Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Angew Chem Int Ed Engl ; 61(46): e202209518, 2022 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-36283971

RESUMEN

Manley and co-workers provide data demonstrating that, at super-pharmacological concentrations (300 µM), a ternary complex between Abl, asciminib, and ATP-competitive inhibitors is possible. The work in our manuscript concerns the interplay of asciminib (and GNF-2) with ATP-competitive inhibitors at pharmacologically relevant concentrations (Cmax =1.6-3.7 µM for asciminib). Manley and co-workers do not question any of the studies that we reported, nor do they provide explanations for how our work fits into their preferred model. Herein, we consider the data presented by Manley and co-workers. In addition, we provide new data supporting the findings in our Communication. Asciminib and ATP-competitive inhibitors do not simultaneously bind Abl at pharmacologically relevant concentrations unless the conformation selectivity for both ligands is matched.


Asunto(s)
Resistencia a Antineoplásicos , Proteínas Proto-Oncogénicas c-abl , Humanos , Adenosina Trifosfato/metabolismo , Conformación Molecular , Mutación , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores
2.
Angew Chem Int Ed Engl ; 60(37): 20196-20199, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34292655

RESUMEN

Allosteric inhibitors of Abl kinase are being explored in the clinic, often in combination with ATP-site inhibitors of Abl kinase. However, there are conflicting data on whether both ATP-competitive inhibitors and myristoyl-site allosteric inhibitors can simultaneously bind Abl kinase. Here, we determine whether there is synergy or antagonism between ATP-competitive inhibitors and allosteric inhibitors of Abl. We observe that clinical ATP-competitive inhibitors are not synergistic with allosteric ABL inhibitors, however, conformation-selective ATP-site inhibitors that modulate the global conformation of Abl can afford synergy. We demonstrate that kinase conformation is the key driver to simultaneously bind two compounds to Abl kinase. Finally, we explore the interaction of allosteric and conformation selective ATP-competitive inhibitors in a series of biochemical and cellular assays.


Asunto(s)
Adenosina Trifosfato/farmacología , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Adenosina Trifosfato/química , Dominio Catalítico/efectos de los fármacos , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Modelos Moleculares , Inhibidores de Proteínas Quinasas/química
3.
ACS Chem Biol ; 19(1): 110-116, 2024 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-38113191

RESUMEN

Using dasatinib linked to E3 ligase ligands, we identified a potent and selective dual Csk/c-Src PROTAC degrader. We then replaced dasatinib, the c-Src-directed ligand, with a conformation-selective analogue that stabilizes the αC-helix-out conformation of c-Src. Using the αC-helix-out ligand, we identified a PROTAC that is potent and selective for c-Src. We demonstrated a high degree of catalysis with our c-Src PROTACs. Using our c-Src PROTACs, we identified pharmacological advantages of c-Src degradation compared to inhibition with respect to cancer cell proliferation.


Asunto(s)
Ubiquitina-Proteína Ligasas , Dasatinib/farmacología , Proteína Tirosina Quinasa CSK/metabolismo , Ligandos , Proliferación Celular , Ubiquitina-Proteína Ligasas/metabolismo , Proteolisis
4.
ACS Med Chem Lett ; 14(6): 860-866, 2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37284689

RESUMEN

The COVID-19 pandemic has highlighted the need for new antiviral approaches because many of the currently approved drugs have proven ineffective against mitigating SARS-CoV-2 infections. The host transmembrane serine protease TMPRSS2 is a promising antiviral target because it plays a role in priming the spike protein before viral entry occurs for the most virulent variants. Further, TMPRSS2 has no established physiological role, thereby increasing its attractiveness as a target for antiviral agents. Here, we utilize virtual screening to curate large libraries into a focused collection of potential inhibitors. Optimization of a recombinant expression and purification protocol for the TMPRSS2 peptidase domain facilitates subsequent biochemical screening and characterization of selected compounds from the curated collection in a kinetic assay. In doing so, we identify new noncovalent TMPRSS2 inhibitors that block SARS-CoV-2 infectivity in a cellular model. One such inhibitor, debrisoquine, has high ligand efficiency, and an initial structure-activity relationship study demonstrates that debrisoquine is a tractable hit compound for TMPRSS2.

5.
Sci Rep ; 10(1): 16551, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33024171

RESUMEN

The neurodegenerative Alzheimer's disease (AD) affects more than 30 million people worldwide. There is thus far no cure or prevention for AD. Aggregation of hyperphosphorylated tau in the brain correlates with the cognitive decline of patients of AD and other neurodegenerative tauopathies. Intracerebral injection of tau aggregates isolated from tauopathy brains causes similar pathology in the recipient mice, demonstrating the pathogenic role of abnormally phosphorylated tau. Compounds controlling the aggregation of hyperphosphorylated tau therefore are probable modulators for the disease. Here we report the use of recombinant hyperphosphorylated tau (p-tau) to identify potential tauopathy therapeutics and risk factors. Hyperphosphorylation renders tau prone to aggregate and to impair cell viability. Taking advantage of these two characters of p-tau, we performed a screen of a 1280-compound library, and tested a selective group of prescription drugs in p-tau aggregation and cytotoxicity assays. R-(-)-apomorphine and raloxifene were found to be p-tau aggregation inhibitors that protected p-tau-treated cells. In contrast, a subset of benzodiazepines exacerbated p-tau cytotoxicity apparently via enhancing p-tau aggregation. R-(-)apomorphine and raloxifene have been shown to improve cognition in animals or in humans, whereas benzodiazepines were linked to increased risks of dementia. Our results demonstrate the feasibility and potential of using hyperphosphorylated tau-based assays for AD drug discovery and risk factor identification.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Apomorfina/farmacología , Cognición/efectos de los fármacos , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Medicamentos bajo Prescripción/farmacología , Agregado de Proteínas/efectos de los fármacos , Clorhidrato de Raloxifeno/farmacología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Apomorfina/uso terapéutico , Benzodiazepinas/efectos adversos , Humanos , Fosforilación/efectos de los fármacos , Medicamentos bajo Prescripción/uso terapéutico , Clorhidrato de Raloxifeno/uso terapéutico , Factores de Riesgo
6.
Mol Neurobiol ; 57(11): 4704-4719, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32780352

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder without a cure or prevention to date. Hyperphosphorylated tau forms the neurofibrillary tangles (NFTs) that correlate well with the progression of cognitive impairments. Animal studies demonstrated the pathogenic role of hyperphosphorylated tau. Understanding how abnormal phosphorylation renders a normal tau prone to form toxic fibrils is key to delineating molecular pathology and to developing efficacious drugs for AD. Production of a tau bearing the disease-relevant hyperphosphorylation and molecular characters is a pivotal step. Here, we report the preparation and characterization of a recombinant hyperphosphorylated tau (p-tau) with strong relevance to disease. P-tau generated by the PIMAX approach resulted in phosphorylation at multiple epitopes linked to the progression of AD neuropathology. In stark contrast to unmodified tau that required an aggregation inducer, and which had minimal effects on cell functions, p-tau formed inducer-free fibrils that triggered a spike of mitochondrial superoxide, induced apoptosis, and caused cell death at sub-micromolar concentrations. P-tau-induced apoptosis was suppressed by inhibitors for reactive oxygen species. Hyperphosphorylation apparently caused rapid formation of a disease-related conformation. In both aggregation and cytotoxicity, p-tau exhibited seeding activities that converted the unmodified tau into a cytotoxic species with an increased propensity for fibrillization. These characters of p-tau are consistent with the emerging view that hyperphosphorylation causes tau to become an aggregation-prone and cytotoxic species that underlies diffusible pathology in AD and other tauopathies. Our results further suggest that p-tau affords a feasible tool for Alzheimer's disease mechanistic and drug discovery studies.


Asunto(s)
Agregado de Proteínas , Proteínas tau/metabolismo , Fenómenos Biofísicos , Muerte Celular , Línea Celular , Supervivencia Celular , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Mitocondrias/metabolismo , Oxidación-Reducción , Fosforilación , Unión Proteica , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/metabolismo , Superóxidos/metabolismo
7.
Anal Biochem ; 392(2): 155-61, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19497294

RESUMEN

Flowcytometric procedures provide distinct advantages over the colorimetric methods currently in use to monitor erythrocytes for exposure of patients to organophosphorus (OP) pesticides and chemical warfare agents; therefore, they warrant exploration. Two types of fluorescent probes-one to detect the total acetylcholinesterase on erythrocytes (RBC-AChE) and the other to distinguish between the active and OP-inhibited RBC-AChE-have been explored. Our studies demonstrate that a fluorescently conjugated fasciculin can be used to monitor total, active, and OP-inhibited RBC-AChE. However, a fluorescently tagged potent inhibitor of AChE, TZ2PIQ-A6 with a K(d) of 33 fM, did not distinguish between the active and OP-inhibited RBC-AChE, nor did three different biotinylated OP compounds. The biotin-fluorescent avidin approach is not a viable procedure for monitoring RBC-AChE. Western blot studies indicate that there are at least 20 serine hydrolases on the surface of red blood cells (RBCs). Plans currently under way for the development of more specific probes to distinguish between active and OP-inhibited RBC-AChE are discussed.


Asunto(s)
Acetilcolinesterasa/análisis , Inhibidores de la Colinesterasa/farmacología , Eritrocitos/efectos de los fármacos , Citometría de Flujo/métodos , Compuestos Organofosforados/farmacología , Acetilcolinesterasa/química , Acetilcolinesterasa/metabolismo , Animales , Biomarcadores/sangre , Inhibidores de la Colinesterasa/química , Eritrocitos/enzimología , Humanos , Ratones , Modelos Moleculares , Estructura Molecular , Compuestos Organofosforados/química
8.
Mol Cell Biol ; 26(17): 6395-402, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16914725

RESUMEN

Histone deacetylase (HDAC) inhibitors are a promising class of anticancer agents for the treatment of solid and hematological malignancies. The precise mechanism by which HDAC inhibitors mediate their effects on tumor cell growth, differentiation, and/or apoptosis is the subject of intense research. Previously we described a family of multiprotein complexes that contain histone deacetylase 1/2 (HDAC1/2) and the histone demethylase BHC110 (LSD1). Here we show that HDAC inhibitors diminish histone H3 lysine 4 (H3K4) demethylation by BHC110 in vitro. In vivo analysis revealed an increased H3K4 methylation concomitant with inhibition of nucleosomal deacetylation by HDAC inhibitors. Reconstitution of recombinant complexes revealed a functional connection between HDAC1 and BHC110 only when nucleosomal substrates were used. Importantly, while the enzymatic activity of BHC110 is required to achieve optimal deacetylation in vitro, in vivo analysis following ectopic expression of an enzymatically dead mutant of BHC110 (K661A) confirmed the functional cross talk between the demethylase and deacetylase enzymes. Our studies not only reveal an intimate link between the histone demethylase and deacetylase enzymes but also identify histone demethylation as a secondary target of HDAC inhibitors.


Asunto(s)
Histona Desacetilasas/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Acetilación/efectos de los fármacos , Proteínas de Unión al ADN/química , Inhibidores Enzimáticos/farmacología , Células HeLa , Histona Desacetilasa 1 , Histona Demetilasas , Humanos , Metilación/efectos de los fármacos , Complejos Multienzimáticos/metabolismo , Nucleosomas/efectos de los fármacos , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo
9.
Mol Endocrinol ; 24(6): 1165-74, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20308527

RESUMEN

The androgen receptor (AR) mediates the effect of androgens through its transcriptional function during both normal prostate development and in the emergence and progression of prostate cancer. AR is known to assemble coactivator complexes at target promoters to facilitate transcriptional activation in response to androgens. Here we identify the ATP-dependent chromatin remodeling factor chromodomain helicase DNA-binding protein 8 (CHD8) as a novel coregulator of androgen-responsive transcription. We demonstrate that CHD8 directly associates with AR and that CHD8 and AR simultaneously localize to the TMPRSS2 enhancer after androgen treatment. In the LNCaP cell line, reduction of CHD8 levels by small interfering RNA treatment severely diminishes androgen-dependent activation of the TMPRSS2 gene. We demonstrate that the recruitment of AR to the TMPRSS2 promoter in response to androgen treatment requires CHD8. Finally, CHD8 facilitates androgen-stimulated proliferation of LNCaP cells, emphasizing the physiological importance of CHD8. Taken together, we present evidence of a functional role for CHD8 in AR-mediated transcriptional regulation of target genes.


Asunto(s)
Andrógenos/farmacología , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Receptores Androgénicos/metabolismo , Elementos de Respuesta/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Factores de Transcripción/genética
10.
FEBS Lett ; 584(4): 689-93, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20085832

RESUMEN

Chromodomain, helicase, DNA-binding protein 8 (CHD8) is an ATP-dependent chromatin remodeling enzyme that has been demonstrated to exist within a large protein complex which includes WDR5, Ash2L, and RbBP5, members of the Mixed Lineage Leukemia (MLL) histone modifying complexes. Here we show that CHD8 relocalizes to the promoter of the MLL regulated gene HOXA2 upon gene activation. Depletion of CHD8 enhances HOXA2 expression under activating conditions. Furthermore, depletion of CHD8 results in a loss of the WDR5/Ash2L/RbBP5 subcomplex, and consequently H3K4 trimethylation, at the HOXA2 promoter. These studies suggest that CHD8 alters HOXA2 gene expression and regulates the recruitment of chromatin modifying enzymes.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lisina/metabolismo , Metilación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Tretinoina/farmacología
11.
Mol Cell Biol ; 28(12): 3894-904, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18378692

RESUMEN

ATP-dependent chromatin remodeling by the CHD family of proteins plays an important role in the regulation of gene transcription. Here we report that full-length CHD8 interacts directly with beta-catenin and that CHD8 is also recruited specifically to the promoter regions of several beta-catenin-responsive genes. Our results indicate that CHD8 negatively regulates beta-catenin-targeted gene expression, since short hairpin RNA against CHD8 results in the activation of several beta-catenin target genes. This regulation is also conserved through evolution; RNA interference against kismet, the apparent Drosophila ortholog of CHD8, results in a similar activation of beta-catenin target genes. We also report the first demonstration of chromatin remodeling activity for a member of the CHD6-9 family of proteins, suggesting that CHD8 functions in transcription through the ATP-dependent modulation of chromatin structure.


Asunto(s)
Adenosina Trifosfato/química , Cromatina/química , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Factores de Transcripción/metabolismo , beta Catenina/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Núcleo Celular/metabolismo , Cromatina/metabolismo , ADN Helicasas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Células HeLa , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Modelos Biológicos , Interferencia de ARN , Transcripción Genética
12.
J Biol Chem ; 282(37): 26717-26724, 2007 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-17613521

RESUMEN

Tryptophan hydroxylase-2 (TPH2) is the rate-limiting enzyme in raphe serotonin biosynthesis, and polymorphisms of TPH2 are implicated in vulnerability to psychiatric disorders. Dynamic transcription regulation of TPH2 may underlie differences in vulnerability. We identified a transcription element in the TPH2 promoter that resembles the binding motif for RE-1 silencer of transcription (REST; also known as NRSF) transcription factor. REST limits tissue expression of non-neuronal genes through a canonical 21-bp motif called the NRSE (neuron-restrictive silencing element). The NRSE in TPH2 is a novel bipartite variant interrupted by a 6-base insertion. We confirmed that this bipartite NRSE permits transcriptional repression by REST identical to canonical NRSE in rat C6-glioma cells. Synthetic permutations of the motif revealed considerable flexibility in the juxtaposition of the two halves of bipartite NRSE. Computational analysis revealed many bipartite NRSE variants conserved between mouse and human genomes. A subgroup of these was found to bind REST by chromatin immunoprecipitation. Messenger RNAs for TPH2 and potassium channel H6, another gene with a bipartite NRSE, were up-regulated by dominant-negative REST in C6-glioma cells. These findings, which indicate that TPH2 expression is part of the developmental program regulated by REST and suggest that many previously unrecognized genes may be regulated by REST through the novel motif, have implications for the mechanism of REST action.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Triptófano Hidroxilasa/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Perros , Humanos , Ratones , Datos de Secuencia Molecular , Canal de Sodio Activado por Voltaje NAV1.2 , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas , Ratas , Proteínas Represoras/química , Canales de Sodio/genética , Factores de Transcripción/química
13.
Cell Cycle ; 4(12): 1854-61, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16294047

RESUMEN

p53 binding protein 1 (53BP1) participates in the repair of DNA double stranded breaks (DSBs) where it is recruited to or near sites of DNA damage. Although little is known about the biochemical functions of 53BP1, the protein possesses several motifs that are likely important for its role as a DNA damage response element. This includes two BRCA1 C-terminal repeats, tandem Tudor domains, and a variety of phosphorylation sites. Here we show that a glycine-arginine rich (GAR) stretch of 53BP1 lying upstream of the Tudor motifs is methylated. We demonstrate that arginine residues within this region are important for asymmetric methylation by the PRMT1 methyltransferase. We further show that sequences upstream of the Tudor domains that do not include the GAR stretch are sufficient for 53BP1 oligomerization in vivo. Thus, although Tudor domains bind methylated proteins, 53BP1 homo-oligomerization occurs independently of Tudor function. Lastly, we find that deficiencies in 53BP1 generate a "hyper-rec" phenotype. Collectively, these data provide new insight into 53BP1, an important component in maintaining genomic stability.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfoproteínas/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Arginina/metabolismo , Biopolímeros/metabolismo , Daño del ADN/genética , Glicina/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Metilación , Datos de Secuencia Molecular , Fosfoproteínas/química , Estructura Terciaria de Proteína , Recombinación Genética , Células Madre/citología , Proteína 1 de Unión al Supresor Tumoral P53
14.
Proc Natl Acad Sci U S A ; 99(11): 7420-5, 2002 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-12032298

RESUMEN

BRAF35, a structural DNA-binding protein, initially was identified as a component of a large BRCA2-containing complex. Biochemical analysis revealed the presence of a smaller core-BRAF35 complex devoid of BRCA2. Here we report the isolation of a six-subunit core-BRAF35 complex with the capacity to deacetylate histones, termed the BRAF-histone deacetylase complex (BHC), from human cells. BHC contains polypeptides reminiscent of the chromatin-remodeling complexes SWI/SNF and NuRD (nucleosome remodeling and deacetylating). Similar to NuRD, BHC contains an Mi2-like subunit, BHC80, and a PHD zinc-finger subunit as well as histone deacetylases 1/2 and an MTA-like subunit, the transcriptional corepressor CoREST. We show that BHC mediates repression of neuron-specific genes through the cis-regulatory element known as the repressor element 1 or neural restrictive silencer (RE1/NRS). Chromatin-immunoprecipitation experiments demonstrate the recruitment of BHC by the neuronal repressor REST. Expression of BRAF35 containing a single point mutation in the HMG domain of the protein abrogated REST-mediated transcriptional repression. These results demonstrate a role for core-BRAF35-containing complex in the regulation of neuron-specific genes through modulation of the chromatin structure.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Histona Desacetilasas/metabolismo , Neuronas/fisiología , Proteínas Represoras/metabolismo , Transcripción Genética/efectos de los fármacos , Núcleo Celular/metabolismo , Cromatina/metabolismo , Cromatina/ultraestructura , Cromatografía por Intercambio Iónico , Proteínas de Unión al ADN/farmacología , Células HeLa , Proteínas del Grupo de Alta Movilidad , Histona Desacetilasas/farmacología , Humanos , Datos de Secuencia Molecular , Subunidades de Proteína , Sinapsinas/genética , Dedos de Zinc
15.
Nature ; 418(6901): 994-8, 2002 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-12198550

RESUMEN

Nucleosomal DNA is arranged in a higher-order structure that presents a barrier to most cellular processes involving protein DNA interactions. The cellular machinery involved in sister chromatid cohesion, the cohesin complex, also requires access to the nucleosomal DNA to perform its function in chromosome segregation. The machineries that provide this accessibility are termed chromatin remodelling factors. Here, we report the isolation of a human ISWI (SNF2h)-containing chromatin remodelling complex that encompasses components of the cohesin and NuRD complexes. We show that the hRAD21 subunit of the cohesin complex directly interacts with the ATPase subunit SNF2h. Mapping of hRAD21, SNF2h and Mi2 binding sites by chromatin immunoprecipitation experiments reveals the specific association of these three proteins with human DNA elements containing Alu sequences. We find a correlation between modification of histone tails and association of the SNF2h/cohesin complex with chromatin. Moreover, we show that the association of the cohesin complex with chromatin can be regulated by the state of DNA methylation. Finally, we present evidence pointing to a role for the ATPase activity of SNF2h in the loading of hRAD21 on chromatin.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromatina/química , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas Humanos/química , Cromosomas Humanos/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Elementos Alu/genética , Cromatina/genética , Cromosomas Humanos/genética , ADN/química , ADN/genética , ADN/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Proteínas Fúngicas , Células HeLa , Humanos , Sustancias Macromoleculares , Pruebas de Precipitina , Unión Proteica , Subunidades de Proteína , Retroelementos/genética , Cohesinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA