Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 14: 1250893, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37841927

RESUMEN

The Wnt/ß-catenin pathway is abnormally activated in most lung cancer tissues and considered to be an accelerator of carcinogenesis and lung cancer progression, which is closely related to increased morbidity rates, malignant progression, and treatment resistance. Although targeting the canonical Wnt/ß-catenin pathway shows significant potential for lung cancer therapy, it still faces challenges owing to its complexity, tumor heterogeneity and wide physiological activity. Therefore, it is necessary to elucidate the role of the abnormal activation of the Wnt/ß-catenin pathway in lung cancer progression. Moreover, Wnt inhibitors used in lung cancer clinical trials are expected to break existing therapeutic patterns, although their adverse effects limit the treatment window. This is the first study to summarize the research progress on various compounds, including natural products and derivatives, that target the canonical Wnt pathway in lung cancer to develop safer and more targeted drugs or alternatives. Various natural products have been found to inhibit Wnt/ß-catenin in various ways, such as through upstream and downstream intervention pathways, and have shown encouraging preclinical anti-tumor efficacy. Their diversity and low toxicity make them a popular research topic, laying the foundation for further combination therapies and drug development.

2.
Res Sq ; 2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36909460

RESUMEN

Missense variants throughout ACTA2, encoding smooth muscle α-actin (αSMA), predispose to adult onset thoracic aortic disease, but variants disrupting arginine 179 (R179) lead to Smooth Muscle Dysfunction Syndrome (SMDS) characterized by childhood-onset diverse vascular diseases. Our data indicate that αSMA localizes to the nucleus in wildtype (WT) smooth muscle cells (SMCs), enriches in the nucleus with SMC differentiation, and associates with chromatin remodeling complexes and SMC contractile gene promotors, and the ACTA2 p.R179 variant decreases nuclear localization of αSMA. SMCs explanted from a SMC-specific conditional knockin mouse model, Acta2SMC-R179/+, are less differentiated than WT SMCs, both in vitro and in vivo, and have global changes in chromatin accessibility. Induced pluripotent stem cells from patients with ACTA2 p.R179 variants fail to fully differentiate from neural crest cells to SMCs, and single cell transcriptomic analyses of an ACTA2 p.R179H patient's aortic tissue shows increased SMC plasticity. Thus, nuclear αSMA participates in SMC differentiation and loss of this nuclear activity occurs with ACTA2 p.R179 pathogenic variants.

3.
Artículo en Inglés | MEDLINE | ID: mdl-36674147

RESUMEN

Covalent organic framework (COF)-TpBD was grafted on the arrayed nanopores of stainless steel fiber (SSF) with (3-aminopropyl) triethoxysilane as the cross-linking agent. The prepared SSF bonded with COF-TpBD showed high thermal and chemical stability and excellent repeatability. The prepared SSF bonded with COF-TpBD was also used for the solid-phase microextraction (SPME) of seven kinds of polycyclic aromatic hydrocarbons (PAHs) in actual water samples, followed by gas chromatography with flame ionization detection (GC-FID) determination, which exhibited low limits of detection (LODs), good relative standard deviation (RSD) and high recoveries.


Asunto(s)
Estructuras Metalorgánicas , Nanoporos , Hidrocarburos Policíclicos Aromáticos , Contaminantes Químicos del Agua , Agua/química , Estructuras Metalorgánicas/química , Acero Inoxidable , Hidrocarburos Policíclicos Aromáticos/análisis , Microextracción en Fase Sólida/métodos , Límite de Detección , Contaminantes Químicos del Agua/análisis
4.
Nat Cardiovasc Res ; 2(10): 937-955, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38919852

RESUMEN

Missense variants throughout ACTA2, encoding smooth muscle α-actin (αSMA), predispose to adult-onset thoracic aortic disease, but variants disrupting arginine 179 (R179) lead to Smooth Muscle Dysfunction Syndrome (SMDS) characterized by diverse childhood-onset vascular diseases. Here we show that αSMA localizes to the nucleus in wildtype (WT) smooth muscle cells (SMCs), enriches in the nucleus with SMC differentiation, and associates with chromatin remodeling complexes and SMC contractile gene promotors. The ACTA2 p.R179 αSMA variant shows decreased nuclear localization. Primary SMCs from Acta2 SMC-R179C/+ mice are less differentiated than WT SMCs in vitro and in vivo and have global changes in chromatin accessibility. Induced pluripotent stem cells from patients with ACTA2 p.R179 variants fail to fully differentiate from neuroectodermal progenitor cells to SMCs, and single-cell transcriptomic analyses of an ACTA2 p.R179H patient's aortic tissue show increased SMC plasticity. Thus, nuclear αSMA participates in SMC differentiation, and loss of this nuclear activity occurs with ACTA2 p.R179 pathogenic variants.

5.
PLoS Pathog ; 17(6): e1009657, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34133468

RESUMEN

GTP-binding protein (G-protein) and regulator of G-protein signaling (RGS) mediated signal transduction are critical in the growth and virulence of the rice blast pathogen Magnaporthe oryzae. We have previously reported that there are eight RGS and RGS-like proteins named MoRgs1 to MoRgs8 playing distinct and shared regulatory functions in M. oryzae and that MoRgs1 has a more prominent role compared to others in the fungus. To further explore the unique regulatory mechanism of MoRgs1, we screened a M. oryzae cDNA library for genes encoding MoRgs1-interacting proteins and identified MoCkb2, one of the two regulatory subunits of the casein kinase (CK) 2 MoCk2. We found that MoCkb2 and the sole catalytic subunit MoCka1 are required for the phosphorylation of MoRgs1 at the plasma membrane (PM) and late endosome (LE). We further found that an endoplasmic reticulum (ER) membrane protein complex (EMC) subunit, MoEmc2, modulates the phosphorylation of MoRgs1 by MoCk2. Interestingly, this phosphorylation is also essential for the GTPase-activating protein (GAP) function of MoRgs1. The balance among MoRgs1, MoCk2, and MoEmc2 ensures normal operation of the G-protein MoMagA-cAMP signaling required for appressorium formation and pathogenicity of the fungus. This has been the first report that an EMC subunit is directly linked to G-protein signaling through modulation of an RGS-casein kinase interaction.


Asunto(s)
Ascomicetos/metabolismo , Ascomicetos/patogenicidad , Proteínas Fúngicas/metabolismo , Interacciones Huésped-Parásitos/fisiología , Virulencia/fisiología , Caseína Quinasas/metabolismo , Fosforilación , Transducción de Señal/fisiología
6.
J Clin Invest ; 130(11): 5951-5966, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33016929

RESUMEN

ARID1A, a component of the chromatin-remodeling complex SWI/SNF, is one of the most frequently mutated genes in human cancer. We sought to develop rational combination therapy to potentiate the efficacy of immune checkpoint blockade in ARID1A-deficient tumors. In a proteomic analysis of a data set from The Cancer Genomic Atlas, we found enhanced expression of Chk2, a DNA damage checkpoint kinase, in ARID1A-mutated/deficient tumors. Surprisingly, we found that ARID1A targets the nonchromatin substrate Chk2 for ubiquitination. Loss of ARID1A increased the Chk2 level through modulating autoubiquitination of the E3-ligase RNF8 and thereby reducing RNF8-mediated Chk2 degradation. Inhibition of the ATM/Chk2 DNA damage checkpoint axis led to replication stress and accumulation of cytosolic DNA, which subsequently activated the DNA sensor STING-mediated innate immune response in ARID1A-deficient tumors. As expected, tumors with mutation or low expression of both ARID1A and ATM/Chk2 exhibited increased tumor-infiltrating lymphocytes and were associated with longer patient survival. Notably, an ATM inhibitor selectively potentiated the efficacy of immune checkpoint blockade in ARID1A-depleted tumors but not in WT tumors. Together, these results suggest that ARID1A's targeting of the nonchromatin substrate Chk2 for ubiquitination makes it possible to selectively modulate cancer cell-intrinsic innate immunity to enhance the antitumor activity of immune checkpoint blockade.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Quinasa de Punto de Control 2/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias , Neoplasias , Nucleotidiltransferasas/metabolismo , Transducción de Señal , Factores de Transcripción/deficiencia , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Quinasa de Punto de Control 2/genética , Proteínas de Unión al ADN/metabolismo , Células HCT116 , Humanos , Proteínas de la Membrana/genética , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Nucleotidiltransferasas/genética , Proteolisis , Factores de Transcripción/metabolismo
7.
Cell Rep ; 32(13): 108172, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32997990

RESUMEN

Nuclear actin has been elusive due to the lack of knowledge about molecular mechanisms. From actin-containing chromatin remodeling complexes, we discovered an arginine mono-methylation mark on an evolutionarily conserved R256 residue of actin (R256me1). Actin R256 mutations in yeast affect nuclear functions and cause diseases in human. Interestingly, we show that an antibody specific for actin R256me1 preferentially stains nuclear actin over cytoplasmic actin in yeast, mouse, and human cells. We also show that actin R256me1 is regulated by protein arginine methyl transferase-5 (PRMT5) in HEK293 cells. A genome-wide survey of actin R256me1 mark provides a landscape for nuclear actin correlated with transcription. Further, gene expression and protein interaction studies uncover extensive correlations between actin R256me1 and active transcription. The discovery of actin R256me1 mark suggests a fundamental mechanism to distinguish nuclear actin from cytoplasmic actin through post-translational modification (PTM) and potentially implicates an actin PTM mark in transcription and human diseases.


Asunto(s)
Actinas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Factores de Transcripción/metabolismo , Animales , Humanos , Metilación , Ratones
8.
Front Genet ; 9: 398, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319687

RESUMEN

Actin is not only one of the most abundant proteins in eukaryotic cells, but also one of the most versatile. In addition to its familiar involvement in enabling contraction and establishing cellular motility and scaffolding in the cytosol, actin has well-documented roles in a variety of processes within the confines of the nucleus, such as transcriptional regulation and DNA repair. Interestingly, monomeric actin as well as actin-related proteins (Arps) are found as stoichiometric subunits of a variety of chromatin remodeling complexes and histone acetyltransferases, raising the question of precisely what roles they serve in these contexts. Actin and Arps are present in unique combinations in chromatin modifiers, helping to establish structural integrity of the complex and enabling a wide range of functions, such as recruiting the complex to nucleosomes to facilitate chromatin remodeling and promoting ATPase activity of the catalytic subunit. Actin and Arps are also thought to help modulate chromatin dynamics and maintain higher-order chromatin structure. Moreover, the presence of actin and Arps in several chromatin modifiers is necessary for promoting genomic integrity and an effective DNA damage response. In this review, we discuss the involvement of actin and Arps in these nuclear complexes that control chromatin remodeling and histone modifications, while also considering avenues for future study to further shed light on their functional importance.

9.
Nat Med ; 24(5): 556-562, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29736026

RESUMEN

ARID1A (the AT-rich interaction domain 1A, also known as BAF250a) is one of the most commonly mutated genes in cancer1,2. The majority of ARID1A mutations are inactivating mutations and lead to loss of ARID1A expression 3 , which makes ARID1A a poor therapeutic target. Therefore, it is of clinical importance to identify molecular consequences of ARID1A deficiency that create therapeutic vulnerabilities in ARID1A-mutant tumors. In a proteomic screen, we found that ARID1A interacts with mismatch repair (MMR) protein MSH2. ARID1A recruited MSH2 to chromatin during DNA replication and promoted MMR. Conversely, ARID1A inactivation compromised MMR and increased mutagenesis. ARID1A deficiency correlated with microsatellite instability genomic signature and a predominant C>T mutation pattern and increased mutation load across multiple human cancer types. Tumors formed by an ARID1A-deficient ovarian cancer cell line in syngeneic mice displayed increased mutation load, elevated numbers of tumor-infiltrating lymphocytes, and PD-L1 expression. Notably, treatment with anti-PD-L1 antibody reduced tumor burden and prolonged survival of mice bearing ARID1A-deficient but not ARID1A-wild-type ovarian tumors. Together, these results suggest ARID1A deficiency contributes to impaired MMR and mutator phenotype in cancer, and may cooperate with immune checkpoint blockade therapy.


Asunto(s)
Inmunoterapia , Mutación/genética , Neoplasias/genética , Neoplasias/inmunología , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Animales , Línea Celular Tumoral , Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos C57BL , Proteína 2 Homóloga a MutS/metabolismo , Unión Proteica
10.
Am J Hum Genet ; 100(1): 21-30, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-27939641

RESUMEN

Fibromuscular dysplasia (FMD) is a heterogeneous group of non-atherosclerotic and non-inflammatory arterial diseases that primarily involves the renal and cerebrovascular arteries. Grange syndrome is an autosomal-recessive condition characterized by severe and early-onset vascular disease similar to FMD and variable penetrance of brachydactyly, syndactyly, bone fragility, and learning disabilities. Exome-sequencing analysis of DNA from three affected siblings with Grange syndrome identified compound heterozygous nonsense variants in YY1AP1, and homozygous nonsense or frameshift YY1AP1 variants were subsequently identified in additional unrelated probands with Grange syndrome. YY1AP1 encodes yin yang 1 (YY1)-associated protein 1 and is an activator of the YY1 transcription factor. We determined that YY1AP1 localizes to the nucleus and is a component of the INO80 chromatin remodeling complex, which is responsible for transcriptional regulation, DNA repair, and replication. Molecular studies revealed that loss of YY1AP1 in vascular smooth muscle cells leads to cell cycle arrest with decreased proliferation and increased levels of the cell cycle regulator p21/WAF/CDKN1A and disrupts TGF-ß-driven differentiation of smooth muscle cells. Identification of YY1AP1 mutations as a cause of FMD indicates that this condition can result from underlying genetic variants that significantly alter the phenotype of vascular smooth muscle cells.


Asunto(s)
Displasia Fibromuscular/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Mutación , Proteínas Nucleares/genética , Factores de Transcripción/genética , Adolescente , Adulto , Huesos/patología , Braquidactilia/genética , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular , Exoma/genética , Femenino , Genes Recesivos , Heterocigoto , Homocigoto , Humanos , Discapacidades para el Aprendizaje/genética , Masculino , Persona de Mediana Edad , Linaje , Sindactilia/genética , Síndrome
11.
Cell ; 167(3): 600-601, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27768882

RESUMEN

Using a reconstituted system containing genomic DNA and purified proteins from yeast, Krietenstein et al. uncover the direct contributions of key factors in nucleosome positioning and conceptualize the process into four distinct stages.


Asunto(s)
Nucleosomas/metabolismo , Saccharomyces cerevisiae/genética , Ensamble y Desensamble de Cromatina , Genoma , Proteínas de Saccharomyces cerevisiae/genética
12.
Nat Commun ; 7: 11655, 2016 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-27229179

RESUMEN

The ataxia-telangiectasia mutated (ATM) protein is an apical kinase that orchestrates the multifaceted DNA-damage response. Normally, ATM kinase is in an inactive, homodimer form and is transformed into monomers upon activation. Besides a conserved kinase domain at the C terminus, ATM contains three other structural modules, referred to as FAT, FATC and N-terminal helical solenoid. Here we report the first cryo-EM structure of ATM kinase, which is an intact homodimeric ATM/Tel1 from Schizosaccharomyces pombe. We show that two monomers directly contact head-to-head through the FAT and kinase domains. The tandem N-terminal helical solenoid tightly packs against the FAT and kinase domains. The structure suggests that ATM/Tel1 dimer interface and the consecutive HEAT repeats inhibit the binding of kinase substrates and regulators by steric hindrance. Our study provides a structural framework for understanding the mechanisms of ATM/Tel1 regulation as well as the development of new therapeutic agents.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/química , Conformación Proteica , Multimerización de Proteína , Proteínas Serina-Treonina Quinasas/química , Proteínas de Schizosaccharomyces pombe/química , Secuencia de Aminoácidos , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Sitios de Unión/genética , Microscopía por Crioelectrón , Modelos Moleculares , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Homología de Secuencia de Aminoácido
14.
Cancer Discov ; 5(7): 752-67, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26069190

RESUMEN

UNLABELLED: ARID1A, SWI/SNF chromatin remodeling complex subunit, is a recently identified tumor suppressor that is mutated in a broad spectrum of human cancers. Thus, it is of fundamental clinical importance to understand its molecular functions and determine whether ARID1A deficiency can be exploited therapeutically. In this article, we report a key function of ARID1A in regulating the DNA damage checkpoint. ARID1A is recruited to DNA double-strand breaks (DSB) via its interaction with the upstream DNA damage checkpoint kinase ATR. At the molecular level, ARID1A facilitates efficient processing of DSB to single-strand ends and sustains DNA damage signaling. Importantly, ARID1A deficiency sensitizes cancer cells to PARP inhibitors in vitro and in vivo, providing a potential therapeutic strategy for patients with ARID1A-mutant tumors. SIGNIFICANCE: ARID1A has been identified as one of the most frequently mutated genes across human cancers. Our data suggest that clinical utility of PARP inhibitors might be extended beyond patients with BRCA mutations to a larger group of patients with ARID1A-mutant tumors, which may exhibit therapeutic vulnerability to PARP inhibitors.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Daño del ADN , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Nucleares/deficiencia , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Factores de Transcripción/deficiencia , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN , Femenino , Células HCT116 , Humanos , Neoplasias Pulmonares/genética , Masculino , Ratones , Proteínas Nucleares/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Factores de Transcripción/química , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Mol Cell ; 58(5): 863-9, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25959398

RESUMEN

ATP-dependent chromatin remodeling complexes such as INO80 have been implicated in checkpoint regulation in response to DNA damage. However, how chromatin remodeling complexes regulate DNA damage checkpoints remain unclear. Here, we identified a mechanism of regulating checkpoint effector kinase Rad53 through a direct interaction with the INO80 chromatin remodeling complex. Rad53 is a key checkpoint kinase downstream of Mec1. Mec1/Tel1 phosphorylates the Ies4 subunit of the INO80 complex in response to DNA damage. We find that the phosphorylated Ies4 binds to the N-terminal FHA domain of Rad53. In vitro, INO80 can activate Rad53 kinase activity in an Ies4-phosphorylation-dependent manner in the absence of known activators such as Rad9. In vivo, Ies4 and Rad9 function synergistically to activate Rad53. These findings establish a direct connection between ATP-dependent chromatin remodeling complexes and checkpoint regulation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Quinasa de Punto de Control 2/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/enzimología , Secuencia de Aminoácidos , Activación Enzimática , Datos de Secuencia Molecular , Fosforilación , Proteolisis
16.
Genes Dev ; 29(6): 591-602, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25792597

RESUMEN

ATP-dependent chromatin remodeling complexes alter chromatin structure through interactions with chromatin substrates such as DNA, histones, and nucleosomes. However, whether chromatin remodeling complexes have the ability to regulate nonchromatin substrates remains unclear. Saccharomyces cerevisiae checkpoint kinase Mec1 (ATR in mammals) is an essential master regulator of genomic integrity. Here we found that the SWI/SNF chromatin remodeling complex is capable of regulating Mec1 kinase activity. In vivo, Mec1 activity is reduced by the deletion of Snf2, the core ATPase subunit of the SWI/SNF complex. SWI/SNF interacts with Mec1, and cross-linking studies revealed that the Snf2 ATPase is the main interaction partner for Mec1. In vitro, SWI/SNF can activate Mec1 kinase activity in the absence of chromatin or known activators such as Dpb11. The subunit requirement of SWI/SNF-mediated Mec1 regulation differs from that of SWI/SNF-mediated chromatin remodeling. Functionally, SWI/SNF-mediated Mec1 regulation specifically occurs in S phase of the cell cycle. Together, these findings identify a novel regulator of Mec1 kinase activity and suggest that ATP-dependent chromatin remodeling complexes can regulate nonchromatin substrates such as a checkpoint kinase.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Factores de Transcripción/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Ensamble y Desensamble de Cromatina , Daño del ADN/fisiología , Activación Enzimática , Activadores de Enzimas/metabolismo , Fase S , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/genética
17.
Trends Cell Biol ; 24(4): 238-46, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24246764

RESUMEN

The mystery of nuclear actin has puzzled biologists for decades largely due to the lack of defined experimental systems. However, the development of actin-containing chromatin-modifying complexes as a defined genetic and biochemical system in the past decade has provided an unprecedented opportunity to dissect the mechanism of actin in the nucleus. Although the established functions of actin mostly rely on its dynamic polymerization, the novel finding of the mechanism of action of actin in the INO80 chromatin-remodeling complex suggests a conceptually distinct mode of actin that functions as a monomer. In this review we highlight the new paradigm and discuss how actin interaction with chromatin suggests a fundamental divergence between conventional cytoplasmic actin and nuclear actin. Furthermore, we provide how this framework could be applied to investigations of nuclear actin in other actin-containing chromatin-modifying complexes.


Asunto(s)
Actinas/metabolismo , Núcleo Celular/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Cromatina/metabolismo , Animales , Histonas/metabolismo , Humanos , Modelos Biológicos
18.
Nat Struct Mol Biol ; 20(4): 426-32, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23524535

RESUMEN

Actin has well-established functions in the cytoplasm, but its roles in the nucleus remain poorly defined. Here, by studying the nuclear actin-containing yeast INO80 chromatin remodeling complex, we provide genetic and biochemical evidence for a role of monomeric actin in INO80 chromatin remodeling. We demonstrate that, in contrast to cytoplasmic actin, nuclear actin is present as a monomer in the INO80 complex, and its barbed end is not accessible for polymerization. We identify an actin mutation in subdomain 2 affecting in vivo nuclear functions and reducing the chromatin remodeling activity of the INO80 complex in vitro. Notably, the highly conserved subdomain 2 at the pointed end of actin contributes to the interaction of INO80 with chromatin. Our results establish an evolutionarily conserved function of nuclear actin in its monomeric form and suggest that nuclear actin can utilize a fundamentally distinct mechanism from that of cytoplasmic actin.


Asunto(s)
Actinas/fisiología , Ensamble y Desensamble de Cromatina , Proteínas de Saccharomyces cerevisiae/química , Actinas/química , Secuencia de Aminoácidos , Animales , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
19.
Front Biosci (Landmark Ed) ; 17(3): 970-5, 2012 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-22201784

RESUMEN

ATP-dependent chromatin remodeling complexes are involved in several nuclear processes. In particular the INO80 remodeling complex is an essential factor during transcription and DNA repair. Recently, several labs have described a novel role for INO80 during DNA replication. Moreover, Falbo et al. have presented evidence linking INO80's activities to the DNA damage tolerance pathways during replication (1). In this review we will discuss and integrate the results obtain by these various research groups to describe a novel role for INO80 in DNA replication.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Replicación del ADN/fisiología , Proteínas de Saccharomyces cerevisiae/metabolismo , Daño del ADN , ADN de Hongos/biosíntesis , Genes Fúngicos , Humanos , Modelos Biológicos , Mutación , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA