Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Commun ; 15(1): 7332, 2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-39187494

RESUMEN

The nucleolus, a recognized biomolecular condensate, serves as the hub for ribosome biogenesis within the cell nucleus. Its quantity and morphology are discernible indicators of cellular functional states. However, precise identification and quantification of nucleoli remain challenging without specific labeling, particularly for suspended cells, tissue-level analysis and high-throughput applications. Here we introduce a single-cell laser emitting cytometry (SLEC) for label-free nucleolus differentiation through light-matter interactions within a Fabry-Perot resonator. The separated gain medium enhances the threshold difference by 36-fold between nucleolus and its surroundings, enabling selective laser emissions at nucleolar area while maintaining lower-order mode. The laser emission image provides insights into structural inhomogeneity, temporal fluid-like dynamics, and pathological application. Lasing spectral fingerprint depicts the quantity and size of nucleoli within a single cell, showcasing the label-free flow cytometry for nucleolus. This approach holds promise for nucleolus-guided cell screening and drug evaluation, advancing the study of diseases such as cancer and neurodegenerative disorders.


Asunto(s)
Nucléolo Celular , Citometría de Flujo , Rayos Láser , Análisis de la Célula Individual , Nucléolo Celular/metabolismo , Análisis de la Célula Individual/métodos , Humanos , Citometría de Flujo/métodos , Células HeLa
2.
Front Immunol ; 11: 640, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32373120

RESUMEN

IRF-7 mediates robust production of type I IFN via MyD88 of the TLR9 pathway in plasmacytoid dendritic cells (pDCs). Previous in vitro studies using bone marrow-derived dendritic cells lacking either Irf7 or Irf3 have demonstrated that only IRF-3 is required for IFN-ß production in the TLR4 pathway. Here, we show that IRF-7 is essential for both type I IFN induction and IL-1ß responses via TLR4 in mice. Mice lacking Irf7 were defective in production of both IFN-ß and IL-1ß, an IFN-ß-induced pro-inflammatory cytokine, after LPS challenge. IFN-ß production in response to LPS was impaired in IRF-7-deficient macrophages, but not dendritic cells. Unlike pDCs, IRF-7 is activated by the TRIF-, but not MyD88-, dependent pathway via TBK-1 in macrophages after LPS stimulation. Like pDCs, resting macrophages constitutively expressed IRF-7 protein. This basal IRF-7 protein was completely abolished in either Ifnar1-/- or Stat1-/- macrophages, which corresponded with the loss of LPS-stimulated IFN-ß induction in these macrophages. These findings demonstrate that macrophage IRF-7 is critical for LPS-induced type I IFN responses, which in turn facilitate IL-1ß production in mice.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Células Dendríticas/inmunología , Endotoxemia/inmunología , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Macrófagos/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Endotoxinas/inmunología , Humanos , Factor 7 Regulador del Interferón/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Receptor de Interferón alfa y beta/genética , Factor de Transcripción STAT1/genética
3.
Immunity ; 48(2): 364-379.e8, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29466759

RESUMEN

Neutrophils are specialized innate cells that require constant replenishment from proliferative bone marrow (BM) precursors as a result of their short half-life. Although it is established that neutrophils are derived from the granulocyte-macrophage progenitor (GMP), the differentiation pathways from GMP to functional mature neutrophils are poorly defined. Using mass cytometry (CyTOF) and cell-cycle-based analysis, we identified three neutrophil subsets within the BM: a committed proliferative neutrophil precursor (preNeu) which differentiates into non-proliferating immature neutrophils and mature neutrophils. Transcriptomic profiling and functional analysis revealed that preNeu require the C/EBPε transcription factor for their generation from the GMP, and their proliferative program is substituted by a gain of migratory and effector function as they mature. preNeus expand under microbial and tumoral stress, and immature neutrophils are recruited to the periphery of tumor-bearing mice. In summary, our study identifies specialized BM granulocytic populations that ensure supply under homeostasis and stress responses.


Asunto(s)
Células de la Médula Ósea/fisiología , Neutrófilos/fisiología , Animales , Células de la Médula Ósea/inmunología , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Ratones , Neoplasias Experimentales/inmunología , Neutrófilos/inmunología
4.
Sci Rep ; 7: 46485, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28429725

RESUMEN

Epidemiological studies have indicated increased risk for breast cancer within 10 years of childbirth. Acute inflammation during mammary involution has been suggested to promote this parity-associated breast cancer. We report here that estrogen exacerbates mammary inflammation during involution. Microarray analysis shows that estrogen induces an extensive proinflammatory gene signature in the involuting mammary tissue. This is associated with estrogen-induced neutrophil infiltration. Furthermore, estrogen induces the expression of protumoral cytokines/chemokines, COX-2 and tissue-remodeling enzymes in isolated mammary neutrophils and systemic neutrophil depletion abolished estrogen-induced expression of these genes in mammary tissue. More interestingly, neutrophil depletion diminished estrogen-induced growth of ERα-negative mammary tumor 4T1 in Balb/c mice. These findings highlight a novel aspect of estrogen action that reprograms the activity of neutrophils to create a pro-tumoral microenvironment during mammary involution. This effect on the microenvironment would conceivably aggravate its known neoplastic effect on mammary epithelial cells.


Asunto(s)
Reprogramación Celular , Estrógenos/metabolismo , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Neutrófilos/metabolismo , Microambiente Tumoral , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/biosíntesis , Neutrófilos/patología
5.
Oncotarget ; 7(24): 37347-37360, 2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-26993608

RESUMEN

Leukocytes undergo frequent phenotypic changes and rapidly infiltrate peripheral and lymphoid tissues in order to carry out immune responses. The recruitment of circulating leukocytes into inflamed tissues depends on integrin-mediated tethering and rolling of these cells on the vascular endothelium, followed by transmigration into the tissues. This dynamic process of migration requires the coordination of large numbers of cytosolic and transmembrane proteins whose functional activities are typically regulated by post-translational modifications (PTMs). Our recent studies have shown that the lysine methyltransferase, Ezh2, critically regulates integrin signalling and governs the adhesion dynamics of leukocytes via direct methylation of talin, a key molecule that controls these processes by linking integrins to the actin cytoskeleton. In this review, we will discuss the various modes of leukocyte migration and examine how PTMs of cytoskeletal/adhesion associated proteins play fundamental roles in the dynamic regulation of leukocyte migration. Furthermore, we will discuss molecular details of the adhesion dynamics controlled by Ezh2-mediated talin methylation and the potential implications of this novel regulatory mechanism for leukocyte migration, immune responses, and pathogenic processes, such as allergic contact dermatitis and tumorigenesis.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Leucocitos/fisiología , Procesamiento Proteico-Postraduccional , Talina/metabolismo , Inmunidad Adaptativa/fisiología , Carcinogénesis/patología , Células Dendríticas/fisiología , Dermatitis Alérgica por Contacto/fisiopatología , Endotelio Vascular , Humanos , Inmunidad Innata/fisiología , Integrinas/metabolismo , Metilación , Transducción de Señal/fisiología
6.
Nat Immunol ; 16(5): 505-16, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25751747

RESUMEN

A cytosolic role for the histone methyltransferase Ezh2 in regulating lymphocyte activation has been suggested, but the molecular mechanisms underpinning this extranuclear function have remained unclear. Here we found that Ezh2 regulated the integrin signaling and adhesion dynamics of neutrophils and dendritic cells (DCs). Ezh2 deficiency impaired the integrin-dependent transendothelial migration of innate leukocytes and restricted disease progression in an animal model of multiple sclerosis. Direct methylation of talin, a key regulatory molecule in cell migration, by Ezh2 disrupted the binding of talin to F-actin and thereby promoted the turnover of adhesion structures. This regulatory effect was abolished by targeted disruption of the interactions of Ezh2 with the cytoskeletal-reorganization effector Vav1. Our studies reveal an unforeseen extranuclear function for Ezh2 in regulating adhesion dynamics, with implications for leukocyte migration, immune responses and potentially pathogenic processes.


Asunto(s)
Núcleo Celular/metabolismo , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Neutrófilos/inmunología , Complejo Represivo Polycomb 2/metabolismo , Talina/metabolismo , Actinas/metabolismo , Animales , Adhesión Celular/genética , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Activación de Linfocitos/genética , Metilación , Ratones , Ratones Noqueados , Complejo Represivo Polycomb 2/genética , Unión Proteica/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Talina/genética , Migración Transendotelial y Transepitelial/genética
7.
J Biol Chem ; 289(46): 31693-31707, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25266721

RESUMEN

The polycomb group protein Ezh2 is a histone methyltransferase that modifies chromatin structure to alter gene expression during embryonic development, lymphocyte activation, and tumorigenesis. The mechanism by which Ezh2 expression is regulated is not well defined. In the current study, we report that c-Rel is a critical activator of Ezh2 transcription in lymphoid cells. In activated primary murine B and T cells, plus human leukemia and multiple myeloma cell lines, recruitment of c-Rel to the first intron of the Ezh2 locus promoted Ezh2 mRNA expression. This up-regulation was abolished in activated c-Rel-deficient lymphocytes and by c-Rel knockdown in Jurkat T cells. Treatment of malignant cells with the c-Rel inhibitor pentoxifylline not only reduced c-Rel nuclear translocation and Ezh2 expression, but also enhanced their sensitivity to the Ezh2-specific drug, GSK126 through increased growth inhibition and cell death. In summary, our demonstration that c-Rel regulates Ezh2 expression in lymphocytes and malignant lymphoid cells reveals a novel transcriptional network in transformed lymphoid cells expressing high levels of Ezh2 that provides a molecular justification for combinatorial drug therapy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Complejo Represivo Polycomb 2/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular , Proteína Potenciadora del Homólogo Zeste 2 , Células HEK293 , Humanos , Indoles/química , Células Jurkat , Activación de Linfocitos , Linfocitos/citología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neoplasias/metabolismo , Pentoxifilina/química , Piridonas/química , Transcripción Genética , Regulación hacia Arriba
8.
J Biol Chem ; 289(30): 20788-801, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24878960

RESUMEN

Polycomb group protein Ezh2 is a histone H3 Lys-27 histone methyltransferase orchestrating an extensive epigenetic regulatory program. Several nervous system-specific genes are known to be repressed by Ezh2 in stem cells and derepressed during neuronal differentiation. However, the molecular mechanisms underlying this regulation remain poorly understood. Here we show that Ezh2 levels are dampened during neuronal differentiation by brain-enriched microRNA miR-124. Expression of miR-124 in a neuroblastoma cells line was sufficient to up-regulate a significant fraction of nervous system-specific Ezh2 target genes. On the other hand, naturally elevated expression of miR-124 in embryonic carcinoma cells undergoing neuronal differentiation correlated with down-regulation of Ezh2 levels. Importantly, overexpression of Ezh2 mRNA with a 3'-untranslated region (3'-UTR) lacking a functional miR-124 binding site, but not with the wild-type Ezh2 3'-UTR, hampered neuronal and promoted astrocyte-specific differentiation in P19 and embryonic mouse neural stem cells. Overall, our results uncover a molecular mechanism that allows miR-124 to balance the choice between alternative differentiation possibilities through fine-tuning the expression of a critical epigenetic regulator.


Asunto(s)
Astrocitos/metabolismo , Diferenciación Celular/fisiología , Regulación de la Expresión Génica/fisiología , MicroARNs/metabolismo , Neuronas/metabolismo , Complejo Represivo Polycomb 2/biosíntesis , Regiones no Traducidas 3'/fisiología , Animales , Astrocitos/citología , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2 , Células HEK293 , Humanos , Ratones , MicroARNs/genética , Neuronas/citología , Complejo Represivo Polycomb 2/genética
9.
J Clin Invest ; 123(12): 5009-22, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24200695

RESUMEN

Protection against deadly pathogens requires the production of high-affinity antibodies by B cells, which are generated in germinal centers (GCs). Alteration of the GC developmental program is common in many B cell malignancies. Identification of regulators of the GC response is crucial to develop targeted therapies for GC B cell dysfunctions, including lymphomas. The histone H3 lysine 27 methyltransferase enhancer of zeste homolog 2 (EZH2) is highly expressed in GC B cells and is often constitutively activated in GC-derived non-Hodgkin lymphomas (NHLs). The function of EZH2 in GC B cells remains largely unknown. Herein, we show that Ezh2 inactivation in mouse GC B cells caused profound impairment of GC responses, memory B cell formation, and humoral immunity. EZH2 protected GC B cells against activation-induced cytidine deaminase (AID) mutagenesis, facilitated cell cycle progression, and silenced plasma cell determinant and tumor suppressor B-lymphocyte-induced maturation protein 1 (BLIMP1). EZH2 inhibition in NHL cells induced BLIMP1, which impaired tumor growth. In conclusion, EZH2 sustains AID function and prevents terminal differentiation of GC B cells, which allows antibody diversification and affinity maturation. Dysregulation of the GC reaction by constitutively active EZH2 facilitates lymphomagenesis and identifies EZH2 as a possible therapeutic target in NHL and other GC-derived B cell diseases.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/enzimología , Linfoma no Hodgkin/etiología , Complejo Represivo Polycomb 2/fisiología , Animales , Apoptosis , Linfocitos B/patología , Ciclo Celular , Citidina Desaminasa/deficiencia , Citidina Desaminasa/genética , Citidina Desaminasa/fisiología , Daño del ADN , Proteína Potenciadora del Homólogo Zeste 2 , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Reordenamiento Génico de Cadena Pesada de Linfocito B , Silenciador del Gen , Centro Germinal/inmunología , Centro Germinal/patología , Inmunidad Humoral , Memoria Inmunológica , Linfoma no Hodgkin/enzimología , Linfoma no Hodgkin/genética , Linfoma no Hodgkin/patología , Linfopoyesis , Metilación , Ratones , Ratones Transgénicos , Complejo Represivo Polycomb 2/deficiencia , Complejo Represivo Polycomb 2/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Procesamiento Proteico-Postraduccional , Factores de Transcripción/fisiología
10.
J Biol Chem ; 284(33): 22005-22011, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19525229

RESUMEN

Lipid rafts are plasma membrane microdomains that are enriched in cholesterol, glycosphingolipids, and glycosylphosphatidylinositol-anchored proteins and play an important role in the signaling of ITAM-bearing lymphocyte antigen receptors. Dectin-1 is a C-type lectin receptor (CLR) that recognizes beta-glucan in the cell walls of fungi and triggers signal transduction via its cytoplasmic hemi-ITAM. However, it is not known if similar to antigen receptors, Dectin-1 would also signal via lipid rafts and if the integrity of lipid raft microdomains is important for the physiological functions mediated by Dectin-1. We demonstrate here using sucrose gradient ultracentrifugation and confocal microscopy that Dectin-1 translocates to lipid rafts upon stimulation of dendritic cells (DCs) with the yeast derivative zymosan or beta-glucan. In addition, two key signaling molecules, Syk and PLCgamma2 are also recruited to lipid rafts upon the activation of Dectin-1, suggesting that lipid raft microdomains facilitate Dectin-1 signaling. Disruption of lipid raft integrity with the synthetic drug, methyl-beta-cyclodextrin (betamD) leads to reduced intracellular Ca2+ flux and defective Syk and ERK phosphorylation in Dectin-1-activated DCs. Furthermore, betamD-treated DCs have significantly attenuated production of IL-2, IL-10, and TNFalpha upon Dectin-1 engagement, and they also exhibit impaired phagocytosis of zymosan particles. Taken together, the data indicate that Dectin-1 and perhaps also other CLRs are recruited to lipid rafts upon activation and that the integrity of lipid rafts is important for the signaling and cellular functions initiated by this class of innate receptors.


Asunto(s)
Citocinas/metabolismo , Células Dendríticas/metabolismo , Microdominios de Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Células de la Médula Ósea/citología , Calcio/metabolismo , Membrana Celular/metabolismo , Células Dendríticas/citología , Lectinas Tipo C , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal/métodos , Modelos Biológicos , Fagocitosis , Unión Proteica , Transducción de Señal
11.
Genes Dev ; 23(8): 975-85, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19390090

RESUMEN

Proliferation of pancreatic islet beta cells is an important mechanism for self-renewal and for adaptive islet expansion. Increased expression of the Ink4a/Arf locus, which encodes the cyclin-dependent kinase inhibitor p16(INK4a) and tumor suppressor p19(Arf), limits beta-cell regeneration in aging mice, but the basis of beta-cell Ink4a/Arf regulation is poorly understood. Here we show that Enhancer of zeste homolog 2 (Ezh2), a histone methyltransferase and component of a Polycomb group (PcG) protein complex, represses Ink4a/Arf in islet beta cells. Ezh2 levels decline in aging islet beta cells, and this attrition coincides with reduced histone H3 trimethylation at Ink4a/Arf, and increased levels of p16(INK4a) and p19(Arf). Conditional deletion of beta-cell Ezh2 in juvenile mice also reduced H3 trimethylation at the Ink4a/Arf locus, leading to precocious increases of p16(INK4a) and p19(Arf). These mutant mice had reduced beta-cell proliferation and mass, hypoinsulinemia, and mild diabetes, phenotypes rescued by germline deletion of Ink4a/Arf. beta-Cell destruction with streptozotocin in controls led to increased Ezh2 expression that accompanied adaptive beta-cell proliferation and re-establishment of beta-cell mass; in contrast, mutant mice treated similarly failed to regenerate beta cells, resulting in lethal diabetes. Our discovery of Ezh2-dependent beta-cell proliferation revealed unique epigenetic mechanisms underlying normal beta-cell expansion and beta-cell regenerative failure in diabetes pathogenesis.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Diabetes Mellitus/metabolismo , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Células Secretoras de Insulina/metabolismo , Envejecimiento/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Complejo Represivo Polycomb 2 , Estreptozocina/farmacología
12.
Cell ; 136(6): 1122-35, 2009 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-19303854

RESUMEN

Although in vitro studies of embryonic stem cells have identified polycomb repressor complexes (PRCs) as key regulators of differentiation, it remains unclear as to how PRC-mediated mechanisms control fates of multipotent progenitors in developing tissues. Here, we show that an essential PRC component, Ezh2, is expressed in epidermal progenitors but diminishes concomitant with embryonic differentiation and with postnatal decline in proliferative activity. We show that Ezh2 controls proliferative potential of basal progenitors by repressing the Ink4A-Ink4B locus and tempers the developmental rate of differentiation by preventing premature recruitment of AP1 transcriptional activator to the structural genes that are required for epidermal differentiation. Together, our studies reveal that PRCs control epigenetic modifications temporally and spatially in tissue-restricted stem cells. They maintain their proliferative potential and globally repressing undesirable differentiation programs while selectively establishing a specific terminal differentiation program in a stepwise fashion.


Asunto(s)
Diferenciación Celular , Células Epidérmicas , Epidermis/metabolismo , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Células Madre/metabolismo , Animales , Núcleo Celular/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Histonas/metabolismo , Humanos , Metilación , Ratones , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Proteínas Represoras/metabolismo
13.
Cell ; 121(3): 425-36, 2005 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-15882624

RESUMEN

Polycomb group protein Ezh2, one of the key regulators of development in organisms from flies to mice, exerts its epigenetic function through regulation of histone methylation. Here, we report the existence of the cytosolic Ezh2-containing methyltransferase complex and tie the function of this complex to regulation of actin polymerization in various cell types. Genetic evidence supports the essential role of cytosolic Ezh2 in actin polymerization-dependent processes such as antigen receptor signaling in T cells and PDGF-induced dorsal circular ruffle formation in fibroblasts. Revealed function of Ezh2 points to a broader usage of lysine methylation in regulation of both nuclear and extra-nuclear signaling processes.


Asunto(s)
Actinas/metabolismo , Proteína Metiltransferasas/metabolismo , Proteínas/fisiología , Transducción de Señal/fisiología , Animales , Presentación de Antígeno/fisiología , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/inmunología , Diferenciación Celular/fisiología , Proliferación Celular , Citoplasma/metabolismo , Proteínas de Unión al ADN , Proteína Potenciadora del Homólogo Zeste 2 , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , N-Metiltransferasa de Histona-Lisina , Humanos , Células Jurkat , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Neoplasias , Proteínas Nucleares , Factor de Crecimiento Derivado de Plaquetas/farmacología , Complejo Represivo Polycomb 2 , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-vav , Receptores de Antígenos de Linfocitos T/agonistas , Proteínas Represoras/metabolismo , Linfocitos T/metabolismo , Timo/inmunología , Timo/fisiología , Factores de Transcripción , Proteína de Unión al GTP cdc42/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA