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.
Nature ; 568(7753): 557-560, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30971822

RESUMEN

The cell cycle is a tightly regulated process that is controlled by the conserved cyclin-dependent kinase (CDK)-cyclin protein complex1. However, control of the G0-to-G1 transition is not completely understood. Here we demonstrate that p38 MAPK gamma (p38γ) acts as a CDK-like kinase and thus cooperates with CDKs, regulating entry into the cell cycle. p38γ shares high sequence homology, inhibition sensitivity and substrate specificity with CDK family members. In mouse hepatocytes, p38γ induces proliferation after partial hepatectomy by promoting the phosphorylation of retinoblastoma tumour suppressor protein at known CDK target residues. Lack of p38γ or treatment with the p38γ inhibitor pirfenidone protects against the chemically induced formation of liver tumours. Furthermore, biopsies of human hepatocellular carcinoma show high expression of p38γ, suggesting that p38γ could be a therapeutic target in the treatment of this disease.


Asunto(s)
Carcinogénesis/patología , Ciclo Celular , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Hígado/enzimología , Hígado/patología , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Anciano , Animales , Carcinogénesis/efectos de los fármacos , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Femenino , Hepatocitos/citología , Hepatocitos/patología , Humanos , Hígado/cirugía , Neoplasias Hepáticas/inducido químicamente , Masculino , Ratones , Persona de Mediana Edad , Proteína Quinasa 12 Activada por Mitógenos/antagonistas & inhibidores , Fosforilación , Piridonas/farmacología , Proteína de Retinoblastoma/química , Proteína de Retinoblastoma/metabolismo , Homología de Secuencia , Especificidad por Sustrato
2.
Immunity ; 41(3): 465-477, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25200712

RESUMEN

Dendritic cells (DCs), monocytes, and macrophages are leukocytes with critical roles in immunity and tolerance. The DC network is evolutionarily conserved; the homologs of human tissue CD141(hi)XCR1⁺ CLEC9A⁺ DCs and CD1c⁺ DCs are murine CD103⁺ DCs and CD64⁻ CD11b⁺ DCs. In addition, human tissues also contain CD14⁺ cells, currently designated as DCs, with an as-yet unknown murine counterpart. Here we have demonstrated that human dermal CD14⁺ cells are a tissue-resident population of monocyte-derived macrophages with a short half-life of <6 days. The decline and reconstitution kinetics of human blood CD14⁺ monocytes and dermal CD14⁺ cells in vivo supported their precursor-progeny relationship. The murine homologs of human dermal CD14⁺ cells are CD11b⁺ CD64⁺ monocyte-derived macrophages. Human and mouse monocytes and macrophages were defined by highly conserved gene transcripts, which were distinct from DCs. The demonstration of monocyte-derived macrophages in the steady state in human tissue supports a conserved organization of human and mouse mononuclear phagocyte system.


Asunto(s)
Receptores de Lipopolisacáridos/metabolismo , Macrófagos/inmunología , Piel/inmunología , Animales , Antígeno CD11b/biosíntesis , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Movimiento Celular/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Femenino , Humanos , Memoria Inmunológica/inmunología , Ratones , Ratones Transgénicos , Receptores de IgG/biosíntesis , Piel/citología , Linfocitos T/inmunología
4.
Genome Res ; 24(10): 1676-85, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25015382

RESUMEN

Global production of chickens has trebled in the past two decades and they are now the most important source of dietary animal protein worldwide. Chickens are subject to many infectious diseases that reduce their performance and productivity. Coccidiosis, caused by apicomplexan protozoa of the genus Eimeria, is one of the most important poultry diseases. Understanding the biology of Eimeria parasites underpins development of new drugs and vaccines needed to improve global food security. We have produced annotated genome sequences of all seven species of Eimeria that infect domestic chickens, which reveal the full extent of previously described repeat-rich and repeat-poor regions and show that these parasites possess the most repeat-rich proteomes ever described. Furthermore, while no other apicomplexan has been found to possess retrotransposons, Eimeria is home to a family of chromoviruses. Analysis of Eimeria genes involved in basic biology and host-parasite interaction highlights adaptations to a relatively simple developmental life cycle and a complex array of co-expressed surface proteins involved in host cell binding.


Asunto(s)
Eimeria/genética , Genoma de Protozoos , Proteínas Protozoarias/genética , Animales , Línea Celular , Pollos , Mapeo Cromosómico , Coccidiosis/parasitología , Coccidiosis/veterinaria , Eimeria/clasificación , Perfilación de la Expresión Génica , Filogenia , Enfermedades de las Aves de Corral/parasitología , Proteoma , Sintenía
5.
Nature ; 464(7287): 367-73, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-20237561

RESUMEN

Fusarium species are among the most important phytopathogenic and toxigenic fungi. To understand the molecular underpinnings of pathogenicity in the genus Fusarium, we compared the genomes of three phenotypically diverse species: Fusarium graminearum, Fusarium verticillioides and Fusarium oxysporum f. sp. lycopersici. Our analysis revealed lineage-specific (LS) genomic regions in F. oxysporum that include four entire chromosomes and account for more than one-quarter of the genome. LS regions are rich in transposons and genes with distinct evolutionary profiles but related to pathogenicity, indicative of horizontal acquisition. Experimentally, we demonstrate the transfer of two LS chromosomes between strains of F. oxysporum, converting a non-pathogenic strain into a pathogen. Transfer of LS chromosomes between otherwise genetically isolated strains explains the polyphyletic origin of host specificity and the emergence of new pathogenic lineages in F. oxysporum. These findings put the evolution of fungal pathogenicity into a new perspective.


Asunto(s)
Cromosomas Fúngicos/genética , Fusarium/genética , Fusarium/patogenicidad , Genoma Fúngico/genética , Genómica , Evolución Molecular , Fusarium/clasificación , Interacciones Huésped-Parásitos/genética , Familia de Multigenes/genética , Fenotipo , Filogenia , Proteoma/genética , Análisis de Secuencia de ADN , Sintenía/genética , Virulencia/genética
6.
Nature ; 465(7298): 617-21, 2010 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-20520714

RESUMEN

Brown algae (Phaeophyceae) are complex photosynthetic organisms with a very different evolutionary history to green plants, to which they are only distantly related. These seaweeds are the dominant species in rocky coastal ecosystems and they exhibit many interesting adaptations to these, often harsh, environments. Brown algae are also one of only a small number of eukaryotic lineages that have evolved complex multicellularity (Fig. 1). We report the 214 million base pair (Mbp) genome sequence of the filamentous seaweed Ectocarpus siliculosus (Dillwyn) Lyngbye, a model organism for brown algae, closely related to the kelps (Fig. 1). Genome features such as the presence of an extended set of light-harvesting and pigment biosynthesis genes and new metabolic processes such as halide metabolism help explain the ability of this organism to cope with the highly variable tidal environment. The evolution of multicellularity in this lineage is correlated with the presence of a rich array of signal transduction genes. Of particular interest is the presence of a family of receptor kinases, as the independent evolution of related molecules has been linked with the emergence of multicellularity in both the animal and green plant lineages. The Ectocarpus genome sequence represents an important step towards developing this organism as a model species, providing the possibility to combine genomic and genetic approaches to explore these and other aspects of brown algal biology further.


Asunto(s)
Proteínas Algáceas/genética , Evolución Biológica , Genoma/genética , Phaeophyceae/citología , Phaeophyceae/genética , Animales , Eucariontes , Evolución Molecular , Datos de Secuencia Molecular , Phaeophyceae/metabolismo , Filogenia , Pigmentos Biológicos/biosíntesis , Transducción de Señal/genética
7.
Nucleic Acids Res ; 42(1): e6, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24137002

RESUMEN

Transcription factors (TFs) combine with co-factors to form transcriptional regulatory modules (TRMs) that regulate gene expression programs with spatiotemporal specificity. Here we present a novel and generic method (rTRM) for the reconstruction of TRMs that integrates genomic information from TF binding, cell type-specific gene expression and protein-protein interactions. rTRM was applied to reconstruct the TRMs specific for embryonic stem cells (ESC) and hematopoietic stem cells (HSC), neural progenitor cells, trophoblast stem cells and distinct types of terminally differentiated CD4(+) T cells. The ESC and HSC TRM predictions were highly precise, yielding 77 and 96 proteins, of which ∼75% have been independently shown to be involved in the regulation of these cell types. Furthermore, rTRM successfully identified a large number of bridging proteins with known roles in ESCs and HSCs, which could not have been identified using genomic approaches alone, as they lack the ability to bind specific DNA sequences. This highlights the advantage of rTRM over other methods that ignore PPI information, as proteins need to interact with other proteins to form complexes and perform specific functions. The prediction and experimental validation of the co-factors that endow master regulatory TFs with the capacity to select specific genomic sites, modulate the local epigenetic profile and integrate multiple signals will provide important mechanistic insights not only into how such TFs operate, but also into abnormal transcriptional states leading to disease.


Asunto(s)
Redes Reguladoras de Genes , Mapeo de Interacción de Proteínas/métodos , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Células Madre Embrionarias/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratas , Transcripción Genética
8.
Crit Rev Biochem Mol Biol ; 48(5): 430-45, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23879520

RESUMEN

Our understanding of the fundamental regulatory roles that tyrosine phosphatases play within cells has advanced significantly in the last two decades. Out-dated ideas that tyrosine phosphatases acts solely as the "off" switch counterbalancing the action of tyrosine kinases has proved to be flawed. PTP1B is the most characterized of all the tyrosine phosphatases and it acts as a critical negative and positive regulator of numerous signaling cascades. PTP1B's direct regulation of the insulin and the leptin receptors makes it an ideal therapeutic target for type II diabetes and obesity. Moreover, the last decade has also seen several reports establishing PTP1B as key player in cancer serving as both tumor suppressor and tumor promoter depending on the cellular context. Despite many key advances in these fields one largely ignored area is what role PTP1B may play in the modulation of immune signaling. The important recognition that PTP1B is a major negative regulator of Janus kinase - signal transducer and activator of transcription (JAK-STAT) signaling throughout evolution places it as a key link between metabolic diseases and inflammation, as well as a unique regulator between immune response and cancer. This review looks at the emergence of PTP1B through evolution, and then explore at the cell and systemic levels how it is controlled physiologically. The second half of the review will focus on the role(s) PTP1B can play in disease and in particular its involvement in metabolic syndromes and cancer. Finally we will briefly examine several novel directions in the development of PTP1B pharmacological inhibitors.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Animales , Enfermedad , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Especificidad por Sustrato/efectos de los fármacos
9.
Methods ; 65(2): 156-64, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23911837

RESUMEN

Reversible tyrosine phosphorylation is a fundamental signaling mechanism controlling a diversity of cellular processes. Whereas protein tyrosine kinases have long been implicated in many diseases, aberrant protein tyrosine phosphatase (PTP) activity is also increasingly being associated with a wide spectrum of conditions. PTPs are now regarded as key regulators of biochemical processes instead of simple "off" switches operating in tyrosine kinase signaling pathways. Despite the central importance that PTPs play in the cell's biochemistry, the tyrosine phosphatomes of most species remain uncharted. Here we present a highly sensitive and specific sequence-based method for the automatic classification of PTPs. As proof of principle we re-annotated the human tyrosine phosphatome, and discovered four new PTP genes that had not been reported before. Our method and the predicted tyrosine phosphatomes of 65 eukaryotic genomes are accessible online through the user-friendly PTP-central resource (http://www.PTP-central.org/), where users can also submit their own sequences for prediction. PTP-central is a comprehensive and continually developing resource that currently integrates the predicted tyrosine phosphatomes with structural data and genetic association disease studies, as well as homology relationships. PTP-central thus fills an important void for the systematic study of PTPs, both in model organisms and from an evolutionary perspective.


Asunto(s)
Bases de Datos como Asunto , Genoma/genética , Proteínas Tirosina Fosfatasas/genética , Animales , Estudios de Asociación Genética , Humanos , Datos de Secuencia Molecular , Proteínas Tirosina Fosfatasas/química
10.
Methods ; 65(2): 239-46, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23994241

RESUMEN

For years, the two main isoforms of PTPN2 have been an interesting yet academic topic of debate for researchers working on this phosphatase. In recent years, several studies were published in which these isoforms were attributed specific functions. Most importantly, differences in their stoichiometry have been reported to be associated with certain diseases such as inflammatory bowel diseases (IBDs). Hence, understanding the evolutionary ontogeny of the main transcripts and the physiological consequences of their expression have now become clinically relevant issues. Herein we describe the genomic controls placed upon PTPN2, the identified splice variants, the encoded PTPN2 proteins, and both the known and putative post-translational modifications that have been reported. Moreover, we examine the expression of PTPN2 isoforms in specific tissues as well as in a disease setting. PTPN2 is an important negative regulator of inflammation. Therefore, the following protocols are effective approaches for its adequate monitoring in inflammatory diseases' progression and outcome.


Asunto(s)
Biomarcadores/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Animales , Biomarcadores/análisis , Dominio Catalítico , Enfermedad de Crohn/diagnóstico , Evolución Molecular , Humanos , Ratones , Filogenia , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/química , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , ARN Mensajero/metabolismo , Alineación de Secuencia , Linfocitos T/metabolismo
11.
Nucleic Acids Res ; 41(4): 2155-70, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23295670

RESUMEN

Transcription factors (TFs) regulate gene expression by binding to short DNA sequence motifs, yet their binding specificities alone cannot explain how certain TFs drive a diversity of biological processes. In order to investigate the factors that control the functions of the pleiotropic TF STAT3, we studied its genome-wide binding patterns in four different cell types: embryonic stem cells, CD4(+) T cells, macrophages and AtT-20 cells. We describe for the first time two distinct modes of STAT3 binding. First, a small cell type-independent mode represented by a set of 35 evolutionarily conserved STAT3-binding sites that collectively regulate STAT3's own functions and cell growth. We show that STAT3 is recruited to sites with E2F1 already pre-bound before STAT3 activation. Second, a series of different transcriptional regulatory modules (TRMs) assemble around STAT3 to drive distinct transcriptional programs in the four cell types. These modules recognize cell type-specific binding sites and are associated with factors particular to each cell type. Our study illustrates the versatility of STAT3 to regulate both universal- and cell type-specific functions by means of distinct TRMs, a mechanism that might be common to other pleiotropic TFs.


Asunto(s)
Regulación de la Expresión Génica , Factor de Transcripción STAT3/metabolismo , Transcripción Genética , Animales , Sitios de Unión , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , ADN/química , ADN/metabolismo , Células Madre Embrionarias/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/química
12.
Mol Biol Evol ; 30(5): 1172-87, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23393154

RESUMEN

Reversible protein ubiquitination regulates virtually all known cellular activities. Here, we present a quantitatively evaluated and broadly applicable method to predict eukaryotic ubiquitinating enzymes (UBE) and deubiquitinating enzymes (DUB) and its application to 50 distinct genomes belonging to four of the five major phylogenetic supergroups of eukaryotes: unikonts (including metazoans, fungi, choanozoa, and amoebozoa), excavates, chromalveolates, and plants. Our method relies on a collection of profile hidden Markov models, and we demonstrate its superior performance (coverage and classification accuracy >99%) by identifying approximately 25% and approximately 35% additional UBE and DUB genes in yeast and human, which had not been reported before. In yeast, we predict 85 UBE and 24 DUB genes, for 814 UBE and 107 DUB genes in the human genome. Most UBE and DUB families are present in all eukaryotic lineages, with plants and animals harboring massively enlarged repertoires of ubiquitin ligases. Unicellular organisms, on the other hand, typically harbor less than 300 UBEs and less than 40 DUBs per genome. Ninety-one UBE/DUB genes are orthologous across all four eukaryotic supergroups, and these likely represent a primordial core of enzymes of the ubiquitination system probably dating back to the first eukaryotes approximately 2 billion years ago. Our genome-wide predictions are available through the Database of Ubiquitinating and Deubiquitinating Enzymes (www.DUDE-db.org), where users can also perform advanced sequence and phylogenetic analyses and submit their own predictions.


Asunto(s)
Genoma Humano/genética , Humanos , Cadenas de Markov , Ubiquitinación/genética , Ubiquitinación/fisiología
13.
Bioinformatics ; 29(13): i80-8, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23813012

RESUMEN

MOTIVATION: Combinatorial interactions of transcription factors with cis-regulatory elements control the dynamic progression through successive cellular states and thus underpin all metazoan development. The construction of network models of cis-regulatory elements, therefore, has the potential to generate fundamental insights into cellular fate and differentiation. Haematopoiesis has long served as a model system to study mammalian differentiation, yet modelling based on experimentally informed cis-regulatory interactions has so far been restricted to pairs of interacting factors. Here, we have generated a Boolean network model based on detailed cis-regulatory functional data connecting 11 haematopoietic stem/progenitor cell (HSPC) regulator genes. RESULTS: Despite its apparent simplicity, the model exhibits surprisingly complex behaviour that we charted using strongly connected components and shortest-path analysis in its Boolean state space. This analysis of our model predicts that HSPCs display heterogeneous expression patterns and possess many intermediate states that can act as 'stepping stones' for the HSPC to achieve a final differentiated state. Importantly, an external perturbation or 'trigger' is required to exit the stem cell state, with distinct triggers characterizing maturation into the various different lineages. By focusing on intermediate states occurring during erythrocyte differentiation, from our model we predicted a novel negative regulation of Fli1 by Gata1, which we confirmed experimentally thus validating our model. In conclusion, we demonstrate that an advanced mammalian regulatory network model based on experimentally validated cis-regulatory interactions has allowed us to make novel, experimentally testable hypotheses about transcriptional mechanisms that control differentiation of mammalian stem cells. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Redes Reguladoras de Genes , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Modelos Genéticos , Animales , Línea Celular , Eritrocitos/citología , Genes Reguladores , Células Madre Hematopoyéticas/citología , Ratones , Factores de Transcripción/metabolismo
14.
Blood ; 119(13): e110-9, 2012 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-22323479

RESUMEN

Inflammation is a powerful response of the immune system against invading pathogens, and must be cancelled when unneeded or otherwise death inevitably follows. In macrophages, the anti-inflammatory response (AIR) is driven by STAT3 upon IL-10 signaling. The role of STAT3 is to stimulate the expression of specific genes that in-turn suppress the transcription of proinflammatory genes. Here we describe a systematic approach to identify the elusive STAT3-controlled effectors of the AIR. In vivo STAT3-binding sites were identified by ChIP-seq, coupled to expression analysis by RNA-seq, both in resting and IL-10-treated peritoneal macrophages. We report the genomic targets of STAT3 and show that STAT3's transcriptional program during the AIR is highly specific to IL-10-stimulated macrophages, that STAT3 is a positive transcriptional regulator, and we predict severalputative AIR factors that merit further investigation. This is the first in-depth study of the AIR by next-generation sequencing and provides an unprecedented degree of detail into this fundamental physiologic response.


Asunto(s)
Antiinflamatorios/metabolismo , Mapeo Cromosómico , Inflamación/genética , Macrófagos Peritoneales/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Sitios de Unión/genética , Biomarcadores Farmacológicos/análisis , Biomarcadores Farmacológicos/metabolismo , Mapeo Cromosómico/métodos , Análisis por Conglomerados , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Inmunidad Celular/fisiología , Inflamación/metabolismo , Interleucina-10/farmacología , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Transcriptoma/genética
15.
Nucleic Acids Res ; 40(10): e77, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22328731

RESUMEN

The chemical modification of histones at specific DNA regulatory elements is linked to the activation, inactivation and poising of genes. A number of tools exist to predict enhancers from chromatin modification maps, but their practical application is limited because they either (i) consider a smaller number of marks than those necessary to define the various enhancer classes or (ii) work with an excessive number of marks, which is experimentally unviable. We have developed a method for chromatin state detection using support vector machines in combination with genetic algorithm optimization, called ChromaGenSVM. ChromaGenSVM selects optimum combinations of specific histone epigenetic marks to predict enhancers. In an independent test, ChromaGenSVM recovered 88% of the experimentally supported enhancers in the pilot ENCODE region of interferon gamma-treated HeLa cells. Furthermore, ChromaGenSVM successfully combined the profiles of only five distinct methylation and acetylation marks from ChIP-seq libraries done in human CD4(+) T cells to predict ∼21,000 experimentally supported enhancers within 1.0 kb regions and with a precision of ∼90%, thereby improving previous predictions on the same dataset by 21%. The combined results indicate that ChromaGenSVM comfortably outperforms previously published methods and that enhancers are best predicted by specific combinations of histone methylation and acetylation marks.


Asunto(s)
Elementos de Facilitación Genéticos , Epigénesis Genética , Máquina de Vectores de Soporte , Linfocitos T CD4-Positivos/metabolismo , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Genoma Humano , Histonas/metabolismo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos
16.
Genomics ; 100(5): 303-13, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22884873

RESUMEN

Despite the routine application of RNA-seq technology to profile cellular transcriptomes and report novel splice variants, the identification and validation of new transcripts remain underexplored. We prepared two RNA-seq libraries from resting and T cell receptor-stimulated mouse CD4(+) T cells. Transcripts unknown to Ensembl represent as much as 5% of the assembled transcripts and are robustly expressed but do not show the same degree of evolutionary conservation or exon distribution of known transcripts, or of novel splice isoforms. Here we present a straightforward and generally applicable computational/experimental workflow that we apply to characterise and experimentally validate 23 mouse transcripts from the RNA-seq libraries that were uncharacterised by Ensembl. Of these, 7 are not supported by any transcript database and therefore are likely to encode new messages. Furthermore, we also report the fast up-regulation of important regulatory molecules only 4 h post-stimulation of the T cell receptor, which calls for a more detailed investigation into early CD4(+) T cell activation mechanisms.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Evolución Molecular , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/inmunología , Análisis de Secuencia de ARN/métodos , Animales , Biología Computacional/métodos , Biblioteca de Genes , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
Mol Microbiol ; 79(1): 205-21, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21166904

RESUMEN

Aurora kinases compose a family of conserved Ser/Thr protein kinases playing essential roles in eukaryotic cell division. To date, Aurora homologues remain uncharacterized in the protozoan phylum Apicomplexa. In malaria parasites, the characterization of Aurora kinases may help understand the cell cycle control during erythrocytic schizogony where asynchronous nuclear divisions occur. In this study, we revisited the kinome of Plasmodium falciparum and identified three Aurora-related kinases, Pfark-1, -2, -3. Among these, Pfark-1 is highly conserved in malaria parasites and also appears to be conserved across Apicomplexa. By tagging the endogenous Pfark-1 gene with the green fluorescent protein (GFP) in live parasites, we show that the Pfark-1-GFP protein forms paired dots associated with only a subset of nuclei within individual schizonts. Immunofluorescence analysis using an anti-α-tubulin antibody strongly suggests a recruitment of Pfark-1 at duplicated spindle pole bodies at the entry of the M phase of the cell cycle. Unsuccessful attempts at disrupting the Pfark-1 gene with a knockout construct further indicate that Pfark-1 is required for parasite growth in red blood cells. Our study provides new insights into the cell cycle control of malaria parasites and reports the importance of Aurora kinases as potential targets for new antimalarials.


Asunto(s)
Eritrocitos/parasitología , Plasmodium falciparum/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Protozoarias/metabolismo , Huso Acromático/metabolismo , Aurora Quinasas , Secuencia Conservada , Eliminación de Gen , Genes Esenciales , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Filogenia , Plasmodium falciparum/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Protozoarias/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido
18.
Blood ; 116(16): 3013-22, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20647567

RESUMEN

Glucocorticoids play a critical role in the therapy of lymphoid malignancies, including pediatric acute lymphoblastic leukemia (ALL), although the mechanisms underlying cellular resistance remain unclear. We report glucocorticoid resistance attributable to epigenetic silencing of the BIM gene in pediatric ALL biopsies and xenografts established in immune-deficient mice from direct patient explants as well as a therapeutic approach to reverse resistance in vivo. Glucocorticoid resistance in ALL xenografts was consistently associated with failure to up-regulate BIM expression after dexamethasone exposure despite confirmation of a functional glucocorticoid receptor. Although a comprehensive assessment of BIM CpG island methylation revealed no consistent changes, glucocorticoid resistance in xenografts and patient biopsies significantly correlated with decreased histone H3 acetylation. Moreover, the histone deacetylase inhibitor vorinostat relieved BIM repression and exerted synergistic antileukemic efficacy with dexamethasone in vitro and in vivo. These findings provide a novel therapeutic strategy to reverse glucocorticoid resistance and improve outcome for high-risk pediatric ALL.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas Reguladoras de la Apoptosis/genética , Resistencia a Antineoplásicos , Silenciador del Gen , Glucocorticoides/uso terapéutico , Inhibidores de Histona Desacetilasas/uso terapéutico , Proteínas de la Membrana/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Proto-Oncogénicas/genética , Animales , Antineoplásicos/farmacología , Antineoplásicos Hormonales/farmacología , Antineoplásicos Hormonales/uso terapéutico , Proteína 11 Similar a Bcl2 , Niño , Dexametasona/farmacología , Dexametasona/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Sitios Genéticos , Glucocorticoides/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Ratones , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Vorinostat
19.
PLoS Genet ; 5(7): e1000549, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19578406

RESUMEN

Rhizopus oryzae is the primary cause of mucormycosis, an emerging, life-threatening infection characterized by rapid angioinvasive growth with an overall mortality rate that exceeds 50%. As a representative of the paraphyletic basal group of the fungal kingdom called "zygomycetes," R. oryzae is also used as a model to study fungal evolution. Here we report the genome sequence of R. oryzae strain 99-880, isolated from a fatal case of mucormycosis. The highly repetitive 45.3 Mb genome assembly contains abundant transposable elements (TEs), comprising approximately 20% of the genome. We predicted 13,895 protein-coding genes not overlapping TEs, many of which are paralogous gene pairs. The order and genomic arrangement of the duplicated gene pairs and their common phylogenetic origin provide evidence for an ancestral whole-genome duplication (WGD) event. The WGD resulted in the duplication of nearly all subunits of the protein complexes associated with respiratory electron transport chains, the V-ATPase, and the ubiquitin-proteasome systems. The WGD, together with recent gene duplications, resulted in the expansion of multiple gene families related to cell growth and signal transduction, as well as secreted aspartic protease and subtilase protein families, which are known fungal virulence factors. The duplication of the ergosterol biosynthetic pathway, especially the major azole target, lanosterol 14alpha-demethylase (ERG11), could contribute to the variable responses of R. oryzae to different azole drugs, including voriconazole and posaconazole. Expanded families of cell-wall synthesis enzymes, essential for fungal cell integrity but absent in mammalian hosts, reveal potential targets for novel and R. oryzae-specific diagnostic and therapeutic treatments.


Asunto(s)
Duplicación de Gen , Genoma Fúngico , Genómica , Mucormicosis/microbiología , Rhizopus/genética , Elementos Transponibles de ADN , Proteínas Fúngicas/genética , Hongos/clasificación , Hongos/genética , Humanos , Filogenia , Rhizopus/química , Rhizopus/clasificación , Rhizopus/aislamiento & purificación
20.
Blood ; 114(9): 1820-30, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19571317

RESUMEN

The discovery of JAK2V617F as an acquired mutation in the majority of patients with myeloproliferative disorders (MPDs) and the key role of the JAK2-STAT5 signaling cascade in normal hematopoiesis has focused attention on the downstream transcriptional targets of STAT5. Despite evidence of its vital role in normal erythropoiesis and its ability to recapitulate many of the features of myeloid malignancies, including the MPDs, few functionally validated targets of STAT5 have been described. Here we used a combination of comparative genomics and chromatin immunoprecipitation assays to identify ID1 as a novel target of the JAK2-STAT5 signaling axis in erythroid cells. STAT5 binds and transactivates a downstream enhancer of ID1, and ID1 expression levels correlate with the JAK2V617F mutation in both retrovirally transfected fetal liver cells and polycythemia vera patients. Knockdown and overexpression studies in a well-characterized erythroid differentiation assay from primary murine fetal liver cells demonstrated a survival-promoting action of ID1. This hitherto unrecognized function implicates ID1 in the expansion of erythroblasts during terminal differentiation and suggests that ID1 plays an important role in the pathogenesis of polycythemia vera. Furthermore, our findings contribute to an increasing body of evidence implicating ID proteins in a wider range of cellular functions than initially appreciated.


Asunto(s)
Células Eritroides/citología , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Janus Quinasa 2/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Separación Celular , Citometría de Flujo , Humanos , Janus Quinasa 2/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Retroviridae/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA