Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.257
Filtrar
1.
J Med Chem ; 67(9): 6938-6951, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38687638

RESUMEN

Nuclear receptor-binding SET domain-containing 2 (NSD2), a methyltransferase that primarily installs the dimethyl mark on lysine 36 of histone 3 (H3K36me2), has been recognized as a promising therapeutic target against cancer. However, existing NSD2 inhibitors suffer from low activity or inferior selectivity, and none of them can simultaneously remove the methyltransferase activity and chromatin binding function of NSD2. Herein we report the discovery of a novel NSD2 degrader LLC0424 by leveraging the proteolysis-targeting chimera technology. LLC0424 potently degraded NSD2 protein with a DC50 value of 20 nM and a Dmax value of 96% in acute lymphoblastic leukemia (ALL) RPMI-8402 cells. Mechanistic studies revealed LLC0424 to selectively induce NSD2 degradation in a cereblon- and proteasome-dependent fashion. LLC0424 also caused continuous downregulation of H3K36me2 and growth inhibition of ALL cell lines with NSD2 mutation. Importantly, intravenous or intraperitoneal injection of LLC0424 showed potent NSD2 degradation in vivo.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Proteolisis , Humanos , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Proteolisis/efectos de los fármacos , Animales , Línea Celular Tumoral , Ratones , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Descubrimiento de Drogas , Complejo de la Endopetidasa Proteasomal/metabolismo , Relación Estructura-Actividad , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Histonas/metabolismo , Proliferación Celular/efectos de los fármacos
2.
Microb Cell Fact ; 22(1): 50, 2023 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-36915090

RESUMEN

BACKGROUND: The lipopeptide herbicolin A (HA) secreted by the biocontrol agent Pantoea agglomerans ZJU23 is a promising antifungal drug to combat fungal pathogens by targeting lipid rafts, both in agricultural and clinical settings. Improvement of HA production would be of great significance in promoting its commercialization. This study aims to enhance the HA production in ZJU23 by combining fermentation optimization and strain engineering. RESULTS: Based on the results in the single-factor experiments, corn steep liquor, temperature and initial pH were identified as the significant affecting factors by the Plackett-Burman design. The fermentation medium and conditions were further optimized using the Box-Behnken response surface method, and the HA production of the wild type strain ZJU23 was improved from ~ 87 mg/mL in King's B medium to ~ 211 mg/mL in HA induction (HAI) medium. A transposon library was constructed in ZJU23 to screen for mutants with higher HA production, and two transcriptional repressors for HA biosynthesis, LrhA and PurR, were identified. Disruption of the LrhA gene led to increased mRNA expression of HA biosynthetic genes, and subsequently improved about twofold HA production. Finally, the HA production reached ~ 471 mg/mL in the ΔLrhA mutant under optimized fermentation conditions, which is about 5.4 times higher than before (~ 87 mg/mL). The bacterial suspension of the ΔLrhA mutant fermented in HAI medium significantly enhanced its biocontrol efficacy against gray mold disease and Fusarium crown rot of wheat, showing equivalent control efficacies as the chemical fungicides used in this study. Furthermore, HA was effective against fungicide resistant Botrytis cinerea. Increased HA production substantially improved the control efficacy against gray mold disease caused by a pyrimethanil resistant strain. CONCLUSIONS: This study reveals that the transcriptional repressor LrhA negatively regulates HA biosynthesis and the defined HAI medium is suitable for HA production. These findings provide an extended basis for large-scale production of HA and promote biofungicide development based on ZJU23 and HA in the future.


Asunto(s)
Antifúngicos , Agentes de Control Biológico , Reactores Biológicos , Fermentación , Ingeniería Genética , Pantoea , Pantoea/clasificación , Pantoea/efectos de los fármacos , Pantoea/genética , Pantoea/metabolismo , Fermentación/efectos de los fármacos , Fermentación/genética , Ingeniería Genética/métodos , Antifúngicos/metabolismo , Agentes de Control Biológico/metabolismo , Temperatura , Concentración de Iones de Hidrógeno , Regulación Bacteriana de la Expresión Génica , Medios de Cultivo/química , Medios de Cultivo/farmacología , Análisis de Regresión , Análisis de Varianza , Reproducibilidad de los Resultados , Proteínas Represoras/antagonistas & inhibidores , Micosis/prevención & control , Micosis/terapia , Productos Agrícolas/microbiología , Enfermedades de las Plantas/microbiología , Enfermedades de las Plantas/prevención & control , Enfermedades de las Plantas/terapia , Humanos , Animales
3.
ChemMedChem ; 18(3): e202200510, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36250286

RESUMEN

Schistosoma mansoni HDAC8 is a reliable target to fight schistosomiasis, and several inhibitors have been reported in the literature up to now. Nevertheless, only a few displayed selectivity over the human deacetylases and some exhibited very low or no activity against parasite larvae and/or adult worms. We report here the in vitro enzyme and biological activity of a small library of HDAC inhibitors from our lab, in many cases exhibiting submicromolar/nanomolar potency against smHDAC8 and diverse degrees of selectivity over hHDAC1 and/or hHDAC6. Such compounds were tested against schistosomula, and a selection of them against the adult forms of S. mansoni, to detect their effect on viability. Some of them showed the highest viability reduction for the larval stage with IC50 values around 1 µM and/or displayed ∼40-50 % activity in adult worms at 10 µM, joined to moderate to no toxicity in human fibroblast MRC-5 cells.


Asunto(s)
Inhibidores de Histona Desacetilasas , Histona Desacetilasas , Schistosoma mansoni , Esquistosomiasis , Adulto , Animales , Humanos , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Histona Desacetilasas/genética , Larva/efectos de los fármacos , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/genética , Esquistosomiasis/tratamiento farmacológico , Esquistosomiasis/genética
4.
Nat Commun ; 13(1): 3739, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35768432

RESUMEN

Tumor-infiltrating CD8 + T cells progressively lose functionality and fail to reject tumors. The underlying mechanism and re-programing induced by checkpoint blockers are incompletely understood. We show here that genetic ablation or pharmacological inhibition of histone lysine methyltransferase Suv39h1 delays tumor growth and potentiates tumor rejection by anti-PD-1. In the absence of Suv39h1, anti-PD-1 induces alternative activation pathways allowing survival and differentiation of IFNγ and Granzyme B producing effector cells that express negative checkpoint molecules, but do not reach final exhaustion. Their transcriptional program correlates with that of melanoma patients responding to immune-checkpoint blockade and identifies the emergence of cytolytic-effector tumor-infiltrating lymphocytes as a biomarker of clinical response. Anti-PD-1 favors chromatin opening in loci linked to T-cell activation, memory and pluripotency, but in the absence of Suv39h1, cells acquire accessibility in cytolytic effector loci. Overall, Suv39h1 inhibition enhances anti-tumor immune responses, alone or combined with anti-PD-1, suggesting that Suv39h1 is an "epigenetic checkpoint" for tumor immunity.


Asunto(s)
Linfocitos T CD8-positivos , Melanoma , Metiltransferasas , Receptor de Muerte Celular Programada 1 , Proteínas Represoras , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Epigénesis Genética , Humanos , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma/genética , Melanoma/inmunología , Melanoma/terapia , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/genética , Metiltransferasas/inmunología , Metiltransferasas/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
5.
Nat Commun ; 13(1): 866, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35165282

RESUMEN

Epstein-Barr virus (EBV) is reportedly the first identified human tumor virus, and is closely related to the occurrence and development of nasopharyngeal carcinoma (NPC), gastric carcinoma (GC), and several lymphomas. PD-L1 expression is elevated in EBV-positive NPC and GC tissues; however, the specific mechanisms underlying the EBV-dependent promotion of PD-L1 expression to induce immune escape warrant clarification. EBV encodes 44 mature miRNAs. In this study, we find that EBV-miR-BART11 and EBV-miR-BART17-3p upregulate the expression of PD-L1 in EBV-associated NPC and GC. Furthermore, EBV-miR-BART11 targets FOXP1, EBV-miR-BART17-3p targets PBRM1, and FOXP1 and PBRM1 bind to the enhancer region of PD-L1 to inhibit its expression. Therefore, EBV-miR-BART11 and EBV-miR-BART17-3p inhibit FOXP1 and PBRM1, respectively, and enhance the transcription of PD-L1 (CD274, http://www.ncbi.nlm.nih.gov/gene/29126 ), resulting in the promotion of tumor immune escape, which provides insights into potential targets for EBV-related tumor immunotherapy.


Asunto(s)
Herpesvirus Humano 4/genética , MicroARNs/genética , Carcinoma Nasofaríngeo/inmunología , Neoplasias Nasofaríngeas/inmunología , Neoplasias Gástricas/inmunología , Escape del Tumor/inmunología , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Herpesvirus Humano 4/inmunología , Humanos , Linfoma/inmunología , Linfoma/virología , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/virología , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/virología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Neoplasias Gástricas/virología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Escape del Tumor/genética , Microambiente Tumoral/inmunología
6.
Development ; 149(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35037688

RESUMEN

A limited BMP signaling range in the stem cell niche of the ovary protects against germ cell tumors and promotes germ cell homeostasis. The canonical repressor of BMP signaling in both the Drosophila embryo and wing disc is the transcription factor Brinker (Brk), yet the expression and potential role of Brk in the germarium has not previously been described. Here, we find that brk expression requires a promoter-proximal element (PPE) to support long-distance enhancer action as well as to drive expression in the germarium. Furthermore, PPE subdomains have different activities; in particular, the proximal portion acts as a damper to regulate brk levels precisely. Using PPE mutants as well as tissue-specific RNA interference and overexpression, we show that altering brk expression within either the soma or the germline affects germ cell homeostasis. Remarkably, we find that Decapentaplegic (Dpp), the main BMP ligand and canonical antagonist of Brk, is upregulated by Brk in the escort cells of the germarium, demonstrating that Brk can positively regulate this pathway.


Asunto(s)
Proteínas de Drosophila/metabolismo , Células Germinativas/metabolismo , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular , Drosophila , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Embrión no Mamífero/metabolismo , Femenino , Células Germinativas/citología , Ovario/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Bicatenario/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transducción de Señal , Regulación hacia Arriba , Alas de Animales/metabolismo
7.
J Chemother ; 34(1): 45-54, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34424139

RESUMEN

A small fraction of cancer cells known as cancer stem cells (CSCs) are considered to give rise to differentiated cancer cells and have been proposed to predict cancer recurrence and metastasis. There is further evidence that CSCs may act as metastatic precursors of epithelial-mesenchymal transition (EMT). In the present study, we investigated the key molecules involved in maintaining the stability of CSCs by inducing ectopic overexpression of CD133 to characterize EMT in human prostate cancer cell lines, including PC-3, DU145, and LnCaP cells. Additionally, we investigated whether a specific inhibitor of concomitantly expressed metastasis-related genes could alleviate EMT properties in CD133-overexpressing prostate cancer cells. Ectopic overexpression of CD133 in PC-3, DU145, and LnCaP cells led to an increase in the expression of HDAC9. Moreover, HDAC9 inhibition led to a decrease in EMT properties along with increased E-cadherin expression, a narrower wound gap distance, and enhanced cell invasiveness through the suppression of ß-catenin activation and its translocation to the nucleus. Overall, these results suggest that HDAC9 inhibition plays a functional role in the modulation of EMT properties in CSC-like prostate cancer cells. Therefore, these findings could facilitate the development of therapeutic strategies for controlling prostate cancer metastasis.


Asunto(s)
Antígeno AC133/genética , Transición Epitelial-Mesenquimal/fisiología , Genes Relacionados con las Neoplasias/genética , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/patología , Proteínas Represoras/antagonistas & inhibidores , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Histona Desacetilasas , Humanos , Masculino
8.
Exp Neurol ; 348: 113948, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34902358

RESUMEN

Retinal ganglion cells (RGCs) are the sole output neurons that carry visual information from the eye to the brain. Due to various retinal and optic nerve diseases, RGC somas and axons are vulnerable to damage and lose their regenerative capacity. A basic question is whether the manipulation of a key regulator of RGC survival can protect RGCs from retinal and optic nerve diseases. Here, we found that Maf1, a general transcriptional regulator, was upregulated in RGCs from embryonic stage to adulthood. We determined that the knockdown of Maf1 promoted the survival of RGCs and their axon regeneration through altering the activity of the PTEN/mTOR pathway, which could be blocked by rapamycin. We further observed that the inhibition of Maf1 prevented the retinal ganglion cell complex from thinning after optic nerve crush. These findings reveal a neuroprotective effect of knocking down Maf1 on RGC survival after injury and provide a potential therapeutic strategy for traumatic optic neuropathy.


Asunto(s)
Axones/fisiología , Regeneración Nerviosa/fisiología , Traumatismos del Nervio Óptico/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Células Ganglionares de la Retina/fisiología , Animales , Supervivencia Celular/fisiología , Técnicas de Silenciamiento del Gen/métodos , Inyecciones Intravítreas , Ratones , Ratones Endogámicos C57BL , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/patología , Proteínas Represoras/biosíntesis
9.
Exp Cell Res ; 411(1): 112985, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34942190

RESUMEN

Deep venous thrombosis (DVT) endangers human health. Endothelial progenitor cells (EPCs) were proven to promote thrombolysis and miR-204-5p was discovered to be low-expressed in DVT patients. This study concentrated on exploring whether miR-204-5p had a regulatory effect on EPCs and DVT. Concretely, the expression of miR-204-5p in DVT patients' blood was detected by qRT-PCR. The target of miR-204-5p was predicted by bioinformatics and verified by dual-luciferase reporter assay. After rat EPCs were isolated, identified, and transfected with miR-204-5p agomiR, antagomiR, or SPRED1 plasmids, the viability, migration, invasion, and tube formation of EPCs were detected by MTT, wound healing, Transwell, and tube formation assays, respectively. MiR-204-5p, SPRED1, p-PI3K, PI3K, p-AKT, AKT, VEGFA, and Ang1 expressions in EPCs were measured by qRT-PCR or Western blot. EPCs transfected with miR-204-5p overexpression lentivirus plasmid were injected into the DVT rat model. The histopathology of the thrombus and the homing of EPCs to thrombus in the DVT rats were observed by hematoxylin-eosin staining and confocal microscopy, respectively. We found that miR-204-5p was low-expressed in DVT patients and SPRED1 was a target gene of miR-204-5p. MiR-204-5p agomiR promoted the viability, migration, invasion, and tube formation of EPCs, the levels of VEGFA and Ang1 and the activation of PI3K/AKT pathway in EPCs, while miR-204-5p antagomiR and SPRED1 worked oppositely. SPRED1 reversed the effect of miR-204-5p agomiR on EPCs. Up-regulated miR-204-5p inhibited thrombosis and promoted EPCs homing to thrombus in DVT rats. Collectively, up-regulated miR-204-5p enhanced the angiogenesis of EPCs and thrombolysis in DVT rats by targeting SPRED1.


Asunto(s)
Células Progenitoras Endoteliales/fisiología , Regulación de la Expresión Génica , MicroARNs/genética , Neovascularización Fisiológica , Proteínas Represoras/antagonistas & inhibidores , Terapia Trombolítica/métodos , Trombosis de la Vena/terapia , Adulto , Animales , Apoptosis , Biomarcadores/metabolismo , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Células Cultivadas , Células Progenitoras Endoteliales/citología , Femenino , Humanos , Masculino , Pronóstico , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Activación Transcripcional , Regulación hacia Arriba , Trombosis de la Vena/metabolismo , Trombosis de la Vena/patología
10.
Life Sci ; 288: 120157, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34801511

RESUMEN

AIM: High risk Human Papillomavirus (HPV) is an infectious pathogen implicated in a variety of cancers with poor clinical outcome. The mechanism of HPV induced cellular transformation and its intervention remains to be elucidated. Human ADA3 (hADA3), a cellular target of HPV16 E6, is an essential and conserved component of the ADA transcriptional coactivator complex. High risk HPV-E6 binds and functionally inactivates hADA3 to initiate oncogenesis. The aim of this study was to identify the interaction interface between hADA3 and HPV16E6 for designing inhibitory peptides that can potentially disrupt the hADA3-E6 interaction. MATERIAL METHODS: The present investigation employed structure-based in silico tools supported by biochemical validation, in vivo interaction studies and analysis of posttranslational modifications. KEY FINDINGS: First 3D-model of hADA3 was proposed and domains involved in the oncogenic interaction between hADA3 and HPV16E6 were delineated. Rationally designed peptide disrupted hADA3-E6 interaction and impeded malignant properties of cervical cancer cells. SIGNIFICANCE: Intervention of hADA3-E6 interaction thus promises to be a potential strategy to combat HPV induced oncogenic conditions like cervical cancer. The investigation provides mechanistic insights into HPV pathogenesis and shows promise in developing novel therapeutics to treat HPV induced cancers.


Asunto(s)
Proteínas Oncogénicas Virales/antagonistas & inhibidores , Infecciones por Papillomavirus/complicaciones , Fragmentos de Péptidos/farmacología , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Proteínas Represoras/antagonistas & inhibidores , Sumoilación , Factores de Transcripción/antagonistas & inhibidores , Neoplasias del Cuello Uterino/tratamiento farmacológico , Comunicación Celular , Transformación Celular Neoplásica , Femenino , Humanos , Proteínas Oncogénicas Virales/metabolismo , Papillomaviridae/aislamiento & purificación , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , Conformación Proteica , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/virología
11.
Chem Biol Interact ; 352: 109760, 2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-34922903

RESUMEN

The present study aimed to gain insight into putative anticancer effect of dietary zinc in rat lung cancer model by targeting ferroptosis. Ferroptosis is an emerging type of programmed cell death, which activates oxidative cell death in an iron and lipid peroxides-dependent manner. Targeting ferroptosis is a novel therapeutic approach for cancer therapy. Circular RNAs (circRNAs), as a form of noncoding RNAs with a specific closed circular sequence are emerging as a new field in cancer research. However, the regulatory mechanisms of circRNAs in ferroptosis during lung cancer development are still elusive. In this work, we elucidate the potential prognostic value and the crucial role of circular RNA circFOXP1 in ferroptosis during lung cancer and modulatory potential of zinc. We found that the expression of circFOXP1 was remarkably up-regulated in clinical tumorous tissues compared with adjacent tissues. The knockdown of circFOXP1 suppressed the cell viability of lung cancer cells. The colony formation counts of lung cancer cells were repressed by the depletion of circFOXP1 as well. Moreover, the treatment of zinc, repressed the cell viability of lung cancer cells and the overexpression of circFOXP1 rescued the phenotype. Meanwhile, the levels of malondialdehyde (MDA), iron, and lipid ROS were enhanced post zinc treatment. Collectively, we concluded that circular RNA circFOXP1 is a potential diagnostic biomarker and contributes to malignant progression by repressing ferroptosis of lung cancer cells. Further, zinc be served as a promising therapeutic approach against lung cancer.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Ferroptosis/efectos de los fármacos , Factores de Transcripción Forkhead/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , ARN Circular/genética , Proteínas Represoras/genética , Zinc/administración & dosificación , Adenocarcinoma del Pulmón/patología , Sistema de Transporte de Aminoácidos y+/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Ferroptosis/genética , Factores de Transcripción Forkhead/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Persona de Mediana Edad , Pronóstico , ARN Circular/metabolismo , Ratas , Proteínas Represoras/antagonistas & inhibidores
12.
Mediators Inflamm ; 2021: 6359652, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34924813

RESUMEN

Ellagic acid (EA) was reported to play protective roles in rheumatoid arthritis (RA). It was found that the level of metastasis-associated gene 1 (MTA1)/histone deacetylase 1 (HDAC1) protein complex was downregulated by polyphenols in several human disorders. Notably, inhibition of MTA1 or HDAC1 has anti-inflammatory effects on RA. Therefore, our study is aimed at investigating whether EA prevents RA progression through regulating the MTA1/HDAC1 complex. Herein, the human fibroblast-like synoviocyte (FLS) cell line MH7A was treated with TNF-α to induce an inflammation model in vitro and then incubated with different concentrations of EA. Western blot analysis showed that EA reduced MTA1 expression in a dose-dependent manner in MH7A cells. Then, TNF-α-treated MH7A cells were incubated with EA alone or together with MTA1 overexpression plasmid (pcDNA-MTA1), and we found that EA inhibited proliferation, inflammation cytokine levels, and oxidative stress marker protein levels and promoted apoptosis in MH7A cells, while MTA1 overexpression abolished these effects. Moreover, coimmunoprecipitation assay verified the interaction between MTA1 and HDAC1. EA downregulated the MTA1/HDAC1 complex in MH7A cells. MTA1 knockdown inhibited proliferation, inflammation, and oxidative stress and promoted apoptosis in MH7A cells, while HDAC1 overexpression reversed these effects. Moreover, chromatin immunoprecipitation assay indicated that EA inhibited HDAC1-mediated Nur77 deacetylation. Rescue experiments demonstrated that Nur77 knockdown reversed the effects of EA on MH7A cell biological behaviors. Additionally, EA treatment attenuated arthritis index, paw swelling, synovial hyperplasia, and inflammation in collagen-induced arthritis (CIA) rats. In conclusion, EA inhibited proliferation, inflammation, and oxidative stress and promoted apoptosis in MH7A cells and alleviated the severity of RA in CIA rats though downregulating MTA1/HDAC1 complex and promoting HDAC1 deacetylation-mediated Nur77 expression.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Ácido Elágico/farmacología , Histona Desacetilasa 1/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Acetilación , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Histona Desacetilasa 1/fisiología , Humanos , Masculino , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Proteínas Represoras/fisiología , Transactivadores/fisiología
13.
Nat Commun ; 12(1): 6662, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795264

RESUMEN

SPOP, an E3 ubiquitin ligase, acts as a prostate-specific tumor suppressor with several key substrates mediating oncogenic function. However, the mechanisms underlying SPOP regulation are largely unknown. Here, we have identified G3BP1 as an interactor of SPOP and functions as a competitive inhibitor of Cul3SPOP, suggesting a distinctive mode of Cul3SPOP inactivation in prostate cancer (PCa). Transcriptomic analysis and functional studies reveal a G3BP1-SPOP ubiquitin signaling axis that promotes PCa progression through activating AR signaling. Moreover, AR directly upregulates G3BP1 transcription to further amplify G3BP1-SPOP signaling in a feed-forward manner. Our study supports a fundamental role of G3BP1 in disabling the tumor suppressive Cul3SPOP, thus defining a PCa cohort independent of SPOP mutation. Therefore, there are significantly more PCa that are defective for SPOP ubiquitin ligase than previously appreciated, and these G3BP1high PCa are more susceptible to AR-targeted therapy.


Asunto(s)
Proteínas Cullin/antagonistas & inhibidores , ADN Helicasas/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Neoplasias de la Próstata/metabolismo , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Receptores Androgénicos/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Antagonistas de Receptores Androgénicos/farmacología , Animales , Carcinogénesis , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular/efectos de los fármacos , Proteínas Cullin/metabolismo , ADN Helicasas/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Helicasas/genética , Proteínas con Motivos de Reconocimiento de ARN/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
14.
Biomolecules ; 11(10)2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-34680125

RESUMEN

CCR4-NOT transcription complex subunit 2 (CNOT2), a subunit of the CCR4-NOT complex, has been described in cancer progression. The CNOT complex plays an important role in multiple cellular functions. Recent studies in our laboratory showed that CNOT2 promotes breast cancer cell proliferation and angiogenesis. In addition, CNOT2 signals are critically related to apoptosis induced by atorvastatin in lung cancer cells. Furthermore, depletion of CNOT2 was shown to enhance the antitumor effect of midline 1 interacting protein 1 (MID1IP1) depletion, thus inhibiting c-Myc expression in liver cancer cells. However, the molecular mechanisms related to its oncogenic role remain unclear. Herein, for the first time, we report that CNOT2 inhibition can induce apoptosis in colorectal cancer cells by activating p53. Inhibition of CNOT2 markedly induced apoptosis in various cancer cells like that of the wild-type p53. Furthermore, inhibition of CNOT2 elongated p53 s half-life. Previously, our laboratory demonstrated that MID1IP1 promoted colocalization with c-Myc mediated by CNOT2. Interestingly, inhibition of CNOT2 cannot induce p53 expression without MID1IP1 or apoptosis in cancer cells. In conclusion, our results demonstrate that CNOT2 inhibition induces apoptosis through MID1IP1 by activating p53.


Asunto(s)
Apoptosis , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteínas del Citoesqueleto/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Doxorrubicina/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Estabilidad Proteica , Proteínas Represoras/metabolismo
15.
Eur J Pharmacol ; 912: 174531, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34710370

RESUMEN

Endothelial cell activation through nuclear factor-kappa-B (NFkB) and mitogen-activated protein kinases leads to increased biosynthesis of pro-inflammatory mediators, cellular injury and vascular inflammation under lipopolysaccharide (LPS) exposure. Recent studies report that LPS up-regulated global methyltransferase activity. In this study, we observed that a combination treatment with metformin (MET) and cholecalciferol (VD) blocked the LPS-induced S-adenosylmethionine (SAM)-dependent methyltransferase (SDM) activity in Eahy926 cells. We found that LPS challenge (i) increased arginine methylation through up-regulated protein arginine methyltransferase-1 (PRMT1) mRNA, intracellular concentrations of asymmetric dimethylarginine (ADMA) and homocysteine (HCY); (ii) up-regulated cell senescence through mitigated sirtuin-1 (SIRT1) mRNA, nicotinamide adenine dinucleotide (NAD+) concentration, telomerase activity and total antioxidant capacity; and (iii) lead to endothelial dysfunction through compromised nitric oxide (NOx) production. However, these LPS-mediated cellular events in Eahy926 cells were restored by the synergistic effect of MET and VD. Taken together, this study identified that the dual compound effect inhibits LPS-induced protein arginine methylation, endothelial senescence and dysfunction through the components of epigenetic machinery, SIRT1 and PRMT1, which is a previously unidentified function of the test compounds. In silico results identified the presence of vitamin D response element (VDRE) sequence on PRMT1 suggesting that VDR could regulate PRMT1 gene expression. Further characterization of the cellular events associated with the dual compound challenge, using gene silencing approach or adenoviral constructs for SIRT1 and/or PRMT1 under inflammatory stress, could identify therapeutic strategies to address the endothelial consequences in vascular inflammation-mediated atherosclerosis.


Asunto(s)
Antioxidantes/farmacología , Colecalciferol/farmacología , Metformina/farmacología , Sustancias Protectoras/farmacología , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Sirtuina 1/metabolismo , Arginina/análogos & derivados , Arginina/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Senescencia Celular/efectos de los fármacos , Endotelio/efectos de los fármacos , Homocisteína/metabolismo , Humanos , Lipopolisacáridos/toxicidad , Metilación/efectos de los fármacos , NAD/metabolismo , Óxido Nítrico/metabolismo , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/química , Proteínas Represoras/genética , S-Adenosilmetionina/metabolismo , Sirtuina 1/genética , Telomerasa/metabolismo , Elemento de Respuesta a la Vitamina D
16.
J Med Chem ; 64(20): 14913-14929, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34488340

RESUMEN

Nuclear receptor-binding SET domain (NSD) proteins are a class of histone lysine methyltransferases (HKMTases) that are amplified, mutated, translocated, or overexpressed in various types of cancers. Several campaigns to develop NSD inhibitors for cancer treatment have begun following recent advances in knowledge of NSD1, NSD2, and NSD3 structures and functions as well as the U.S. FDA approval of the first HKMTase inhibitor (tazemetostat, an EZH2 inhibitor) to treat follicular lymphoma and epithelioid sarcoma. This perspective highlights recent findings on the structures of catalytic su(var), enhancer-of-zeste, trithorax (SET) domains and other functional domains of NSD methyltransferases. In addition, recent progress and efforts to discover NSD-specific small molecule inhibitors against cancer-targeting catalytic SET domains, plant homeodomains, and proline-tryptophan-tryptophan-proline domains are summarized.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Neoplasias/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Antineoplásicos/química , Inhibidores Enzimáticos/química , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Estructura Molecular , Neoplasias/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Bibliotecas de Moléculas Pequeñas/química
17.
Bioorg Chem ; 116: 105350, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34547645

RESUMEN

In the present study, two novel series of compounds incorporating naphthyl and pyridyl linker were synthesized and biological assays revealed 5-((6-(2-(5-(2-chlorophenyl)-3-(4-fluorophenyl)-4,5-dihydro-1H-pyrazol-1-yl)-2-oxoethoxy) naphthalene-2-yl)methylene)thiazolidine-2,4-dione (14b) as the most potent dual inhibitors of vascular endothelial growth factors receptor-2 (VEGFR-2) and histone deacetylase 4 (HDAC4). Compounds 13b, 14b, 17f, and 21f were found to stabilize HDAC4; where, pyridyl linker swords were endowed with higher stabilization effects than naphthyl linker. Also, 13b and 14b showed best inhibitory activity on VEGFR-2 as compared to others. Compound 14b was most potent as evident by in-vitro and in-vivo biological assessments. It displayed anti-angiogenic potential by inhibiting endothelial cell proliferation, migration, tube formation and also suppressed new capillary formation in the growing chick chorioallantoic membranes (CAMs). It showed selectivity and potency towards HDAC4 as compared to other HDAC isoforms. Compound 14b (25 mg/kg, i.p.) also indicated exceptional antitumor efficacy on in-vivo animal xenograft model of human colorectal adenocarcinoma (HT-29). The mechanism of action of 14b was also confirmed by western blot.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Tiazolidinedionas/farmacología , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/química , Histona Desacetilasas/metabolismo , Humanos , Ratones , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Pirazoles/síntesis química , Pirazoles/química , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Relación Estructura-Actividad , Tiazolidinedionas/síntesis química , Tiazolidinedionas/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
Mol Microbiol ; 116(4): 1173-1188, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34468051

RESUMEN

The quorum-sensing signaling systems in Vibrio bacteria converge to control levels of the master transcription factors LuxR/HapR, a family of highly conserved proteins that regulate gene expression for bacterial behaviors. A compound library screen identified 2-thiophenesulfonamide compounds that specifically inhibit Vibrio campbellii LuxR but do not affect cell growth. We synthesized a panel of 50 thiophenesulfonamide compounds to examine the structure-activity relationship effects on Vibrio quorum sensing. The most potent molecule identified, PTSP (3-phenyl-1-(thiophen-2-ylsulfonyl)-1H-pyrazole), inhibits quorum sensing in multiple strains of V. vulnificus, V. parahaemolyticus, and V. campbellii at nanomolar concentrations. However, thiophenesulfonamide inhibition efficacy varies significantly among Vibrio species: PTSP is most inhibitory against V. vulnificus SmcR, but V. cholerae HapR is completely resistant to all thiophenesulfonamides tested. Reverse genetics experiments show that PTSP efficacy is dictated by amino acid sequence in the putative ligand-binding pocket: F75Y and C170F SmcR substitutions are each sufficient to eliminate PTSP inhibition. Further, in silico modeling distinguished the most potent thiophenesulfonamides from less-effective derivatives. Our results revealed the previously unknown differences in LuxR/HapR proteins that control quorum sensing in Vibrio species and underscore the potential for developing thiophenesulfonamides as specific quorum sensing-directed treatments for Vibrio infections.


Asunto(s)
Percepción de Quorum/efectos de los fármacos , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Sulfonamidas/metabolismo , Sulfonamidas/farmacología , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo , Vibrio/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Represoras/química , Especificidad de la Especie , Relación Estructura-Actividad , Sulfonamidas/química , Transactivadores/química , Vibrio/química , Vibrio/genética
19.
Phys Chem Chem Phys ; 23(32): 17576-17590, 2021 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-34369509

RESUMEN

The rational design of selective histone deacetylase 2 (HDAC2) inhibitors is beneficial for the therapeutic treatment of liver cancer, though HDAC2 is highly homologous to HDAC8, which may lead to undesired side effects due to the pan-inhibition towards HDAC2 and HDAC8. To clarify the structural basis of selective inhibition towards HDAC2 over HDAC8, we utilized multiple in silico strategies, including sequence alignment, structural comparison, molecular docking, molecular dynamics simulations, free energy calculations, alanine scanning mutagenesis, pharmacophore modeling, protein contacts atlas analysis and QM/MM calculations to study the binding patterns of HDAC2/8 selective inhibitors. Through the whole process described above, it is found that although HDAC2 has conserved GLY154 and PHE210 that also exist within HDAC8, namely GLY151 and PHE208, the two isoforms exhibit diverse binding modes towards their inhibitors. Typically, HDAC2 inhibitors interact with the Zn2+ ions through the core chelate group, while HDAC8 inhibitors adopt a bent conformation within the HDAC8 pocket that inclines to be in contact with the Zn2+ ions through the terminal hydroxamic acid group. In summary, our data comprehensively elucidate the selectivity mechanism towards HDAC2 over HDAC8, which would guide the rational design of selective HDAC2 inhibitors for liver cancer treatment.


Asunto(s)
Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Diseño de Fármacos , Histona Desacetilasa 2/química , Histona Desacetilasa 2/genética , Inhibidores de Histona Desacetilasas/química , Histona Desacetilasas/química , Histona Desacetilasas/metabolismo , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutagénesis , Mutación , Unión Proteica , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Termodinámica
20.
Cancer Lett ; 520: 243-254, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34339801

RESUMEN

Hepatocellular carcinoma (HCC) lacks effective treatment, and the patients rapidly develop the acquired resistance to sorafenib with less defined mechanisms. Here, we demonstrate that transcriptional factor myocyte enhancer factor 2D (MEF2D) overexpression is detected in sorafenib-resistant HCC specimens and HCC cell lines and predicts poor prognosis of sorafenib-treated HCC patients. Mechanistically, MEF2D in complex with histone deacetylase HDAC4 directly binds to the SPRY4 promoter regions and suppresses the transcriptional expression of SPRY4, which is a negative regulator of MAPK/ERK signaling pathway. Inhibition of HDAC4 with its clinically used inhibitor induces SPRY4 expression and inhibition of ERK activity, resulting in sensitization of HCC cells to sorafenib-induced apoptosis and greatly improved inhibition of liver tumor growth in mice with sorafenib treatment. These findings highlight the critical role of coupling HDAC4 with MEF2D in activation of ERK by suppressing SPRY4 and underscore the great potential to improve HCC treatment by combined administration of sorafenib with HDAC4 inhibitors.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Histona Desacetilasas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas del Tejido Nervioso/genética , Proteínas Represoras/genética , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factores de Transcripción MEF2/genética , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Represoras/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sorafenib/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA