Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 610(7933): 783-790, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36224385

RESUMEN

Around birth, globin expression in human red blood cells (RBCs) shifts from γ-globin to ß-globin, which results in fetal haemoglobin (HbF, α2γ2) being gradually replaced by adult haemoglobin (HbA, α2ß2)1. This process has motivated the development of innovative approaches to treat sickle cell disease and ß-thalassaemia by increasing HbF levels in postnatal RBCs2. Here we provide therapeutically relevant insights into globin gene switching obtained through a CRISPR-Cas9 screen for ubiquitin-proteasome components that regulate HbF expression. In RBC precursors, depletion of the von Hippel-Lindau (VHL) E3 ubiquitin ligase stabilized its ubiquitination target, hypoxia-inducible factor 1α (HIF1α)3,4, to induce γ-globin gene transcription. Mechanistically, HIF1α-HIF1ß heterodimers bound cognate DNA elements in BGLT3, a long noncoding RNA gene located 2.7 kb downstream of the tandem γ-globin genes HBG1 and HBG2. This was followed by the recruitment of transcriptional activators, chromatin opening and increased long-range interactions between the γ-globin genes and their upstream enhancer. Similar induction of HbF occurred with hypoxia or with inhibition of prolyl hydroxylase domain enzymes that target HIF1α for ubiquitination by the VHL E3 ubiquitin ligase. Our findings link globin gene regulation with canonical hypoxia adaptation, provide a mechanism for HbF induction during stress erythropoiesis and suggest a new therapeutic approach for ß-haemoglobinopathies.


Asunto(s)
gamma-Globinas , Humanos , Cromatina , Hemoglobina Fetal/biosíntesis , Hemoglobina Fetal/genética , gamma-Globinas/biosíntesis , gamma-Globinas/genética , Hipoxia/genética , Prolil Hidroxilasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Largo no Codificante , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Eritropoyesis
2.
Proc Natl Acad Sci U S A ; 120(16): e2220134120, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37036970

RESUMEN

Bromo- and extra-terminal domain inhibitors (BETi) have exhibited therapeutic activities in many cancers. However, the mechanisms controlling BETi response and resistance are not well understood. We conducted genome-wide loss-of-function CRISPR screens using BETi-treated KMT2A-rearranged (KMT2A-r) cell lines. We revealed that Speckle-type POZ protein (SPOP) gene (Speckle Type BTB/POZ Protein) deficiency caused significant BETi resistance, which was further validated in cell lines and xenograft models. Proteomics analysis and a kinase-vulnerability CRISPR screen indicated that cells treated with BETi are sensitive to GSK3 perturbation. Pharmaceutical inhibition of GSK3 reversed the BETi-resistance phenotype. Based on this observation, a combination therapy regimen inhibiting both BET and GSK3 was developed to impede KMT2A-r leukemia progression in patient-derived xenografts in vivo. Our results revealed molecular mechanisms underlying BETi resistance and a promising combination treatment regimen of ABBV-744 and CHIR-98014 by utilizing unique ex vivo and in vivo KMT2A-r PDX models.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Leucemia , Humanos , Glucógeno Sintasa Quinasa 3/metabolismo , Línea Celular Tumoral , Leucemia/tratamiento farmacológico , Leucemia/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo
3.
Small ; : e2402793, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38757420

RESUMEN

Developing a new end group for synthesizing asymmetric small molecule acceptors (SMAs) is crucial for achieving high-performance organic photovoltaics (OPVs). Herein, an asymmetric small molecule acceptor, BTP-BO-4FO, featuring a new difluoro-methoxylated end-group is reported. Compared to its symmetric counterpart L8-BO, BTP-BO-4FO exhibits an upshifted energy level, larger dipole moment, and more sequential crystallinity. By adopting two representative and widely available solvent additives (1-chloronaphthalene (CN) and 1,8-diiodooctane (DIO)), the device based on PM6:BTP-BO-4FO (CN) photovoltaic blend demonstrates a power conversion efficiency (PCE) of 18.62% with an excellent open-circuit voltage (VOC) of 0.933 V, which surpasses the optimal result of L8-BO. The PCE of 18.62% realizes the best efficiencies for binary OPVs based on SMAs with asymmetric end groups. A series of investigations reveal that optimized PM6:BTP-BO-4FO film demonstrates similar molecular packing motif and fibrillar phase distribution as PM6:L8-BO (DIO) does, resulting in comparable recombination dynamics, thus, similar fill factor. Besides, it is found PM6:BTP-BO-4FO possesses more efficient charge generation, which yields better VOC-JSC balance. This study provides a new ending group that enables a cutting-edge efficiency in asymmetric SMA-based OPVs, enriching the material library and shed light on further design ideas.

4.
Blood ; 137(2): 155-167, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-33156908

RESUMEN

The histone mark H3K27me3 and its reader/writer polycomb repressive complex 2 (PRC2) mediate widespread transcriptional repression in stem and progenitor cells. Mechanisms that regulate this activity are critical for hematopoietic development but are poorly understood. Here we show that the E3 ubiquitin ligase F-box only protein 11 (FBXO11) relieves PRC2-mediated repression during erythroid maturation by targeting its newly identified substrate bromo adjacent homology domain-containing 1 (BAHD1), an H3K27me3 reader that recruits transcriptional corepressors. Erythroblasts lacking FBXO11 are developmentally delayed, with reduced expression of maturation-associated genes, most of which harbor bivalent histone marks at their promoters. In FBXO11-/- erythroblasts, these gene promoters bind BAHD1 and fail to recruit the erythroid transcription factor GATA1. The BAHD1 complex interacts physically with PRC2, and depletion of either component restores FBXO11-deficient erythroid gene expression. Our studies identify BAHD1 as a novel effector of PRC2-mediated repression and reveal how a single E3 ubiquitin ligase eliminates PRC2 repression at many developmentally poised bivalent genes during erythropoiesis.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Eritropoyesis/fisiología , Proteínas F-Box/metabolismo , Regulación de la Expresión Génica/fisiología , Complejo Represivo Polycomb 2/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Línea Celular , Eritroblastos/metabolismo , Humanos , Proteolisis
5.
Proc Natl Acad Sci U S A ; 116(52): 26644-26652, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31818950

RESUMEN

Loss of function of CDKN2A/B, also known as INK4/ARF [encoding p16INK4A, p15INK4B, and p14ARF (mouse p19Arf)], confers susceptibility to cancers, whereas its up-regulation during organismal aging provokes cellular senescence and tissue degenerative disorders. To better understand the transcriptional regulation of p16INK4A, a CRISPR screen targeting open, noncoding chromatin regions adjacent to p16INK4A was performed in a human p16INK4A-P2A-mCherry reporter cell line. We identified a repressive element located in the 3' region adjacent to the ARF promoter that controls p16INK4A expression via long-distance chromatin interactions. Coinfection of lentiviral dCas9-KRAB with selected single-guide RNAs against the repressive element abrogated the ARF/p16INK4A chromatin contacts, thus reactivating p16INK4A expression. Genetic CRISPR screening identified candidate transcription factors inhibiting p16INK4A regulation, including ZNF217, which was confirmed to bind the ARF/p16INK4A interaction loop. In summary, direct physical interactions between p16INK4A and ARF genes provide mechanistic insights into their cross-regulation.

6.
Nucleic Acids Res ; 47(13): 6699-6713, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31127282

RESUMEN

Numerous pieces of evidence support the complex, 3D spatial organization of the genome dictates gene expression. CTCF is essential to define topologically associated domain boundaries and to facilitate the formation of insulated chromatin loop structures. To understand CTCF's direct role in global transcriptional regulation, we integrated the miniAID-mClover3 cassette to the endogenous CTCF locus in a human pediatric B-ALL cell line, SEM, and an immortal erythroid precursor cell line, HUDEP-2, to allow for acute depletion of CTCF protein by the auxin-inducible degron system. In SEM cells, CTCF loss notably disrupted intra-TAD loops and TAD integrity in concurrence with a reduction in CTCF-binding affinity, while showing no perturbation to nuclear compartment integrity. Strikingly, the overall effect of CTCF's loss on transcription was minimal. Whole transcriptome analysis showed hundreds of genes differentially expressed in CTCF-depleted cells, among which MYC and a number of MYC target genes were specifically downregulated. Mechanically, acute depletion of CTCF disrupted the direct interaction between the MYC promoter and its distal enhancer cluster residing ∼1.8 Mb downstream. Notably, MYC expression was not profoundly affected upon CTCF loss in HUDEP-2 cells suggesting that CTCF could play a B-ALL cell line specific role in maintaining MYC expression.


Asunto(s)
Factor de Unión a CCCTC/fisiología , Cromatina/ultraestructura , Elementos de Facilitación Genéticos/genética , Regulación Leucémica de la Expresión Génica , Genes myc , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Factor de Unión a CCCTC/deficiencia , Línea Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Cromatina/genética , Regulación hacia Abajo , Células Precursoras Eritroides/metabolismo , Técnicas de Sustitución del Gen , Genes Reporteros , Humanos , Conformación de Ácido Nucleico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Transcriptoma
7.
Proc Natl Acad Sci U S A ; 114(28): 7420-7425, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28652370

RESUMEN

The mouse p19Arf (human p14ARF) tumor suppressor protein, encoded in part from an alternative reading frame of the Ink4a (Cdkn2a) gene, inhibits the Mdm2 E3 ubiquitin ligase to activate p53. Arf is not expressed in most normal tissues of young mice but is induced by high thresholds of aberrant hyperproliferative signals, thereby activating p53 in incipient tumor cells that have experienced oncogene activation. The single Arf mRNA encodes two distinct polypeptides, including full-length p19Arf and N-terminally truncated and unstable p15smArf ("small mitochondrial Arf") initiated from an internal in-frame AUG codon specifying methionine-45. Interactions of p19Arf with Mdm2, or separately with nucleophosmin (NPM, B23) that localizes and stabilizes p19Arf within the nucleolus, require p19Arf N-terminal amino acids that are not present within p15smArf We have generated mice that produce either smARF alone or M45A-mutated (smArf-deficient) full-length p19Arf proteins. BCR-ABL-expressing pro/pre-B cells producing smArf alone are as oncogenic as their Arf-null counterparts in generating acute lymphoblastic leukemia when infused into unconditioned syngeneic mice. In contrast, smArf-deficient cells from mice of the ArfM45A strain are as resistant as wild-type Arf+/+ cells to comparable oncogenic challenge and do not produce tumors. Apart from being prone to tumor development, Arf-null mice are blind, and their male germ cells exhibit defects in meiotic maturation and sperm production. Although ArfM45A mice manifest the latter defects, smArf alone remarkably rescues both of these p53-independent developmental phenotypes.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteína p53 Supresora de Tumor/genética , Células 3T3 , Animales , Ceguera/genética , Proliferación Celular , Codón , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Desoxirribonucleasas/metabolismo , Femenino , Fibroblastos/metabolismo , Proteínas de Fusión bcr-abl/metabolismo , Genes Supresores de Tumor , Células Germinativas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Fenotipo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Dominios Proteicos , Espermatogénesis , Proteína p53 Supresora de Tumor/metabolismo
8.
Angew Chem Int Ed Engl ; 58(43): 15372-15376, 2019 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-31449724

RESUMEN

Microcoil nuclear magnetic resonance (NMR) has been interfaced with digital microfluidics (DMF) and is applied to monitor organic reactions in organic solvents as a proof of concept. DMF permits droplets to be moved and mixed inside the NMR spectrometer to initiate reactions while using sub-microliter volumes of reagent, opening up the potential to follow the reactions of scarce or expensive reagents. By setting up the spectrometer shims on a reagent droplet, data acquisition can be started immediately upon droplet mixing and is only limited by the rate at which NMR data can be collected, allowing the monitoring of fast reactions. Here we report a cyclohexene carbonate hydrolysis in dimethylformamide and a Knoevenagel condensation in methanol/water. This is to our knowledge the first time rapid organic reactions in organic solvents have been monitored by high field DMF-NMR. The study represents a key first step towards larger DMF-NMR arrays that could in future serve as discovery platforms, where computer controlled DMF automates mixing/titration of chemical libraries and NMR is used to study the structures formed and kinetics in real time.

9.
J Cell Physiol ; 234(1): 802-815, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-30078221

RESUMEN

Tumor cell motility is the essential step in cancer metastasis. Previously, we showed that oxytocin and epidermal growth factor (EGF) effects on cell migration in prostate cancer cells require Giα2 protein. In the current study, we investigated the interactions among G-protein coupled receptor (GPCR), Giα2, PI3-kinase, and Rac1 activation in the induction of migratory and invasive behavior by diverse stimuli. Knockdown and knockout of endogenous Giα2 in PC3 cells resulted in attenuation of transforming growth factor ß1 (TGFß1), oxytocin, SDF-1α, and EGF effects on cell migration and invasion. In addition, knockdown of Giα2 in E006AA cells attenuated cell migration and overexpression of Giα2 in LNCaP cells caused significant increase in basal and EGF-stimulated cell migration. Pretreatment of PC3 cells with Pertussis toxin resulted in attenuation of TGFß1- and oxytocin-induced migratory behavior and PI3-kinase activation without affecting EGF-induced PI3-kinase activation and cell migration. Basal- and EGF-induced activation of Rac1 in PC3 and DU145 cells were not affected in cells after Giα2 knockdown. On the other hand, Giα2 knockdown abolished the migratory capability of PC3 cells overexpressing constitutively active Rac1. The knockdown or knockout of Giα2 resulted in impaired formation of lamellipodia at the leading edge of the migrating cells. We conclude that Giα2 protein acts at two different levels which are both dependent and independent of GPCR signaling to induce cell migration and invasion in prostate cancer cells and its action is downstream of PI3-kinase-AKT-Rac1 axis.


Asunto(s)
Movimiento Celular/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Neoplasias de la Próstata/genética , Proteína de Unión al GTP rac1/genética , Línea Celular Tumoral , Proliferación Celular/genética , Quimiocina CXCL12/genética , Factor de Crecimiento Epidérmico/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteína Oncogénica v-akt/genética , Oxitocina/genética , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/patología , Factor de Crecimiento Transformador beta1/genética
10.
J BUON ; 23(6): 1862-1866, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30610814

RESUMEN

PURPOSE: Esophageal cancer (EC) is the 9th most common carcinoma worldwide with poor prognosis. Specific biomarkers can help predicting the development of esophageal squamous cell carcinoma (ESCC), which can improve the assessment of prognosis. This study aimed to explore long noncoding RNA (lncRNA) ANRIL expression and its potential value in ESCC prognosis. METHODS: Quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) was utilized to detect lncRNA ANRIL expression in 50 pairs of ESCC and matched normal samples in order to explore the role of lncRNA ANRIL in ESCC. Moreover, the association was investigated between clinical characteristics of ESCC and the expression level of ANRIL. RESULTS: Disease-free survival (DFS) and overall survival (OS) were significantly shorter in ESCC patients with higher expression level of lncRNA ANRIL. ESCC tissues examined showed an obvious increment in ANRIL expression when compared to normal tissues. Furthermore, ANRIL was positively related to lymph nodes metastasis, TNM stage and tumor clinical stage. Moreover, upregulated ANRIL expression was remarkably associated with shorter survival in ESCC patients,which was also an independent prognostic factor for both OS and DFS. CONCLUSIONS: This study suggested that lncRNA ANRIL could be a potential oncogene of ESCC. ANRIL expression might be served as another potential therapeutic target and prognostic biomarker for ESCC.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/secundario , Neoplasias Esofágicas/patología , Esofagectomía/mortalidad , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/cirugía , Estudios de Casos y Controles , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/cirugía , Femenino , Estudios de Seguimiento , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia , Regulación hacia Arriba
11.
Proc Natl Acad Sci U S A ; 110(12): E1112-21, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23487795

RESUMEN

Induction of the Arf tumor suppressor (encoded by the alternate reading frame of the Cdkn2a locus) following oncogene activation engages a p53-dependent transcriptional program that limits the expansion of incipient cancer cells. Although the p19(Arf) protein is not detected in most tissues of fetal or young adult mice, it is physiologically expressed in the fetal yolk sac, a tissue derived from the extraembryonic endoderm (ExEn). Expression of the mouse p19(Arf) protein marks late stages of ExEn differentiation in cultured embryoid bodies (EBs) derived from either embryonic stem cells or induced pluripotent stem cells. Arf inactivation delays differentiation of the ExEn lineage within EBs, but not the formation of other germ cell lineages from pluripotent progenitors. Arf is required for the timely induction of ExEn cells in response to Ras/Erk signaling and, in turn, acts through p53 to ensure the development, but not maintenance, of the ExEn lineage. Remarkably, a significant temporal delay in ExEn differentiation detected during the maturation of Arf-null EBs is rescued by enforced expression of mouse microRNA-205 (miR-205), a microRNA up-regulated by p19(Arf) and p53 that controls ExEn cell migration and adhesion. The noncanonical and canonical roles of Arf in ExEn development and tumor suppression, respectively, may be conceptually linked through mechanisms that govern cell attachment and migration.


Asunto(s)
Movimiento Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Endodermo/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , MicroARNs/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Adhesión Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular , Linaje de la Célula/fisiología , Endodermo/citología , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Noqueados , MicroARNs/genética , Células Madre Pluripotentes/citología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/fisiología
12.
Angew Chem Int Ed Engl ; 55(38): 11572-6, 2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27529815

RESUMEN

The alternating copolymerization of biorenewable limonene dioxide with carbon dioxide (CO2 ) catalyzed by a zinc ß-diiminate complex is reported. The chemoselective reaction results in linear amorphous polycarbonates that carry pendent methyloxiranes and exhibit glass transition temperatures (Tg ) up to 135 °C. These polycarbonates can be efficiently modified by thiols or carboxylic acids in combination with lithium hydroxide or tetrabutylphosphonium bromide as catalysts, respectively, without destruction of the main chain. Moreover, polycarbonates bearing pendent cyclic carbonates can be quantitatively prepared by CO2 insertion catalyzed by lithium bromide.

13.
Neural Comput ; 27(8): 1738-65, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26079748

RESUMEN

The abundance of real-world data and limited labeling budget calls for active learning, an important learning paradigm for reducing human labeling efforts. Many recently developed active learning algorithms consider both uncertainty and representativeness when making querying decisions. However, exploiting representativeness with uncertainty concurrently usually requires tackling sophisticated and challenging learning tasks, such as clustering. In this letter, we propose a new active learning framework, called hinted sampling, which takes both uncertainty and representativeness into account in a simpler way. We design a novel active learning algorithm within the hinted sampling framework with an extended support vector machine. Experimental results validate that the novel active learning algorithm can result in a better and more stable performance than that achieved by state-of-the-art algorithms. We also show that the hinted sampling framework allows improving another active learning algorithm designed from the transductive support vector machine.


Asunto(s)
Algoritmos , Inteligencia Artificial , Reconocimiento de Normas Patrones Automatizadas/métodos , Análisis por Conglomerados , Humanos
15.
Genome Biol ; 25(1): 16, 2024 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-38216972

RESUMEN

BACKGROUND: The oncogenic protein HOXA9 plays a critical role in leukemia transformation and maintenance, and its aberrant expression is a hallmark of most aggressive acute leukemia. Although inhibiting the upstream regulators of HOXA9 has been proven as a significant therapeutic intervention, the comprehensive regulation network controlling HOXA9 expression in leukemia has not been systematically investigated. RESULTS: Here, we perform genome-wide CRISPR/Cas9 screening in the HOXA9-driven reporter acute leukemia cells. We identify a poorly characterized RNA-binding protein, RBM5, as the top candidate gene required to maintain leukemia cell fitness. RBM5 is highly overexpressed in acute myeloid leukemia (AML) patients compared to healthy individuals. RBM5 loss triggered by CRISPR knockout and shRNA knockdown significantly impairs leukemia maintenance in vitro and in vivo. Through domain CRISPR screening, we reveal that RBM5 functions through a noncanonical transcriptional regulation circuitry rather than RNA splicing, such an effect depending on DNA-binding domains. By integrative analysis and functional assays, we identify HOXA9 as the downstream target of RBM5. Ectopic expression of HOXA9 rescues impaired leukemia cell proliferation upon RBM5 loss. Importantly, acute protein degradation of RBM5 through auxin-inducible degron system immediately reduces HOXA9 transcription. CONCLUSIONS: We identify RBM5 as a new upstream regulator of HOXA9 and reveal its essential role in controlling the survival of AML. These functional and molecular mechanisms further support RBM5 as a promising therapeutic target for myeloid leukemia treatment.


Asunto(s)
Proteínas de Homeodominio , Leucemia Mieloide Aguda , Humanos , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Supresoras de Tumor/metabolismo
16.
J Exp Med ; 221(9)2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-38935023

RESUMEN

Nuclear factor Foxp3 determines regulatory T (Treg) cell fate and function via mechanisms that remain unclear. Here, we investigate the nature of Foxp3-mediated gene regulation in suppressing autoimmunity and antitumor immune response. Contrasting with previous models, we find that Foxp3-chromatin binding is regulated by Treg activation states, tumor microenvironment, and antigen and cytokine stimulations. Proteomics studies uncover dynamic proteins within Foxp3 proximity upon TCR or IL-2 receptor signaling in vitro, reflecting intricate interactions among Foxp3, signal transducers, and chromatin. Pharmacological inhibition and genetic knockdown experiments indicate that NFAT and AP-1 protein Batf are required for enhanced Foxp3-chromatin binding in activated Treg cells and tumor-infiltrating Treg cells to modulate target gene expression. Furthermore, mutations at the Foxp3 DNA-binding domain destabilize the Foxp3-chromatin association. These representative settings delineate context-dependent Foxp3-chromatin interaction, suggesting that Foxp3 associates with chromatin by hijacking DNA-binding proteins resulting from Treg activation or differentiation, which is stabilized by direct Foxp3-DNA binding, to dynamically regulate Treg cell function according to immunological contexts.


Asunto(s)
Cromatina , Factores de Transcripción Forkhead , Linfocitos T Reguladores , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Cromatina/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Transducción de Señal , Unión Proteica , Humanos , Regulación de la Expresión Génica , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina-2/metabolismo , Receptores de Interleucina-2/genética , Diferenciación Celular
17.
J Biol Chem ; 287(46): 38970-9, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23019320

RESUMEN

Klinefelter syndrome (KS) is the most common male chromosome aneuploidy. Its pathophysiology is largely unexplained due to the lack of adequate models. Here, we report the derivation of induced pluripotent stem cell (iPSCs) lines from a KS patient with a karyotype of 47, XXY. Derived KS-iPSCs meet all criteria of normal iPSCs with the potential for germ cell differentiation. Although X chromosome inactivation occurs in all KS-iPSCs, genome-wide transcriptome analysis identifies aberrantly expressed genes associated with the clinical features of KS. Our KS-iPSCs can serve as a cellular model for KS research. Identified genes may become biomarkers for early diagnosis or potential therapeutic targets for KS and significantly accelerate the understanding, diagnosis, and treatment of Klinefelter syndrome.


Asunto(s)
Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Síndrome de Klinefelter/metabolismo , Aneuploidia , Diferenciación Celular , Bandeo Cromosómico , Perfilación de la Expresión Génica/métodos , Humanos , Cariotipificación , Masculino , Microscopía Fluorescente/métodos , Regeneración , Transcripción Genética
18.
Proc Natl Acad Sci U S A ; 107(4): 1402-7, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20080709

RESUMEN

Self-renewal and differentiation of embryonic stem cells (ESCs) are controlled by intracellular transcriptional factors and extracellular factor-activated signaling pathways. Transcription factor Oct4 is a key player maintaining ESCs in an undifferentiated state, whereas the Erk/MAPK pathway is known to be important for ESC differentiation. However, the manner in which intracellular pluripotency factors modulate extracellular factor-activated signaling pathways in ESCs is not well understood. Here, we report identification of a target gene of Oct4, serine/threonine kinase 40 (Stk40), which is able to activate the Erk/MAPK pathway and induce extraembryonic-endoderm (ExEn) differentiation in mouse ESCs. Interestingly, cells overexpressing Stk40 exclusively contribute to the ExEn layer of chimeric embryos when injected into host blastocysts. In contrast, deletion of Stk40 in ESCs markedly reduces ExEn differentiation in vitro. Mechanistically, Stk40 interacts with Rcn2, which also activates Erk1/2 to induce ExEn specification in mouse ESCs. Moreover, Rcn2 proteins are specifically located in the cytoplasm of the ExEn layer of early mouse embryos. Importantly, knockdown of Rcn2 blocks Stk40-activated Erk1/2 and ESC differentiation. Therefore, our study establishes a link between the pluripotency factor Oct4 and the Erk/MAPK signaling pathway, and it uncovers cooperating signals in the Erk/MAPK activation that control ExEn differentiation.


Asunto(s)
Diferenciación Celular , Endodermo/citología , Endodermo/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Línea Celular , Linaje de la Célula , Células Madre Embrionarias/metabolismo , Activación Enzimática , Regulación de la Expresión Génica , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN
19.
FEBS J ; 290(2): 321-339, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-34743404

RESUMEN

Dysregulation of the oncogenic transcription factor HOXA9 is a prominent feature for most aggressive acute myeloid leukemia cases and a strong indicator of poor prognosis in patients. Leukemia subtypes with hallmark overexpression of HOXA9 include those carrying MLL gene rearrangements, NPM1c mutations, and other genetic alternations. A growing body of evidence indicates that HOXA9 dysregulation is both sufficient and necessary for leukemic transformation. The HOXA9 mRNA and protein regulation includes multilayered controls by transcription factors (such as CDX2/4 and USF2/1), epigenetic factors (such as MLL-menin-LEDGF, DOT1L, ENL, HBO1, NPM1c-XPO1, and polycomb proteins), microRNAs (such as miR-126 and miR-196b), long noncoding RNAs (such as HOTTIP), three-dimensional chromatin interactions, and post-translational protein modifications. Recently, insights into the dynamic regulation of HOXA9 have led to an advanced understanding of the HOXA9 regulome and provided new cancer therapeutic opportunities, including developing inhibitors targeting DOT1L, menin, and ENL proteins. This review summarizes recent advances in understanding the molecular mechanisms controlling HOXA9 regulation and the pharmacological approaches that target HOXA9 regulators to treat HOXA9-driven acute myeloid leukemia.


Asunto(s)
Proteínas de Homeodominio , Leucemia Mieloide Aguda , MicroARNs , Humanos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/genética , MicroARNs/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Procesamiento Proteico-Postraduccional , Factores de Transcripción/genética
20.
Genome Biol ; 24(1): 14, 2023 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-36698211

RESUMEN

BACKGROUND: CTCF is a well-established chromatin architectural protein that also plays various roles in transcriptional regulation. While CTCF biology has been extensively studied, how the domains of CTCF function to regulate transcription remains unknown. Additionally, the original auxin-inducible degron 1 (AID1) system has limitations in investigating the function of CTCF. RESULTS: We employ an improved auxin-inducible degron technology, AID2, to facilitate the study of acute depletion of CTCF while overcoming the limitations of the previous AID system. As previously observed through the AID1 system and steady-state RNA analysis, the new AID2 system combined with SLAM-seq confirms that CTCF depletion leads to modest nascent and steady-state transcript changes. A CTCF domain sgRNA library screening identifies the zinc finger (ZF) domain as the region within CTCF with the most functional relevance, including ZFs 1 and 10. Removal of ZFs 1 and 10 reveals genomic regions that independently require these ZFs for DNA binding and transcriptional regulation. Notably, loci regulated by either ZF1 or ZF10 exhibit unique CTCF binding motifs specific to each ZF. CONCLUSIONS: By extensively comparing the AID1 and AID2 systems for CTCF degradation in SEM cells, we confirm that AID2 degradation is superior for achieving miniAID-tagged protein degradation without the limitations of the AID1 system. The model we create that combines AID2 depletion of CTCF with exogenous overexpression of CTCF mutants allows us to demonstrate how peripheral ZFs intricately orchestrate transcriptional regulation in a cellular context for the first time.


Asunto(s)
Regulación de la Expresión Génica , Ácidos Indolacéticos , Factor de Unión a CCCTC/metabolismo , Dedos de Zinc , Genoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA