Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 381
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2297: 83-93, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33656672

RESUMEN

The UV RESISTANCE LOCUS 8 (UVR8) is a photoreceptor mediating photomorphogenic responses to UV-B. UVR8 exists as homodimer in plants and UV-B induces dissociation of dimeric UVR8 into monomers to initiate responses. The monomer/dimer status of UVR8 is reversible and a dynamic photo-equilibrium is established in plants according to the ambient light conditions. Here we describe a method to detect UVR8 homodimer and monomer by immunoblotting method from tomato (Solanum lycopersicum) plants. The feature of this method is that protein samples are not boiled prior to loading on an SDS-PAGE gel, which allows the detection of UVR8 homodimer and monomers simultaneously with a single antibody.


Asunto(s)
Fotorreceptores de Plantas/química , Fotorreceptores de Plantas/aislamiento & purificación , Solanum lycopersicum/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/aislamiento & purificación , Electroforesis en Gel de Poliacrilamida , Luz , Proteínas de Plantas/química , Proteínas de Plantas/aislamiento & purificación , Multimerización de Proteína/efectos de la radiación
2.
Curr Biol ; 30(24): 4869-4881.e5, 2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33035484

RESUMEN

The nanoscale protein architecture of the kinetochore plays an integral role in specifying the mechanisms underlying its functions in chromosome segregation. However, defining this architecture in human cells remains challenging because of the large size and compositional complexity of the kinetochore. Here, we use Förster resonance energy transfer to reveal the architecture of individual kinetochore-microtubule attachments in human cells. We find that the microtubule-binding domains of the Ndc80 complex cluster at the microtubule plus end. This clustering occurs only after microtubule attachment, and it increases proportionally with centromeric tension. Surprisingly, Ndc80 complex clustering is independent of the organization and number of its centromeric receptors. Moreover, this clustering is similar in yeast and human kinetochores despite significant differences in their centromeric organizations. These and other data suggest that the microtubule-binding interface of the human kinetochore behaves like a flexible "lawn" despite being nucleated by repeating biochemical subunits.


Asunto(s)
Centrómero/metabolismo , Segregación Cromosómica , Cinetocoros/ultraestructura , Microtúbulos/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/ultraestructura , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/aislamiento & purificación , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/ultraestructura , Transferencia Resonante de Energía de Fluorescencia , Técnicas de Silenciamiento del Gen , Genes Reporteros/genética , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Cinetocoros/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Saccharomyces cerevisiae
3.
EMBO Rep ; 20(12): e47952, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31701657

RESUMEN

In most eukaryotes, constitutive heterochromatin is associated with H3K9me3 and HP1α. The latter has been shown to play a role in heterochromatin formation through liquid-liquid phase separation. However, many other proteins are known to regulate and/or interact with constitutive heterochromatic regions in several species. We postulate that some of these heterochromatic proteins may play a role in the regulation of heterochromatin formation by liquid-liquid phase separation. Indeed, an analysis of the constitutive heterochromatin proteome shows that proteins associated with constitutive heterochromatin are significantly more disordered than a random set or a full nucleome set of proteins. Interestingly, their expression begins low and increases during preimplantation development. These observations suggest that the preimplantation embryo is a useful model to address the potential role for phase separation in heterochromatin formation, anticipating exciting research in the years to come.


Asunto(s)
Blastocisto/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Heterocromatina/metabolismo , Histonas/metabolismo , Animales , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/aislamiento & purificación , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Heterocromatina/genética , Código de Histonas , Histonas/aislamiento & purificación , Proteínas Intrínsecamente Desordenadas/aislamiento & purificación , Proteínas Intrínsecamente Desordenadas/metabolismo , Espectrometría de Masas , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Nat Commun ; 10(1): 1673, 2019 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-30975984

RESUMEN

Accurate chromosome segregation relies on microtubule end conversion, the ill-understood ability of kinetochores to transit from lateral microtubule attachment to durable association with dynamic microtubule plus-ends. The molecular requirements for this conversion and the underlying biophysical mechanisms are elusive. We reconstituted end conversion in vitro using two kinetochore components: the plus end-directed kinesin CENP-E and microtubule-binding Ndc80 complex, combined on the surface of a microbead. The primary role of CENP-E is to ensure close proximity between Ndc80 complexes and the microtubule plus-end, whereas Ndc80 complexes provide lasting microtubule association by diffusing on the microtubule wall near its tip. Together, these proteins mediate robust plus-end coupling during several rounds of microtubule dynamics, in the absence of any specialized tip-binding or regulatory proteins. Using a Brownian dynamics model, we show that end conversion is an emergent property of multimolecular ensembles of microtubule wall-binding proteins with finely tuned force-dependent motility characteristics.


Asunto(s)
Segregación Cromosómica , Cinesinas/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Animales , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas Cromosómicas no Histona/metabolismo , Proteínas del Citoesqueleto , Microscopía Fluorescente , Modelos Biológicos , Dinámicas no Lineales , Proteínas Nucleares/genética , Proteínas Nucleares/aislamiento & purificación , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Células Sf9 , Imagen Individual de Molécula , Procesos Estocásticos , Proteínas de Xenopus/genética , Proteínas de Xenopus/aislamiento & purificación , Proteínas de Xenopus/metabolismo
5.
Cell Mol Biol (Noisy-le-grand) ; 65(3): 25-31, 2019 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-30942153

RESUMEN

Flowering is a very important developmental stage in the plant life cycle. LIKE HETEROCHROMATIN PROTEIN 1 (LHP1) has been shown to participate in epigenetic silencing of flowering genes. Here, for the first time, we isolated and characterized six CmLHP1 homolog genes from the important day-neutral ornamental Chrysanthemum morifolium cultivar 'Jin budiao'. These homolog genes were most likely generated by whole-genome duplication. Bioinformatic analysis showed that chrysanthemum LHP1 homologs present low similarity to other plant LHP1-like genes. However, three nuclear localization signals and two domains were highly conserved among them. The secondary structures of the CmLHP1 homologs mainly include α-helices and random coils, indicating that the proteins are mixed proteins. Phylogenetic tree analysis indicated that the six CmLHP1 genes constituted a small clade and had the closest relationship with LsLHP1 (Lactuca sativa LHP1). Quantitative RT-PCR analysis showed that the CmLHP1 homologs were expressed in different tissues during the developmental period of chrysanthemum, but they were highly expressed in the buds, especially during the key S1 stage of the inflorescence. Furthermore, the expression patterns of CmLHP1 homologs showed divergence under different photoperiods. Both CmLHP1b and CmLHP1e exhibited photoperiod sensitivity in leaves. Intriguingly, CmLHP1c was insensitive to photoperiod in both the shoot apexes and the leaves. Subcellular localization revealed that the six CmLHP1 proteins were located in the nucleus. These results reveal that CmLHP1 homolog genes could be strong candidates as important regulators of flowering time in chrysanthemum.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Chrysanthemum/genética , Clonación Molecular/métodos , Regulación de la Expresión Génica de las Plantas , Homología de Secuencia de Aminoácido , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas Cromosómicas no Histona/metabolismo , Filogenia , Proteínas de Plantas/química , Proteínas de Plantas/genética , Proteínas de Plantas/aislamiento & purificación , Proteínas de Plantas/metabolismo , Estructura Secundaria de Proteína , Transporte de Proteínas , Fracciones Subcelulares/metabolismo
6.
Mol Biol (Mosk) ; 51(3): 490-501, 2017.
Artículo en Ruso | MEDLINE | ID: mdl-28707666

RESUMEN

Chimeric transcription factor E2A-PBX1 induces the development of acute lymphoblastic B-cell leukemia in children. Using a transgenic mouse model, we previously demonstrated that homeobox (HOX) gene HOXA9 genetically interact with E2A-PBX1 gene in the development of B-cell leukemia in mice. HOXA9 itself is a potent oncogene resulting in myeloid leukemia when overexpressed, which is strongly accelerated by its collaborator Meis1. HOX, PBX1 and MEIS1 proteins have been shown to form hetero dimeric or trimeric complexes in different combinations. Cooperative interaction between PBX1 and HOX proteins enhances their DNA binding specificity, essential for HOX dependent developmental programs. PBX1 is retained in E2A-PBX1, and thus the strong transcriptional activator properties of E2A-PBX1 may lead to aberrant activation of normally repressed targets of HOX-PBX complexes. However, although there is evidence that E2A-PBX1 could bind to HOX and MEIS1 proteins it is still unclear whether such complexes are actually required for leukemic transformation or whether E2A-PBX1 and HOXA9 are each part of larger protein complexes acting in independent complementing oncogenic pathways. In this study we aim to search for other HOXA9 and E2A-PBX1 interacting proteins. To identify novel proteins interacting with human E2A-PBX1 or HOXA9 we used tandem affinity purification (TAP) of protein complexes from 697 pre-B leukemic and HeLa cell lines transduced to express E2A-PBX1 or HOXA9, respectively, with covalently attached FLAG/HA peptides. The protein composition of each complex was determined using tandem mass-spectrometry. In the E2A-PBX1 containing complex we identified lymphoid transcription factor IKAROS, chromatin remodeling factors of SWI/SNF family while multiple subunits of translation initiation factor eIF3, E3 ubiquitin ligase UBR5 emerged from the HOXA9 complex as potential critical protein partners. This is the first time the protein partners of either E2A-PBX1 or HOXA9 oncoproteins were identified using an unbiased biochemical approach. The identification of translation initiation factors associated with HOXA9 might indicate a novel function for HOX proteins independent of their transcriptional activity.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Leucemia de Células B/genética , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas de Unión al ADN/metabolismo , Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/aislamiento & purificación , Leucemia de Células B/patología , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/genética , Proteínas de Fusión Oncogénica/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Unión Proteica , Mapas de Interacción de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Espectrometría de Masas en Tándem , Factores de Transcripción/genética , Factores de Transcripción/aislamiento & purificación , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/aislamiento & purificación
7.
Methods Enzymol ; 586: 115-142, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28137559

RESUMEN

Chromatin architecture in mammalian spermatogenesis undergoes extensive structural and functional reorganization during which several testis-specific histone variants and other chromatin proteins are expressed in a stage-dependent manner. The most dramatic change in chromatin composition is observed during spermiogenesis where nucleosomal chromatin is transformed into nucleoprotamine fiber. Role of posttranslational modification (PTM) of somatic canonical histones and histone variants is well documented and effect several chromatin-templated events. PTM of testis-specific chromatin proteins is proposed to orchestrate chromatin-templated events during mammalian spermatogenesis and their identification and subsequent functional characterization is key to understand chromatin restructuring events and establishment of sperm epigenome. Here, we present protocols for the purification of endogenous testis chromatin proteins from different stages of spermatogenesis and identification of their PTM repertoire by mass spectrometry through examples of testis-specific histone variants (TH2B and HILS1), and transition proteins (TP1 and TP2).


Asunto(s)
Proteínas Cromosómicas no Histona/química , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Animales , Cromatina/química , Cromatina/metabolismo , Cromatografía Líquida de Alta Presión , Cromatografía de Fase Inversa , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas Cromosómicas no Histona/metabolismo , Masculino , Ratas , Espermatogénesis , Espectrometría de Masas en Tándem , Testículo/citología
8.
Methods Mol Biol ; 1515: 23-35, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27797071

RESUMEN

The cohesin complex is involved in a broad range of chromosomal biology, including DNA repair, gene transcription as well as sister chromatid cohesion. Cohesin is a large, ring-shaped protein complex and is thought to entrap DNA molecules inside of its ring. The unique DNA association is central to cohesin function and requires its ATPase and another heterodimer complex called the cohesin loader. Here we describe the biochemical reconstitution of topological cohesin loading onto DNA using the purified fission yeast cohesin proteins.


Asunto(s)
Proteínas de Ciclo Celular/aislamiento & purificación , Proteínas Cromosómicas no Histona/aislamiento & purificación , ADN/genética , Biología Molecular/métodos , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación , Adenosina Trifosfatasas/genética , Proteínas de Ciclo Celular/genética , Cromátides/genética , Proteínas Cromosómicas no Histona/genética , Segregación Cromosómica/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Saccharomyces cerevisiae/genética , Intercambio de Cromátides Hermanas/genética , Cohesinas
9.
Methods Mol Biol ; 1515: 37-53, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27797072

RESUMEN

During the cell cycle, duplicated sister chromatids become physically connected during S phase through a process called sister-chromatid cohesion. Cohesion is terminated during the metaphase-to-anaphase transition to trigger sister-chromatid segregation. The establishment and dissolution of cohesion are highly regulated by the cohesin complex and its multitude of regulators. In particular, the cohesin regulator Wapl promotes the release of cohesin from chromosomes during both interphase and mitosis. Here, we describe in vitro protein binding assays between Wapl and a cohesin subcomplex, and cellular assays in human cells that probe the functions of Wapl in cohesin release.


Asunto(s)
Proteínas Portadoras/aislamiento & purificación , Proteínas de Ciclo Celular/aislamiento & purificación , Proteínas Cromosómicas no Histona/aislamiento & purificación , Segregación Cromosómica/genética , Biología Molecular/métodos , Proteínas Nucleares/aislamiento & purificación , Proteínas Proto-Oncogénicas/aislamiento & purificación , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Cromátides/genética , Proteínas Cromosómicas no Histona/genética , Humanos , Mitosis/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/genética , Saccharomyces cerevisiae/genética , Cohesinas
10.
Methods Mol Biol ; 1515: 55-64, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27797073

RESUMEN

Cohesin is a protein complex with key roles in chromosome biology, from chromatid segregation to DNA repair. Cohesin function is regulated by several posttranslational modifications, including phosphorylation, acetylation, ubiquitylation, and SUMOylation. Recent studies have shown that cohesin SUMOylation is essential for sister chromatid cohesion during normal cell cycle and in response to DNA damage. Posttranslational modification by the small ubiquitin-like modifier (SUMO) is a field in expansion, however, detecting SUMOylation can be challenging because the amount of modified substrates are usually low and de-conjugation during sample preparation often occurs. In this chapter we describe a method that can be adapted to different model organisms, and substrates to detect SUMOylation. We focus on cohesin and show that SUMOylation indeed occurs in most of the subunits of budding yeast cohesin.


Asunto(s)
Proteínas de Ciclo Celular/aislamiento & purificación , Proteínas Cromosómicas no Histona/aislamiento & purificación , Biología Molecular/métodos , Intercambio de Cromátides Hermanas/genética , Sumoilación/genética , Proteínas de Ciclo Celular/genética , Cromátides/genética , Proteínas Cromosómicas no Histona/genética , Segregación Cromosómica/genética , Daño del ADN/genética , Reparación del ADN/genética , Saccharomyces cerevisiae/genética , Cohesinas
11.
Methods Mol Biol ; 1515: 65-95, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27797074

RESUMEN

In sexually reproducing organisms, the formation of healthy gametes (sperm and eggs) requires the proper establishment and release of meiotic sister chromatid cohesion (SCC). SCC tethers replicated sisters from their formation in premeiotic S phase until the stepwise removal of cohesion in anaphase of meiosis I and II allows the separation of homologs and then sisters. Defects in the establishment or release of meiotic cohesion cause chromosome segregation errors that lead to the formation of aneuploid gametes and inviable embryos. The nematode Caenorhabditis elegans is an attractive model for studies of meiotic sister chromatid cohesion due to its genetic tractability and the excellent cytological properties of the hermaphrodite gonad. Moreover, mutants defective in the establishment or maintenance of meiotic SCC nevertheless produce abundant gametes, allowing analysis of the pattern of chromosome segregation. Here I describe two approaches for analysis of meiotic cohesion in C. elegans. The first approach relies on cytology to detect and quantify defects in SCC. The second approach relies on PCR and restriction digests to identify embryos that inherited an incorrect complement of chromosomes due to aberrant meiotic chromosome segregation. Both approaches are sensitive enough to identify rare errors and precise enough to reveal distinctive phenotypes resulting from mutations that perturb meiotic SCC in different ways. The robust, quantitative nature of these assays should strengthen phenotypic comparisons of different meiotic mutants and enhance the reproducibility of data generated by different investigators.


Asunto(s)
Proteínas de Ciclo Celular/aislamiento & purificación , Cromátides/genética , Proteínas Cromosómicas no Histona/aislamiento & purificación , Biología Molecular/métodos , Intercambio de Cromátides Hermanas/genética , Anafase/genética , Animales , Caenorhabditis elegans/genética , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Segregación Cromosómica/genética , Meiosis/genética , Cohesinas
12.
Mol Cell Proteomics ; 15(3): 854-65, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26560067

RESUMEN

In recent years, cross-linking mass spectrometry has proven to be a robust and effective method of interrogating macromolecular protein complex topologies at peptide resolution. Traditionally, cross-linking mass spectrometry workflows have utilized homogenous complexes obtained through time-limiting reconstitution, tandem affinity purification, and conventional chromatography workflows. Here, we present cross-linking immunoprecipitation-MS (xIP-MS), a simple, rapid, and efficient method for structurally probing chromatin-associated protein complexes using small volumes of mammalian whole cell lysates, single affinity purification, and on-bead cross-linking followed by LC-MS/MS analysis. We first benchmarked xIP-MS using the structurally well-characterized phosphoribosyl pyrophosphate synthetase complex. We then applied xIP-MS to the chromatin-associated cohesin (SMC1A/3), XRCC5/6 (Ku70/86), and MCM complexes, and we provide novel structural and biological insights into their architectures and molecular function. Of note, we use xIP-MS to perform topological studies under cell cycle perturbations, showing that the xIP-MS protocol is sufficiently straightforward and efficient to allow comparative cross-linking experiments. This work, therefore, demonstrates that xIP-MS is a robust, flexible, and widely applicable methodology for interrogating chromatin-associated protein complex architectures.


Asunto(s)
Cromatina/metabolismo , Inmunoprecipitación/métodos , Espectrometría de Masas/métodos , Complejos Multiproteicos/química , Complejos Multiproteicos/aislamiento & purificación , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/aislamiento & purificación , Cromatografía Liquida , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/aislamiento & purificación , Reactivos de Enlaces Cruzados , Células HeLa , Humanos , Proteínas de Mantenimiento de Minicromosoma/química , Proteínas de Mantenimiento de Minicromosoma/aislamiento & purificación , Modelos Moleculares , Estructura Cuaternaria de Proteína , Ribosa-Fosfato Pirofosfoquinasa/química , Ribosa-Fosfato Pirofosfoquinasa/aislamiento & purificación , Cohesinas
13.
Nat Rev Mol Cell Biol ; 16(7): 443-9, 2015 07.
Artículo en Inglés | MEDLINE | ID: mdl-25991376

RESUMEN

The kinetochore is a complex molecular machine that directs chromosome segregation during mitosis. It is one of the most elaborate subcellular protein structures in eukaryotes, comprising more than 100 different proteins. Inner kinetochore proteins associate with specialized centromeric chromatin containing the histone H3 variant centromere protein A (CENP-A) in place of H3. Outer kinetochore proteins bind to microtubules and signal to delay anaphase onset when microtubules are absent. Since the first kinetochore proteins were discovered and cloned 30 years ago using autoimmune sera from patients with scleroderma-spectrum disease, much has been learnt about the composition, functions and regulation of this remarkable structure.


Asunto(s)
Autoantígenos/aislamiento & purificación , Proteína B del Centrómero/aislamiento & purificación , Centrómero/metabolismo , Proteínas Cromosómicas no Histona/aislamiento & purificación , Animales , Autoantígenos/metabolismo , Proteína A Centromérica , Proteína B del Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Humanos , Cinetocoros/metabolismo
14.
Methods Mol Biol ; 1288: 245-72, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25827884

RESUMEN

The highly dynamic nucleoprotein structure of eukaryotic genome is organized in an ordered fashion, the unit of which is the nucleosome. The nucleosome is composed of core histones and DNA of variable size wrapped around it. Apart from the histone proteins, several nonhistone proteins also interact with the complex consisting of the DNA, the core and linker histones conferring highly regulated fluidity on the chromatin and permitting fine tuning of its functions. The nonhistone proteins are multifunctional and accentuate diverse cellular outcomes. In spite of the technical challenges, the architectural role of the nonhistone proteins altering the topology of the chromatin has been studied extensively. To appreciate the significance of the chromatin for genome function, it is essential to examine the role of the nonhistone proteins in different physiological conditions. Here, taking the example of a highly abundant chromatin protein, PC4 (Positive coactivator 4), we describe strategies for the identification of the chromatin-associated proteins and their structural and functional characterization.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Fraccionamiento Celular/métodos , Línea Celular , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Unión al ADN/metabolismo , Técnicas de Silenciamiento del Gen , Histonas/metabolismo , Humanos , Técnicas In Vitro , Microscopía de Fuerza Atómica , Unión Proteica , Interferencia de ARN , Factores de Transcripción/genética , Factores de Transcripción/aislamiento & purificación , Factores de Transcripción/metabolismo
15.
Methods Mol Biol ; 1170: 589-615, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24906339

RESUMEN

Histone variants such as H3.3, macroH2A, H2A.Z, and CENP-A are important epigenetic modifiers of the chromatin state in eukaryotic genomes. The centromeric histone H3 variant CENP-A/CENH3 epigenetically marks centromeres and is required for assembly of the kinetochore complex, a region of the chromosome that is responsible for proper genome segregation during mitosis. Several diverse techniques using biochemical, cell biology, and biophysical approaches have been utilized to study the nature of the CENP-A nucleosome across the cell cycle. In this chapter, we describe methods for CENP-A nucleosome purification and separation of CENP-A from other core histones using traditional SDS-PAGE and more resolving techniques such as Triton acid urea (TAU) and two-dimensional gels. We also discuss methods for observation of CENP-A on chromatin fibers using immunofluorescence. Finally, we provide a detailed description of analysis of chromatin structures using atomic force microscopy.


Asunto(s)
Autoantígenos/análisis , Ciclo Celular , Proteínas Cromosómicas no Histona/análisis , Histonas/análisis , Nucleosomas/química , Autoantígenos/aislamiento & purificación , Western Blotting/métodos , Proteína A Centromérica , Cromatina/ultraestructura , Proteínas Cromosómicas no Histona/aislamiento & purificación , Electroforesis en Gel de Poliacrilamida/métodos , Técnica del Anticuerpo Fluorescente/métodos , Histonas/aislamiento & purificación , Humanos , Inmunoprecipitación/métodos , Microscopía de Fuerza Atómica/métodos
16.
Protein Expr Purif ; 100: 54-60, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24859675

RESUMEN

Spt4/5 is a hetero-dimeric transcription elongation factor that can both inhibit and promote transcription elongation by RNA polymerase II (RNAPII). However, Spt4/5's mechanism of action remains elusive. Spt5 is an essential protein and the only universally-conserved RNAP-associated transcription elongation factor. The protein contains multiple Kyrpides, Ouzounis and Woese (KOW) domains. These domains, in other proteins, are thought to bind RNA although there is little direct evidence in the literature to support such a function in Spt5. This could be due, at least in part, to difficulties in expressing and purifying recombinant Spt5. When expressed in Escherichia coli (E. coli), Spt5 is innately insoluble. Here we report a new approach for the successful expression and purification of milligram quantities of three different multi-KOW domain complexes of Saccharomyces cerevisiae Spt4/5 for use in future functional studies. Using the E. coli strain Rosetta2 (DE3) we have developed strategies for co-expression of Spt4 and multi-KOW domain Spt5 complexes from the bi-cistronic pET-Duet vector. In a second strategy, Spt4/5 was expressed via co-transformation of Spt4 in the vector pET-M11 with Spt5 ubiquitin fusion constructs in the vector pHUE. We characterized the multi-KOW domain Spt4/5 complexes by Western blot, limited proteolysis, circular dichroism, SDS-PAGE and size exclusion chromatography-multiangle light scattering and found that the proteins are folded with a Spt4:Spt5 hetero-dimeric stoichiometry of 1:1. These expression constructs encompass a larger region of Spt5 than has previously been reported, and will provide the opportunity to elucidate the biological function of the multi-KOW containing Spt5.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas Nucleares/genética , Proteínas Nucleares/aislamiento & purificación , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/aislamiento & purificación , Ubiquitina/genética , Ubiquitina/aislamiento & purificación , Proteínas Cromosómicas no Histona/química , Clonación Molecular/métodos , Escherichia coli/genética , Proteínas Nucleares/química , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas de Saccharomyces cerevisiae/química , Elongación de la Transcripción Genética , Factores de Elongación Transcripcional/química , Ubiquitina/química
17.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 5): 656-8, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24817731

RESUMEN

Interactions between cohesin and dockerin modules are critical for the formation of the cellulosome, which is responsible for the efficient degradation of plant cell-wall carbohydrates by anaerobes. Type I dockerin modules found in modular enzymatic components interact with type I cohesins in primary scaffoldins, enabling the assembly of the multi-enzyme complex. In contrast, type II dockerins located in primary scaffoldins bind to type II cohesins in adaptor scaffoldins or anchoring scaffoldins located at the bacterial envelope, contributing to the cell-surface attachment of the entire complex. Acetivibrio cellulolyticus possesses an extremely complex cellulosome arrangement which is organized by a primary enzyme-binding scaffoldin (ScaA), two anchoring scaffoldins (ScaC and ScaD) and an unusual adaptor scaffoldin (ScaB). An ScaA X-dockerin mutated to inactivate one of the two putative cohesin-binding interfaces complexed with the third ScaB cohesin from A. cellulolyticus has been purified and crystallized and data were collected to a resolution of 2.41 Å.


Asunto(s)
Proteínas Bacterianas/química , Proteínas de Ciclo Celular/química , Proteínas Cromosómicas no Histona/química , Bacterias Grampositivas , Proteínas de la Membrana/química , Proteínas Bacterianas/aislamiento & purificación , Proteínas de Ciclo Celular/aislamiento & purificación , Proteínas Cromosómicas no Histona/aislamiento & purificación , Cristalización , Cristalografía por Rayos X , Proteínas de la Membrana/aislamiento & purificación , Cohesinas
18.
Proteomics ; 14(19): 2167-78, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24841622

RESUMEN

Centromeres are chromosomal regions crucial for correct chromosome segregation during mitosis and meiosis. They are epigenetically defined by centromeric proteins such as the centromere-specific histone H3-variant centromere protein A (CENP-A). In humans, 16 additional proteins have been described to be constitutively associated with centromeres throughout the cell cycle, known as the constitutive centromere-associated network (CCAN). In contrast, only one additional constitutive centromeric protein is known in Drosophila melanogaster (D.mel), the conserved CCAN member CENP-C. To gain further insights into D.mel centromere composition and biology, we analyzed affinity-purified chromatin prepared from D.mel cell lines expressing green fluorescent protein tagged histone three variants by MS. In addition to already-known centromeric proteins, we identified novel factors that were repeatedly enriched in affinity purification-MS experiments. We analyzed the cellular localization of selected candidates by immunocytochemistry and confirmed localization to the centromere and other genomic regions for ten factors. Furthermore, RNA interference mediated depletion of CG2051, CG14480, and hyperplastic discs, three of our strongest candidates, leads to elevated mitotic defects. Knockdowns of these candidates neither impair the localization of several known kinetochore proteins nor CENP-A(CID) loading, suggesting their involvement in alternative pathways that contribute to proper centromere function. In summary, we provide a comprehensive analysis of the proteomic composition of Drosophila centromeres. All MS data have been deposited in the ProteomeXchange with identifier PXD000758 (http://proteomecentral.proteomexchange.org/dataset/PXD000758).


Asunto(s)
Centrómero/química , Proteínas Cromosómicas no Histona/química , Proteínas de Drosophila/química , Animales , Línea Celular , Centrómero/genética , Centrómero/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/aislamiento & purificación , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Técnicas de Silenciamiento del Gen , Cinetocoros/metabolismo , Microscopía Fluorescente
19.
Nat Cell Biol ; 16(3): 281-93, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24561620

RESUMEN

To maintain genome function and stability, DNA sequence and its organization into chromatin must be duplicated during cell division. Understanding how entire chromosomes are copied remains a major challenge. Here, we use nascent chromatin capture (NCC) to profile chromatin proteome dynamics during replication in human cells. NCC relies on biotin-dUTP labelling of replicating DNA, affinity purification and quantitative proteomics. Comparing nascent chromatin with mature post-replicative chromatin, we provide association dynamics for 3,995 proteins. The replication machinery and 485 chromatin factors such as CAF-1, DNMT1 and SUV39h1 are enriched in nascent chromatin, whereas 170 factors including histone H1, DNMT3, MBD1-3 and PRC1 show delayed association. This correlates with H4K5K12diAc removal and H3K9me1 accumulation, whereas H3K27me3 and H3K9me3 remain unchanged. Finally, we combine NCC enrichment with experimentally derived chromatin probabilities to predict a function in nascent chromatin for 93 uncharacterized proteins, and identify FAM111A as a replication factor required for PCNA loading. Together, this provides an extensive resource to understand genome and epigenome maintenance.


Asunto(s)
Cromatina/metabolismo , Replicación del ADN , Proteoma/metabolismo , Receptores Virales/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Unión al ADN/metabolismo , Células HeLa , Histonas/aislamiento & purificación , Histonas/metabolismo , Humanos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Transporte de Proteínas , Proteoma/aislamiento & purificación , Proteómica , Puntos de Control de la Fase S del Ciclo Celular
20.
Mol Cell Biol ; 33(21): 4198-211, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23979598

RESUMEN

Spt2/Sin1 is a DNA binding protein with HMG-like domains. It plays a role in chromatin modulations associated with transcription elongation in Saccharomyces cerevisiae. Spt2 maintains the nucleosome level in coding regions and is important for the inhibition of spurious transcription in yeast. In this work, we undertook a biochemical approach to identify Spt2-interacting partners. Interestingly, casein kinase 2 (CK2) interacts with Spt2 and phosphorylates it in vitro as well as in vivo on two small regions, region I (RI) (amino acids 226 to 230) and RII (amino acids 277 to 281), located in its essential C-terminal domain. Mutation of the phosphorylation sites in RI and RII to acidic residues, thereby mimicking CK2 phosphorylation, leads to the inhibition of Spt2 function in the repression of spurious transcription and to a loss of its recruitment to coding regions. Inversely, depleting cells of CK2 activity leads to an increased Spt2 association with genes. We further show that Spt2 physically interacts with the essential histone chaperone Spt6 and that this association is inhibited in vitro and in vivo by CK2-dependent phosphorylation. Taken together, our data suggest that CK2 regulates the function of Spt2 by modulating its interaction with chromatin and the histone chaperone Spt6.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Fúngica de la Expresión Génica , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Transcripción Genética , Quinasa de la Caseína II/química , Quinasa de la Caseína II/aislamiento & purificación , Dominio Catalítico , Cromatina/genética , Cromatina/metabolismo , Cromatografía de Afinidad , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/aislamiento & purificación , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/aislamiento & purificación , Sistemas de Lectura Abierta , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Subunidades de Proteína/aislamiento & purificación , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación , Serina/metabolismo , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...