Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 5(1): 110, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115654

RESUMEN

Somatic mutation in TET2 gene is one of the most common clonal genetic events detected in age-related clonal hematopoiesis as well as in chronic myelomonocytic leukemia (CMML). In addition to being a pre-malignant state, TET2 mutated clones are associated with an increased risk of death from cardiovascular disease, which could involve cytokine/chemokine overproduction by monocytic cells. Here, we show in mice and in human cells that, in the absence of any inflammatory challenge, TET2 downregulation promotes the production of MIF (macrophage migration inhibitory factor), a pivotal mediator of atherosclerotic lesion formation. In healthy monocytes, TET2 is recruited to MIF promoter and interacts with the transcription factor EGR1 and histone deacetylases. Disruption of these interactions as a consequence of TET2-decreased expression favors EGR1-driven transcription of MIF gene and its secretion. MIF favors monocytic differentiation of myeloid progenitors. These results designate MIF as a chronically overproduced chemokine and a potential therapeutic target in patients with clonal TET2 downregulation in myeloid cells.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Monocitos/metabolismo , Animales , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Dioxigenasas/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Regulación de la Expresión Génica/fisiología , Humanos , Recién Nacido , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones
2.
Nat Commun ; 12(1): 634, 2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33504775

RESUMEN

The SARS-COV-2 pandemic has put pressure on intensive care units, so that identifying predictors of disease severity is a priority. We collect 58 clinical and biological variables, and chest CT scan data, from 1003 coronavirus-infected patients from two French hospitals. We train a deep learning model based on CT scans to predict severity. We then construct the multimodal AI-severity score that includes 5 clinical and biological variables (age, sex, oxygenation, urea, platelet) in addition to the deep learning model. We show that neural network analysis of CT-scans brings unique prognosis information, although it is correlated with other markers of severity (oxygenation, LDH, and CRP) explaining the measurable but limited 0.03 increase of AUC obtained when adding CT-scan information to clinical variables. Here, we show that when comparing AI-severity with 11 existing severity scores, we find significantly improved prognosis performance; AI-severity can therefore rapidly become a reference scoring approach.


Asunto(s)
COVID-19/diagnóstico , COVID-19/fisiopatología , Aprendizaje Profundo , Redes Neurales de la Computación , Tomografía Computarizada por Rayos X/métodos , Inteligencia Artificial , COVID-19/clasificación , Humanos , Modelos Biológicos , Análisis Multivariante , Pronóstico , Radiólogos , Índice de Severidad de la Enfermedad
3.
Nat Commun ; 11(1): 3877, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32747659

RESUMEN

Deep learning methods for digital pathology analysis are an effective way to address multiple clinical questions, from diagnosis to prediction of treatment outcomes. These methods have also been used to predict gene mutations from pathology images, but no comprehensive evaluation of their potential for extracting molecular features from histology slides has yet been performed. We show that HE2RNA, a model based on the integration of multiple data modes, can be trained to systematically predict RNA-Seq profiles from whole-slide images alone, without expert annotation. Through its interpretable design, HE2RNA provides virtual spatialization of gene expression, as validated by CD3- and CD20-staining on an independent dataset. The transcriptomic representation learned by HE2RNA can also be transferred on other datasets, even of small size, to increase prediction performance for specific molecular phenotypes. We illustrate the use of this approach in clinical diagnosis purposes such as the identification of tumors with microsatellite instability.


Asunto(s)
Biología Computacional/métodos , Aprendizaje Profundo , Regulación Neoplásica de la Expresión Génica , Procesamiento de Imagen Asistido por Computador/métodos , Neoplasias/genética , RNA-Seq/métodos , Algoritmos , Perfilación de la Expresión Génica/métodos , Humanos , Inestabilidad de Microsatélites , Modelos Genéticos , Neoplasias/diagnóstico , Neoplasias/metabolismo
4.
Hepatology ; 72(6): 2000-2013, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32108950

RESUMEN

BACKGROUND AND AIMS: Standardized and robust risk-stratification systems for patients with hepatocellular carcinoma (HCC) are required to improve therapeutic strategies and investigate the benefits of adjuvant systemic therapies after curative resection/ablation. APPROACH AND RESULTS: In this study, we used two deep-learning algorithms based on whole-slide digitized histological slides (whole-slide imaging; WSI) to build models for predicting survival of patients with HCC treated by surgical resection. Two independent series were investigated: a discovery set (Henri Mondor Hospital, n = 194) used to develop our algorithms and an independent validation set (The Cancer Genome Atlas [TCGA], n = 328). WSIs were first divided into small squares ("tiles"), and features were extracted with a pretrained convolutional neural network (preprocessing step). The first deep-learning-based algorithm ("SCHMOWDER") uses an attention mechanism on tumoral areas annotated by a pathologist whereas the second ("CHOWDER") does not require human expertise. In the discovery set, c-indices for survival prediction of SCHMOWDER and CHOWDER reached 0.78 and 0.75, respectively. Both models outperformed a composite score incorporating all baseline variables associated with survival. Prognostic value of the models was further validated in the TCGA data set, and, as observed in the discovery series, both models had a higher discriminatory power than a score combining all baseline variables associated with survival. Pathological review showed that the tumoral areas most predictive of poor survival were characterized by vascular spaces, the macrotrabecular architectural pattern, and a lack of immune infiltration. CONCLUSIONS: This study shows that artificial intelligence can help refine the prediction of HCC prognosis. It highlights the importance of pathologist/machine interactions for the construction of deep-learning algorithms that benefit from expert knowledge and allow a biological understanding of their output.


Asunto(s)
Carcinoma Hepatocelular/mortalidad , Aprendizaje Profundo , Hepatectomía/métodos , Neoplasias Hepáticas/mortalidad , Anciano , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/cirugía , Estudios de Factibilidad , Femenino , Estudios de Seguimiento , Humanos , Hígado/patología , Hígado/cirugía , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/cirugía , Masculino , Persona de Mediana Edad , Pronóstico , Medición de Riesgo/métodos , Análisis de Supervivencia , Resultado del Tratamiento
5.
Nat Med ; 25(10): 1519-1525, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31591589

RESUMEN

Malignant mesothelioma (MM) is an aggressive cancer primarily diagnosed on the basis of histological criteria1. The 2015 World Health Organization classification subdivides mesothelioma tumors into three histological types: epithelioid, biphasic and sarcomatoid MM. MM is a highly complex and heterogeneous disease, rendering its diagnosis and histological typing difficult and leading to suboptimal patient care and decisions regarding treatment modalities2. Here we have developed a new approach-based on deep convolutional neural networks-called MesoNet to accurately predict the overall survival of mesothelioma patients from whole-slide digitized images, without any pathologist-provided locally annotated regions. We validated MesoNet on both an internal validation cohort from the French MESOBANK and an independent cohort from The Cancer Genome Atlas (TCGA). We also demonstrated that the model was more accurate in predicting patient survival than using current pathology practices. Furthermore, unlike classical black-box deep learning methods, MesoNet identified regions contributing to patient outcome prediction. Strikingly, we found that these regions are mainly located in the stroma and are histological features associated with inflammation, cellular diversity and vacuolization. These findings suggest that deep learning models can identify new features predictive of patient survival and potentially lead to new biomarker discoveries.


Asunto(s)
Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/patología , Mesotelioma/diagnóstico , Mesotelioma/patología , Pronóstico , Aprendizaje Profundo , Femenino , Humanos , Neoplasias Pulmonares/clasificación , Masculino , Mesotelioma/clasificación , Mesotelioma Maligno , Clasificación del Tumor , Redes Neurales de la Computación
6.
Clin Cancer Res ; 25(10): 2956-2962, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30655313

RESUMEN

In 2013, two seminal studies identified gain-of-function mutations in the Calreticulin (CALR) gene in a subset of JAK2/MPL-negative myeloproliferative neoplasm (MPN) patients. CALR is an endoplasmic reticulum (ER) chaperone protein that normally binds misfolded proteins in the ER and prevents their export to the Golgi and had never previously been reported mutated in cancer or to be associated with hematologic disorders. Further investigation determined that mutated CALR is able to achieve oncogenic transformation primarily through constitutive activation of the MPL-JAK-STAT signaling axis. Here we review our current understanding of the role of CALR mutations in MPN pathogenesis and how these insights can lead to innovative therapeutics approaches.


Asunto(s)
Calreticulina/genética , Transformación Celular Neoplásica/metabolismo , Neoplasias Hematológicas/genética , Mutación , Trastornos Mieloproliferativos/genética , Animales , Calreticulina/metabolismo , Transformación Celular Neoplásica/genética , Aparato de Golgi/metabolismo , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Transducción de Señal
7.
JCI Insight ; 3(22)2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30429377

RESUMEN

Mutations in the ER chaperone calreticulin (CALR) are common in myeloproliferative neoplasm (MPN) patients, activate the thrombopoietin receptor (MPL), and mediate constitutive JAK/STAT signaling. The mechanisms by which CALR mutations cause myeloid transformation are incompletely defined. We used mass spectrometry proteomics to identify CALR-mutant interacting proteins. Mutant CALR caused mislocalization of binding partners and increased recruitment of FLI1, ERP57, and CALR to the MPL promoter to enhance transcription. Consistent with a critical role for CALR-mediated JAK/STAT activation, we confirmed the efficacy of JAK2 inhibition on CALR-mutant cells in vitro and in vivo. Due to the altered interactome induced by CALR mutations, we hypothesized that CALR-mutant MPNs may be vulnerable to disruption of aberrant CALR protein complexes. A synthetic peptide designed to competitively inhibit the carboxy terminal of CALR specifically abrogated MPL/JAK/STAT signaling in cell lines and primary samples and improved the efficacy of JAK kinase inhibitors. These findings reveal what to our knowledge is a novel potential therapeutic approach for patients with CALR-mutant MPN.


Asunto(s)
Antineoplásicos/farmacología , Calreticulina/genética , Leucemia/genética , Trastornos Mieloproliferativos/genética , Animales , Calreticulina/antagonistas & inhibidores , Calreticulina/metabolismo , Línea Celular , Cromatina/metabolismo , Sistemas de Liberación de Medicamentos , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Quinasas Janus/antagonistas & inhibidores , Leucemia/tratamiento farmacológico , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Trastornos Mieloproliferativos/tratamiento farmacológico , Receptores de Trombopoyetina/genética , Transducción de Señal
8.
Blood ; 132(12): 1265-1278, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30064973

RESUMEN

Genetic studies have identified recurrent somatic mutations in acute myeloid leukemia (AML) patients, including in the Wilms' tumor 1 (WT1) gene. The molecular mechanisms by which WT1 mutations contribute to leukemogenesis have not yet been fully elucidated. We investigated the role of Wt1 gene dosage in steady-state and pathologic hematopoiesis. Wt1 heterozygous loss enhanced stem cell self-renewal in an age-dependent manner, which increased stem cell function over time and resulted in age-dependent leukemic transformation. Wt1-haploinsufficient leukemias were characterized by progressive genetic and epigenetic alterations, including those in known leukemia-associated alleles, demonstrating a requirement for additional events to promote hematopoietic transformation. Consistent with this observation, we found that Wt1 depletion cooperates with Flt3-ITD mutation to induce fully penetrant AML. Our studies provide insight into mechanisms of Wt1-loss leukemogenesis and into the evolutionary events required to induce transformation of Wt1-haploinsufficient stem/progenitor cells.


Asunto(s)
Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Mutación , Proteínas Represoras/genética , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Autorrenovación de las Células , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/patología , Leucopoyesis , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Células Mieloides/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Proteínas WT1 , Tirosina Quinasa 3 Similar a fms/genética
9.
Cancer Cell ; 33(1): 44-59.e8, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29275866

RESUMEN

Mutations in epigenetic modifiers and signaling factors often co-occur in myeloid malignancies, including TET2 and NRAS mutations. Concurrent Tet2 loss and NrasG12D expression in hematopoietic cells induced myeloid transformation, with a fully penetrant, lethal chronic myelomonocytic leukemia (CMML), which was serially transplantable. Tet2 loss and Nras mutation cooperatively led to decrease in negative regulators of mitogen-activated protein kinase (MAPK) activation, including Spry2, thereby causing synergistic activation of MAPK signaling by epigenetic silencing. Tet2/Nras double-mutant leukemia showed preferential sensitivity to MAPK kinase (MEK) inhibition in both mouse model and patient samples. These data provide insights into how epigenetic and signaling mutations cooperate in myeloid transformation and provide a rationale for mechanism-based therapy in CMML patients with these high-risk genetic lesions.


Asunto(s)
Proteínas de Unión al ADN/genética , GTP Fosfohidrolasas/genética , Leucemia Mielomonocítica Crónica/genética , Proteínas de la Membrana/genética , Proteínas de Unión al GTP Monoméricas/genética , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Animales , Transformación Celular Neoplásica/genética , Dioxigenasas , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones Transgénicos , Trastornos Mieloproliferativos/genética , Proteínas Serina-Treonina Quinasas , Transducción de Señal/genética
10.
Blood ; 129(13): 1779-1790, 2017 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-28077417

RESUMEN

Recent studies have reported that activation-induced cytidine deaminase (AID) and ten-eleven-translocation (TET) family members regulate active DNA demethylation. Genetic alterations of TET2 occur in myeloid malignancies, and hematopoietic-specific loss of Tet2 induces aberrant hematopoietic stem cell (HSC) self-renewal/differentiation, implicating TET2 as a master regulator of normal and malignant hematopoiesis. Despite the functional link between AID and TET in epigenetic gene regulation, the role of AID loss in hematopoiesis and myeloid transformation remains to be investigated. Here, we show that Aid loss in mice leads to expansion of myeloid cells and reduced erythroid progenitors resulting in anemia, with dysregulated expression of Cebpa and Gata1, myeloid/erythroid lineage-specific transcription factors. Consistent with data in the murine context, silencing of AID in human bone marrow cells skews differentiation toward myelomonocytic lineage. However, in contrast to Tet2 loss, Aid loss does not contribute to enhanced HSC self-renewal or cooperate with Flt3-ITD to induce myeloid transformation. Genome-wide transcription and differential methylation analysis uncover the critical role of Aid as a key epigenetic regulator. These results indicate that AID and TET2 share common effects on myeloid and erythroid lineage differentiation, however, their role is nonredundant in regulating HSC self-renewal and in myeloid transformation.


Asunto(s)
Diferenciación Celular , Citidina Desaminasa/fisiología , Metilación de ADN , Células Madre Hematopoyéticas/metabolismo , Animales , Linaje de la Célula , Autorrenovación de las Células , Transformación Celular Neoplásica , Citidina Desaminasa/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Dioxigenasas , Células Eritroides/citología , Silenciador del Gen , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Células Mieloides/citología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología
11.
Nat Med ; 22(12): 1488-1495, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27841873

RESUMEN

Although the majority of patients with acute myeloid leukemia (AML) initially respond to chemotherapy, many of them subsequently relapse, and the mechanistic basis for AML persistence following chemotherapy has not been determined. Recurrent somatic mutations in DNA methyltransferase 3A (DNMT3A), most frequently at arginine 882 (DNMT3AR882), have been observed in AML and in individuals with clonal hematopoiesis in the absence of leukemic transformation. Patients with DNMT3AR882 AML have an inferior outcome when treated with standard-dose daunorubicin-based induction chemotherapy, suggesting that DNMT3AR882 cells persist and drive relapse. We found that Dnmt3a mutations induced hematopoietic stem cell expansion, cooperated with mutations in the FMS-like tyrosine kinase 3 gene (Flt3ITD) and the nucleophosmin gene (Npm1c) to induce AML in vivo, and promoted resistance to anthracycline chemotherapy. In patients with AML, the presence of DNMT3AR882 mutations predicts minimal residual disease, underscoring their role in AML chemoresistance. DNMT3AR882 cells showed impaired nucleosome eviction and chromatin remodeling in response to anthracycline treatment, which resulted from attenuated recruitment of histone chaperone SPT-16 following anthracycline exposure. This defect led to an inability to sense and repair DNA torsional stress, which resulted in increased mutagenesis. Our findings identify a crucial role for DNMT3AR882 mutations in driving AML chemoresistance and highlight the importance of chromatin remodeling in response to cytotoxic chemotherapy.


Asunto(s)
Antraciclinas/uso terapéutico , Ensamble y Desensamble de Cromatina/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Resistencia a Antineoplásicos/genética , Leucemia Mieloide Aguda/genética , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular , ADN Metiltransferasa 3A , Daunorrubicina/uso terapéutico , Células Madre Hematopoyéticas , Humanos , Immunoblotting , Inmunoprecipitación , Leucemia Mieloide Aguda/tratamiento farmacológico , Espectrometría de Masas , Ratones , Mutación , Proteínas Nucleares/genética , Nucleofosmina , Nucleosomas/metabolismo , Tirosina Quinasa 3 Similar a fms/genética
12.
Nat Commun ; 7: 12475, 2016 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27534895

RESUMEN

In acute myeloid leukaemia (AML) initiating pre-leukaemic lesions can be identified through three major hallmarks: their early occurrence in the clone, their persistence at relapse and their ability to initiate multilineage haematopoietic repopulation and leukaemia in vivo. Here we analyse the clonal composition of a series of AML through these characteristics. We find that not only DNMT3A mutations, but also TET2, ASXL1 mutations, core-binding factor and MLL translocations, as well as del(20q) mostly fulfil these criteria. When not eradicated by AML treatments, pre-leukaemic cells with these lesions can re-initiate the leukaemic process at various stages until relapse, with a time-dependent increase in clonal variegation. Based on the nature, order and association of lesions, we delineate recurrent genetic hierarchies of AML. Our data indicate that first lesions, variegation and treatment selection pressure govern the expansion and adaptive behaviour of the malignant clone, shaping AML in a time-dependent manner.


Asunto(s)
Evolución Clonal/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Animales , Secuencia de Bases , Células Clonales , Epigénesis Genética , Reordenamiento Génico/genética , Hematopoyesis , Humanos , Ratones , Mutación/genética , Análisis de la Célula Individual , Factores de Tiempo
13.
Cancer Cell ; 27(3): 323-5, 2015 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-25759018

RESUMEN

There is a pressing need to develop novel, mechanism-based therapeutic approaches that can be used to improve therapies for genetically defined tumor subtypes. Chan and colleagues have demonstrated recently that BCL-2 inhibitors can target IDH1/2 mutant cancers through a mutant-specific dependency in metabolic regulation.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Resistencia a Antineoplásicos/genética , Isocitrato Deshidrogenasa/genética , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Sulfonamidas/farmacología , Humanos
14.
Cell Rep ; 9(5): 1841-1855, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25482556

RESUMEN

Somatic mutations in IDH1/IDH2 and TET2 result in impaired TET2-mediated conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). The observation that WT1 inactivating mutations anticorrelate with TET2/IDH1/IDH2 mutations in acute myeloid leukemia (AML) led us to hypothesize that WT1 mutations may impact TET2 function. WT1 mutant AML patients have reduced 5hmC levels similar to TET2/IDH1/IDH2 mutant AML. These mutations are characterized by convergent, site-specific alterations in DNA hydroxymethylation, which drive differential gene expression more than alterations in DNA promoter methylation. WT1 overexpression increases global levels of 5hmC, and WT1 silencing reduced 5hmC levels. WT1 physically interacts with TET2 and TET3, and WT1 loss of function results in a similar hematopoietic differentiation phenotype as observed with TET2 deficiency. These data provide a role for WT1 in regulating DNA hydroxymethylation and suggest that TET2 IDH1/IDH2 and WT1 mutations define an AML subtype defined by dysregulated DNA hydroxymethylation.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas WT1/genética , 5-Metilcitosina/análogos & derivados , Animales , Diferenciación Celular , Citosina/análogos & derivados , Citosina/fisiología , Dioxigenasas , Elementos de Facilitación Genéticos , Hematopoyesis , Humanos , Leucemia Mieloide Aguda/metabolismo , Ratones Noqueados , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Análisis de Secuencia de ADN
15.
PLoS One ; 8(3): e59045, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527084

RESUMEN

Bone Morphogenetic Proteins (BMPs) are morphogens that play a major role in regulating development and homeostasis. Although BMPs are used for the treatment of bone and kidney disorders, their clinical use is limited due to the supra-physiological doses required for therapeutic efficacy causing severe side effects. Because recombinant BMPs are expensive to produce, small molecule activators of BMP signaling would be a cost-effective alternative with the added benefit of being potentially more easily deliverable. Here, we report our efforts to identify small molecule activators of BMP signaling. We have developed a cell-based assay to monitor BMP signaling by stably transfecting a BMP-responsive human cervical carcinoma cell line (C33A) with a reporter construct in which the expression of luciferase is driven by a multimerized BMP-responsive element from the Id1 promoter. A BMP-responsive clone C33A-2D2 was used to screen a bioactive library containing ∼5,600 small molecules. We identified four small molecules of the family of flavonoids all of which induced luciferase activity in a dose-dependent manner and ventralized zebrafish embryos. Two of the identified compounds induced Smad1, 5 phosphorylation (P-Smad), Id1 and Id2 expression in a dose-dependent manner demonstrating that our assays identified small molecule activators of BMP signaling.


Asunto(s)
Proteínas Morfogenéticas Óseas/agonistas , Proteínas Morfogenéticas Óseas/metabolismo , Descubrimiento de Drogas , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas , Animales , Línea Celular Tumoral , Chalcona/farmacología , Embrión no Mamífero/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Flavonas/farmacología , Genes Reporteros , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Ratones Noqueados , Mioblastos/citología , Mioblastos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Pez Cebra
16.
Curr Hematol Malig Rep ; 7(1): 57-64, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22200996

RESUMEN

Recently, 5-hydroxymethylcytosine (5-hmC), the 6th base of DNA, was discovered as the product of the hydroxylation of 5-methylcytosine (5-mC) by the ten-eleven translocation (TET) oncogene family members. One of them, TET oncogene family member 2 (TET2), is mutated in a variety of myeloid malignancies, including in 15% of myeloproliferative neoplasms (MPNs). Recent studies tried to go further into the biological and epigenetic function of TET2 protein and 5-hmC marks in the pathogenesis of myeloid malignancies. Although its precise function remains partially unknown, TET2 appears to be an important regulator of hematopoietic stem cell biology. In both mouse and human cells, its inactivation leads to a dramatic deregulation of hematopoiesis that ultimately triggers blood malignancies. Understanding this leukemogenic process will provide tools to develop new epigenetic therapies against blood cancers.


Asunto(s)
Proteínas de Unión al ADN/genética , Mutación/genética , Trastornos Mieloproliferativos/genética , Proteínas Proto-Oncogénicas/genética , Animales , Proteínas de Unión al ADN/fisiología , Dioxigenasas , Modelos Animales de Enfermedad , Frecuencia de los Genes , Hematopoyesis/genética , Humanos , Ratones , Trastornos Mieloproliferativos/fisiopatología
17.
Blood ; 118(9): 2551-5, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21734233

RESUMEN

TET2 converts 5-methylcytosine to 5-hydroxymethylcytosine (5-hmC) in DNA and is frequently mutated in myeloid malignancies, including myeloproliferative neoplasms. Here we show that the level of 5-hmC is decreased in granulocyte DNA from myeloproliferative neoplasm patients with TET2 mutations compared with granulocyte DNA from healthy patients. Inhibition of TET2 by RNA interference decreases 5-hmC levels in both human leukemia cell lines and cord blood CD34(+) cells. These results confirm the enzymatic function of TET2 in human hematopoietic cells. Knockdown of TET2 in cord blood CD34(+) cells skews progenitor differentiation toward the granulomonocytic lineage at the expense of lymphoid and erythroid lineages. In addition, by monitoring in vitro granulomonocytic development we found a decreased granulocytic differentiation and an increase in monocytic cells. Our results indicate that TET2 disruption affects 5-hmC levels in human myeloid cells and participates in the pathogenesis of myeloid malignancies through the disturbance of myeloid differentiation.


Asunto(s)
5-Metilcitosina/metabolismo , Citosina/análogos & derivados , Metilación de ADN/genética , Proteínas de Unión al ADN/fisiología , Eritropoyesis/genética , Células Madre Hematopoyéticas/citología , Mielopoyesis/genética , Proteínas Proto-Oncogénicas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , Línea Celular Tumoral , Linaje de la Célula , Ensayo de Unidades Formadoras de Colonias , Citosina/biosíntesis , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Dioxigenasas , Eritropoyesis/fisiología , Sangre Fetal/citología , Vectores Genéticos/genética , Granulocitos/metabolismo , Granulocitos/patología , Humanos , Lentivirus/genética , Monocitos/metabolismo , Monocitos/patología , Mutación , Mielopoyesis/fisiología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA