Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
J Biol Chem ; 298(10): 102433, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36041630

RESUMEN

TFIIH is an evolutionarily conserved complex that plays central roles in both RNA polymerase II (pol II) transcription and DNA repair. As an integral component of the pol II preinitiation complex, TFIIH regulates pol II enzyme activity in numerous ways. The TFIIH subunit XPB/Ssl2 is an ATP-dependent DNA translocase that stimulates promoter opening prior to transcription initiation. Crosslinking-mass spectrometry and cryo-EM results have shown a conserved interaction network involving XPB/Ssl2 and the C-terminal Hub region of the TFIIH p52/Tfb2 subunit, but the functional significance of specific residues is unclear. Here, we systematically mutagenized the HubA region of Tfb2 and screened for growth phenotypes in a TFB6 deletion background in Saccharomyces cerevisiae. We identified six lethal and 12 conditional mutants. Slow growth phenotypes of all but three conditional mutants were relieved in the presence of TFB6, thus identifying a functional interaction between Tfb2 HubA mutants and Tfb6, a protein that dissociates Ssl2 from TFIIH. Our biochemical analysis of Tfb2 mutants with severe growth phenotypes revealed defects in Ssl2 association, with similar results in human cells. Further characterization of these tfb2 mutant cells revealed defects in GAL gene induction, and reduced occupancy of TFIIH and pol II at GAL gene promoters, suggesting that functionally competent TFIIH is required for proper pol II recruitment to preinitiation complexes in vivo. Consistent with recent structural models of TFIIH, our results identify key residues in the p52/Tfb2 HubA domain that are required for stable incorporation of XPB/Ssl2 into TFIIH and for pol II transcription.


Asunto(s)
ADN Helicasas , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Factor de Transcripción TFIIH , Humanos , ADN Helicasas/genética , ADN Helicasas/metabolismo , Reparación del ADN , Mutagénesis , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Factor de Transcripción TFIIH/genética , Factor de Transcripción TFIIH/metabolismo , Transcripción Genética
2.
Curr Opin Hematol ; 28(3): 150-157, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33714970

RESUMEN

PURPOSE OF REVIEW: Erythropoiesis is a hierarchical process by which hematopoietic stem cells give rise to red blood cells through gradual cell fate restriction and maturation. Deciphering this process requires the establishment of dynamic gene regulatory networks (GRNs) that predict the response of hematopoietic cells to signals from the environment. Although GRNs have historically been derived from transcriptomic data, recent proteomic studies have revealed a major role for posttranscriptional mechanisms in regulating gene expression during erythropoiesis. These new findings highlight the need to integrate proteomic data into GRNs for a refined understanding of erythropoiesis. RECENT FINDINGS: Here, we review recent proteomic studies that have furthered our understanding of erythropoiesis with a focus on quantitative mass spectrometry approaches to measure the abundance of transcription factors and cofactors during differentiation. Furthermore, we highlight challenges that remain in integrating transcriptomic, proteomic, and other omics data into a predictive model of erythropoiesis, and discuss the future prospect of single-cell proteomics. SUMMARY: Recent proteomic studies have considerably expanded our knowledge of erythropoiesis beyond the traditional transcriptomic-centric perspective. These findings have both opened up new avenues of research to increase our understanding of erythroid differentiation, while at the same time presenting new challenges in integrating multiple layers of information into a comprehensive gene regulatory model.


Asunto(s)
Eritropoyesis/fisiología , Perfilación de la Expresión Génica , Proteoma , Proteómica , Transcriptoma , Animales , Biomarcadores , Diferenciación Celular/genética , Perfilación de la Expresión Génica/métodos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Proteómica/métodos , Análisis de la Célula Individual
3.
mSphere ; 6(1)2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33627513

RESUMEN

Unlike most other eukaryotes, Leishmania and other trypanosomatid protozoa have largely eschewed transcriptional control of gene expression, relying instead on posttranscriptional regulation of mRNAs derived from polycistronic transcription units (PTUs). In these parasites, a novel modified nucleotide base (ß-d-glucopyranosyloxymethyluracil) known as J plays a critical role in ensuring that transcription termination occurs only at the end of each PTU, rather than at the polyadenylation sites of individual genes. To further understand the biology of J-associated processes, we used tandem affinity purification (TAP) tagging and mass spectrometry to reveal proteins that interact with the glucosyltransferase performing the final step in J synthesis. These studies identified four proteins reminiscent of subunits in the PTW/PP1 complex that controls transcription termination in higher eukaryotes. Moreover, bioinformatic analyses identified the DNA-binding subunit of Leishmania PTW/PP1 as a novel J-binding protein (JBP3), which is also part of another complex containing proteins with domains suggestive of a role in chromatin modification/remodeling. Additionally, JBP3 associates (albeit transiently and/or indirectly) with the trypanosomatid equivalent of the PAF1 complex involved in the regulation of transcription in other eukaryotes. The downregulation of JBP3 expression levels in Leishmania resulted in a substantial increase in transcriptional readthrough at the 3' end of most PTUs. We propose that JBP3 recruits one or more of these complexes to the J-containing regions at the end of PTUs, where they halt the progression of the RNA polymerase. This decoupling of transcription termination from the splicing of individual genes enables the parasites' unique reliance on polycistronic transcription and posttranscriptional regulation of gene expression.IMPORTANCELeishmania parasites cause a variety of serious human diseases, with no effective vaccine and emerging resistance to current drug therapy. We have previously shown that a novel DNA base called J is critical for transcription termination at the ends of the polycistronic gene clusters that are a hallmark of Leishmania and related trypanosomatids. Here, we describe a new J-binding protein (JBP3) associated with three different protein complexes that are reminiscent of those involved in the control of transcription in other eukaryotes. However, the parasite complexes have been reprogrammed to regulate transcription and gene expression in trypanosomatids differently than in the mammalian hosts, providing new opportunities to develop novel chemotherapeutic treatments against these important pathogens.


Asunto(s)
Cromatina/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Leishmania/genética , Proteínas Protozoarias/genética , Terminación de la Transcripción Genética , Cromatina/metabolismo , ADN Protozoario/metabolismo , Regulación de la Expresión Génica , ARN Mensajero
4.
STAR Protoc ; 1(3): 100216, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33377109

RESUMEN

Quantitative changes in transcription factor (TF) abundance regulate dynamic cellular processes, including cell fate decisions. Protein copy number provides information about the relative stoichiometry of TFs that can be used to determine how quantitative changes in TF abundance influence gene regulatory networks. In this protocol, we describe a targeted selected reaction monitoring (SRM)-based mass-spectrometry method to systematically measure the absolute protein concentration of nuclear TFs as human hematopoietic stem and progenitor cells differentiate along the erythropoietic lineage. For complete details on the use and execution of this protocol, please refer to Gillespie et al. (2020).


Asunto(s)
Eritropoyesis/fisiología , Espectrometría de Masas/métodos , Factores de Transcripción/análisis , Diferenciación Celular/genética , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Proteómica/métodos
5.
Cell ; 183(3): 802-817.e24, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33053319

RESUMEN

Mammalian SWI/SNF complexes are ATP-dependent chromatin remodeling complexes that regulate genomic architecture. Here, we present a structural model of the endogenously purified human canonical BAF complex bound to the nucleosome, generated using cryoelectron microscopy (cryo-EM), cross-linking mass spectrometry, and homology modeling. BAF complexes bilaterally engage the nucleosome H2A/H2B acidic patch regions through the SMARCB1 C-terminal α-helix and the SMARCA4/2 C-terminal SnAc/post-SnAc regions, with disease-associated mutations in either causing attenuated chromatin remodeling activities. Further, we define changes in BAF complex architecture upon nucleosome engagement and compare the structural model of endogenous BAF to those of related SWI/SNF-family complexes. Finally, we assign and experimentally interrogate cancer-associated hot-spot mutations localizing within the endogenous human BAF complex, identifying those that disrupt BAF subunit-subunit and subunit-nucleosome interfaces in the nucleosome-bound conformation. Taken together, this integrative structural approach provides important biophysical foundations for understanding the mechanisms of BAF complex function in normal and disease states.


Asunto(s)
Enfermedad , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Ensamble y Desensamble de Cromatina , Microscopía por Crioelectrón , ADN Helicasas/química , ADN Helicasas/genética , ADN Helicasas/metabolismo , Enfermedad/genética , Humanos , Mutación Missense/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleosomas/metabolismo , Unión Proteica , Dominios Proteicos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/metabolismo , Homología Estructural de Proteína , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Mol Cell ; 78(5): 960-974.e11, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32330456

RESUMEN

Dynamic cellular processes such as differentiation are driven by changes in the abundances of transcription factors (TFs). However, despite years of studies, our knowledge about the protein copy number of TFs in the nucleus is limited. Here, by determining the absolute abundances of 103 TFs and co-factors during the course of human erythropoiesis, we provide a dynamic and quantitative scale for TFs in the nucleus. Furthermore, we establish the first gene regulatory network of cell fate commitment that integrates temporal protein stoichiometry data with mRNA measurements. The model revealed quantitative imbalances in TFs' cross-antagonistic relationships that underlie lineage determination. Finally, we made the surprising discovery that, in the nucleus, co-repressors are dramatically more abundant than co-activators at the protein level, but not at the RNA level, with profound implications for understanding transcriptional regulation. These analyses provide a unique quantitative framework to understand transcriptional regulation of cell differentiation in a dynamic context.


Asunto(s)
Eritropoyesis/genética , Redes Reguladoras de Genes/genética , Factores de Transcripción/genética , Bases de Datos Factuales , Regulación de la Expresión Génica/genética , Hematopoyesis/genética , Humanos , Proteómica/métodos , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo
7.
Proc Natl Acad Sci U S A ; 117(18): 10055-10066, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32312822

RESUMEN

Synaptic activity in neurons leads to the rapid activation of genes involved in mammalian behavior. ATP-dependent chromatin remodelers such as the BAF complex contribute to these responses and are generally thought to activate transcription. However, the mechanisms keeping such "early activation" genes silent have been a mystery. In the course of investigating Mendelian recessive autism, we identified six families with segregating loss-of-function mutations in the neuronal BAF (nBAF) subunit ACTL6B (originally named BAF53b). Accordingly, ACTL6B was the most significantly mutated gene in the Simons Recessive Autism Cohort. At least 14 subunits of the nBAF complex are mutated in autism, collectively making it a major contributor to autism spectrum disorder (ASD). Patient mutations destabilized ACTL6B protein in neurons and rerouted dendrites to the wrong glomerulus in the fly olfactory system. Humans and mice lacking ACTL6B showed corpus callosum hypoplasia, indicating a conserved role for ACTL6B in facilitating neural connectivity. Actl6b knockout mice on two genetic backgrounds exhibited ASD-related behaviors, including social and memory impairments, repetitive behaviors, and hyperactivity. Surprisingly, mutation of Actl6b relieved repression of early response genes including AP1 transcription factors (Fos, Fosl2, Fosb, and Junb), increased chromatin accessibility at AP1 binding sites, and transcriptional changes in late response genes associated with early response transcription factor activity. ACTL6B loss is thus an important cause of recessive ASD, with impaired neuron-specific chromatin repression indicated as a potential mechanism.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Hipocampo/patología , Actinas/genética , Adenosina Trifosfato/genética , Animales , Trastorno del Espectro Autista/patología , Conducta Animal/fisiología , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Emparejamiento Cromosómico/genética , Emparejamiento Cromosómico/fisiología , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Dendritas/genética , Dendritas/fisiología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Hipocampo/metabolismo , Humanos , Ratones , Ratones Noqueados , Mutación/genética , Neuronas/metabolismo , Neuronas/patología , Factores de Transcripción/genética
8.
Cell Stem Cell ; 24(5): 812-820.e5, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30880026

RESUMEN

Hematopoiesis provides an accessible system for studying the principles underlying cell-fate decisions in stem cells. Proposed models of hematopoiesis suggest that quantitative changes in lineage-specific transcription factors (LS-TFs) underlie cell-fate decisions. However, evidence for such models is lacking as TF levels are typically measured via RNA expression rather than by analyzing temporal changes in protein abundance. Here, we used single-cell mass cytometry and absolute quantification by mass spectrometry to capture the temporal dynamics of TF protein expression in individual cells during human erythropoiesis. We found that LS-TFs from alternate lineages are co-expressed, as proteins, in individual early progenitor cells and quantitative changes of LS-TFs occur gradually rather than abruptly to direct cell-fate decisions. Importantly, upregulation of a megakaryocytic TF in early progenitors is sufficient to deviate cells from an erythroid to a megakaryocyte trajectory, showing that quantitative changes in protein abundance of LS-TFs in progenitors can determine alternate cell fates.


Asunto(s)
Eritropoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Proteómica/métodos , Antígenos CD34/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Regulación de la Expresión Génica , Hematopoyesis , Humanos , Espectrometría de Masas , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Activación Transcripcional , Cordón Umbilical/citología
9.
Cell ; 175(5): 1272-1288.e20, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30343899

RESUMEN

Mammalian SWI/SNF (mSWI/SNF) ATP-dependent chromatin remodeling complexes are multi-subunit molecular machines that play vital roles in regulating genomic architecture and are frequently disrupted in human cancer and developmental disorders. To date, the modular organization and pathways of assembly of these chromatin regulators remain unknown, presenting a major barrier to structural and functional determination. Here, we elucidate the architecture and assembly pathway across three classes of mSWI/SNF complexes-canonical BRG1/BRM-associated factor (BAF), polybromo-associated BAF (PBAF), and newly defined ncBAF complexes-and define the requirement of each subunit for complex formation and stability. Using affinity purification of endogenous complexes from mammalian and Drosophila cells coupled with cross-linking mass spectrometry (CX-MS) and mutagenesis, we uncover three distinct and evolutionarily conserved modules, their organization, and the temporal incorporation of these modules into each complete mSWI/SNF complex class. Finally, we map human disease-associated mutations within subunits and modules, defining specific topological regions that are affected upon subunit perturbation.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Factores de Transcripción/metabolismo , Animales , Cromatina/química , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/genética , Drosophila/metabolismo , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Espectrometría de Masas , Mutagénesis , Subunidades de Proteína/análisis , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Factores de Transcripción/análisis , Factores de Transcripción/genética
10.
Nat Genet ; 49(11): 1613-1623, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28945250

RESUMEN

Perturbations to mammalian SWI/SNF (mSWI/SNF or BAF) complexes contribute to more than 20% of human cancers, with driving roles first identified in malignant rhabdoid tumor, an aggressive pediatric cancer characterized by biallelic inactivation of the core BAF complex subunit SMARCB1 (BAF47). However, the mechanism by which this alteration contributes to tumorigenesis remains poorly understood. We find that BAF47 loss destabilizes BAF complexes on chromatin, absent significant changes in complex assembly or integrity. Rescue of BAF47 in BAF47-deficient sarcoma cell lines results in increased genome-wide BAF complex occupancy, facilitating widespread enhancer activation and opposition of Polycomb-mediated repression at bivalent promoters. We demonstrate differential regulation by two distinct mSWI/SNF assemblies, BAF and PBAF complexes, enhancers and promoters, respectively, suggesting that each complex has distinct functions that are perturbed upon BAF47 loss. Our results demonstrate collaborative mechanisms of mSWI/SNF-mediated gene activation, identifying functions that are co-opted or abated to drive human cancers and developmental disorders.


Asunto(s)
Carcinogénesis/genética , Proteínas Cromosómicas no Histona/genética , Regulación Neoplásica de la Expresión Génica , Tumor Rabdoide/genética , Proteína SMARCB1/genética , Sarcoma/genética , Factores de Transcripción/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Cromatina/química , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Elementos de Facilitación Genéticos , Prueba de Complementación Genética , Humanos , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Regiones Promotoras Genéticas , Tumor Rabdoide/metabolismo , Tumor Rabdoide/patología , Proteína SMARCB1/deficiencia , Sarcoma/metabolismo , Sarcoma/patología , Factores de Transcripción/metabolismo
11.
Mol Syst Biol ; 13(3): 919, 2017 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-28320772

RESUMEN

Managing trade-offs through gene regulation is believed to confer resilience to a microbial community in a fluctuating resource environment. To investigate this hypothesis, we imposed a fluctuating environment that required the sulfate-reducer Desulfovibrio vulgaris to undergo repeated ecologically relevant shifts between retaining metabolic independence (active capacity for sulfate respiration) and becoming metabolically specialized to a mutualistic association with the hydrogen-consuming Methanococcus maripaludis Strikingly, the microbial community became progressively less proficient at restoring the environmentally relevant physiological state after each perturbation and most cultures collapsed within 3-7 shifts. Counterintuitively, the collapse phenomenon was prevented by a single regulatory mutation. We have characterized the mechanism for collapse by conducting RNA-seq analysis, proteomics, microcalorimetry, and single-cell transcriptome analysis. We demonstrate that the collapse was caused by conditional gene regulation, which drove precipitous decline in intracellular abundance of essential transcripts and proteins, imposing greater energetic burden of regulation to restore function in a fluctuating environment.


Asunto(s)
Desulfovibrio vulgaris/crecimiento & desarrollo , Methanococcus/crecimiento & desarrollo , Biología de Sistemas/métodos , Desulfovibrio vulgaris/genética , Evolución Molecular Dirigida , Perfilación de la Expresión Génica , Methanococcus/genética , Oxidación-Reducción , Fenotipo , Proteómica , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Sulfatos/metabolismo
13.
Mol Cell Biol ; 36(19): 2464-75, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27381459

RESUMEN

TFIIH is a 10-subunit RNA polymerase II basal transcription factor with a dual role in DNA repair. TFIIH contains three enzymatic functions and over 30 conserved subdomains and topological regions. We systematically tested the function of these regions in three TFIIH core module subunits, i.e., Ssl1, Tfb4, and Tfb2, in the DNA translocase subunit Ssl2, and in the kinase module subunit Tfb3. Our results are consistent with previously predicted roles for the Tfb2 Hub, Ssl2 Lock, and Tfb3 Latch regions, with mutations in these elements typically having severe defects in TFIIH subunit association. We also found unexpected roles for other domains whose function had not previously been defined. First, the Ssl1-Tfb4 Ring domains are important for TFIIH assembly. Second, the Tfb2 Hub and HEAT domains have an unexpected role in association with Tfb3. Third, the Tfb3 Ring domain is important for association with many other TFIIH subunits. Fourth, a partial deletion of the Ssl1 N-terminal extension (NTE) domain inhibits TFIIH function without affecting subunit association. Finally, we used site-specific cross-linking to localize the Tfb3-binding surface on the Rad3 Arch domain. Our cross-linking results suggest that Tfb3 and Rad3 have an unusual interface, with Tfb3 binding on two opposite faces of the Arch.


Asunto(s)
Saccharomyces cerevisiae/metabolismo , Factor de Transcripción TFIIH/química , Factor de Transcripción TFIIH/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , Mutación , Unión Proteica , Multimerización de Proteína , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Factor de Transcripción TFIIH/metabolismo , Factores de Transcripción TFII/genética , Factores de Transcripción TFII/metabolismo
14.
EMBO J ; 34(9): 1244-58, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25755249

RESUMEN

LXR-cofactor complexes activate the gene expression program responsible for cholesterol efflux in macrophages. Inflammation antagonizes this program, resulting in foam cell formation and atherosclerosis; however, the molecular mechanisms underlying this antagonism remain to be fully elucidated. We use promoter enrichment-quantitative mass spectrometry (PE-QMS) to characterize the composition of gene regulatory complexes assembled at the promoter of the lipid transporter Abca1 following downregulation of its expression. We identify a subset of proteins that show LXR ligand- and binding-dependent association with the Abca1 promoter and demonstrate they differentially control Abca1 expression. We determine that NCOA5 is linked to inflammatory Toll-like receptor (TLR) signaling and establish that NCOA5 functions as an LXR corepressor to attenuate Abca1 expression. Importantly, TLR3-LXR signal crosstalk promotes recruitment of NCOA5 to the Abca1 promoter together with loss of RNA polymerase II and reduced cholesterol efflux. Together, these data significantly expand our knowledge of regulatory inputs impinging on the Abca1 promoter and indicate a central role for NCOA5 in mediating crosstalk between pro-inflammatory and anti-inflammatory pathways that results in repression of macrophage cholesterol efflux.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/genética , Colesterol/metabolismo , Macrófagos/metabolismo , Coactivadores de Receptor Nuclear/genética , Receptores Nucleares Huérfanos/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Femenino , Regulación de la Expresión Génica , Inflamación/genética , Inflamación/metabolismo , Receptores X del Hígado , Espectrometría de Masas/métodos , Ratones Endogámicos C57BL , Ratones Noqueados , Coactivadores de Receptor Nuclear/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , Transducción de Señal , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo
15.
EMBO J ; 33(21): 2534-46, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25216679

RESUMEN

The conserved transcription coactivator SAGA is comprised of several modules that are involved in activator binding, TBP binding, histone acetylation (HAT) and deubiquitination (DUB). Crosslinking and mass spectrometry, together with genetic and biochemical analyses, were used to determine the molecular architecture of the SAGA-TBP complex. We find that the SAGA Taf and Taf-like subunits form a TFIID-like core complex at the center of SAGA that makes extensive interactions with all other SAGA modules. SAGA-TBP binding involves a network of interactions between subunits Spt3, Spt8, Spt20, and Spt7. The HAT and DUB modules are in close proximity, and the DUB module modestly stimulates HAT function. The large activator-binding subunit Tra1 primarily connects to the TFIID-like core via its FAT domain. These combined results were used to derive a model for the arrangement of the SAGA subunits and its interactions with TBP. Our results provide new insight into SAGA function in gene regulation, its structural similarity with TFIID, and functional interactions between the SAGA modules.


Asunto(s)
Regulación Fúngica de la Expresión Génica/fisiología , Modelos Biológicos , Complejos Multiproteicos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transactivadores/metabolismo , Factor de Transcripción TFIID/metabolismo , Complejos Multiproteicos/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Transactivadores/genética , Factor de Transcripción TFIID/genética
16.
Nat Struct Mol Biol ; 21(9): 810-6, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25132180

RESUMEN

Core Factor (CF) is a conserved RNA polymerase (Pol) I general transcription factor comprising Rrn6, Rrn11 and the TFIIB-related subunit Rrn7. CF binds TATA-binding protein (TBP), Pol I and the regulatory factors Rrn3 and upstream activation factor. We used chemical cross-linking-MS to determine the molecular architecture of CF and its interactions with TBP. The CF subunits assemble through an interconnected network of interactions between five structural domains that are conserved in orthologous subunits of the human Pol I factor SL1. We validated the cross-linking-derived model through a series of genetic and biochemical assays. Our combined results show the architecture of CF and the functions of the CF subunits in assembly of the complex. We extend these findings to model how CF assembles into the Pol I preinitiation complex, providing new insight into the roles of CF, TBP and Rrn3.


Asunto(s)
Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , ARN Polimerasa I/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Modelos Moleculares , Proteínas del Complejo de Iniciación de Transcripción Pol1/química , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , ARN Polimerasa I/química , Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/química
17.
J Mol Biol ; 426(21): 3479-99, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25073102

RESUMEN

Faithful repair of DNA double-strand breaks by homologous recombination is crucial to maintain functional genomes. The major Escherichia coli pathway of DNA break repair requires RecBCD enzyme, a complex protein machine with multiple activities. Upon encountering a Chi recombination hotspot (5' GCTGGTGG 3') during DNA unwinding, RecBCD's unwinding, nuclease, and RecA-loading activities change dramatically, but the physical basis for these changes is unknown. Here, we identify, during RecBCD's DNA unwinding, two Chi-stimulated conformational changes involving RecC. One produced a marked, long-lasting, Chi-dependent increase in protease sensitivity of a small patch, near the Chi recognition domain, on the solvent-exposed RecC surface. The other change was identified by crosslinking of an artificial amino acid inserted in this RecC patch to RecB. Small-angle X-ray scattering analysis confirmed a major conformational change upon binding of DNA to the enzyme and is consistent with these two changes. We propose that, upon DNA binding, the RecB nuclease domain swings from one side of RecC to the other; when RecBCD encounters Chi, the nuclease domain returns to its initial position determined by crystallography, where it nicks DNA exiting from RecC and loads RecA onto the newly generated 3'-ended single-stranded DNA during continued unwinding; a crevice between RecB and RecC increasingly narrows during these steps. This model provides a physical basis for the intramolecular "signal transduction" from Chi to RecC to RecD to RecB inferred previously from genetic and enzymatic analyses, and it accounts for the enzymatic changes that accompany Chi's stimulation of recombination.


Asunto(s)
Proteínas de Escherichia coli/fisiología , Escherichia coli/metabolismo , Exodesoxirribonucleasa V/fisiología , Regulación Bacteriana de la Expresión Génica , Secuencia de Aminoácidos , Cristalografía por Rayos X , Reparación del ADN , ADN de Cadena Simple/química , Magnesio/química , Espectrometría de Masas , Datos de Secuencia Molecular , Péptido Hidrolasas/química , Unión Proteica , Estructura Terciaria de Proteína , Recombinación Genética , Dispersión de Radiación , Tripsina/química , Rayos X
18.
J Proteome Res ; 12(5): 2034-44, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23540550

RESUMEN

Blood is an ideal window for viewing our health and disease status. Because blood circulates throughout the entire body and carries secreted, shed, and excreted signature proteins from every organ and tissue type, it is thus possible to use the blood proteome to achieve a comprehensive assessment of multiple-organ physiology and pathology. To date, the blood proteome has been frequently examined for diseases of individual organs; studies on compound insults impacting multiple organs are, however, elusive. We believe that a characterization of peripheral blood for organ-specific proteins affords a powerful strategy to allow early detection, staging, and monitoring of diseases and their treatments at a whole-body level. In this paper we test this hypothesis by examining a mouse model of acetaminophen (APAP)-induced hepatic and extra-hepatic toxicity. We used a glycocapture-assisted global quantitative proteomics (gagQP) approach to study serum proteins and validated our results using Western blot. We discovered in mouse sera both hepatic and extra-hepatic organ-specific proteins. From our validation, it was determined that selected organ-specific proteins had changed their blood concentration during the course of toxicity development and recovery. Interestingly, the peak responding time of proteins specific to different organs varied in a time-course study. The collected molecular information shed light on a complex, dynamic, yet interweaving, multiorgan-enrolled APAP toxicity. The developed technique as well as the identified protein markers is translational to human studies. We hope our work can broaden the utility of blood proteomics in diagnosis and research of the whole-body response to pathogenic cues.


Asunto(s)
Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Proteínas Sanguíneas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , Proteoma/metabolismo , Alanina Transaminasa/metabolismo , Animales , Proteínas Sanguíneas/química , Proteínas Sanguíneas/aislamiento & purificación , Glicopéptidos/química , Glicopéptidos/aislamiento & purificación , Glicosilación , Humanos , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Anotación de Secuencia Molecular , Especificidad de Órganos , Mapas de Interacción de Proteínas , Proteoma/química , Proteoma/aislamiento & purificación
19.
Proc Natl Acad Sci U S A ; 110(9): 3645-50, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23388641

RESUMEN

Regulation of gene expression involves the orchestrated interaction of a large number of proteins with transcriptional regulatory elements in the context of chromatin. Our understanding of gene regulation is limited by the lack of a protein measurement technology that can systematically detect and quantify the ensemble of proteins associated with the transcriptional regulatory elements of specific genes. Here, we introduce a set of selected reaction monitoring (SRM) assays for the systematic measurement of 464 proteins with known or suspected roles in transcriptional regulation at RNA polymerase II transcribed promoters in Saccharomyces cerevisiae. Measurement of these proteins in nuclear extracts by SRM permitted the reproducible quantification of 42% of the proteins over a wide range of abundances. By deploying the assay to systematically identify DNA binding transcriptional regulators that interact with the environmentally regulated FLO11 promoter in cell extracts, we identified 15 regulators that bound specifically to distinct regions along ∼600 bp of the regulatory sequence. Importantly, the dataset includes a number of regulators that have been shown to either control FLO11 expression or localize to these regulatory regions in vivo. We further validated the utility of the approach by demonstrating that two of the SRM-identified factors, Mot3 and Azf1, are required for proper FLO11 expression. These results demonstrate the utility of SRM-based targeted proteomics to guide the identification of gene-specific transcriptional regulators.


Asunto(s)
ADN de Hongos/metabolismo , Regulación Fúngica de la Expresión Génica , Estudios de Asociación Genética , Espectrometría de Masas/métodos , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Factores de Transcripción/metabolismo , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Proteoma/genética , Proteoma/metabolismo , Proteínas Represoras/metabolismo , Reproducibilidad de los Resultados , Saccharomyces cerevisiae/crecimiento & desarrollo , Transactivadores/metabolismo
20.
PLoS Genet ; 9(2): e1003216, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23408894

RESUMEN

The kinetochore is the macromolecular complex that assembles onto centromeric DNA and orchestrates the segregation of duplicated chromosomes. More than 60 components make up the budding yeast kinetochore, including inner kinetochore proteins that bind to centromeric chromatin and outer proteins that directly interact with microtubules. However, little is known about how these components assemble into a functional kinetochore and whether there are quality control mechanisms that monitor kinetochore integrity. We previously developed a method to isolate kinetochore particles via purification of the conserved Dsn1 kinetochore protein. We find that the Mub1/Ubr2 ubiquitin ligase complex associates with kinetochore particles through the CENP-C(Mif2) protein. Although Mub1/Ubr2 are not stable kinetochore components in vivo, they regulate the levels of the conserved outer kinetochore protein Dsn1 via ubiquitylation. Strikingly, a deletion of Mub1/Ubr2 restores the levels and viability of a mutant Dsn1 protein, reminiscent of quality control systems that target aberrant proteins for degradation. Consistent with this, Mub1/Ubr2 help to maintain viability when kinetochores are defective. Together, our data identify a previously unknown regulatory mechanism for the conserved Dsn1 kinetochore protein. We propose that Mub1/Ubr2 are part of a quality control system that monitors kinetochore integrity, thus ensuring genomic stability.


Asunto(s)
Proteínas Portadoras , Proteínas Cromosómicas no Histona , Cinetocoros/metabolismo , Proteínas de Saccharomyces cerevisiae , Ubiquitina-Proteína Ligasas , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Supervivencia Celular/genética , Centrómero/genética , Centrómero/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...