Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cell ; 166(6): 1423-1435.e12, 2016 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-27594426

RESUMEN

Apicomplexan parasites are leading causes of human and livestock diseases such as malaria and toxoplasmosis, yet most of their genes remain uncharacterized. Here, we present the first genome-wide genetic screen of an apicomplexan. We adapted CRISPR/Cas9 to assess the contribution of each gene from the parasite Toxoplasma gondii during infection of human fibroblasts. Our analysis defines ∼200 previously uncharacterized, fitness-conferring genes unique to the phylum, from which 16 were investigated, revealing essential functions during infection of human cells. Secondary screens identify as an invasion factor the claudin-like apicomplexan microneme protein (CLAMP), which resembles mammalian tight-junction proteins and localizes to secretory organelles, making it critical to the initiation of infection. CLAMP is present throughout sequenced apicomplexan genomes and is essential during the asexual stages of the malaria parasite Plasmodium falciparum. These results provide broad-based functional information on T. gondii genes and will facilitate future approaches to expand the horizon of antiparasitic interventions.


Asunto(s)
Apicomplexa/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Estudio de Asociación del Genoma Completo , Interacciones Huésped-Parásitos , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Toxoplasma/genética , Células Cultivadas , Claudinas/genética , Claudinas/metabolismo , Fibroblastos/parasitología , Genoma de Protozoos/genética , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/fisiopatología , Plasmodium falciparum/genética , Toxoplasmosis/parasitología , Toxoplasmosis/fisiopatología
2.
Cell ; 158(5): 1094-1109, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25171410

RESUMEN

It is increasingly appreciated that oncogenic transformation alters cellular metabolism to facilitate cell proliferation, but less is known about the metabolic changes that promote cancer cell aggressiveness. Here, we analyzed metabolic gene expression in cancer cell lines and found that a set of high-grade carcinoma lines expressing mesenchymal markers share a unique 44 gene signature, designated the "mesenchymal metabolic signature" (MMS). A FACS-based shRNA screen identified several MMS genes as essential for the epithelial-mesenchymal transition (EMT), but not for cell proliferation. Dihydropyrimidine dehydrogenase (DPYD), a pyrimidine-degrading enzyme, was highly expressed upon EMT induction and was necessary for cells to acquire mesenchymal characteristics in vitro and for tumorigenic cells to extravasate into the mouse lung. This role of DPYD was mediated through its catalytic activity and enzymatic products, the dihydropyrimidines. Thus, we identify metabolic processes essential for the EMT, a program associated with the acquisition of metastatic and aggressive cancer cell traits.


Asunto(s)
Transición Epitelial-Mesenquimal , Pirimidinas/metabolismo , Animales , Carcinoma/metabolismo , Línea Celular Tumoral , Dihidrouracilo Deshidrogenasa (NADP)/genética , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Mesodermo/citología , Mesodermo/metabolismo , Ratones , ARN Interferente Pequeño/metabolismo
3.
PLoS Genet ; 20(6): e1011241, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38870220

RESUMEN

Although introns are typically tens to thousands of nucleotides, there are notable exceptions. In flies as well as humans, a small number of genes contain introns that are more than 1000 times larger than typical introns, exceeding hundreds of kilobases (kb) to megabases (Mb). It remains unknown why gigantic introns exist and how cells overcome the challenges associated with their transcription and RNA processing. The Drosophila Y chromosome contains some of the largest genes identified to date: multiple genes exceed 4Mb, with introns accounting for over 99% of the gene span. Here we demonstrate that co-transcriptional splicing of these gigantic Y-linked genes is important to ensure successful transcription: perturbation of splicing led to the attenuation of transcription, leading to a failure to produce mature mRNA. Cytologically, defective splicing of the Y-linked gigantic genes resulted in disorganization of transcripts within the nucleus suggestive of entanglement of transcripts, likely resulting from unspliced long RNAs. We propose that co-transcriptional splicing maintains the length of nascent transcripts of gigantic genes under a critical threshold, preventing their entanglement and ensuring proper gene expression. Our study reveals a novel biological significance of co-transcriptional splicing.


Asunto(s)
Drosophila melanogaster , Intrones , Empalme del ARN , Transcripción Genética , Empalme del ARN/genética , Animales , Intrones/genética , Drosophila melanogaster/genética , Cromosoma Y/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Masculino , Humanos
4.
Nature ; 543(7647): 681-686, 2017 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-28329758

RESUMEN

Post-mitotic, differentiated cells exhibit a variety of characteristics that contrast with those of actively growing neoplastic cells, such as the expression of cell-cycle inhibitors and differentiation factors. We hypothesized that the gene expression profiles of these differentiated cells could reveal the identities of genes that may function as tumour suppressors. Here we show, using in vitro and in vivo studies in mice and humans, that the mitochondrial protein LACTB potently inhibits the proliferation of breast cancer cells. Its mechanism of action involves alteration of mitochondrial lipid metabolism and differentiation of breast cancer cells. This is achieved, at least in part, through reduction of the levels of mitochondrial phosphatidylserine decarboxylase, which is involved in the synthesis of mitochondrial phosphatidylethanolamine. These observations uncover a novel mitochondrial tumour suppressor and demonstrate a connection between mitochondrial lipid metabolism and the differentiation program of breast cancer cells, thereby revealing a previously undescribed mechanism of tumour suppression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Diferenciación Celular , Metabolismo de los Lípidos , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Supresoras de Tumor/metabolismo , beta-Lactamasas/metabolismo , Animales , Neoplasias de la Mama/genética , Carboxiliasas/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metabolismo de los Lípidos/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/enzimología , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Fosfatidiletanolaminas/metabolismo , Proteínas Supresoras de Tumor/genética , beta-Lactamasas/genética
5.
Proc Natl Acad Sci U S A ; 116(15): 7353-7362, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30910979

RESUMEN

Carcinoma cells residing in an intermediate phenotypic state along the epithelial-mesenchymal (E-M) spectrum are associated with malignant phenotypes, such as invasiveness, tumor-initiating ability, and metastatic dissemination. Using the recently described CD104+/CD44hi antigen marker combination, we isolated highly tumorigenic breast cancer cells residing stably-both in vitro and in vivo-in an intermediate phenotypic state and coexpressing both epithelial (E) and mesenchymal (M) markers. We demonstrate that tumorigenicity depends on individual cells residing in this E/M hybrid state and cannot be phenocopied by mixing two cell populations that reside stably at the two ends of the spectrum, i.e., in the E and in the M state. Hence, residence in a specific intermediate state along the E-M spectrum rather than phenotypic plasticity appears critical to the expression of tumor-initiating capacity. Acquisition of this E/M hybrid state is facilitated by the differential expression of EMT-inducing transcription factors (EMT-TFs) and is accompanied by the expression of adult stem cell programs, notably, active canonical Wnt signaling. Furthermore, transition from the highly tumorigenic E/M state to a fully mesenchymal phenotype, achieved by constitutive ectopic expression of Zeb1, is sufficient to drive cells out of the E/M hybrid state into a highly mesenchymal state, which is accompanied by a substantial loss of tumorigenicity and a switch from canonical to noncanonical Wnt signaling. Identifying the gatekeepers of the various phenotypic states arrayed along the E-M spectrum is likely to prove useful in developing therapeutic approaches that operate by shifting cancer cells between distinct states along this spectrum.


Asunto(s)
Células Madre Adultas/metabolismo , Neoplasias de la Mama/metabolismo , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Neoplasias Basocelulares/metabolismo , Células Madre Neoplásicas/metabolismo , Vía de Señalización Wnt , Células Madre Adultas/patología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Basocelulares/genética , Neoplasias Basocelulares/patología , Células Madre Neoplásicas/patología
7.
Nat Chem Biol ; 15(7): 681-689, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31133756

RESUMEN

The mechanisms by which cells adapt to proteotoxic stress are largely unknown, but are key to understanding how tumor cells, particularly in vivo, are largely resistant to proteasome inhibitors. Analysis of cancer cell lines, mouse xenografts and patient-derived tumor samples all showed an association between mitochondrial metabolism and proteasome inhibitor sensitivity. When cells were forced to use oxidative phosphorylation rather than glycolysis, they became proteasome-inhibitor resistant. This mitochondrial state, however, creates a unique vulnerability: sensitivity to the small molecule compound elesclomol. Genome-wide CRISPR-Cas9 screening showed that a single gene, encoding the mitochondrial reductase FDX1, could rescue elesclomol-induced cell death. Enzymatic function and nuclear-magnetic-resonance-based analyses further showed that FDX1 is the direct target of elesclomol, which promotes a unique form of copper-dependent cell death. These studies explain a fundamental mechanism by which cells adapt to proteotoxic stress and suggest strategies to mitigate proteasome inhibitor resistance.


Asunto(s)
Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Inhibidores de Proteasoma/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Ratones , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Proteasoma/química , Bibliotecas de Moléculas Pequeñas/química
8.
Nature ; 508(7494): 108-12, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24670634

RESUMEN

As the concentrations of highly consumed nutrients, particularly glucose, are generally lower in tumours than in normal tissues, cancer cells must adapt their metabolism to the tumour microenvironment. A better understanding of these adaptations might reveal cancer cell liabilities that can be exploited for therapeutic benefit. Here we developed a continuous-flow culture apparatus (Nutrostat) for maintaining proliferating cells in low-nutrient media for long periods of time, and used it to undertake competitive proliferation assays on a pooled collection of barcoded cancer cell lines cultured in low-glucose conditions. Sensitivity to low glucose varies amongst cell lines, and an RNA interference (RNAi) screen pinpointed mitochondrial oxidative phosphorylation (OXPHOS) as the major pathway required for optimal proliferation in low glucose. We found that cell lines most sensitive to low glucose are defective in the OXPHOS upregulation that is normally caused by glucose limitation as a result of either mitochondrial DNA (mtDNA) mutations in complex I genes or impaired glucose utilization. These defects predict sensitivity to biguanides, antidiabetic drugs that inhibit OXPHOS, when cancer cells are grown in low glucose or as tumour xenografts. Notably, the biguanide sensitivity of cancer cells with mtDNA mutations was reversed by ectopic expression of yeast NDI1, a ubiquinone oxidoreductase that allows bypass of complex I function. Thus, we conclude that mtDNA mutations and impaired glucose utilization are potential biomarkers for identifying tumours with increased sensitivity to OXPHOS inhibitors.


Asunto(s)
Biguanidas/farmacología , Medios de Cultivo/metabolismo , Medios de Cultivo/farmacología , Glucosa/deficiencia , Neoplasias/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Medios de Cultivo/química , ADN Mitocondrial/genética , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Glucosa/metabolismo , Glucosa/farmacología , Humanos , Hipoglucemiantes/farmacología , Masculino , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Tipificación Molecular , Mutación , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fosforilación Oxidativa/efectos de los fármacos , Fenformina/farmacología , Interferencia de ARN , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Proc Natl Acad Sci U S A ; 114(12): E2337-E2346, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28270621

RESUMEN

Neoplastic cells within individual carcinomas often exhibit considerable phenotypic heterogeneity in their epithelial versus mesenchymal-like cell states. Because carcinoma cells with mesenchymal features are often more resistant to therapy and may serve as a source of relapse, we sought to determine whether such cells could be further stratified into functionally distinct subtypes. Indeed, we find that a basal epithelial marker, integrin-ß4 (ITGB4), can be used to enable stratification of mesenchymal-like triple-negative breast cancer (TNBC) cells that differ from one another in their relative tumorigenic abilities. Notably, we demonstrate that ITGB4+ cancer stem cell (CSC)-enriched mesenchymal cells reside in an intermediate epithelial/mesenchymal phenotypic state. Among patients with TNBC who received chemotherapy, elevated ITGB4 expression was associated with a worse 5-year probability of relapse-free survival. Mechanistically, we find that the ZEB1 (zinc finger E-box binding homeobox 1) transcription factor activity in highly mesenchymal SUM159 TNBC cells can repress expression of the epithelial transcription factor TAp63α (tumor protein 63 isoform 1), a protein that promotes ITGB4 expression. In addition, we demonstrate that ZEB1 and ITGB4 are important in modulating the histopathological phenotypes of tumors derived from mesenchymal TNBC cells. Hence, mesenchymal carcinoma cell populations are internally heterogeneous, and ITGB4 is a mechanistically driven prognostic biomarker that can be used to identify the more aggressive subtypes of mesenchymal carcinoma cells in TNBC. The ability to rapidly isolate and mechanistically interrogate the CSC-enriched, partially mesenchymal carcinoma cells should further enable identification of novel therapeutic opportunities to improve the prognosis for high-risk patients with TNBC.


Asunto(s)
Carcinoma/metabolismo , Integrina beta4/metabolismo , Mesodermo/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Carcinoma/genética , Carcinoma/mortalidad , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Humanos , Integrina beta4/genética , Mesodermo/citología , Pronóstico , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/mortalidad , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
10.
Proc Natl Acad Sci U S A ; 114(2): 382-387, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28028240

RESUMEN

The use of proteasome inhibitors to target cancer's dependence on altered protein homeostasis has been greatly limited by intrinsic and acquired resistance. Analyzing data from thousands of cancer lines and tumors, we find that those with suppressed expression of one or more 19S proteasome subunits show intrinsic proteasome inhibitor resistance. Moreover, such proteasome subunit suppression is associated with poor outcome in myeloma patients, where proteasome inhibitors are a mainstay of treatment. Beyond conferring resistance to proteasome inhibitors, proteasome subunit suppression also serves as a sentinel of a more global remodeling of the transcriptome. This remodeling produces a distinct gene signature and new vulnerabilities to the proapoptotic drug, ABT-263. This frequent, naturally arising imbalance in 19S regulatory complex composition is achieved through a variety of mechanisms, including DNA methylation, and marks the emergence of a heritably altered and therapeutically relevant state in diverse cancers.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos
11.
Nature ; 499(7456): 92-6, 2013 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-23748442

RESUMEN

Stem cells and progenitors in many lineages undergo self-renewing divisions, but the extracellular and intracellular proteins that regulate this process are largely unknown. Glucocorticoids stimulate red blood cell formation by promoting self-renewal of early burst-forming unit-erythroid (BFU-E) progenitors. Here we show that the RNA-binding protein ZFP36L2 is a transcriptional target of the glucocorticoid receptor (GR) in BFU-Es and is required for BFU-E self-renewal. ZFP36L2 is normally downregulated during erythroid differentiation from the BFU-E stage, but its expression is maintained by all tested GR agonists that stimulate BFU-E self-renewal, and the GR binds to several potential enhancer regions of ZFP36L2. Knockdown of ZFP36L2 in cultured BFU-E cells did not affect the rate of cell division but disrupted glucocorticoid-induced BFU-E self-renewal, and knockdown of ZFP36L2 in transplanted erythroid progenitors prevented expansion of erythroid lineage progenitors normally seen following induction of anaemia by phenylhydrazine treatment. ZFP36L2 preferentially binds to messenger RNAs that are induced or maintained at high expression levels during terminal erythroid differentiation and negatively regulates their expression levels. ZFP36L2 therefore functions as part of a molecular switch promoting BFU-E self-renewal and a subsequent increase in the total numbers of colony-forming unit-erythroid (CFU-E) progenitors and erythroid cells that are generated.


Asunto(s)
División Celular , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Tristetraprolina/metabolismo , Animales , Recuento de Células , División Celular/efectos de los fármacos , Linaje de la Célula , Regulación hacia Abajo , Eritropoyesis/genética , Técnicas de Silenciamiento del Gen , Glucocorticoides/farmacología , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/metabolismo , Estrés Fisiológico , Tristetraprolina/deficiencia , Tristetraprolina/genética
12.
Nucleic Acids Res ; 44(D1): D560-6, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26626150

RESUMEN

The oncogenic transformation of normal cells into malignant, rapidly proliferating cells requires major alterations in cell physiology. For example, the transformed cells remodel their metabolic processes to supply the additional demand for cellular building blocks. We have recently demonstrated essential metabolic processes in tumor progression through the development of a methodological analysis of gene expression. Here, we present the Metabolic gEne RApid Visualizer (MERAV, http://merav.wi.mit.edu), a web-based tool that can query a database comprising ∼4300 microarrays, representing human gene expression in normal tissues, cancer cell lines and primary tumors. MERAV has been designed as a powerful tool for whole genome analysis which offers multiple advantages: one can search many genes in parallel; compare gene expression among different tissue types as well as between normal and cancer cells; download raw data; and generate heatmaps; and finally, use its internal statistical tool. Most importantly, MERAV has been designed as a unique tool for analyzing metabolic processes as it includes matrixes specifically focused on metabolic genes and is linked to the Kyoto Encyclopedia of Genes and Genomes pathway search.


Asunto(s)
Bases de Datos Genéticas , Perfilación de la Expresión Génica , Neoplasias/genética , Programas Informáticos , Línea Celular , Línea Celular Tumoral , Perfilación de la Expresión Génica/normas , Humanos , Internet , Redes y Vías Metabólicas/genética , Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/normas
13.
Proc Natl Acad Sci U S A ; 112(42): E5679-88, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26438848

RESUMEN

Reactive oxygen species (ROS) such as hydrogen peroxide (H2O2) govern cellular homeostasis by inducing signaling. H2O2 modulates the activity of phosphatases and many other signaling molecules through oxidation of critical cysteine residues, which led to the notion that initiation of ROS signaling is broad and nonspecific, and thus fundamentally distinct from other signaling pathways. Here, we report that H2O2 signaling bears hallmarks of a regular signal transduction cascade. It is controlled by hierarchical signaling events resulting in a focused response as the results place the mitochondrial respiratory chain upstream of tyrosine-protein kinase Lyn, Lyn upstream of tyrosine-protein kinase SYK (Syk), and Syk upstream of numerous targets involved in signaling, transcription, translation, metabolism, and cell cycle regulation. The active mediators of H2O2 signaling colocalize as H2O2 induces mitochondria-associated Lyn and Syk phosphorylation, and a pool of Lyn and Syk reside in the mitochondrial intermembrane space. Finally, the same intermediaries control the signaling response in tissues and species responsive to H2O2 as the respiratory chain, Lyn, and Syk were similarly required for H2O2 signaling in mouse B cells, fibroblasts, and chicken DT40 B cells. Consistent with a broad role, the Syk pathway is coexpressed across tissues, is of early metazoan origin, and displays evidence of evolutionary constraint in the human. These results suggest that H2O2 signaling is under control of a signal transduction pathway that links the respiratory chain to the mitochondrial intermembrane space-localized, ubiquitous, and ancient Syk pathway in hematopoietic and nonhematopoietic cells.


Asunto(s)
Transporte de Electrón , Peróxido de Hidrógeno/metabolismo , Membranas Mitocondriales/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Pollos , Activación Enzimática , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Quinasa Syk , Tirosina/metabolismo
14.
Proc Natl Acad Sci U S A ; 111(11): 4103-8, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24591581

RESUMEN

Mouse models have been used extensively for decades and have been instrumental in improving our understanding of mammalian erythropoiesis. Nonetheless, there are several examples of variation between human and mouse erythropoiesis. We performed a comparative global gene expression study using data from morphologically identical stage-matched sorted populations of human and mouse erythroid precursors from early to late erythroblasts. Induction and repression of major transcriptional regulators of erythropoiesis, as well as major erythroid-important proteins, are largely conserved between the species. In contrast, at a global level we identified a significant extent of divergence between the species, both at comparable stages and in the transitions between stages, especially for the 500 most highly expressed genes during development. This suggests that the response of multiple developmentally regulated genes to key erythroid transcriptional regulators represents an important modification that has occurred in the course of erythroid evolution. In developing a systematic framework to understand and study conservation and divergence between human and mouse erythropoiesis, we show how mouse models can fail to mimic specific human diseases and provide predictions for translating findings from mouse models to potential therapies for human disease.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Eritropoyesis/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Transcriptoma/genética , Animales , Western Blotting , Eritropoyesis/genética , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Ratones , Análisis por Micromatrices , Especificidad de la Especie
15.
Proc Natl Acad Sci U S A ; 108(4): 1519-24, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21205891

RESUMEN

Many human aneuploidy syndromes have unique phenotypic consequences, but in most instances it is unclear whether these phenotypes are attributable to alterations in the dosage of specific genes. In human trisomy 13, there is delayed switching and persistence of fetal hemoglobin (HbF) and elevation of embryonic hemoglobin in newborns. Using partial trisomy cases, we mapped this trait to chromosomal band 13q14; by examining the genes in this region, two microRNAs, miR-15a and -16-1, appear as top candidates for the elevated HbF levels. Indeed, increased expression of these microRNAs in primary human erythroid progenitor cells results in elevated fetal and embryonic hemoglobin gene expression. Moreover, we show that a direct target of these microRNAs, MYB, plays an important role in silencing the fetal and embryonic hemoglobin genes. Thus we demonstrate how the developmental regulation of a clinically important human trait can be better understood through the genetic and functional study of aneuploidy syndromes and suggest that miR-15a, -16-1, and MYB may be important therapeutic targets to increase HbF levels in patients with sickle cell disease and ß-thalassemia.


Asunto(s)
Cromosomas Humanos Par 13/genética , Hemoglobina Fetal/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas c-myb/genética , Trisomía , Algoritmos , Antígenos CD/metabolismo , Secuencia de Bases , Western Blotting , Células Precursoras Eritroides/metabolismo , Femenino , Hemoglobina Fetal/metabolismo , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Lactante , Recién Nacido , Células K562 , Masculino , Modelos Genéticos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Transferrina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Cell Stem Cell ; 29(5): 795-809.e11, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35452598

RESUMEN

To understand the mechanisms regulating the in vitro maturation of hPSC-derived hepatocytes, we developed a 3D differentiation system and compared gene regulatory elements in human primary hepatocytes with those in hPSC-hepatocytes that were differentiated in 2D or 3D conditions by RNA-seq, ATAC-seq, and H3K27Ac ChIP-seq. Regulome comparisons showed a reduced enrichment of thyroid receptor THRB motifs in accessible chromatin and active enhancers without a reduced transcription of THRB. The addition of thyroid hormone T3 increased the binding of THRB to the CYP3A4 proximal enhancer, restored the super-enhancer status and gene expression of NFIC, and reduced the expression of AFP. The resultant hPSC-hepatocytes showed gene expression, epigenetic status, and super-enhancer landscape closer to primary hepatocytes and activated regulatory regions including non-coding SNPs associated with liver-related diseases. Transplanting the hPSC-hepatocytes resulted in the engraftment of human hepatocytes into the mouse liver without disrupting normal liver histology. This work implicates the environmental factor-nuclear receptor axis in regulating the maturation of hPSC-hepatocytes.


Asunto(s)
Cromatina , Hepatocitos , Animales , Diferenciación Celular , Cromatina/metabolismo , Hepatocitos/metabolismo , Humanos , Ratones , Polimorfismo de Nucleótido Simple , Receptores Citoplasmáticos y Nucleares/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos
17.
Nat Cell Biol ; 24(4): 554-564, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35411083

RESUMEN

Epithelial-mesenchymal transition (EMT) programs operate within carcinoma cells, where they generate phenotypes associated with malignant progression. In their various manifestations, EMT programs enable epithelial cells to enter into a series of intermediate states arrayed along the E-M phenotypic spectrum. At present, we lack a coherent understanding of how carcinoma cells control their entrance into and continued residence in these various states, and which of these states favour the process of metastasis. Here we characterize a layer of EMT-regulating machinery that governs E-M plasticity (EMP). This machinery consists of two chromatin-modifying complexes, PRC2 and KMT2D-COMPASS, which operate as critical regulators to maintain a stable epithelial state. Interestingly, loss of these two complexes unlocks two distinct EMT trajectories. Dysfunction of PRC2, but not KMT2D-COMPASS, yields a quasi-mesenchymal state that is associated with highly metastatic capabilities and poor survival of patients with breast cancer, suggesting that great caution should be applied when PRC2 inhibitors are evaluated clinically in certain patient cohorts. These observations identify epigenetic factors that regulate EMP, determine specific intermediate EMT states and, as a direct consequence, govern the metastatic ability of carcinoma cells.


Asunto(s)
Neoplasias de la Mama , Carcinoma , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Metástasis de la Neoplasia/patología
18.
Cancer Discov ; 11(5): 1286-1305, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33328216

RESUMEN

The epithelial-to-mesenchymal transition, which conveys epithelial (E) carcinoma cells to quasi-mesenchymal (qM) states, enables them to metastasize and acquire resistance to certain treatments. Murine tumors composed of qM mammary carcinoma cells assemble an immunosuppressive tumor microenvironment (TME) and develop resistance to anti-CTLA4 immune-checkpoint blockade (ICB) therapy, unlike their E counterparts. Importantly, minority populations of qM cells within a tumor can cross-protect their more E neighbors from immune attack. The underlying mechanisms of immunosuppression and cross-protection have been unclear. We demonstrate that abrogation of qM carcinoma cell-derived factors (CD73, CSF1, or SPP1) prevents the assembly of an immunosuppressive TME and sensitizes otherwise refractory qM tumors partially or completely to anti-CTLA4 ICB. Most strikingly, mixed tumors in which minority populations of carcinoma cells no longer express CD73 are now sensitized to anti-CTLA4 ICB. Finally, loss of CD73 also enhances the efficacy of anti-CTLA4 ICB during the process of metastatic colonization. SIGNIFICANCE: Minority populations of qM carcinoma cells, which likely reside in human breast carcinomas, can cross-protect their E neighbors from immune attack. Understanding the mechanisms by which qM carcinoma cells resist antitumor immune attack can help identify signaling channels that can be interrupted to potentiate the efficacy of checkpoint blockade immunotherapies.This article is highlighted in the In This Issue feature, p. 995.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias de la Mama/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Terapia de Inmunosupresión , Ratones , Ratones Endogámicos C57BL , Microambiente Tumoral
19.
Genome Biol ; 19(1): 152, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30286781

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) are short regulatory RNAs that derive from hairpin precursors. Important for understanding the functional roles of miRNAs is the ability to predict the messenger RNA (mRNA) targets most responsive to each miRNA. Progress towards developing quantitative models of miRNA targeting in Drosophila and other invertebrate species has lagged behind that of mammals due to the paucity of datasets measuring the effects of miRNAs on mRNA levels. RESULTS: We acquired datasets suitable for the quantitative study of miRNA targeting in Drosophila. Analyses of these data expanded the types of regulatory sites known to be effective in flies, expanded the mRNA regions with detectable targeting to include 5' untranslated regions, and identified features of site context that correlate with targeting efficacy in fly cells. Updated evolutionary analyses evaluated the probability of conserved targeting for each predicted site and indicated that more than a third of the Drosophila genes are preferentially conserved targets of miRNAs. Based on these results, a quantitative model was developed to predict targeting efficacy in insects. This model performed better than existing models, and it drives the most recent version, v7, of TargetScanFly. CONCLUSIONS: Our evolutionary and functional analyses expand the known scope of miRNA targeting in flies and other insects. The existence of a quantitative model that has been developed and trained using Drosophila data will provide a valuable resource for placing miRNAs into gene regulatory networks of this important experimental organism.


Asunto(s)
Drosophila melanogaster/genética , MicroARNs/metabolismo , Regiones no Traducidas 3'/genética , Regiones no Traducidas 5'/genética , Algoritmos , Animales , Secuencia de Bases , Secuencia Conservada/genética , Evolución Molecular , MicroARNs/genética , Modelos Genéticos , Anotación de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
Mol Biol Cell ; 29(26): 3168-3182, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30332327

RESUMEN

Heat shock factor 1 is the master transcriptional regulator of molecular chaperones and binds to the same cis-acting heat shock element (HSE) across the eukaryotic lineage. In budding yeast, Hsf1 drives the transcription of ∼20 genes essential to maintain proteostasis under basal conditions, yet its specific targets and extent of inducible binding during heat shock remain unclear. Here we combine Hsf1 chromatin immunoprecipitation sequencing (seq), nascent RNA-seq, and Hsf1 nuclear depletion to quantify Hsf1 binding and transcription across the yeast genome. We find that Hsf1 binds 74 loci during acute heat shock, and these are linked to 46 genes with strong Hsf1-dependent expression. Notably, Hsf1's induced DNA binding leads to a disproportionate (∼7.5-fold) increase in nascent transcription. Promoters with high basal Hsf1 occupancy have nucleosome-depleted regions due to the presence of "pioneer factors." These accessible sites are likely critical for Hsf1 occupancy as the activator is incapable of binding HSEs within a stably positioned, reconstituted nucleosome. In response to heat shock, however, Hsf1 accesses nucleosomal sites and promotes chromatin disassembly in concert with the Remodels Structure of Chromatin (RSC) complex. Our data suggest that the interplay between nucleosome positioning, HSE strength, and active Hsf1 levels allows cells to precisely tune expression of the proteostasis network.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Factores de Transcripción del Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/fisiología , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Proteínas de Unión al ADN/metabolismo , Regulación Fúngica de la Expresión Génica/genética , Factores de Transcripción del Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Respuesta al Choque Térmico/genética , Chaperonas Moleculares/metabolismo , Nucleosomas/metabolismo , Regiones Promotoras Genéticas/genética , Secuencias Reguladoras de Ácidos Nucleicos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda