Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 185
Filtrar
1.
Mol Cell ; 76(4): 617-631.e4, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31564557

RESUMEN

Spt5 is a conserved and essential transcription elongation factor that promotes promoter-proximal pausing, promoter escape, elongation, and mRNA processing. Spt5 plays specific roles in the transcription of inflammation and stress-induced genes and tri-nucleotide expanded-repeat genes involved in inherited neurological pathologies. Here, we report the identification of Spt5-Pol II small-molecule inhibitors (SPIs). SPIs faithfully reproduced Spt5 knockdown effects on promoter-proximal pausing, NF-κB activation, and expanded-repeat huntingtin gene transcription. Using SPIs, we identified Spt5 target genes that responded with profoundly diverse kinetics. SPIs uncovered the regulatory role of Spt5 in metabolism via GDF15, a food intake- and body weight-inhibitory hormone. SPIs further unveiled a role for Spt5 in promoting the 3' end processing of histone genes. While several SPIs affect all Spt5 functions, a few inhibit a single one, implying uncoupling and selective targeting of Spt5 activities. SPIs expand the understanding of Spt5-Pol II functions and are potential drugs against metabolic and neurodegenerative diseases.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , ARN Polimerasa II/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Factores de Elongación Transcripcional/antagonistas & inhibidores , Regiones no Traducidas 3' , Animales , Núcleo Celular/enzimología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Descubrimiento de Drogas/métodos , Metabolismo Energético/efectos de los fármacos , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/metabolismo , Células HEK293 , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Histonas/genética , Histonas/metabolismo , Humanos , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Células Jurkat , Células MCF-7 , Ratones Transgénicos , Mutación , FN-kappa B/biosíntesis , FN-kappa B/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN Polimerasa II/genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
2.
Cell ; 143(5): 737-49, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21111234

RESUMEN

Sister chromatid cohesion is essential for chromosome segregation and is mediated by cohesin bound to DNA. Cohesin-DNA interactions can be reversed by the cohesion-associated protein Wapl, whereas a stably DNA-bound form of cohesin is thought to mediate cohesion. In vertebrates, Sororin is essential for cohesion and stable cohesin-DNA interactions, but how Sororin performs these functions is unknown. We show that DNA replication and cohesin acetylation promote binding of Sororin to cohesin, and that Sororin displaces Wapl from its binding partner Pds5. In the absence of Wapl, Sororin becomes dispensable for cohesion. We propose that Sororin maintains cohesion by inhibiting Wapl's ability to dissociate cohesin from DNA. Sororin has only been identified in vertebrates, but we show that many invertebrate species contain Sororin-related proteins, and that one of these, Dalmatian, is essential for cohesion in Drosophila. The mechanism we describe here may therefore be widely conserved among different species.


Asunto(s)
Cromátides/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Replicación del ADN , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/química , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/química , Humanos , Fase S , Xenopus/metabolismo , Cohesinas
3.
Genes Dev ; 31(3): 241-246, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28270516

RESUMEN

Chromobox homolog 3 (Cbx3/heterochromatin protein 1γ [HP1γ]) stimulates cell differentiation, but its mechanism is unknown. We found that Cbx3 binds to gene promoters upon differentiation of murine embryonic stem cells (ESCs) to neural progenitor cells (NPCs) and recruits the Mediator subunit Med26. RNAi knockdown of either Cbx3 or Med26 inhibits neural differentiation while up-regulating genes involved in mesodermal lineage decisions. Thus, Cbx3 and Med26 together ensure the fidelity of lineage specification by enhancing the expression of neural genes and down-regulating genes specific to alternative fates.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Proteínas Cromosómicas no Histona/metabolismo , Células Madre Embrionarias/citología , Regulación de la Expresión Génica , Complejo Mediador/metabolismo , Células-Madre Neurales/citología , Animales , Células Cultivadas , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/genética , Quinasa 8 Dependiente de Ciclina/genética , Quinasa 8 Dependiente de Ciclina/metabolismo , Células Madre Embrionarias/metabolismo , Complejo Mediador/genética , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética
4.
Org Biomol Chem ; 22(23): 4651-4655, 2024 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-38787760

RESUMEN

An arylazopyrazole-based covalent inhibitor targeting the mitotic motor protein of centromere-associated protein E (CENP-E) was developed. Using this photoswitchable inhibitor, a photoswitchable CENP-E was chemically constructed in cells, which enabled to local control of mitotic cell division with light illumination.


Asunto(s)
Proteínas Cromosómicas no Histona , Humanos , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/metabolismo , Procesos Fotoquímicos , Luz , Pirazoles/química , Pirazoles/farmacología , Pirazoles/síntesis química , Células HeLa , Estructura Molecular , Mitosis/efectos de los fármacos
5.
J Virol ; 95(18): e0057421, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34191584

RESUMEN

Noncanonical poly(A) polymerases PAPD5 and PAPD7 (PAPD5/7) stabilize hepatitis B virus (HBV) RNA via the interaction with the viral posttranscriptional regulatory element (PRE), representing new antiviral targets to control HBV RNA metabolism, hepatitis B surface antigen (HBsAg) production, and viral replication. Inhibitors targeting these proteins are being developed as antiviral therapies; therefore, it is important to understand how PAPD5/7 coordinate to stabilize HBV RNA. Here, we utilized a potent small-molecule AB-452 as a chemical probe, along with genetic analyses to dissect the individual roles of PAPD5/7 in HBV RNA stability. AB-452 inhibits PAPD5/7 enzymatic activities and reduces HBsAg both in vitro (50% effective concentration [EC50] ranged from 1.4 to 6.8 nM) and in vivo by 0.94 log10. Our genetic studies demonstrate that the stem-loop alpha sequence within PRE is essential for both maintaining HBV poly(A) tail integrity and determining sensitivity toward the inhibitory effect of AB-452. Although neither single knockout (KO) of PAPD5 nor PAPD7 reduces HBsAg RNA and protein production, PAPD5 KO does impair poly(A) tail integrity and confers partial resistance to AB-452. In contrast, PAPD7 KO did not result in any measurable changes within the HBV poly(A) tails, but cells with both PAPD5 and PAPD7 KO show reduced HBsAg production and conferred complete resistance to AB-452 treatment. Our results indicate that PAPD5 plays a dominant role in stabilizing viral RNA by protecting the integrity of its poly(A) tail, while PAPD7 serves as a second line of protection. These findings inform PAPD5-targeted therapeutic strategies and open avenues for further investigating PAPD5/7 in HBV replication. IMPORTANCE Chronic hepatitis B affects more than 250 million patients and is a major public health concern worldwide. HBsAg plays a central role in maintaining HBV persistence, and as such, therapies that aim at reducing HBsAg through destabilizing or degrading HBV RNA have been extensively investigated. Besides directly degrading HBV transcripts through antisense oligonucleotides or RNA silencing technologies, small-molecule compounds targeting host factors such as the noncanonical poly(A) polymerase PAPD5 and PAPD7 have been reported to interfere with HBV RNA metabolism. Herein, our antiviral and genetic studies using relevant HBV infection and replication models further characterize the interplays between the cis element within the viral sequence and the trans elements from the host factors. PAPD5/7-targeting inhibitors, with oral bioavailability, thus represent an opportunity to reduce HBsAg through destabilizing HBV RNA.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Virus de la Hepatitis B/genética , Hepatitis B/virología , ARN Nucleotidiltransferasas/metabolismo , Estabilidad del ARN , ARN Viral/química , Replicación Viral , Animales , Antivirales/farmacología , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/genética , ADN Polimerasa Dirigida por ADN/genética , Inhibidores Enzimáticos/farmacología , Células Hep G2 , Hepatitis B/genética , Hepatitis B/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Nucleotidiltransferasas/antagonistas & inhibidores , ARN Nucleotidiltransferasas/genética , ARN Viral/genética
6.
Biochem J ; 478(4): 799-810, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33522570

RESUMEN

A key nucleolar protein, fibrillarin, has emerged as an important pharmacological target as its aberrant expression and localization are related to tumorigenesis, chemoresistance and poor survival in breast cancer patients. Fibrillarin contains a N-terminal low complexity sequence (LC) domain with a skewed amino acid distribution, which is known to undergo a phase transition to liquid-like droplets. However, the underlying mechanism of the phase transition of the fibrillarin LC domain and its physiological function are still elusive. In this study, we show that the localization of fibrillarin and its association with RNA binding proteins is regulated by this phase transition. Phenylalanine-to-serine substitutions of the phenylalanine:glycine repeats in the fibrillarin LC domain impede its phase transition into liquid-like droplets, as well as the hydrogel-like state composed of polymers, and also its incorporation into hydrogel or liquid-like droplets composed of wild-type LC domains. When expressed in cultured cells, fibrillarin containing the mutant LC domain fails to localize to the dense fibrillar component of nucleoli in the same way as intact fibrillarin. Moreover, the phase transition of the fibrillarin LC domain is required for the interaction of fibrillarin with other RNA binding proteins, such as FUS, TAF15, DDX5 and DHX9. Taken together, the results suggest that the phenylalanine residues in the LC domain are critical for the phase transition of fibrillarin, which in turn regulates the sub-nucleolar localization of fibrillarin and its interaction with RNA binding proteins, providing a useful framework for regulating the function of fibrillarin.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Sustitución de Aminoácidos , Nucléolo Celular/metabolismo , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Hidrogeles , Mutación Missense , Transición de Fase , Fenilalanina/química , Mutación Puntual , Unión Proteica , Dominios Proteicos , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes/metabolismo
7.
Toxicol Appl Pharmacol ; 423: 115568, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-33965371

RESUMEN

N-methyl pyrrolidone (NMP) is an FDA approved molecule used as an excipient in pharmaceutical industry. Besides having a central role in formulation of drugs, the most important function of any excipient is to guarantee the safety of the medicine during and after its administration. Several studies have shown that exposure to NMP and especially in rats produce a gonadotoxic effect leading to infertility. However, the mechanisms underlying the effect of NMP on male reproduction are unknown. The aim of this study was to assess the reproductive toxicity of NMP in male rats and to elucidate the underlying mechanism. Male Sprague Dawley rats were injected intraperitoneally, twice/ week, at a dose of 108 mg/ 100 g of body weight with NMP. Analysis of reproductive parameters revealed testicular atrophy in NMP treated animals compared to control animals. Germ cell composition within the seminiferous tubules was disturbed and manifested in an increase in number of cells with fragmented DNA. A subsequent decrease in number of spermatocytes and spermatids was observed. Alpha screen assay shows that NMP acts at the concentrations we applied in vivo as a low affinity inhibitor for BRDT (testis specific bromodomain protein). BRDT inhibition is mirrored by a significant decrease in the expression of early stage spermatocyte markers (lmna, aurkc and ccna1), during which BRDT expression predominates. A significant decrease in testosterone levels was also observed. Since NMP interferes with spermatogenesis on various levels, its use in humans must be carefully monitored.


Asunto(s)
Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/metabolismo , Pirrolidinonas/toxicidad , Espermatogénesis/efectos de los fármacos , Teratógenos/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Hormona Folículo Estimulante/sangre , Masculino , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Espermatogénesis/fisiología , Testosterona/sangre
8.
Bioorg Med Chem Lett ; 33: 127745, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33333161

RESUMEN

BAZ1A is a bromodomain-containing protein, and has been recognized as a potential target for multiple diseases, particularly cancer. However, there is no BAZ1A inhibitor reported so far. In this study, we used a consensus docking/scoring strategy to screen for BAZ1A bromodomain inhibitors from commercial chemical libraries and an in-house chemical database. The retrieved hit compounds were evaluated experimentally and four compounds were found to be active against BAZ1A bromodomain. To the most active compounds, similarity and substructure searches were used to find more BAZ1A bromodomain inhibitors. Among all the obtained active compounds, Cpd-2 is the most potent one, which showed a KD value of 0.52 µM. The interaction model of Cpd-2 with BAZ1A bromodomain was revealed by molecular docking. In a cellular assay, Cpd-2 displayed good anti-viability activity against cancer cell lines expressing a high level of BAZ1A. Overall, we discovered a number of BAZ1A bromodomain inhibitors for the first time, which can be a good starting point for subsequent drug discovery targeting BAZ1A bromodomain.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Descubrimiento de Drogas , Compuestos Orgánicos/farmacología , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Compuestos Orgánicos/química , Relación Estructura-Actividad
9.
J Cell Mol Med ; 24(23): 13739-13750, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33124760

RESUMEN

This study is to investigate the inhibitory effects and mechanisms of DEK-targeting aptamer (DTA-64) on epithelial mesenchymaltransition (EMT)-mediated airway remodelling in mice and human bronchial epithelial cell line BEAS-2B. In the ovalbumin (OVA)-induced asthmatic mice, DTA-64 significantly reduced the infiltration of eosinophils and neutrophils in lung tissue, attenuated the airway resistance and the proliferation of goblet cells. In addition, DTA-64 reduced collagen deposition, transforming growth factor 1 (TGF-ß1) level in BALF and IgE levels in serum, balanced Th1/Th2/Th17 ratio, and decreased mesenchymal proteins (vimentin and α-SMA), as well as weekend matrix metalloproteinases (MMP-2 and MMP-9) and NF-κB p65 activity. In the in vitro experiments, we used TGF-ß1 to induce EMT in the human epithelial cell line BEAS-2B. DEK overexpression (ovDEK) or silencing (shDEK) up-regulated or down-regulated TGF-ß1 expression, respectively, on the contrary, TGF-ß1 exposure had no effect on DEK expression. Furthermore, ovDEK and TGF-ß1 synergistically promoted EMT, whereas shDEK significantly reduced mesenchymal markers and increased epithelial markers, thus inhibiting EMT. Additionally, shDEK inhibited key proteins in TGF-ß1-mediated signalling pathways, including Smad2/3, Smad4, p38 MAPK, ERK1/2, JNK and PI3K/AKT/mTOR. In conclusion, the effects of DTA-64 against EMT of asthmatic mice and BEAS-2B might partially be achieved through suppressing TGF-ß1/Smad, MAPK and PI3K signalling pathways. DTA-64 may be a new therapeutic option for the management of airway remodelling in asthma patients.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Asma/etiología , Asma/metabolismo , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Transición Epitelial-Mesenquimal/efectos de los fármacos , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Animales , Asma/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores , Susceptibilidad a Enfermedades , Transición Epitelial-Mesenquimal/genética , Femenino , Silenciador del Gen , Humanos , Inmunoglobulina E/inmunología , Inmunoglobulina E/metabolismo , Inmunomodulación/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Ovalbúmina/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Proteínas Smad/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
10.
J Pharmacol Exp Ther ; 373(2): 279-289, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32102917

RESUMEN

Cholangiocarcinoma (CCA) is a malignant tumor that arises from the epithelial cells of the bile duct and is notorious for its poor prognosis. The clinical outcome remains disappointing, and thus more effective therapeutic options are urgently required. Cordycepin, a traditional Chinese medicine, provides multiple pharmacological strategies in antitumors, but its mechanisms have not been fully elucidated. In this study, we reported that cordycepin inhibited the viability and proliferation capacity of CCA cells in a time- and dose-dependent manner determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and colony formation assay. Flow cytometry and Hoechst dye showed that cordycepin induced cancer cell apoptosis via extracellular signal-regulated kinase (ERK) 1/2 deactivation. Moreover, cordycepin significantly reduced the angiogenetic capabilities of CCA in vitro as examined by tube formation assay. We also discovered that cordycepin inhibited DEK expression by using Western blot assay. DEK serves as an oncogenic protein that is overexpressed in various gastrointestinal tumors. DEK silencing inhibited CCA cell viability and angiogenesis but not apoptosis induction determined by Western blot and flow cytometry. Furthermore, cordycepin significantly inhibited tumor growth and angiogenic capacities in a xenograft model by downregulating the expression of DEK, phosphorylated ERK1/2 CD31 and von Willebrand factor (vWF). Taken together, we demonstrated that cordycepin inhibited CCA cell proliferation and angiogenesis with a DEK interaction via downregulation in ERK signaling. These data indicate that cordycepin may serve as a novel agent for CCA clinical treatment and prognosis improvement. SIGNIFICANCE STATEMENT: Cordycepin provides multiple strategies in antitumors, but its mechanisms are not fully elucidated, especially on cholangiocarcinoma (CCA). We reported that cordycepin inhibited the viability of CCA cells, induced apoptosis via extracellular signal-regulated kinase 1/2 deactivation and DEK inhibition, and reduced the angiogenetic capabilities of CCA both in vivo and in vitro.


Asunto(s)
Neoplasias de los Conductos Biliares/tratamiento farmacológico , Colangiocarcinoma/tratamiento farmacológico , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Desoxiadenosinas/farmacología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neovascularización Patológica/prevención & control , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Animales , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colangiocarcinoma/patología , Humanos , Masculino , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Control ; 27(1): 1073274820968914, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33153318

RESUMEN

Opa-interacting protein 5 (OIP5) is a member of the cancer-testis antigen (CTA) family that elicits a spontaneous antitumor immune response. The failure of current immunotherapies for glioma has prompted the search for novel biomarkers that may be utilized as therapeutic targets. This study aimed to investigate whether OIP5 serves as a target for malignant glioma immunotherapy. Glioma specimens from 53 adult patients were evaluated for OIP5 expression by immunohistochemical (IHC) staining, and the correlation of OIP5 expression with World Health Organization (WHO) tumor grade was analyzed. Endogenous expression of OIP5 in glioma cell lines was determined via real-time polymerase chain reaction (RT-PCR). Using lentiviral siOIP5, the effect of OIP5 gene knockdown on proliferation, cell cycle, and apoptosis in U251 glioma cells was studied. The results show that OIP5 is overexpressed in glioma tissues and is correlated with WHO tumor grade (P < 0.001). However, OIP5 protein expression is barely detectable in normal adult brain tissues. MTT assays and analysis using the Celigo Imaging Cytometry System reveal that the silencing of OIP5 inhibits U251 cell growth. Cell cycle assays and Annexin V staining show that OIP5 silencing disrupts the balance of the cell cycle and increases U251 cell death. These results indicate that OIP5 is upregulated in malignant glioma specimens but barely detected in normal brain tissues. OIP5 knockdown inhibits the biological function of glioma cells, reinforcing that OIP5 may serve as an immunotherapeutic target for malignant glioma.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Encéfalo/patología , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Glioma/diagnóstico , Adulto , Anciano , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/cirugía , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/cirugía , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Técnicas de Silenciamiento del Gen , Glioma/tratamiento farmacológico , Glioma/inmunología , Glioma/cirugía , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología , Adulto Joven
12.
Mol Ther ; 27(12): 2182-2194, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31451355

RESUMEN

Long noncoding RNAs (lncRNAs) are an important class of pervasive noncoding RNA involved in a variety of biological functions. Numerous studies have demonstrated their important regulatory role in human disease, especially cancer. However, the mechanism underlying the transcription of lncRNAs is not fully elucidated. Here, a comparison of local chromatin structure of the ROR lncRNA locus revealed a cohesin-complex-mediated intrachromosomal loop that is juxtaposed with an upstream enhancer to the ROR promoter, enabling activation of endogenous ROR lncRNA in tumor cells. This chromosomal interaction was not observed in normal control cells. Knockdown of SMC1 by RNAi or deletion of the enhancer DNA by CRISPR/Cas9 abolished the intrachromosomal interaction, resulting in ROR lncRNA silencing and inhibition of the tumor progression in animals carrying tumor xenografts. Our results reveal a novel mechanism by which the cohesin-orchestrated intrachromosomal looping may serve as a critical epigenetic driver to activate transcription of ROR lncRNA, subsequently inducing tumorigenesis. Our data represent a novel chromosomal folding pattern of lncRNA regulation, thereby providing a novel alternative concept of chromosomal interaction in lncRNA-triggered tumorigenesis.


Asunto(s)
Carcinogénesis/patología , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Cromosomas Humanos , Complejo Mediador/genética , Neoplasias/patología , ARN Largo no Codificante/genética , ARN Interferente Pequeño/genética , Animales , Apoptosis , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proliferación Celular , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Complejo Mediador/antagonistas & inhibidores , Ratones , Ratones Desnudos , Neoplasias/genética , Neoplasias/metabolismo , Regiones Promotoras Genéticas , ARN Largo no Codificante/antagonistas & inhibidores , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Trends Biochem Sci ; 40(8): 468-79, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26145250

RESUMEN

Bromodomains (BRDs) are evolutionarily conserved protein interaction modules that specifically recognise acetyl-lysine on histones and other proteins, facilitating roles in regulating gene transcription. BRD-containing proteins bound to chromatin loci such as enhancers are often deregulated in disease leading to aberrant expression of proinflammatory cytokines and growth-promoting genes. Recent developments targeting the bromo and extraterminal (BET) subset of BRD proteins demonstrated remarkable efficacy in murine models providing a compelling rationale for drug development and translation to the clinic. Here we summarise recent advances in our understanding of the roles of BETs in regulating gene transcription in normal and diseased tissue as well as the current status of their clinical translation.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Enfermedad , Estructura Terciaria de Proteína , Transcripción Genética , Animales , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Humanos , Modelos Moleculares , Transcripción Genética/genética
14.
J Biol Chem ; 293(10): 3663-3674, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29358331

RESUMEN

The pluripotency-controlling stem-cell protein SRY-box 2 (SOX2) plays a pivotal role in maintaining the self-renewal and pluripotency of embryonic stem cells and also of teratocarcinoma or embryonic carcinoma cells. SOX2 is monomethylated at lysine 119 (Lys-119) in mouse embryonic stem cells by the SET7 methyltransferase, and this methylation triggers ubiquitin-dependent SOX2 proteolysis. However, the molecular regulators and mechanisms controlling SET7-induced SOX2 proteolysis are unknown. Here, we report that in human ovarian teratocarcinoma PA-1 cells, methylation-dependent SOX2 proteolysis is dynamically regulated by the LSD1 lysine demethylase and a methyl-binding protein, PHD finger protein 20-like 1 (PHF20L1). We found that LSD1 not only removes the methyl group from monomethylated Lys-117 (equivalent to Lys-119 in mouse SOX2), but it also demethylates monomethylated Lys-42 in SOX2, a reaction that SET7 also regulated and that also triggered SOX2 proteolysis. Our studies further revealed that PHF20L1 binds both monomethylated Lys-42 and Lys-117 in SOX2 and thereby prevents SOX2 proteolysis. Down-regulation of either LSD1 or PHF20L1 promoted SOX2 proteolysis, which was prevented by SET7 inactivation in both PA-1 and mouse embryonic stem cells. Our studies also disclosed that LSD1 and PHF20L1 normally regulate the growth of pluripotent mouse embryonic stem cells and PA-1 cells by preventing methylation-dependent SOX2 proteolysis. In conclusion, our findings reveal an important mechanism by which the stability of the pluripotency-controlling stem-cell protein SOX2 is dynamically regulated by the activities of SET7, LSD1, and PHF20L1 in pluripotent stem cells.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Histona Demetilasas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/metabolismo , Procesamiento Proteico-Postraduccional , Factores de Transcripción SOXB1/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Células Cultivadas , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Células HEK293 , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/química , Histona Demetilasas/genética , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Metilación , Ratones Endogámicos C57BL , Mutación , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Estabilidad Proteica , Proteolisis , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción SOXB1/química , Factores de Transcripción SOXB1/genética , Teratocarcinoma/enzimología , Teratocarcinoma/metabolismo , Teratocarcinoma/patología
15.
J Am Chem Soc ; 141(29): 11497-11505, 2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31246451

RESUMEN

Bromodomains, epigenetic "readers" of lysine acetylation marks, exist in different nuclear proteins with diverse biological functions in chromatin biology. Malfunctions of bromodomains are associated with the pathogenesis of human diseases, such as cancer. Bromodomains have therefore emerged as therapeutic targets for drug discovery. Given the high structural similarity of bromodomains, a critical step in the development of bromodomain inhibitors is the evaluation of their selectivity to avoid off-target effects. While numerous bromodomain inhibitors have been identified, new methods to evaluate the inhibitor selectivity toward endogenous bromodomains in living cells remain needed. Here we report the development of a photoaffinity probe, photo-bromosporine (photo-BS), that enables the wide-spectrum profiling of bromodomain inhibitors in living cells. Photo-BS allowed light-induced cross-linking of recombinant bromodomains and endogenous bromodomain-containing proteins (BCPs) both in vitro and in living cells. The photo-BS-induced labeling of the bromodomains was selectively competed by the corresponding bromodomain inhibitors. Proteomics analysis revealed that photo-BS captured 28 out of the 42 known BCPs from the living cells. Assessment of the two bromodomain inhibitors, bromosporine and GSK6853, resulted in the identification of known as well as previously uncharacterized bromodomain targets. Collectively, we established a chemical proteomics platform to comprehensively evaluate bromodomain inhibitors in terms of their selectivity against endogenous BCPs in living cells.


Asunto(s)
Carbamatos/química , Evaluación Preclínica de Medicamentos/métodos , Etiquetas de Fotoafinidad/química , Dominios Proteicos , Proteínas/química , Proteómica/métodos , Piridazinas/química , Triazoles/química , Carbamatos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/química , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/química , Reactivos de Enlaces Cruzados/química , Células HEK293 , Humanos , Espectrometría de Masas/métodos , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Piridazinas/farmacología , Proteínas Recombinantes/química , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química , Triazoles/farmacología
16.
Biochem Biophys Res Commun ; 519(3): 505-511, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31530389

RESUMEN

Kinesin centromere-associated protein E (CENP-E) has emerged as a potential target for the development of anticancer drugs due to its involvement in the mitotic progression of the cell cycle. Although several CENP-E inhibitors have been reported, more knowledge of chemical structures and inhibitory mechanisms is necessary for developing CENP-E inhibitors. Here, we describe the identification of new CENP-E inhibitors. Screening of a small-molecule chemical library identified benzo[d]pyrrolo[2,1-b]thiazole derivatives, including 1, as compounds with inhibitory activity against the microtubule-stimulated ATPase of the CENP-E motor domain. Among the mitotic kinesins examined, 1 selectively inhibited the kinesin ATPase activity of CENP-E. In a steady-state ATPase assay, 1 exhibited ATP-competitive behavior, which was different from the CENP-E inhibitor GSK923295. Compound 1 inhibited the proliferation of tumor-derived HeLa and HCT116 cells more efficiently than that of non-cancerous WI-38 cells. The inhibition of cell proliferation was attributed to the ability of 1 to induce apoptotic cell death. The compound showed antimitotic activity, which caused cell cycle arrest at mitosis via interference with proper chromosome alignment. We identified 1 and its derivatives as the lead compounds that target CENP-E, thus providing a new opportunity for the development of anticancer agents targeting kinesins.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Sarcosina/análogos & derivados , Antineoplásicos/química , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Cromosómicas no Histona/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HCT116 , Células HeLa , Humanos , Estructura Molecular , Sarcosina/química , Sarcosina/farmacología , Relación Estructura-Actividad , Células Tumorales Cultivadas
17.
Biochem Biophys Res Commun ; 516(3): 999-1006, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31277942

RESUMEN

Regulator of chromosome condensation 2 (RCC2) is a regulator of cell-cycle progression linked in multiple cancers to pro-tumorigenic phenomena including promotion of tumor growth, tumor metastases and poorer patient prognoses. However, the role of RCC2 in GBM remains under-investigated. Here, we sought to determine the relevance of RCC2 in GBM, as well as its roles in GBM development, progression and prognosis. Initial clinical evaluation determined significant RCC2 enrichment in GBM when compared to normal brain tissue, and elevated expression was closely associated with a poorer prognosis in glioma patients. Via shRNA inhibition, we determined that RCC2 is essential to tumor proliferation and tumorigenicity in vitro and in vivo. Additionally, RCC2 was determined to promote radioresistance of GBM tumor cells. Investigation of the underlying mechanisms implicated DNA mismatch repair, JAK-STAT pathway and activated transcription of DNA methyltransferase 1 (DNMT1). For validation, pharmacologic inhibition via administration of a DNMT1 inhibitor demonstrated attenuated GBM tumor growth both in vitro and in vivo. Collectively, this study determined a novel therapeutic target for GBM in the form of RCC2, which plays a pivotal role in GBM proliferation and radio-resistance via regulation of DNMT1 expression in a p-STAT3 dependent manner.


Asunto(s)
Neoplasias Encefálicas/genética , Proteínas Cromosómicas no Histona/genética , ADN (Citosina-5-)-Metiltransferasa 1/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Factores de Intercambio de Guanina Nucleótido/genética , Tolerancia a Radiación/genética , Animales , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Decitabina/farmacología , Progresión de la Enfermedad , Inhibidores Enzimáticos/farmacología , Glioblastoma/mortalidad , Glioblastoma/patología , Glioblastoma/terapia , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/metabolismo , Xenoinjertos , Humanos , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Ratones , Ratones SCID , Clasificación del Tumor , Neuroglía/metabolismo , Neuroglía/patología , Neuroglía/efectos de la radiación , Pronóstico , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Análisis de Supervivencia , Transcripción Genética
18.
Biochem Biophys Res Commun ; 517(3): 491-498, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31376935

RESUMEN

Dexamethasone (DEX) induces significant cytotoxicity to human osteoblasts. cPWWP2A is recently-indentified novel circular RNA (circRNA), acting as an endogenous sponge of microRNA-579 (miR-579). The present study tested the expression and potential functions of the cPWWP2A-miR-579 axis in DEX-treated osteoblasts. We show that cPWWP2A is downregulated in the necrotic femoral head tissues of DEX-taking human patients as well as in DEX-treated human osteoblasts. In OB-6 osteoblastic cells and primary human osteoblasts ectopic overexpression of cPWWP2A potently inhibited DEX-induced miR-579 accumulation, cell death, apoptosis and programmed necrosis. Silencing miR-579, by targeted siRNAs, also attenuated DEX-induced cytotoxicity in human osteoblasts. Significantly, mimicking DEX-induced actions, cPWWP2A silencing or forced miR-579 overexpression induced significant cytotoxicity in human osteoblasts. Further analyses demonstrated that miR-579's targets, including SIRT1 and PDK1 (phosphoinositide-dependent protein kinase 1), were downregulated in DEX-treated osteoblasts. Their levels were decreased as well in the necrotic femoral head tissues of DEX-taking human patients. Taken together we show that dysregulation of the cPWWP2A-miR-579 axis is involved in DEX-induced cytotoxicity in human osteoblasts.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Dexametasona/toxicidad , MicroARNs/genética , Necrosis/genética , Osteoblastos/efectos de los fármacos , ARN Circular/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Secuencia de Bases , Línea Celular , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/metabolismo , Fémur/efectos de los fármacos , Fémur/metabolismo , Fémur/patología , Regulación de la Expresión Génica , Humanos , MicroARNs/metabolismo , Necrosis/inducido químicamente , Necrosis/metabolismo , Necrosis/patología , Osteoblastos/metabolismo , Osteoblastos/patología , Cultivo Primario de Células , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , ARN Circular/antagonistas & inhibidores , ARN Circular/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Transducción de Señal , Sirtuina 1/genética , Sirtuina 1/metabolismo
19.
RNA Biol ; 16(6): 742-753, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30794054

RESUMEN

In the fission yeast Schizosaccharomyces pombe (S.pombe), heterochromatin domains are established and maintained by protein complexes that contain numerous RNA binding domains among their components. The fission yeast HP1 protein Swi6 is one such component and contains an unstructured RNA-binding hinge, which is important for the integrity and silencing of heterochromatin. In this study, we have used an RNA aptamer that likely binds to the Swi6 hinge with high affinity, as a tool to perturb the natural interactions mediated by this domain. When the hinge is blocked by the aptamer RNA, Swi6 appears to become less restricted to the pericentromeres and is enriched at specific euchromatic loci. This suggests a role for the Swi6 hinge, along with the chromoshadow domain (previously shown) in controlling the spread of heterochromatin in S.pombe. The study also highlights the potential of using a synthetic aptamer RNA as a tool to perturb nucleic acid - protein interaction in vivo with the objective of understanding the functional relevance of such an interaction.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Heterocromatina , Proteínas de Schizosaccharomyces pombe/antagonistas & inhibidores , Aptámeros de Nucleótidos/química , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Motivos de Nucleótidos , Dominios Proteicos , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/metabolismo
20.
J Immunol ; 198(3): 1093-1103, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28039300

RESUMEN

Experimental autoimmune uveitis (EAU), in which CD4+ Th1 and/or Th17 cells are immunopathogenic, mimics various clinical features of noninfectious uveitis in humans. The impact of bromodomain extraterminal (BET) inhibitors on Th17 cell function was studied in a mouse model of EAU in vivo and in mouse and human Th17 cells in vitro. Two BET inhibitors (GSK151 and JQ1) were able to ameliorate the progression of inflammation in EAU and in mouse CD4+ T cells in vitro, downregulating levels of Th17 cells. Additionally, the uveitogenic capacity of Th17 cells to transfer EAU was abrogated by BET inhibitors in an adoptive transfer model. In human CD4+ T cells, a 5-d exposure to BET inhibitors was accompanied by a significant downregulation of Th17-associated genes IL-17A, IL-22, and retinoic acid-related orphan receptor γt. However, in vitro, the inhibitors had no effect on already polarized Th17 cells. The key finding is that, in response to BET inhibitors, Th17-enriched cultures developed a regulatory phenotype, upregulated FOXP3 expression and IL-10 secretion, and lost pathogenicity in vivo. We conclude that BET targeting of Th17 cells is a potential therapeutic opportunity for a wide range of inflammatory and autoimmune diseases, including uveitis.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Retina/efectos de los fármacos , Células Th17/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Uveítis/tratamiento farmacológico , Animales , Citocinas/biosíntesis , Regulación hacia Abajo , Femenino , Factores de Transcripción Forkhead/análisis , Ratones , Proteínas Nucleares/fisiología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/análisis , Receptores CCR6/análisis , Retina/inmunología , Células TH1/inmunología , Células Th17/inmunología , Factores de Transcripción/fisiología , Factor de Necrosis Tumoral alfa/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA