Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 152(5): 1008-20, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23452850

RESUMEN

Metazoan evolution involves increasing protein domain complexity, but how this relates to control of biological decisions remains uncertain. The Ras guanine nucleotide exchange factor (RasGEF) Sos1 and its adaptor Grb2 are multidomain proteins that couple fibroblast growth factor (FGF) signaling to activation of the Ras-Erk pathway during mammalian development and drive embryonic stem cells toward the primitive endoderm (PrE) lineage. We show that the ability of Sos1/Grb2 to appropriately regulate pluripotency and differentiation factors and to initiate PrE development requires collective binding of multiple Sos1/Grb2 domains to their protein and phospholipid ligands. This provides a cooperative system that only allows lineage commitment when all ligand-binding domains are occupied. Furthermore, our results indicate that the interaction domains of Sos1 and Grb2 have evolved so as to bind ligands not with maximal strength but with specificities and affinities that maintain cooperativity. This optimized system ensures that PrE lineage commitment occurs in a timely and selective manner during embryogenesis.


Asunto(s)
Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Proteína Adaptadora GRB2/metabolismo , Proteína SOS1/metabolismo , Secuencia de Aminoácidos , Animales , Linaje de la Célula , Endodermo/metabolismo , Eucariontes/genética , Eucariontes/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Factores de Intercambio de Guanina Nucleótido ras/metabolismo
2.
Brain Behav Immun ; 119: 637-647, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38663773

RESUMEN

Obesity is a major modifiable risk factor for Alzheimer's disease (AD), characterized by progressive atrophy of the cerebral cortex. The neurobiology of obesity contributions to AD is poorly understood. Here we show with in vivo MRI that diet-induced obesity decreases cortical volume in mice, and that higher body adiposity associates with lower cortical volume in humans. Single-nuclei transcriptomics of the mouse cortex reveals that dietary obesity promotes an array of neuron-adverse transcriptional dysregulations, which are mediated by an interplay of excitatory neurons and glial cells, and which involve microglial activation and lowered neuronal capacity for neuritogenesis and maintenance of membrane potential. The transcriptional dysregulations of microglia, more than of other cell types, are like those in AD, as assessed with single-nuclei cortical transcriptomics in a mouse model of AD and two sets of human donors with the disease. Serial two-photon tomography of microglia demonstrates microgliosis throughout the mouse cortex. The spatial pattern of adiposity-cortical volume associations in human cohorts interrogated together with in silico bulk and single-nucleus transcriptomic data from the human cortex implicated microglia (along with other glial cells and subtypes of excitatory neurons), and it correlated positively with the spatial profile of cortical atrophy in patients with mild cognitive impairment and AD. Thus, multi-cell neuron-adverse dysregulations likely contribute to the loss of cortical tissue in obesity. The dysregulations of microglia may be pivotal to the obesity-related risk of AD.


Asunto(s)
Enfermedad de Alzheimer , Corteza Cerebral , Obesidad , Animales , Obesidad/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Humanos , Ratones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Masculino , Microglía/metabolismo , Neuronas/metabolismo , Femenino , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Imagen por Resonancia Magnética , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/etiología , Atrofia , Dieta Alta en Grasa/efectos adversos , Anciano , Adiposidad , Transcriptoma
3.
Blood ; 136(1): 81-92, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32299104

RESUMEN

Through a clustered regularly insterspaced short palindromic repeats (CRISPR) screen to identify mitochondrial genes necessary for the growth of acute myeloid leukemia (AML) cells, we identified the mitochondrial outer membrane protein mitochondrial carrier homolog 2 (MTCH2). In AML, knockdown of MTCH2 decreased growth, reduced engraftment potential of stem cells, and induced differentiation. Inhibiting MTCH2 in AML cells increased nuclear pyruvate and pyruvate dehydrogenase (PDH), which induced histone acetylation and subsequently promoted the differentiation of AML cells. Thus, we have defined a new mechanism by which mitochondria and metabolism regulate AML stem cells and gene expression.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Proteínas de Neoplasias/fisiología , Acetilación , Animales , Sistemas CRISPR-Cas , Diferenciación Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Sangre Fetal/citología , Regulación Leucémica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Histonas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Proteínas de Fusión Oncogénica/fisiología , Procesamiento Proteico-Postraduccional , Ácido Pirúvico/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología
4.
Mol Pharmacol ; 95(6): 638-651, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30979813

RESUMEN

Evofosfamide (TH-302) is a hypoxia-activated DNA-crosslinking prodrug currently in clinical development for cancer therapy. Oxygen-sensitive activation of evofosfamide depends on one-electron reduction, yet the reductases that catalyze this process in tumors are unknown. We used RNA sequencing, whole-genome CRISPR knockout, and reductase-focused short hairpin RNA screens to interrogate modifiers of evofosfamide activation in cancer cell lines. Involvement of mitochondrial electron transport in the activation of evofosfamide and the related nitroaromatic compounds EF5 and FSL-61 was investigated using 143B ρ 0 (ρ zero) cells devoid of mitochondrial DNA and biochemical assays in UT-SCC-74B cells. The potency of evofosfamide in 30 genetically diverse cancer cell lines correlated with the expression of genes involved in mitochondrial electron transfer. A whole-genome CRISPR screen in KBM-7 cells identified the DNA damage-response factors SLX4IP, C10orf90 (FATS), and SLFN11, in addition to the key regulator of mitochondrial function, YME1L1, and several complex I constituents as modifiers of evofosfamide sensitivity. A reductase-focused shRNA screen in UT-SCC-74B cells similarly identified mitochondrial respiratory chain factors. Surprisingly, 143B ρ 0 cells showed enhanced evofosfamide activation and sensitivity but had global transcriptional changes, including increased expression of nonmitochondrial flavoreductases. In UT-SCC-74B cells, evofosfamide oxidized cytochromes a, b, and c and inhibited respiration at complexes I, II, and IV without quenching reactive oxygen species production. Our results suggest that the mitochondrial electron transport chain contributes to evofosfamide activation and that predicting evofosfamide sensitivity in patients by measuring the expression of canonical bioreductive enzymes such as cytochrome P450 oxidoreductase is likely to be futile.


Asunto(s)
Transporte de Electrón/efectos de los fármacos , Mitocondrias/genética , Neoplasias/genética , Nitroimidazoles/farmacología , Mostazas de Fosforamida/farmacología , Análisis de Secuencia de ARN/métodos , Sistemas CRISPR-Cas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Células HCT116 , Humanos , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Profármacos , ARN Interferente Pequeño/farmacología
5.
Am J Physiol Renal Physiol ; 317(6): F1593-F1604, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31566424

RESUMEN

Podocyte function is tightly linked to the complex organization of its cytoskeleton and adhesion to the underlying glomerular basement membrane. Adhesion of cultured podocytes to a variety of substrates is reported to correlate with podocyte health. To identify novel genes that are important for podocyte function, we designed an in vitro genetic screen based on podocyte adhesion to plates coated with either fibronectin or soluble Fms-like tyrosine kinase-1 (sFLT1)/Fc. A genome-scale pooled RNA interference screen on immortalized human podocytes identified 77 genes that increased adhesion to fibronectin, 101 genes that increased adhesion to sFLT1/Fc, and 44 genes that increased adhesion to both substrates when knocked down. Multiple shRNAs against diphthamide biosynthesis protein 1-4 (DPH1-DPH4) were top hits for increased adhesion. Immortalized human podocyte cells stably expressing these hairpins displayed increased adhesion to both substrates. We then used CRISPR-Cas9 to generate podocyte knockout cells for DPH1, DPH2, or DPH3, which also displayed increased adhesion to both fibronectin and sFLT1/Fc, as well as a spreading defect. Finally, we showed that Drosophila nephrocyte-specific knockdown of Dph1, Dph2, and Dph4 resulted in altered nephrocyte function. In summary, we report here a novel high-throughput method to identify genes important for podocyte function. Given the central role of podocyte adhesion as a marker of podocyte health, these data are a rich source of candidate regulators of glomerular disease.


Asunto(s)
Adhesión Celular/genética , Adhesión Celular/fisiología , Histidina/análogos & derivados , Riñón/metabolismo , Podocitos/metabolismo , Animales , Línea Celular , Drosophila , Fibronectinas/metabolismo , Proteínas del Choque Térmico HSP40/genética , Ensayos Analíticos de Alto Rendimiento , Histidina/biosíntesis , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Antígenos de Histocompatibilidad Menor/genética , Proteínas/genética , ARN Interferente Pequeño , Proteínas Supresoras de Tumor/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
6.
PLoS Genet ; 12(11): e1006452, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27870871

RESUMEN

Fungal biofilms are complex, structured communities that can form on surfaces such as catheters and other indwelling medical devices. Biofilms are of particular concern with Candida albicans, one of the leading opportunistic fungal pathogens of humans. C. albicans biofilms include yeast and filamentous cells that are surrounded by an extracellular matrix, and they are intrinsically resistant to antifungal drugs such that resolving biofilm infections often requires surgery to remove the contaminated device. C. albicans biofilms form through a regulated process of adhesion to surfaces, filamentation, maturation, and ultimately dispersion. To uncover new strategies to block the initial stages of biofilm formation, we utilized a functional genomic approach to identify genes that modulate C. albicans adherence. We screened a library of 1,481 double barcoded doxycycline-repressible conditional gene expression strains covering ~25% of the C. albicans genome. We identified five genes for which transcriptional repression impaired adherence, including: ARC18, PMT1, MNN9, SPT7, and orf19.831. The most severe adherence defect was observed upon transcriptional repression of ARC18, which encodes a member of the Arp2/3 complex that is involved in regulation of the actin cytoskeleton and endocytosis. Depletion of components of the Arp2/3 complex not only impaired adherence, but also caused reduced biofilm formation, increased cell surface hydrophobicity, and increased exposure of cell wall chitin and ß-glucans. Reduced function of the Arp2/3 complex led to impaired cell wall integrity and activation of Rho1-mediated cell wall stress responses, thereby causing cell wall remodelling and reduced adherence. Thus, we identify important functional relationships between cell wall stress responses and a novel mechanism that controls adherence and biofilm formation, thereby illuminating novel strategies to cripple a leading fungal pathogen of humans.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/genética , Biopelículas/crecimiento & desarrollo , Candida albicans/genética , Farmacorresistencia Fúngica/genética , Proteínas Fúngicas/biosíntesis , Citoesqueleto de Actina/genética , Complejo 2-3 Proteico Relacionado con la Actina/efectos de los fármacos , Biopelículas/efectos de los fármacos , Candida albicans/efectos de los fármacos , Candida albicans/crecimiento & desarrollo , Candida albicans/patogenicidad , Candidiasis/tratamiento farmacológico , Candidiasis/genética , Candidiasis/microbiología , Adhesión Celular/genética , Pared Celular/genética , Endocitosis/genética , Proteínas Fúngicas/genética , Regulación Fúngica de la Expresión Génica , Genoma Fúngico , Genómica , Humanos , Redes y Vías Metabólicas/genética , Estrés Fisiológico/genética
7.
Proteomics ; 15(2-3): 419-33, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25311528

RESUMEN

Aberrant expression and activation of FGFR3 is associated with disease states including bone dysplasia and malignancies of bladder, cervix, and bone marrow. MS analysis of protein-phosphotyrosine in multiple myeloma cells revealed a prevalent phosphorylated motif, D/EYYR/K, derived from the kinase domain activation loops of tyrosine kinases including FGFR3 corresponding to a recognition sequence of protein-tyrosine phosphatase PTPN1. Knockdown of PTPN1 or the related enzyme PTPN2 by RNAi resulted in ligand-independent activation of FGFR3. Modulation of FGFR3 activation loop phosphorylation by both PTPN1 and PTPN2 was a function of receptor trafficking and phosphotyrosine phosphatase (PTP) compartmentalization. The FGFR3 activation loop motif DYYKK(650) is altered to DYYKE(650) in the oncogenic variant FGFR3(K650E) , and consequently it is constitutively fully activated and unaffected by activation loop phosphorylation. FGFR3(K650E) was nevertheless remarkably sensitive to negative regulation by PTPN1 and PTPN2. This suggests that in addition to modulating FGFR3 phosphorylation, PTPN1 and PTPN2 constrain the kinase domain by fostering an inactive-state. Loss of this constraint in response to ligand or impaired PTPN1/N2 may initiate FGFR3 activation. These results suggest a model wherein PTP expression levels may define conditions that select for ectopic FGFR3 expression and activation during tumorigenesis.


Asunto(s)
Mieloma Múltiple/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Endocitosis , Regulación Neoplásica de la Expresión Génica , Glicosilación , Humanos , Datos de Secuencia Molecular , Mieloma Múltiple/genética , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/análisis , Proteína Tirosina Fosfatasa no Receptora Tipo 2/análisis , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Interferencia de ARN , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/análisis , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética
8.
Mol Syst Biol ; 9: 696, 2013 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-24104479

RESUMEN

Improved efforts are necessary to define the functional product of cancer mutations currently being revealed through large-scale sequencing efforts. Using genome-scale pooled shRNA screening technology, we mapped negative genetic interactions across a set of isogenic cancer cell lines and confirmed hundreds of these interactions in orthogonal co-culture competition assays to generate a high-confidence genetic interaction network of differentially essential or differential essentiality (DiE) genes. The network uncovered examples of conserved genetic interactions, densely connected functional modules derived from comparative genomics with model systems data, functions for uncharacterized genes in the human genome and targetable vulnerabilities. Finally, we demonstrate a general applicability of DiE gene signatures in determining genetic dependencies of other non-isogenic cancer cell lines. For example, the PTEN(-/-) DiE genes reveal a signature that can preferentially classify PTEN-dependent genotypes across a series of non-isogenic cell lines derived from the breast, pancreas and ovarian cancers. Our reference network suggests that many cancer vulnerabilities remain to be discovered through systematic derivation of a network of differentially essential genes in an isogenic cancer cell model.


Asunto(s)
Neoplasias de la Mama/genética , Epistasis Genética , Genes Esenciales , Proteínas de Neoplasias/genética , Neoplasias Ováricas/genética , Fosfohidrolasa PTEN/genética , Neoplasias Pancreáticas/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Redes Reguladoras de Genes , Genoma Humano , Humanos , Mutación , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosfohidrolasa PTEN/deficiencia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
9.
Nucleic Acids Res ; 40(Database issue): D957-63, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22102578

RESUMEN

Genome-wide pooled shRNA screens enable global identification of the genes essential for cancer cell survival and proliferation and provide a 'functional genetic' map of human cancer to complement genomic studies. Using a lentiviral shRNA library targeting approximately 16,000 human genes and a newly developed scoring approach, we identified essential gene profiles in more than 70 breast, pancreatic and ovarian cancer cell lines. We developed a web-accessible database system for capturing information from each step in our standardized screening pipeline and a gene-centric search tool for exploring shRNA activities within a given cell line or across multiple cell lines. The database consists of a laboratory information and management system for tracking each step of a pooled shRNA screen as well as a web interface for querying and visualization of shRNA and gene-level performance across multiple cancer cell lines. COLT-Cancer Version 1.0 is currently accessible at http://colt.ccbr.utoronto.ca/cancer.


Asunto(s)
Bases de Datos Genéticas , Genes Esenciales , Genes Relacionados con las Neoplasias , Neoplasias/genética , Interferencia de ARN , Línea Celular Tumoral , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño
10.
Methods Mol Biol ; 2658: 145-165, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37024700

RESUMEN

Candida albicans is a prevalent fungal pathogen of humans that can cause both superficial and life-threatening disease, primarily in immunocompromised populations. Currently, antifungal drug classes available to treat fungal infections remain limited and the emergence of drug-resistant strains threatens antifungal efficacy, necessitating the discovery and development of additional therapeutics. The construction of the C. albicans double-barcoded heterozygous deletion collection (DBC) enables the rapid and systematic assessment of haploinsufficiency phenotypes in a pooled format. Specifically, this functional genomics resource can be used to identify heterozygous deletion mutants that are hypersensitive to compounds in order to define putative cellular targets and/or other modifiers of compound activity. Here, we describe protocols to characterize the mode of action of small molecules using the C. albicans DBC, including how to prepare compound-treated cultures, isolate genomic DNA, amplify strain-specific barcodes, and prepare DNA libraries for high-throughput sequencing. This technique provides a powerful approach to elucidate the compound mechanism of action in order to bolster the antifungal pipeline.


Asunto(s)
Candida albicans , Micosis , Humanos , Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Micosis/tratamiento farmacológico , Genómica , Fenotipo , Pruebas de Sensibilidad Microbiana
11.
Commun Biol ; 6(1): 770, 2023 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-37481629

RESUMEN

Low blood flow through the fetal left heart is often conjectured as an etiology for hypoplastic left heart syndrome (HLHS). To investigate if a decrease in left heart flow results in growth failure, we generate left ventricular inflow obstruction (LVIO) in mid-gestation fetal lambs by implanting coils in their left atrium using an ultrasound-guided percutaneous technique. Significant LVIO recapitulates important clinical features of HLHS: decreased antegrade aortic valve flow, compensatory retrograde perfusion of the brain and ascending aorta (AAo) from the arterial duct, severe left heart hypoplasia, a non-apex forming LV, and a thickened endocardial layer. The hypoplastic AAo have miRNA-gene pairs annotating to cell proliferation that are inversely differentially expressed by bulk RNA-seq. Single-nucleus RNA-seq of the hypoplastic LV myocardium shows an increase in fibroblasts with a reciprocal decrease in cardiomyocyte nuclei proportions. Fibroblasts, cardiomyocytes and endothelial cells from hypoplastic myocardium have increased expression of extracellular matrix component or fibrosis genes with dysregulated fibroblast growth factor signaling. Hence, a severe sustained ( ~ 1/3 gestation) reduction in fetal left heart flow is sufficient to cause left heart hypoplasia. This is accompanied by changes in cellular composition and gene expression consistent with a pro-fibrotic environment and aberrant induction of mesenchymal programs.


Asunto(s)
Células Endoteliales , Oveja Doméstica , Ovinos , Animales , Feto , Miocardio , Ventrículos Cardíacos
12.
Commun Biol ; 6(1): 538, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-37202533

RESUMEN

During cancer development, tumor cells acquire changes that enable them to invade surrounding tissues and seed metastasis at distant sites. These changes contribute to the aggressiveness of metastatic cancer and interfere with success of therapy. Our comprehensive analysis of "matched" pairs of HNSCC lines derived from primary tumors and corresponding metastatic sites identified several components of Notch3 signaling that are differentially expressed and/or altered in metastatic lines and confer a dependency on this pathway. These components were also shown to be differentially expressed between early and late stages of tumors in a TMA constructed from over 200 HNSCC patients. Finally, we show that suppression of Notch3 improves survival in mice in both subcutaneous and orthotopic models of metastatic HNSCC. Novel treatments targeting components of this pathway may prove effective in targeting metastatic HNSCC cells alone or in combination with conventional therapies.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Animales , Ratones , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello , Humanos
13.
bioRxiv ; 2023 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-38187588

RESUMEN

The understanding of how different cell types contribute to amyotrophic lateral sclerosis (ALS) pathogenesis is limited. Here we generated a single-nucleus transcriptomic and epigenomic atlas of the frontal cortex of ALS cases with C9orf72 (C9) hexanucleotide repeat expansions and sporadic ALS (sALS). Our findings reveal shared pathways in C9-ALS and sALS, characterized by synaptic dysfunction in excitatory neurons and a disease-associated state in microglia. The disease subtypes diverge with loss of astrocyte homeostasis in C9-ALS, and a more substantial disturbance of inhibitory neurons in sALS. Leveraging high depth 3'-end sequencing, we found a widespread switch towards distal polyadenylation (PA) site usage across ALS subtypes relative to controls. To explore this differential alternative PA (APA), we developed APA-Net, a deep neural network model that uses transcript sequence and expression levels of RNA-binding proteins (RBPs) to predict cell-type specific APA usage and RBP interactions likely to regulate APA across disease subtypes.

14.
Physiol Genomics ; 44(2): 183-97, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22147266

RESUMEN

Myogenesis is a tightly controlled process involving the transcriptional activation and repression of thousands of genes. Although many components of the transcriptional network regulating the later phases of myogenesis have been identified, relatively few studies have described the transcriptional landscape during the first 24 h, when myoblasts commit to differentiate. Through dense temporal profiling of differentiating C2C12 myoblasts, we identify 193 transcriptional regulators (TRs) whose expression is significantly altered within the first 24 h of myogenesis. A high-content shRNA screen of 77 TRs involving 427 stable lines identified 42 genes whose knockdown significantly inhibits differentiation of C2C12 myoblasts. Of the TRs that were differentially expressed within the first 24 h, over half inhibited differentiation when knocked down, including known regulators of myogenesis (Myod1, Myog, and Myf5), as well as 19 TRs not previously associated with this process. Surprisingly, a similar proportion (55%) of shRNAs targeting TRs whose expression did not change also inhibited C2C12 myogenesis. We further show that a subset of these TRs inhibits myogenesis by downregulating expression of known regulatory and structural proteins. Our findings clearly illustrate that several TRs critical for C2C12 myogenesis are not differentially regulated, suggesting that approaches that focus functional studies on differentially-expressed transcripts will fail to provide a comprehensive view of this complex process.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Desarrollo de Músculos/genética , Mioblastos/citología , Animales , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Ratones , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
J Biol Chem ; 286(47): 41046-56, 2011 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-21937449

RESUMEN

The AC133 epitope expressed on the CD133 glycoprotein has been widely used as a cell surface marker of numerous stem cell and cancer stem cell types. It has been recently proposed that posttranslational modification and regulation of CD133 may govern cell surface AC133 recognition. Therefore, we performed a large scale pooled RNA interference (RNAi) screen to identify genes involved in cell surface AC133 expression. Gene hits could be validated at a rate of 70.5% in a secondary assay using an orthogonal RNAi system, demonstrating that our primary RNAi screen served as a powerful genetic screening approach. Within the list of hits from the primary screen, genes involved in N-glycan biosynthesis were significantly enriched as determined by Ingenuity Canonical Pathway analyses. Indeed, inhibiting biosynthesis of the N-glycan precursor using the small molecule tunicamycin or inhibiting its transfer to CD133 by generating N-glycan-deficient CD133 mutants resulted in undetectable cell surface AC133. Among the screen hits involved in N-glycosylation were genes involved in complex N-glycan processing, including the poorly characterized MGAT4C, which we demonstrate to be a positive regulator of cell surface AC133 expression. Our study identifies a set of genes involved in CD133 N-glycosylation as a direct contributing factor to cell surface AC133 recognition and provides biochemical evidence for the function and structure of CD133 N-glycans.


Asunto(s)
Antígenos CD/inmunología , Antígenos CD/metabolismo , Epítopos/inmunología , Glicoproteínas/inmunología , Glicoproteínas/metabolismo , Nitrógeno/metabolismo , Péptidos/inmunología , Péptidos/metabolismo , Procesamiento Proteico-Postraduccional , Antígeno AC133 , Biomarcadores/metabolismo , Glicosilación , Glicosiltransferasas/deficiencia , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Células HEK293 , Humanos , Polisacáridos/biosíntesis , Unión Proteica , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Interferencia de ARN , Reproducibilidad de los Resultados , Tunicamicina/farmacología
16.
Apoptosis ; 17(7): 666-78, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22488300

RESUMEN

Acquisition of resistance to anchorage dependant cell death, a process termed anoikis, is a requirement for cancer cell metastasis. However, the molecular determinants of anoikis resistance and sensitivity are poorly understood. To better understand resistance to anoikis we conducted a genome wide lentiviral shRNA screen to identify genes whose knockdown render anoikis-sensitive RWPE-1 prostate cells resistant to anoikis. RWPE-1 cells were infected with a pooled lentiviral shRNA library with 54,021 shRNA targeting 11,255 genes. After infection, an anoikis-resistant cell population was selected and shRNA sequences were amplified and sequenced. Thirty-four shRNA sequences reproducibly protected RWPE-1 cells from anoikis after culture under suspension conditions including the top validated hit, α/ß hydrolase domain containing 4 (ABHD4). In validation studies, ABHD4 knockdown inhibited anoikis in RWPE-1 cells as well as anoikis sensitive NP69 nasopharyngeal and OVCAR3 ovarian cancer cells, while over-expression of the gene increased sensitivity. Induction of anoikis after ABHD4 knockdown was associated with cleavage of PARP and activation of caspases-3, but was independent in changes of FLIP, FAK and Src expression. Interestingly, induction of anoikis after ABHD4 knockdown was independent of the known role of ABHD4 in the anandamide synthesis pathway and the generation of glycerophospho-N-acyl ethanolamines. Thus, ABHD4 is a novel genetic regulator of anoikis sensitivity.


Asunto(s)
Anoicis/genética , Pruebas Genéticas , Genoma Humano/genética , Hidrolasas/metabolismo , ARN Interferente Pequeño/metabolismo , Ácidos Araquidónicos/biosíntesis , Ácidos Araquidónicos/química , Línea Celular Tumoral , Endocannabinoides/biosíntesis , Endocannabinoides/química , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Hidrolasas/genética , Lentivirus/genética , Lisofosfolipasa , Masculino , Alcamidas Poliinsaturadas/química , Estructura Terciaria de Proteína
17.
Blood ; 115(11): 2251-9, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-20075161

RESUMEN

The proteasomal pathway of protein degradation involves 2 discrete steps: ubiquitination and degradation. Here, we evaluated the effects of inhibiting the ubiquitination pathway at the level of the ubiquitin-activating enzyme UBA1 (E1). By immunoblotting, leukemia cell lines and primary patient samples had increased protein ubiquitination. Therefore, we examined the effects of genetic and chemical inhibition of the E1 enzyme. Knockdown of E1 decreased the abundance of ubiquitinated proteins in leukemia and myeloma cells and induced cell death. To further investigate effects of E1 inhibition in malignancy, we discovered a novel small molecule inhibitor, 3,5-dioxopyrazolidine compound, 1-(3-chloro-4-fluorophenyl)-4-[(5-nitro-2-furyl)methylene]-3,5-pyrazolidinedione (PYZD-4409). PYZD-4409 induced cell death in malignant cells and preferentially inhibited the clonogenic growth of primary acute myeloid leukemia cells compared with normal hematopoietic cells. Mechanistically, genetic or chemical inhibition of E1 increased expression of E1 stress markers. Moreover, BI-1 overexpression blocked cell death after E1 inhibition, suggesting ER stress is functionally important for cell death after E1 inhibition. Finally, in a mouse model of leukemia, intraperitoneal administration of PYZD-4409 decreased tumor weight and volume compared with control without untoward toxicity. Thus, our work highlights the E1 enzyme as a novel target for the treatment of hematologic malignancies.


Asunto(s)
Leucemia/enzimología , Leucemia/terapia , Mieloma Múltiple/enzimología , Mieloma Múltiple/terapia , Enzimas Activadoras de Ubiquitina/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D3/metabolismo , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/patología , Inhibidores Enzimáticos/farmacología , Técnicas de Silenciamiento del Gen , Sistema Hematopoyético/citología , Sistema Hematopoyético/efectos de los fármacos , Humanos , Ratones , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Estrés Fisiológico/efectos de los fármacos , Factores de Tiempo , Proteína p53 Supresora de Tumor/metabolismo , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Ubiquitinación/efectos de los fármacos
18.
EMBO Rep ; 11(10): 777-83, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20829881

RESUMEN

ΔNp63α is a critical pro-survival protein overexpressed in 80% of head and neck squamous cell carcinomas (HNSCCs) where it inhibits TAp73ß transcription of p53-family target genes, which is thought to increase HNSCC resistance to chemotherapy-induced cell death. However, the mechanisms governing ΔNp63α function are largely unknown. In this study, we identify special AT-rich-binding protein 2 (SATB2) as a new ΔNp63α-binding protein that is preferentially expressed in advanced-stage primary HNSCC and show that SATB2 promotes chemoresistance by enhancing ΔNp63α-mediated transrepression by augmenting ΔNp63α engagement to p53-family responsive elements. Furthermore, SATB2 expression positively correlates with HNSCC chemoresistance, and RNA interference-mediated knockdown of endogenous SATB2 re-sensitizes HNSCC cells to chemotherapy- and γ-irradiation-induced apoptosis, irrespective of p53 status. These findings unveil SATB2 as a pivotal modulator of ΔNp63α that governs HNSCC cell survival.


Asunto(s)
Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Apoptosis , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos , Genes p53 , Humanos , Transactivadores/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética
19.
Sci Adv ; 8(35): eabo7792, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-36054348

RESUMEN

Circulating tumor cells (CTCs) break free from primary tumors and travel through the circulation system to seed metastatic tumors, which are the major cause of death from cancer. The identification of the major genetic factors that enhance production and persistence of CTCs in the bloodstream at a whole genome level would enable more comprehensive molecular mechanisms of metastasis to be elucidated and the identification of novel therapeutic targets, but this remains a challenging task due to the heterogeneity and extreme rarity of CTCs. Here, we describe an in vivo genome-wide CRISPR knockout screen using CTCs directly isolated from a mouse xenograft. This screen elucidated SLIT2-a gene encoding a secreted protein acting as a cellular migration cue-as the most significantly represented gene knockout in the CTC population. SLIT2 knockout cells are highly metastatic with hypermigratory and mesenchymal phenotype, resulting in enhanced cancer progression in xenograft models.


Asunto(s)
Células Neoplásicas Circulantes , Animales , Transición Epitelial-Mesenquimal , Xenoinjertos , Humanos , Ratones , Metástasis de la Neoplasia/patología , Células Neoplásicas Circulantes/patología
20.
EJHaem ; 3(2): 426-433, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35846029

RESUMEN

Internal tandem duplication of the Feline McDonough Sarcoma (FMS)-like tyrosine kinase 3 (FLT3-ITD) is one of the most clinically relevant mutations in acute myeloid leukemia (AML), with a high FLT3-ITD allelic ratio (AR) (≥0.5) being strongly associated with poor prognosis. FLT3-ITDs are heterogeneous, varying in size and location, with some patients having multiple FLT3-ITDs. Bulk cell-based approaches are limited in their ability to reveal the clonal structure in such cases. Using single-cell proteogenomic sequencing (ScPGseq), we attempted to identify a relapse-fated subclone in an AML case with mutations in WT1, NPM1, and FLT3 tyrosine kinase domain and two FLT3-ITDs (21 bp and 39 bp) (low AR) at presentation, then relapsed only with WT1 and NPM1 mutations and one FLT3-ITD (high AR). This relapse-fated subclone at presentation (∼2.1% of sequenced cells) was characterized by the presence of a homozygous 21 bp FLT3-ITD resulting from copy neutral loss of heterozygosity (CN-LOH) of chr13q and an aberrant, immature myeloid cell surface signature, contrast to the cell surface phenotype at presentation. In contrast to results from multicolor flow-cytometry, ScPGseq not only enabled the early detection of rare relapse-fated subclone showing immature myeloid signature but also highlighted the presence of homozygous 21 bp FLT3-ITDs in the clone at presentation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA