined by increased chromatin accessibility of non-coding elements near genes with increased expression during regeneration. Several of these candidate TREEs were incorporated into stable transgenic lines, with five out of six elements directing injury-induced epicardial expression but not ontogenetic epicardial expression in larval hearts. Whereas two independent TREEs linked to the gene gnai3 showed similar functional features of gene regulation in transgenic lines, two independent ncam1a-linked TREEs directed distinct spatiotemporal domains of epicardial gene expression. Thus, multiple TREEs linked to a regeneration gene can possess either matching or complementary regulatory controls. Our study provides a new resource and principles for understanding the regulation of epicardial genetic programs during heart regeneration. This article has an associated 'The people behind the papers' interview.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Corazón/fisiología , Pericardio/metabolismo , Regeneración/fisiología , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Cromatina/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Regulación de la Expresión Génica , Larva/crecimiento & desarrollo , Larva/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Pericardio/citología , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
Mol Cell ; 62(4): 479-90, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-27184077

RESUMEN

Recurrent mutations in the splicing factor U2AF35 are found in several cancers and myelodysplastic syndrome (MDS). How oncogenic U2AF35 mutants promote transformation remains to be determined. Here we derive cell lines transformed by the oncogenic U2AF35(S34F) mutant and identify aberrantly processed pre-mRNAs by deep sequencing. We find that in U2AF35(S34F)-transformed cells the autophagy-related factor 7 (Atg7) pre-mRNA is abnormally processed, which unexpectedly is not due to altered splicing but rather selection of a distal cleavage and polyadenylation (CP) site. This longer Atg7 mRNA is translated inefficiently, leading to decreased ATG7 levels and an autophagy defect that predisposes cells to secondary mutations, resulting in transformation. MDS and acute myeloid leukemia patient samples harboring U2AF35(S34F) have a similar increased use of the ATG7 distal CP site, and previous studies have shown that mice with hematopoietic cells lacking Atg7 develop an MDS-like syndrome. Collectively, our results reveal a basis for U2AF35(S34F) oncogenic activity.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/genética , Transformación Celular Neoplásica/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Procesamiento de Término de ARN 3' , Precursores del ARN/genética , ARN Mensajero/genética , Factor de Empalme U2AF/genética , Anciano , Anciano de 80 o más Años , Animales , Autofagia , Proteína 7 Relacionada con la Autofagia/metabolismo , Línea Celular Transformada , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Mitocondrias/metabolismo , Mitocondrias/patología , Mutación , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Poliadenilación , Interferencia de ARN , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Factor de Empalme U2AF/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral
7.
Circulation ; 146(1): 48-63, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35652354

RESUMEN

BACKGROUND: Certain nonmammalian species such as zebrafish have an elevated capacity for innate heart regeneration. Understanding how heart regeneration occurs in these contexts can help illuminate cellular and molecular events that can be targets for heart failure prevention or treatment. The epicardium, a mesothelial tissue layer that encompasses the heart, is a dynamic structure that is essential for cardiac regeneration in zebrafish. The extent to which different cell subpopulations or states facilitate heart regeneration requires research attention. METHODS: To dissect epicardial cell states and associated proregenerative functions, we performed single-cell RNA sequencing and identified 7 epicardial cell clusters in adult zebrafish, 3 of which displayed enhanced cell numbers during regeneration. We identified paralogs of hapln1 as factors associated with the extracellular matrix and largely expressed in cluster 1. We assessed HAPLN1 expression in published single-cell RNA sequencing data sets from different stages and injury states of murine and human hearts, and we performed molecular genetics to determine the requirements for hapln1-expressing cells and functions of each hapln1 paralog. RESULTS: A particular cluster of epicardial cells had the strongest association with regeneration and was marked by expression of hapln1a and hapln1b. The hapln1 paralogs are expressed in epicardial cells that enclose dedifferentiated and proliferating cardiomyocytes during regeneration. Induced genetic depletion of hapln1-expressing cells or genetic inactivation of hapln1b altered deposition of the key extracellular matrix component hyaluronic acid, disrupted cardiomyocyte proliferation, and inhibited heart regeneration. We also found that hapln1-expressing epicardial cells first emerge at the juvenile stage, when they associate with and are required for focused cardiomyocyte expansion events that direct maturation of the ventricular wall. CONCLUSIONS: Our findings identify a subset of epicardial cells that emerge in postembryonic zebrafish and sponsor regions of active cardiomyogenesis during cardiac growth and regeneration. We provide evidence that, as the heart achieves its mature structure, these cells facilitate hyaluronic acid deposition to support formation of the compact muscle layer of the ventricle. They are also required, along with the function of hapln1b paralog, in the production and organization of hyaluronic acid-containing matrix in cardiac injury sites, enabling normal cardiomyocyte proliferation and muscle regeneration.


Asunto(s)
Proteínas de la Matriz Extracelular , Corazón , Miocitos Cardíacos , Proteoglicanos , Animales , Proliferación Celular , Proteínas de la Matriz Extracelular/metabolismo , Corazón/fisiología , Humanos , Ácido Hialurónico/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Organogénesis , Proteoglicanos/metabolismo , Regeneración/fisiología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
8.
Development ; 147(14)2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32665240

RESUMEN

To identify candidate tissue regeneration enhancer elements (TREEs) important for zebrafish fin regeneration, we performed ATAC-seq from bulk tissue or purified fibroblasts of uninjured and regenerating caudal fins. We identified tens of thousands of DNA regions from each sample type with dynamic accessibility during regeneration, and assigned these regions to proximal genes with corresponding expression changes by RNA-seq. To determine whether these profiles reveal bona fide TREEs, we tested the sufficiency and requirements of several sequences in stable transgenic lines and mutant lines with homozygous deletions. These experiments validated new non-coding regulatory sequences near induced and/or essential genes during fin regeneration, including fgf20a, mdka and cx43, identifying distinct domains of directed expression for each confirmed TREE. Whereas deletion of the previously identified LEN enhancer abolished detectable induction of the nearby leptin b gene during regeneration, deletions of enhancers linked to fgf20a, mdka and cx43 had no effect or partially reduced gene expression. Our study generates a new resource for dissecting the regulatory mechanisms of appendage generation and reveals a range of requirements for individual TREEs in control of regeneration programs.


Asunto(s)
Aletas de Animales/metabolismo , Elementos de Facilitación Genéticos/genética , Regeneración/fisiología , Pez Cebra/metabolismo , Aletas de Animales/fisiología , Animales , Animales Modificados Genéticamente/metabolismo , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Conexina 43/genética , Conexina 43/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Expresión Génica , Leptina/genética , Leptina/metabolismo , Midkina/genética , Midkina/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
9.
Mol Cell ; 58(5): 819-31, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25921069

RESUMEN

The mechanisms that regulate alternative precursor mRNA (pre-mRNA) splicing are largely unknown. Here, we perform an RNAi screen to identify factors required for alternative splicing regulation by RBFOX2, an RNA-binding protein that promotes either exon inclusion or exclusion. Unexpectedly, we find that two mRNA 3' end formation factors, cleavage and polyadenylation specificity factor (CPSF) and SYMPK, are RBFOX2 cofactors for both inclusion and exclusion of internal exons. RBFOX2 interacts with CPSF/SYMPK and recruits it to the pre-mRNA. RBFOX2 and CPSF/SYMPK then function together to regulate binding of the early intron recognition factors U2AF and U1 small nuclear ribonucleoprotein particle (snRNP). Genome-wide analysis reveals that CPSF also mediates alternative splicing of many internal exons in the absence of RBFOX2. Accordingly, we show that CPSF/SYMPK is also a cofactor of NOVA2 and heterologous nuclear ribonucleoprotein A1 (HNRNPA1), RNA-binding proteins that also regulate alternative splicing. Collectively, our results reveal an unanticipated role for mRNA 3' end formation factors in global promotion of alternative splicing.


Asunto(s)
Empalme Alternativo , Factor de Especificidad de Desdoblamiento y Poliadenilación/fisiología , Proteínas Nucleares/metabolismo , ARN Mensajero/genética , Secuencia de Bases , Exones , Genes Reporteros , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Unión Proteica , Sitios de Empalme de ARN , Factores de Empalme de ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Ribonucleoproteínas/metabolismo , Análisis de Secuencia de ARN , Factor de Empalme U2AF
10.
PLoS Genet ; 16(4): e1008600, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32343701

RESUMEN

Upon exposure to environmental stressors, cells transiently arrest the cell cycle while they adapt and restore homeostasis. A challenge for all cells is to distinguish between stress signals and coordinate the appropriate adaptive response with cell cycle arrest. Here we investigate the role of the phosphatase calcineurin (CN) in the stress response and demonstrate that CN activates the Hog1/p38 pathway in both yeast and human cells. In yeast, the MAPK Hog1 is transiently activated in response to several well-studied osmostressors. We show that when a stressor simultaneously activates CN and Hog1, CN disrupts Hog1-stimulated negative feedback to prolong Hog1 activation and the period of cell cycle arrest. Regulation of Hog1 by CN also contributes to inactivation of multiple cell cycle-regulatory transcription factors (TFs) and the decreased expression of cell cycle-regulated genes. CN-dependent downregulation of G1/S genes is dependent upon Hog1 activation, whereas CN inactivates G2/M TFs through a combination of Hog1-dependent and -independent mechanisms. These findings demonstrate that CN and Hog1 act in a coordinated manner to inhibit multiple nodes of the cell cycle-regulatory network. Our results suggest that crosstalk between CN and stress-activated MAPKs helps cells tailor their adaptive responses to specific stressors.


Asunto(s)
Calcineurina/metabolismo , Ciclo Celular , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , Estrés Fisiológico/fisiología , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Regulación hacia Abajo , Retroalimentación Fisiológica , Regulación Fúngica de la Expresión Génica , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Estrés Fisiológico/genética , Factores de Transcripción/química , Factores de Transcripción/metabolismo
11.
Genome Res ; 29(8): 1235-1249, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31201210

RESUMEN

In interphase eukaryotic cells, almost all heterochromatin is located adjacent to the nucleolus or to the nuclear lamina, thus defining nucleolus-associated domains (NADs) and lamina-associated domains (LADs), respectively. Here, we determined the first genome-scale map of murine NADs in mouse embryonic fibroblasts (MEFs) via deep sequencing of chromatin associated with purified nucleoli. We developed a Bioconductor package called NADfinder and demonstrated that it identifies NADs more accurately than other peak-calling tools, owing to its critical feature of chromosome-level local baseline correction. We detected two distinct classes of NADs. Type I NADs associate frequently with both the nucleolar periphery and the nuclear lamina, and generally display characteristics of constitutive heterochromatin, including late DNA replication, enrichment of H3K9me3, and little gene expression. In contrast, Type II NADs associate with nucleoli but do not overlap with LADs. Type II NADs tend to replicate earlier, display greater gene expression, and are more often enriched in H3K27me3 than Type I NADs. The nucleolar associations of both classes of NADs were confirmed via DNA-FISH, which also detected Type I but not Type II probes enriched at the nuclear lamina. Type II NADs are enriched in distinct gene classes, including factors important for differentiation and development. In keeping with this, we observed that a Type II NAD is developmentally regulated, and present in MEFs but not in undifferentiated embryonic stem (ES) cells.


Asunto(s)
Nucléolo Celular/metabolismo , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genoma , Heterocromatina/clasificación , Animales , Nucléolo Celular/ultraestructura , Células Cultivadas , Mapeo Cromosómico/métodos , Replicación del ADN , Embrión de Mamíferos , Fibroblastos/ultraestructura , Heterocromatina/química , Heterocromatina/ultraestructura , Histonas/genética , Histonas/metabolismo , Hibridación Fluorescente in Situ , Ratones , Lámina Nuclear/metabolismo , Lámina Nuclear/ultraestructura
12.
Mol Cell ; 55(6): 904-915, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25219500

RESUMEN

Most colorectal cancers (CRCs) containing activated BRAF (BRAF[V600E]) have a CpG island methylator phenotype (CIMP) characterized by aberrant hypermethylation of many genes, including the mismatch repair gene MLH1. MLH1 silencing results in microsatellite instability and a hypermutable phenotype. Through an RNAi screen, here we identify the transcriptional repressor MAFG as the pivotal factor required for MLH1 silencing and CIMP in CRCs containing BRAF(V600E). In BRAF-positive human CRC cell lines and tumors, MAFG is bound at the promoters of MLH1 and other CIMP genes, and recruits a corepressor complex that includes its heterodimeric partner BACH1, the chromatin remodeling factor CHD8, and the DNA methyltransferase DNMT3B, resulting in hypermethylation and transcriptional silencing. BRAF(V600E) increases BRAF/MEK/ERK signaling resulting in phosphorylation and elevated levels of MAFG, which drives DNA binding. Analysis of transcriptionally silenced CIMP genes in KRAS-positive CRCs indicates that different oncoproteins direct the assembly of distinct repressor complexes on common promoters.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias Colorrectales/genética , Islas de CpG/genética , Factor de Transcripción MafG/metabolismo , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Represoras/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/patología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Ratones Endogámicos BALB C , Homólogo 1 de la Proteína MutL , Mutación , Neoplasias Experimentales , Fenotipo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas B-raf/genética , Transducción de Señal , Factores de Transcripción/metabolismo , ADN Metiltransferasa 3B
13.
Proc Natl Acad Sci U S A ; 116(21): 10482-10487, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31068472

RESUMEN

A major obstacle to curing chronic myeloid leukemia (CML) is the intrinsic resistance of CML stem cells (CMLSCs) to the drug imatinib mesylate (IM). Prosurvival genes that are preferentially expressed in CMLSCs compared with normal hematopoietic stem cells (HSCs) represent potential therapeutic targets for selectively eradicating CMLSCs. However, the discovery of such preferentially expressed genes has been hampered by the inability to completely separate CMLSCs from HSCs, which display a very similar set of surface markers. To overcome this challenge, and to minimize confounding effects of individual differences in gene expression profiles, we performed single-cell RNA-seq on CMLSCs and HSCs that were isolated from the same patient and distinguished based on the presence or absence of BCR-ABL. Among genes preferentially expressed in CMLSCs is PIM2, which encodes a prosurvival serine-threonine kinase that phosphorylates and inhibits the proapoptotic protein BAD. We show that IM resistance of CMLSCs is due, at least in part, to maintenance of BAD phosphorylation by PIM2. We find that in CMLSCs, PIM2 expression is promoted by both a BCR-ABL-dependent (IM-sensitive) STAT5-mediated pathway and a BCR-ABL-independent (IM-resistant) STAT4-mediated pathway. Combined treatment with IM and a PIM inhibitor synergistically increases apoptosis of CMLSCs, suppresses colony formation, and significantly prolongs survival in a mouse CML model, with a negligible effect on HSCs. Our results reveal a therapeutically targetable mechanism of IM resistance in CMLSCs. The experimental approach that we describe can be generally applied to other malignancies that harbor oncogenic fusion proteins or other characteristic genetic markers.


Asunto(s)
Compuestos de Bifenilo/uso terapéutico , Resistencia a Antineoplásicos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Células Madre Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Tiazolidinas/uso terapéutico , Animales , Ensayos de Selección de Medicamentos Antitumorales , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Mesilato de Imatinib , Leucemia Experimental/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Terapia Molecular Dirigida , Fosforilación , Inhibidores de Proteínas Quinasas , Factores de Transcripción STAT/metabolismo , Proteína Letal Asociada a bcl/metabolismo
14.
Nature ; 516(7529): 116-20, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25470042

RESUMEN

The TRIM37 (also known as MUL) gene is located in the 17q23 chromosomal region, which is amplified in up to ∼ 40% of breast cancers. TRIM37 contains a RING finger domain, a hallmark of E3 ubiquitin ligases, but its protein substrate(s) is unknown. Here we report that TRIM37 mono-ubiquitinates histone H2A, a chromatin modification associated with transcriptional repression. We find that in human breast cancer cell lines containing amplified 17q23, TRIM37 is upregulated and, reciprocally, the major H2A ubiquitin ligase RNF2 (also known as RING1B) is downregulated. Genome-wide chromatin immunoprecipitation (ChIP)-chip experiments in 17q23-amplified breast cancer cells identified many genes, including multiple tumour suppressors, whose promoters were bound by TRIM37 and enriched for ubiquitinated H2A. However, unlike RNF2, which is a subunit of polycomb repressive complex 1 (PRC1), we find that TRIM37 associates with polycomb repressive complex 2 (PRC2). TRIM37, PRC2 and PRC1 are co-bound to specific target genes, resulting in their transcriptional silencing. RNA-interference-mediated knockdown of TRIM37 results in loss of ubiquitinated H2A, dissociation of PRC1 and PRC2 from target promoters, and transcriptional reactivation of silenced genes. Knockdown of TRIM37 in human breast cancer cells containing amplified 17q23 substantially decreases tumour growth in mouse xenografts. Conversely, ectopic expression of TRIM37 renders non-transformed cells tumorigenic. Collectively, our results reveal TRIM37 as an oncogenic H2A ubiquitin ligase that is overexpressed in a subset of breast cancers and promotes transformation by facilitating silencing of tumour suppressors and other genes.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/genética , Complejo Represivo Polycomb 1/genética , Animales , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Xenoinjertos , Histonas/metabolismo , Humanos , Células MCF-7 , Ratones , Células 3T3 NIH , Proteínas Oncogénicas/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas
15.
Proc Natl Acad Sci U S A ; 114(44): 11751-11756, 2017 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-29078326

RESUMEN

Developing tools to accurately predict the clinical prevalence of drug-resistant mutations is a key step toward generating more effective therapeutics. Here we describe a high-throughput CRISPR-Cas9-based saturated mutagenesis approach to generate comprehensive libraries of point mutations at a defined genomic location and systematically study their effect on cell growth. As proof of concept, we mutagenized a selected region within the leukemic oncogene BCR-ABL1 Using bulk competitions with a deep-sequencing readout, we analyzed hundreds of mutations under multiple drug conditions and found that the effects of mutations on growth in the presence or absence of drug were critical for predicting clinically relevant resistant mutations, many of which were cancer adaptive in the absence of drug pressure. Using this approach, we identified all clinically isolated BCR-ABL1 mutations and achieved a prediction score that correlated highly with their clinical prevalence. The strategy described here can be broadly applied to a variety of oncogenes to predict patient mutations and evaluate resistance susceptibility in the development of new therapeutics.


Asunto(s)
Sistemas CRISPR-Cas/genética , Resistencia a Antineoplásicos/genética , Mutagénesis/genética , Animales , Antineoplásicos/farmacología , Sistemas CRISPR-Cas/efectos de los fármacos , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/efectos de los fármacos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas de Fusión bcr-abl/genética , Leucemia/tratamiento farmacológico , Leucemia/genética , Ratones , Mutagénesis/efectos de los fármacos , Oncogenes/genética , Mutación Puntual/efectos de los fármacos , Mutación Puntual/genética
16.
BMC Genomics ; 19(1): 169, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29490630

RESUMEN

BACKGROUND: ATAC-seq (Assays for Transposase-Accessible Chromatin using sequencing) is a recently developed technique for genome-wide analysis of chromatin accessibility. Compared to earlier methods for assaying chromatin accessibility, ATAC-seq is faster and easier to perform, does not require cross-linking, has higher signal to noise ratio, and can be performed on small cell numbers. However, to ensure a successful ATAC-seq experiment, step-by-step quality assurance processes, including both wet lab quality control and in silico quality assessment, are essential. While several tools have been developed or adopted for assessing read quality, identifying nucleosome occupancy and accessible regions from ATAC-seq data, none of the tools provide a comprehensive set of functionalities for preprocessing and quality assessment of aligned ATAC-seq datasets. RESULTS: We have developed a Bioconductor package, ATACseqQC, for easily generating various diagnostic plots to help researchers quickly assess the quality of their ATAC-seq data. In addition, this package contains functions to preprocess aligned ATAC-seq data for subsequent peak calling. Here we demonstrate the utilities of our package using 25 publicly available ATAC-seq datasets from four studies. We also provide guidelines on what the diagnostic plots should look like for an ideal ATAC-seq dataset. CONCLUSIONS: This software package has been used successfully for preprocessing and assessing several in-house and public ATAC-seq datasets. Diagnostic plots generated by this package will facilitate the quality assessment of ATAC-seq data, and help researchers to evaluate their own ATAC-seq experiments as well as select high-quality ATAC-seq datasets from public repositories such as GEO to avoid generating hypotheses or drawing conclusions from low-quality ATAC-seq experiments. The software, source code, and documentation are freely available as a Bioconductor package at https://bioconductor.org/packages/release/bioc/html/ATACseqQC.html .


Asunto(s)
Biología Computacional/métodos , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Sitios de Unión , Elementos Transponibles de ADN , Proteínas de Unión al ADN , Estudio de Asociación del Genoma Completo , Secuenciación de Nucleótidos de Alto Rendimiento , Mutagénesis Insercional , Sitio de Iniciación de la Transcripción , Transposasas/genética , Transposasas/metabolismo , Navegador Web
18.
Proc Natl Acad Sci U S A ; 111(35): 12591-8, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25136103

RESUMEN

X-chromosome inactivation (XCI), the random transcriptional silencing of one X chromosome in somatic cells of female mammals, is a mechanism that ensures equal expression of X-linked genes in both sexes. XCI is initiated in cis by the noncoding Xist RNA, which coats the inactive X chromosome (Xi) from which it is produced. However, trans-acting factors that mediate XCI remain largely unknown. Here, we perform a large-scale RNA interference screen to identify trans-acting XCI factors (XCIFs) that comprise regulators of cell signaling and transcription, including the DNA methyltransferase, DNMT1. The expression pattern of the XCIFs explains the selective onset of XCI following differentiation. The XCIFs function, at least in part, by promoting expression and/or localization of Xist to the Xi. Surprisingly, we find that DNMT1, which is generally a transcriptional repressor, is an activator of Xist transcription. Small-molecule inhibitors of two of the XCIFs can reversibly reactivate the Xi, which has implications for treatment of Rett syndrome and other dominant X-linked diseases. A homozygous mouse knockout of one of the XCIFs, stanniocalcin 1 (STC1), has an expected XCI defect but surprisingly is phenotypically normal. Remarkably, X-linked genes are not overexpressed in female Stc1(-/-) mice, revealing the existence of a mechanism(s) that can compensate for a persistent XCI deficiency to regulate X-linked gene expression.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Glicoproteínas/genética , Proteína 2 de Unión a Metil-CpG/genética , ARN Largo no Codificante/genética , Síndrome de Rett/genética , Inactivación del Cromosoma X/genética , Animales , Corteza Cerebral/citología , Cromonas/farmacología , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Células Madre Embrionarias/fisiología , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/genética , Femenino , Fibroblastos/citología , Fibroblastos/fisiología , Biblioteca de Genes , Terapia Genética/métodos , Humanos , Mamíferos , Ratones , Ratones Noqueados , Morfolinas/farmacología , Neuronas/citología , Neuronas/fisiología , Pirazoles/farmacología , ARN Interferente Pequeño/genética , Síndrome de Rett/terapia , Sulfonamidas/farmacología , Transcriptoma , Inactivación del Cromosoma X/efectos de los fármacos
19.
Genome Res ; 23(6): 928-40, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23471540

RESUMEN

Cys2-His2 zinc finger proteins (ZFPs) are the largest group of transcription factors in higher metazoans. A complete characterization of these ZFPs and their associated target sequences is pivotal to fully annotate transcriptional regulatory networks in metazoan genomes. As a first step in this process, we have characterized the DNA-binding specificities of 129 zinc finger sets from Drosophila using a bacterial one-hybrid system. This data set contains the DNA-binding specificities for at least one encoded ZFP from 70 unique genes and 23 alternate splice isoforms representing the largest set of characterized ZFPs from any organism described to date. These recognition motifs can be used to predict genomic binding sites for these factors within the fruit fly genome. Subsets of fingers from these ZFPs were characterized to define their orientation and register on their recognition sequences, thereby allowing us to define the recognition diversity within this finger set. We find that the characterized fingers can specify 47 of the 64 possible DNA triplets. To confirm the utility of our finger recognition models, we employed subsets of Drosophila fingers in combination with an existing archive of artificial zinc finger modules to create ZFPs with novel DNA-binding specificity. These hybrids of natural and artificial fingers can be used to create functional zinc finger nucleases for editing vertebrate genomes.


Asunto(s)
Sitios de Unión , Proteínas de Drosophila/genética , Drosophila/genética , Motivos de Nucleótidos , Dedos de Zinc/genética , Empalme Alternativo , Animales , Secuencia de Bases , Análisis por Conglomerados , Biología Computacional/métodos , Proteínas de Drosophila/química , Proteínas de Drosophila/clasificación , Modelos Moleculares , Filogenia , Posición Específica de Matrices de Puntuación , Unión Proteica , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA