Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 4943, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37582831

RESUMEN

Metabolic flexibility of muscle tissue describes the adaptive capacity to use different energy substrates according to their availability. The disruption of this ability associates with metabolic disease. Here, using a Drosophila model of systemic metabolic dysfunction triggered by yorkie-induced gut tumors, we show that the transcription factor REPTOR is an important regulator of energy metabolism in muscles. We present evidence that REPTOR is activated in muscles of adult flies with gut yorkie-tumors, where it modulates glucose metabolism. Further, in vivo studies indicate that sustained activity of REPTOR is sufficient in wildtype muscles to repress glycolysis and increase tricarboxylic acid (TCA) cycle metabolites. Consistent with the fly studies, higher levels of CREBRF, the mammalian ortholog of REPTOR, reduce glycolysis in mouse myotubes while promoting oxidative metabolism. Altogether, our results define a conserved function for REPTOR and CREBRF as key regulators of muscle energy metabolism.


Asunto(s)
Proteínas de Drosophila , Drosophila , Metabolismo Energético , Factores de Transcripción , Proteínas Supresoras de Tumor , Animales , Ratones , Ciclo del Ácido Cítrico/fisiología , Glucólisis , Músculos/metabolismo , Neoplasias/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas de Drosophila/genética , Factores de Transcripción/genética
2.
bioRxiv ; 2023 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-36712038

RESUMEN

Adipose thermogenesis involves specialized mitochondrial function that counteracts metabolic disease through dissipation of chemical energy as heat. However, inflammation present in obese adipose tissue can impair oxidative metabolism. Here, we show that PGC1α, a key governor of mitochondrial biogenesis and thermogenesis, is negatively regulated at the level of mRNA translation by the little-known RNA-binding protein RBM43. Rbm43 is expressed selectively in white adipose depots that have low thermogenic potential, and is induced by inflammatory cytokines. RBM43 suppresses mitochondrial and thermogenic gene expression in a PGC1α-dependent manner and its loss protects cells from cytokine-induced mitochondrial impairment. In mice, adipocyte-selective Rbm43 disruption increases PGC1α translation, resulting in mitochondrial biogenesis and adipose thermogenesis. These changes are accompanied by improvements in glucose homeostasis during diet-induced obesity that are independent of body weight. The action of RBM43 suggests a translational mechanism by which inflammatory signals associated with metabolic disease dampen mitochondrial function and thermogenesis.

3.
Cell Metab ; 35(3): 535-549.e7, 2023 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-36681077

RESUMEN

Proteins are secreted from cells to send information to neighboring cells or distant tissues. Because of the highly integrated nature of energy balance systems, there has been particular interest in myokines and adipokines. These are challenging to study through proteomics because serum or plasma contains highly abundant proteins that limit the detection of proteins with lower abundance. We show here that extracellular fluid (EF) from muscle and fat tissues of mice shows a different protein composition than either serum or tissues. Mass spectrometry analyses of EFs from mice with physiological perturbations, like exercise or cold exposure, allowed the quantification of many potentially novel myokines and adipokines. Using this approach, we identify prosaposin as a secreted product of muscle and fat. Prosaposin expression stimulates thermogenic gene expression and induces mitochondrial respiration in primary fat cells. These studies together illustrate the utility of EF isolation as a discovery tool for adipokines and myokines.


Asunto(s)
Líquido Extracelular , Saposinas , Ratones , Animales , Líquido Extracelular/metabolismo , Saposinas/metabolismo , Músculos/metabolismo , Tejido Adiposo/metabolismo , Adipoquinas
4.
Cell ; 185(8): 1444-1444.e1, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35427500

RESUMEN

The peroxisome proliferator-activated receptor γ coactivator-1α (Ppargc1a) gene encodes several PGC-1α isoforms that regulate mitochondrial bioenergetics and cellular adaptive processes. Expressing specific PGC-1α isoforms in mice can confer protection in different disease models. This SnapShot summarizes how regulation of Ppargc1a transcription, splicing, translation, protein stability, and activity underlies its multifaceted functions. To view this SnapShot, open or download the PDF.


Asunto(s)
Regulación de la Expresión Génica , Mitocondrias , Animales , Biología , Metabolismo Energético , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
5.
Nature ; 593(7860): 580-585, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33981039

RESUMEN

Adaptive thermogenesis has attracted much attention because of its ability to increase systemic energy expenditure and to counter obesity and diabetes1-3. Recent data have indicated that thermogenic fat cells use creatine to stimulate futile substrate cycling, dissipating chemical energy as heat4,5. This model was based on the super-stoichiometric relationship between the amount of creatine added to mitochondria and the quantity of oxygen consumed. Here we provide direct evidence for the molecular basis of this futile creatine cycling activity in mice. Thermogenic fat cells have robust phosphocreatine phosphatase activity, which is attributed to tissue-nonspecific alkaline phosphatase (TNAP). TNAP hydrolyses phosphocreatine to initiate a futile cycle of creatine dephosphorylation and phosphorylation. Unlike in other cells, TNAP in thermogenic fat cells is localized to the mitochondria, where futile creatine cycling occurs. TNAP expression is powerfully induced when mice are exposed to cold conditions, and its inhibition in isolated mitochondria leads to a loss of futile creatine cycling. In addition, genetic ablation of TNAP in adipocytes reduces whole-body energy expenditure and leads to rapid-onset obesity in mice, with no change in movement or feeding behaviour. These data illustrate the critical role of TNAP as a phosphocreatine phosphatase in the futile creatine cycle.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Mitocondrias/enzimología , Fosfocreatina/metabolismo , Termogénesis , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Animales , Frío , Metabolismo Energético , Hidrólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Mitocondrias/ultraestructura , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo
6.
Mol Cell ; 79(1): 127-139.e4, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32437639

RESUMEN

C.neoformans Dnmt5 is an unusually specific maintenance-type CpG methyltransferase (DNMT) that mediates long-term epigenome evolution. It harbors a DNMT domain and SNF2 ATPase domain. We find that the SNF2 domain couples substrate specificity to an ATPase step essential for DNA methylation. Coupling occurs independent of nucleosomes. Hemimethylated DNA preferentially stimulates ATPase activity, and mutating Dnmt5's ATP-binding pocket disproportionately reduces ATPase stimulation by hemimethylated versus unmethylated substrates. Engineered DNA substrates that stabilize a reaction intermediate by mimicking a "flipped-out" conformation of the target cytosine bypass the SNF2 domain's requirement for hemimethylation. This result implies that ATP hydrolysis by the SNF2 domain is coupled to the DNMT domain conformational changes induced by preferred substrates. These findings establish a new role for a SNF2 ATPase: controlling an adjoined enzymatic domain's substrate recognition and catalysis. We speculate that this coupling contributes to the exquisite specificity of Dnmt5 via mechanisms related to kinetic proofreading.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , ADN de Hongos/metabolismo , Proteínas Fúngicas/metabolismo , Nucleosomas/metabolismo , Adenosina Trifosfatasas/genética , Cryptococcus neoformans/genética , Cryptococcus neoformans/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN de Hongos/química , ADN de Hongos/genética , Proteínas Fúngicas/genética , Hidrólisis , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidad por Sustrato , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Cell ; 180(2): 263-277.e20, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31955845

RESUMEN

Cytosine methylation of DNA is a widespread modification of DNA that plays numerous critical roles. In the yeast Cryptococcus neoformans, CG methylation occurs in transposon-rich repeats and requires the DNA methyltransferase Dnmt5. We show that Dnmt5 displays exquisite maintenance-type specificity in vitro and in vivo and utilizes similar in vivo cofactors as the metazoan maintenance methylase Dnmt1. Remarkably, phylogenetic and functional analysis revealed that the ancestral species lost the gene for a de novo methylase, DnmtX, between 50-150 mya. We examined how methylation has persisted since the ancient loss of DnmtX. Experimental and comparative studies reveal efficient replication of methylation patterns in C. neoformans, rare stochastic methylation loss and gain events, and the action of natural selection. We propose that an epigenome has been propagated for >50 million years through a process analogous to Darwinian evolution of the genome.


Asunto(s)
Cryptococcus neoformans/genética , Metilación de ADN/genética , Metiltransferasas/genética , Evolución Biológica , Cryptococcus neoformans/metabolismo , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/fisiología , Metilasas de Modificación del ADN/genética , Elementos Transponibles de ADN/genética , Epigenómica/métodos , Evolución Molecular , Genoma/genética , Metiltransferasas/metabolismo , Filogenia
9.
Cell Metab ; 30(1): 190-200.e6, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31105043

RESUMEN

Mitochondrial abundance and function are tightly controlled during metabolic adaptation but dysregulated in pathological states such as diabetes, neurodegeneration, cancer, and kidney disease. We show here that translation of PGC1α, a key governor of mitochondrial biogenesis and oxidative metabolism, is negatively regulated by an upstream open reading frame (uORF) in the 5' untranslated region of its gene (PPARGC1A). We find that uORF-mediated translational repression is a feature of PPARGC1A orthologs from human to fly. Strikingly, whereas multiple inhibitory uORFs are broadly present in fish PPARGC1A orthologs, they are completely absent in the Atlantic bluefin tuna, an animal with exceptionally high mitochondrial content. In mice, an engineered mutation disrupting the PPARGC1A uORF increases PGC1α protein levels and oxidative metabolism and confers protection from acute kidney injury. These studies identify a translational regulatory element governing oxidative metabolism and highlight its potential contribution to the evolution of organismal mitochondrial function.


Asunto(s)
Regiones no Traducidas 5'/genética , Sistemas de Lectura Abierta/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Animales , Dípteros , Femenino , Células HEK293 , Humanos , Inmunoprecipitación , Masculino , Ratones , Mutación/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Filogenia , Procesamiento Proteico-Postraduccional/genética , Atún , Pez Cebra
10.
PLoS Pathog ; 12(12): e1006051, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27977806

RESUMEN

The opportunistic fungal pathogen Cryptococcus neoformans is a major cause of mortality in immunocompromised individuals, resulting in more than 600,000 deaths per year. Many human fungal pathogens secrete peptidases that influence virulence, but in most cases the substrate specificity and regulation of these enzymes remains poorly understood. The paucity of such information is a roadblock to our understanding of the biological functions of peptidases and whether or not these enzymes are viable therapeutic targets. We report here an unbiased analysis of secreted peptidase activity and specificity in C. neoformans using a mass spectrometry-based substrate profiling strategy and subsequent functional investigations. Our initial studies revealed that global peptidase activity and specificity are dramatically altered by environmental conditions. To uncover the substrate preferences of individual enzymes and interrogate their biological functions, we constructed and profiled a ten-member gene deletion collection of candidate secreted peptidases. Through this deletion approach, we characterized the substrate specificity of three peptidases within the context of the C. neoformans secretome, including an enzyme known to be important for fungal entry into the brain. We selected a previously uncharacterized peptidase, which we term Major aspartyl peptidase 1 (May1), for detailed study due to its substantial contribution to extracellular proteolytic activity. Based on the preference of May1 for proteolysis between hydrophobic amino acids, we screened a focused library of aspartyl peptidase inhibitors and identified four high-affinity antagonists. Finally, we tested may1Δ strains in a mouse model of C. neoformans infection and found that strains lacking this enzyme are significantly attenuated for virulence. Our study reveals the secreted peptidase activity and specificity of an important human fungal pathogen, identifies responsible enzymes through genetic tests of their function, and demonstrates how this information can guide the development of high affinity small molecule inhibitors.


Asunto(s)
Proteasas de Ácido Aspártico/metabolismo , Criptococosis/enzimología , Cryptococcus neoformans/patogenicidad , Proteínas Fúngicas/metabolismo , Animales , Cryptococcus neoformans/enzimología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Concentración de Iones de Hidrógeno , Immunoblotting , Espectrometría de Masas , Ratones , Péptido Hidrolasas/metabolismo , Proteómica , Reacción en Cadena en Tiempo Real de la Polimerasa , Virulencia , Factores de Virulencia/metabolismo
11.
Nucleic Acids Res ; 43(18): 9017-27, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26275773

RESUMEN

Despite conservation of the signal recognition particle (SRP) from bacteria to man, computational approaches have failed to identify SRP components from genomes of many lower eukaryotes, raising the possibility that they have been lost or altered in those lineages. We report purification and analysis of SRP in the human pathogen Cryptococcus neoformans, providing the first description of SRP in basidiomycetous yeast. The C. neoformans SRP RNA displays a predicted structure in which the universally conserved helix 8 contains an unprecedented stem-loop insertion. Guided by this sequence, we computationally identified 152 SRP RNAs throughout the phylum Basidiomycota. This analysis revealed additional helix 8 alterations including single and double stem-loop insertions as well as loop diminutions affecting RNA structural elements that are otherwise conserved from bacteria to man. Strikingly, these SRP RNA features in Basidiomycota are accompanied by phylum-specific alterations in the RNA-binding domain of Srp54, the SRP protein subunit that directly interacts with helix 8. Our findings reveal unexpected fungal SRP diversity and suggest coevolution of the two most conserved SRP features-SRP RNA helix 8 and Srp54-in basidiomycetes. Because members of this phylum include important human and plant pathogens, these noncanonical features provide new targets for antifungal compound development.


Asunto(s)
Cryptococcus neoformans/genética , ARN de Hongos/química , Partícula de Reconocimiento de Señal/química , Basidiomycota/genética , Proteínas Fúngicas/química , Humanos , Conformación de Ácido Nucleico , Estructura Terciaria de Proteína , ARN de Hongos/aislamiento & purificación , Partícula de Reconocimiento de Señal/aislamiento & purificación
12.
Cell ; 160(1-2): 204-18, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25533783

RESUMEN

We characterize the Polycomb system that assembles repressive subtelomeric domains of H3K27 methylation (H3K27me) in the yeast Cryptococcus neoformans. Purification of this PRC2-like protein complex reveals orthologs of animal PRC2 components as well as a chromodomain-containing subunit, Ccc1, which recognizes H3K27me. Whereas removal of either the EZH or EED ortholog eliminates H3K27me, disruption of mark recognition by Ccc1 causes H3K27me to redistribute. Strikingly, the resulting pattern of H3K27me coincides with domains of heterochromatin marked by H3K9me. Indeed, additional removal of the C. neoformans H3K9 methyltransferase Clr4 results in loss of both H3K9me and the redistributed H3K27me marks. These findings indicate that the anchoring of a chromatin-modifying complex to its product suppresses its attraction to a different chromatin type, explaining how enzymes that act on histones, which often harbor product recognition modules, may deposit distinct chromatin domains despite sharing a highly abundant and largely identical substrate-the nucleosome.


Asunto(s)
Cryptococcus neoformans/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Secuencia de Aminoácidos , Centrómero/metabolismo , Cryptococcus neoformans/genética , Heterocromatina/metabolismo , Código de Histonas , N-Metiltransferasa de Histona-Lisina/metabolismo , Datos de Secuencia Molecular , Alineación de Secuencia
13.
Trends Biochem Sci ; 39(1): 25-34, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24280023

RESUMEN

How do cells distinguish normal genes from transposons? Although much has been learned about RNAi-related RNA silencing pathways responsible for genome defense, this fundamental question remains. The literature points to several classes of mechanisms. In some cases, double-stranded RNA (dsRNA) structures produced by transposon inverted repeats or antisense integration trigger endogenous small interfering RNA (siRNA) biogenesis. In other instances, DNA features associated with transposons--such as their unusual copy number, chromosomal arrangement, and/or chromatin environment--license RNA silencing. Finally, recent studies have identified improper transcript processing events, such as stalled pre-mRNA splicing, as signals for siRNA production. Thus, the suboptimal gene expression properties of selfish elements can enable their identification by RNA silencing pathways.


Asunto(s)
Elementos Transponibles de ADN , Interferencia de ARN , ARN Interferente Pequeño/genética , Animales , Inestabilidad Genómica , Humanos , Mutagénesis Insercional , Empalme del ARN , ARN Bicatenario/genética , ARN Bicatenario/metabolismo
14.
Cell ; 152(5): 957-68, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23415457

RESUMEN

Using the yeast Cryptococcus neoformans, we describe a mechanism by which transposons are initially targeted for RNAi-mediated genome defense. We show that intron-containing mRNA precursors template siRNA synthesis. We identify a Spliceosome-Coupled And Nuclear RNAi (SCANR) complex required for siRNA synthesis and demonstrate that it physically associates with the spliceosome. We find that RNAi target transcripts are distinguished by suboptimal introns and abnormally high occupancy on spliceosomes. Functional investigations demonstrate that the stalling of mRNA precursors on spliceosomes is required for siRNA accumulation. Lariat debranching enzyme is also necessary for siRNA production, suggesting a requirement for processing of stalled splicing intermediates. We propose that recognition of mRNA precursors by the SCANR complex is in kinetic competition with splicing, thereby promoting siRNA production from transposon transcripts stalled on spliceosomes. Disparity in the strength of expression signals encoded by transposons versus host genes offers an avenue for the evolution of genome defense.


Asunto(s)
Cryptococcus neoformans/genética , Elementos Transponibles de ADN , Interferencia de ARN , Empalmosomas/metabolismo , Genoma Fúngico , Intrones , Cinética , ARN Mensajero/metabolismo , ARN Nuclear/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo
15.
RNA Biol ; 10(11): 1653-60, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24418889

RESUMEN

The ability to distinguish self from non-self nucleic acids enables eukaryotes to suppress mobile elements and maintain genome integrity. In organisms from protist to human, this function is performed by RNA silencing pathways. There have been major advances in our understanding of the RNA silencing machinery, but the mechanisms by which these pathways distinguish self from non-self remain unclear. Recent studies in the yeast C. neoformans indicate that transposon-derived transcripts encode suboptimal introns and tend to stall in spliceosomes, which promotes the biogenesis of siRNA that targets these transcripts. These findings identify gene expression signal strength as a metric by which a foreign element can be distinguished from a host gene, and reveal a new function for introns and the spliceosome in genome defense. Anticipating that these principles may apply to RNA silencing in other systems, we discuss strong hints in the literature suggesting that the spliceosome may guide small RNA biogenesis in the siRNA and piRNA pathways of plants and animals.


Asunto(s)
Plantas/genética , Interferencia de ARN , Empalme del ARN , ARN Interferente Pequeño/genética , Empalmosomas/genética , Animales , Caenorhabditis elegans/genética , Hongos/genética , Humanos , Intrones , Modelos Genéticos , ARN de Planta
16.
Proc Natl Acad Sci U S A ; 109(28): 11258-63, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-22733737

RESUMEN

Pericentromeric heterochromatin formation is mediated by repressive histone H3 lysine 9 methylation (H3K9Me) and its recognition by HP1 proteins. Intriguingly, in many organisms, RNAi is coupled to this process through poorly understood mechanisms. In Schizosaccharomyces pombe, the H3-K9 methyltransferase Clr4 and the heterochromatin protein 1 (HP1) ortholog Swi6 are critical for RNAi, whereas RNAi stimulates H3K9Me. In addition to the endoribonuclease Dcr1, RNAi in S. pombe requires two interacting protein complexes, the RITS complex, which contains an Argonaute subunit, and the RDRC complex, which contains an RNA-dependent RNA polymerase subunit. We previously identified Ers1 (essential for RNAi-dependent silencing) as an orphan protein that genetically acts in the RNAi pathway. Using recombinant proteins, we show here that Ers1 directly and specifically interacts with HP1/Swi6. Two-hybrid assays indicate that Ers1 also directly interacts with several RNAi factors. Consistent with these interactions, Ers1 associates in vivo with the RITS complex, the RDRC complex, and Dcr1, and it promotes interactions between these factors. Ers1, like Swi6, is also required for RNAi complexes to associate with pericentromeric noncoding RNAs. Overexpression of Ers1 results in a dominant-negative phenotype that can be specifically suppressed by increasing levels of the RDRC subunit Hrr1 or of Dcr1, further supporting a functional role for Ers1 in promoting the assembly of the RNAi machinery. Through the interactions described here, Ers1 may promote RNAi by tethering the corresponding enzyme complexes to HP1-coated chromatin, thereby placing them in proximity to the nascent noncoding RNA substrate.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Regulación Fúngica de la Expresión Génica , Interferencia de ARN , Proteínas Recombinantes/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Proteínas Portadoras/metabolismo , Homólogo de la Proteína Chromobox 5 , Endorribonucleasas/metabolismo , Escherichia coli/metabolismo , Silenciador del Gen , Heterocromatina/metabolismo , Fenotipo , Schizosaccharomyces/genética , Técnicas del Sistema de Dos Híbridos
17.
Genes Dev ; 24(16): 1758-71, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20675407

RESUMEN

High-resolution nucleosome occupancy maps of heterochromatic regions of wild-type and silencing-defective mutants of the fission yeast Schizosaccharomyces pombe revealed that heterochromatin induces the elimination of nucleosome-free regions (NFRs). NFRs associated with transcription initiation sites as well as those not associated with promoters are affected. We dissected the roles of the histone H3K9 methyltransferase Clr4 and the HP1 proteins Swi6 and Chp2, as well as the two catalytic activities of the SHREC histone deacetylase (HDAC)/ATPase effector complex. Strikingly, different DNA sites have distinct combinatorial requirements for these factors: Five classes of NFRs were identified that are eliminated by silencing factors through a mechanistic hierarchy governed by Clr4. The SHREC HDAC activity plays a major role in the elimination of class I-IV NFRs by antagonizing the action of RSC, a remodeling complex implicated in NFR formation. We propose that heterochromatin formation involves the deployment in several sequence-specific mechanisms to eliminate gaps between nucleosomes, thereby blocking access to the DNA.


Asunto(s)
Silenciador del Gen , Heterocromatina/genética , Nucleosomas/genética , Nucleosomas/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , Proteínas de Ciclo Celular/metabolismo , Cromosomas Fúngicos/genética , N-Metiltransferasa de Histona-Lisina , Metiltransferasas/metabolismo , Schizosaccharomyces/enzimología
18.
J Cell Biol ; 185(3): 409-22, 2009 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-19414607

RESUMEN

Erk1/2 mitogen-activated protein kinases (MAPKs) are often hyperactivated in human cancers, where they affect multiple processes, including proliferation. However, the effects of Erk1/2 loss in normal epithelial tissue, the setting of most extracellular signal-regulated kinase (Erk)-associated neoplasms, are unknown. In epidermis, loss of Erk1 or Erk2 individually has no effect, whereas simultaneous Erk1/2 depletion inhibits cell division, demonstrating that these MAPKs are necessary for normal tissue self-renewal. Growth inhibition caused by Erk1/2 loss is rescued by reintroducing Erk2, but not by activating Erk effectors that promote G1 cell cycle progression. Unlike fibroblasts, in which Erk1/2 loss decreases cyclin D1 expression and induces G1/S arrest, Erk1/2 loss in epithelial cells reduces cyclin B1 and c-Fos expression and induces G2/M arrest while disrupting a gene regulatory network centered on cyclin B1-Cdc2. Thus, the cell cycle stages at which Erk1/2 activity is required vary by cell type, with Erk1/2 functioning in epithelial cells to enable progression through G2/M.


Asunto(s)
Ciclo Celular/fisiología , División Celular/fisiología , Fase G2/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Animales , Proteína Quinasa CDC2/genética , Ciclina B/genética , Ciclina B1 , Activación Enzimática , Células Epidérmicas , Epidermis/fisiología , Células Epiteliales/citología , Células Epiteliales/enzimología , Fibroblastos/citología , Fibroblastos/enzimología , Fibroblastos/fisiología , Regulación de la Expresión Génica , Humanos , Queratinocitos/citología , Queratinocitos/fisiología , Queratinocitos/trasplante , Ratones , Ratones SCID , Neoplasias/enzimología , Neoplasias/patología , ARN Interferente Pequeño/genética , Trasplante Heterólogo
19.
Cancer Res ; 69(9): 3772-8, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19383924

RESUMEN

The Ras/Raf/Mek/Erk mitogen-activated protein kinase pathway regulates fundamental processes in normal and malignant cells, including proliferation, differentiation, and cell survival. Mutations in this pathway have been associated with carcinogenesis and developmental disorders, making Mek1 and Mek2 prime therapeutic targets. In this study, we examined the requirement for Mek1 and Mek2 in skin neoplasia using the two-step 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate (DMBA/TPA) skin carcinogenesis model. Mice lacking epidermal Mek1 protein develop fewer papillomas than both wild-type and Mek2-null mice following DMBA/TPA treatment. Mek1 knockout mice had smaller papillomas, delayed tumor onset, and half the tumor burden of wild-type mice. Loss of one Mek1 allele, however, did not affect tumor development, indicating that one Mek1 allele is sufficient for normal papilloma formation. No difference in TPA-induced hyperproliferation, inflammation, or Erk activation was observed between wild-type, conditional Mek1 knockout, and Mek2-null mice, indicating that Mek1 findings were not due to a general failure of these processes. These data show that Mek1 is important for skin tumor development and that Mek2 cannot compensate for the loss of Mek1 function in this setting.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , MAP Quinasa Quinasa 1/deficiencia , MAP Quinasa Quinasa 2/deficiencia , Neoplasias Cutáneas/enzimología , 9,10-Dimetil-1,2-benzantraceno , Animales , Diferenciación Celular/fisiología , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Genotipo , Hiperplasia , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 2/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Oncogénica v-akt/metabolismo , Papiloma/inducido químicamente , Papiloma/enzimología , Papiloma/genética , Fosforilación , Piel/efectos de los fármacos , Piel/enzimología , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Acetato de Tetradecanoilforbol
20.
Dev Cell ; 12(4): 615-29, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17419998

RESUMEN

The p42/p44 mitogen-activated protein kinase (MAPK) cascade includes Ras, Raf, Mek, and Erk MAPK. To determine the effect of a full knockout at a single level of this signaling pathway in mammals, and to investigate functional redundancy between Mek1 and Mek2, we disrupted these genes in murine and human epidermis. Loss of either protein alone produced no phenotype, whereas combined Mek1/2 deletion in development or adulthood abolished Erk1/2 phosphorylation and led to hypoproliferation, apoptosis, skin barrier defects, and death. Conversely, a single copy of either allele was sufficient for normal development. Combined Mek1/2 loss also abolished Raf-induced hyperproliferation. Human tissue deficient in either Mek isoform was normal, whereas loss of both proteins led to hypoplasia, which was rescued by active Erk2 expression. These data indicate that Mek1/2 are functionally redundant in the epidermis, where they act as a linear relay in the MAPK pathway to mediate development and homeostasis.


Asunto(s)
Homeostasis , Hiperplasia/metabolismo , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Muerte , Epidermis/embriología , Epidermis/metabolismo , Epidermis/patología , Humanos , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 2/genética , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Piel/embriología , Piel/metabolismo , Piel/patología , Lengua/embriología , Lengua/metabolismo , Lengua/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...