Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 249
Filtrar
1.
Inflamm Res ; 71(2): 243-253, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35059772

RESUMEN

OBJECTIVE: Inflammation and proliferation of vascular smooth muscle cells (VSMCs), induced by angiotensin II (AngII) and other growth factors, play important roles in the pathogenesis of hypertension, restenosis, and atherosclerosis. Dihydroartemisinin (DHA) exhibits broad protective effects. However, the effects of DHA on AngII-induced inflammation and proliferation of VSMCs remain unknown. MATERIALS AND METHODS: AngII was used to construct VSMCs and vascular inflammation model in vitro and in vivo. The protective roles of DHA in inflammatory response and proliferation were evaluated through CCK-8, BrdU assay and immunofluorescence staining. The level of mRNA N6-methyladenosine was measured by m6A-RNA immunoprecipitation (MeRIP) assay. Western blot and quantitative real-time PCR were used to investigate the relationship between FTO and its potential downstream signaling molecules. RESULTS: In the present study, we found that DHA significantly suppressed AngII-induced proliferation of VSMCs and the expression of IL-6 and Ccl2 in a dose-dependent manner. Additionally, we confirmed that fat mass and obesity-associated (FTO) plays a critical role in AngII-induced VSMC proliferation and inflammation. FTO knockdown increased the methylation level of NR4A3 mRNA, whereas FTO, but not mutated FTO overexpression, reduced the methylation level of NR4A3 mRNA. These results suggest that DHA plays a protective role in AngII-induced VSMC proliferation and the associated inflammation by inhibiting the FTO/NR4A3 axis. CONCLUSION: Our findings provide new insight into the mechanisms of DHA and its critical role in the pathogenesis of hypertension-related vascular complications.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/antagonistas & inhibidores , Angiotensina II/farmacología , Artemisininas/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Inflamación/prevención & control , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Hormona Tiroidea/antagonistas & inhibidores , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/fisiología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Ratones , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Proteínas del Tejido Nervioso/fisiología , Receptores de Esteroides/fisiología , Receptores de Hormona Tiroidea/fisiología , Transducción de Señal/efectos de los fármacos
2.
Nat Commun ; 12(1): 4662, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34341345

RESUMEN

Impaired cellular cholesterol efflux is a key factor in the progression of renal, cardiovascular, and autoimmune diseases. Here we describe a class of 5-arylnicotinamide compounds, identified through phenotypic drug discovery, that upregulate ABCA1-dependent cholesterol efflux by targeting Oxysterol Binding Protein Like 7 (OSBPL7). OSBPL7 was identified as the molecular target of these compounds through a chemical biology approach, employing a photoactivatable 5-arylnicotinamide derivative in a cellular cross-linking/immunoprecipitation assay. Further evaluation of two compounds (Cpd A and Cpd G) showed that they induced ABCA1 and cholesterol efflux from podocytes in vitro and normalized proteinuria and prevented renal function decline in mouse models of proteinuric kidney disease: Adriamycin-induced nephropathy and Alport Syndrome. In conclusion, we show that small molecule drugs targeting OSBPL7 reveal an alternative mechanism to upregulate ABCA1, and may represent a promising new therapeutic strategy for the treatment of renal diseases and other disorders of cellular cholesterol homeostasis.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , Colesterol/metabolismo , Nefropatías Diabéticas/metabolismo , Compuestos Orgánicos/farmacología , Podocitos/metabolismo , Proteinuria/metabolismo , Receptores de Esteroides/antagonistas & inhibidores , Transportador 1 de Casete de Unión a ATP/genética , Animales , Transporte Biológico/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Ratones de la Cepa 129 , Ratones Noqueados , Estructura Molecular , Niacinamida/química , Niacinamida/farmacología , Compuestos Orgánicos/síntesis química , Compuestos Orgánicos/química , Podocitos/citología , Interferencia de ARN , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Células THP-1
3.
Oncol Rep ; 45(3): 963-974, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33650661

RESUMEN

The human testicular nuclear receptor 4 (TR4) is a critical regulatory gene for the progression of prostate cancer (PCa). Although it has been revealed that TR4 causes chemoresistance in PCa via the activation of octamer­binding transcription factor 4 (OCT4), the detailed mechanism remains unexplored. In the present study, it was revealed that inhibition of TR4 by shRNA in PCa enhanced the sensitivity to docetaxel in vitro and in vivo. TR4 induced the downregulation of miR­145 by directly binding it to the promoter of miR­145, which was confirmed by chromatin immunoprecipitation analysis and luciferase assay. The overexpression of miR­145 suppressed both the chemoresistance and the expression of OCT4 mRNA and protein. Additionally, the TR4 shRNA mediated re­sensitization to docetaxel, along with the downregulated expression of OCT4, were reversed by the concurrent inhibition of miR­145. The luciferase assay revealed that the activity of the wild­type OCT4 3' untranslated region reporter was suppressed. This suppression diminished when the miR­145 response element mutated. These findings suggest an undescribed regulatory pathway in PCa, by which TR4 directly suppressed the expression of miR­145, thereby inhibiting its direct target OCT4, leading to the promotion of chemoresistance in PCa.


Asunto(s)
Docetaxel/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , MicroARNs/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Hormona Tiroidea/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Docetaxel/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , MicroARNs/genética , Trasplante de Neoplasias , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Transducción de Señal
4.
J Insect Sci ; 20(5)2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-33057682

RESUMEN

Trehalose plays a crucial role in the diapause process of many insects, serving as an energy source and a stress protectant. Trehalose accumulation has been reported in diapause pupae of Antheraea pernyi; however, trehalose metabolic regulatory mechanisms associated with diapause termination remain unclear. Here, we showed that the enhanced trehalose catabolism was associated with an increase in endogenous 20-hydroxyecdysone (20E) in hemolymph of A. pernyi pupae during their diapause termination and posttermination period. Injection of 20E increased the mRNA level of trehalase 1A (ApTre-1A) and trehalase 2 (ApTre-2) of A. pernyi diapause pupae in a dose-dependent manner but did not affect the mRNA level of trehalase 1B (ApTre-1B). Meanwhile, exogenous 20E increased the enzyme activities of soluble and membrane-bound trehalase, leading to a decline in hemolymph trehalose. Conversely, the expression of ApTre-1A and ApTre-2 were down-regulated after the ecdysone receptor gene (ApEcRB1) was silenced by RNA interference or by injection of an ecdysone receptor antagonist cucurbitacin B (CucB), which inhibits the 20E pathway. Moreover, CucB treatment delayed adult emergence, which suggests that ApEcRB1 might be involved in regulating pupal-adult development of A. pernyi by mediating ApTre-1A and ApTre-2 expressions. This study provides an overview of the changes in the expression and activity of different trehalase enzymes in A. pernyi in response to 20E, confirming the important role of 20E in controlling trehalose catabolism during A. pernyi diapause termination and posttermination period.


Asunto(s)
Ecdisterona , Mariposas Nocturnas/metabolismo , Animales , Diapausa de Insecto/efectos de los fármacos , Ecdisterona/metabolismo , Ecdisterona/farmacología , Metabolismo Energético/efectos de los fármacos , Genes de Insecto , Hemolinfa/efectos de los fármacos , Hemolinfa/metabolismo , Estadios del Ciclo de Vida/efectos de los fármacos , Mariposas Nocturnas/efectos de los fármacos , Mariposas Nocturnas/crecimiento & desarrollo , Pupa/efectos de los fármacos , Pupa/crecimiento & desarrollo , Pupa/metabolismo , Interferencia de ARN , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Trehalasa/efectos de los fármacos , Trehalasa/metabolismo , Trehalosa/metabolismo , Triterpenos
5.
Cancer Lett ; 495: 100-111, 2020 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-32768524

RESUMEN

Early studies indicated that the testicular nuclear receptor 4 (TR4) might play key roles in altering prostate cancer (PCa) progression; however, its ability to alter PCa radiosensitivity remains unclear. Here, we found that suppressing TR4 expression promoted radiosensitivity and better suppressed PCa by modulating the protein quaking (QKI)/circZEB1/miR-141-3p/ZEB1 signaling pathway. Mechanism dissection studies revealed that TR4 could transcriptionally increase the RNA-binding protein QKI to increase circZEB1 levels, which then sponges the miR-141-3p to increase the expression of its host gene ZEB1. Preclinical studies with an in vivo mouse model further proved that combining radiation therapy (RT) with metformin promoted radiosensitivity to suppress PCa progression. Together, these results suggest that TR4 may play key roles in altering PCa radiosensitivity and show that targeting this newly identified TR4-mediated QKI/circZEB1/miR-141-3p/ZEB1 signaling pathway may help in the development of a novel RT to better suppress the progression of PCa.


Asunto(s)
Metformina/administración & dosificación , Neoplasias de la Próstata/terapia , Tolerancia a Radiación/efectos de los fármacos , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Línea Celular Tumoral , Humanos , Masculino , Metformina/farmacología , Ratones , MicroARNs/genética , Trasplante de Neoplasias , Células PC-3 , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , ARN Circular/genética , Proteínas de Unión al ARN/genética , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Hormona Tiroidea/antagonistas & inhibidores , Transducción de Señal/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
6.
Development ; 147(14)2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32631830

RESUMEN

The activation of a neuroendocrine system that induces a surge in steroid production is a conserved initiator of the juvenile-to-adult transition in many animals. The trigger for maturation is the secretion of brain-derived neuropeptides, yet the mechanisms controlling the timely onset of this event remain ill-defined. Here, we show that a regulatory feedback circuit controlling the Drosophila neuropeptide Prothoracicotropic hormone (PTTH) triggers maturation onset. We identify the Ecdysone Receptor (EcR) in the PTTH-expressing neurons (PTTHn) as a regulator of developmental maturation onset. Loss of EcR in these PTTHn impairs PTTH signaling, which delays maturation. We find that the steroid ecdysone dose-dependently affects Ptth transcription, promoting its expression at lower concentrations and inhibiting it at higher concentrations. Our findings indicate the existence of a feedback circuit in which rising ecdysone levels trigger, via EcR activity in the PTTHn, the PTTH surge that generates the maturation-inducing ecdysone peak toward the end of larval development. Because steroid feedback is also known to control the vertebrate maturation-inducing hypothalamic-pituitary-gonadal axis, our findings suggest an overall conservation of the feedback-regulatory neuroendocrine circuitry that controls the timing of maturation initiation.


Asunto(s)
Proteínas de Drosophila/metabolismo , Hormonas de Insectos/metabolismo , Receptores de Esteroides/metabolismo , Animales , Tamaño Corporal , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Ecdisterona/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hormonas de Insectos/antagonistas & inhibidores , Hormonas de Insectos/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Metamorfosis Biológica , Microscopía Fluorescente , Neuronas/metabolismo , Interferencia de ARN , ARN Guía de Kinetoplastida/metabolismo , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/genética , Transducción de Señal
8.
Oncogene ; 39(9): 1891-1903, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31748715

RESUMEN

Prostate cancer (PCa) is the second leading cause of cancer death in men in America, and there are no curative options for metastatic castration-resistant prostate cancer (mCRPC). Docetaxel (DTX) has been used as a standard chemotherapy for the mCRPC. However, resistance to DTX is a significant clinical problem as half of patients fail to respond to therapy. The TR4 nuclear receptor has been reported to play an important role in PCa progression, however, its linkage to the DTX resistance remains unclear. Here we found that TR4 was upregulated after DTX chemotherapy in the mCRPC cells and patients, and TR4 expression is correlated with DTX sensitivity with a higher level conferring chemo-resistance. Targeting TR4 with an antagonist bexarotene (Bex, a derivative of retinoid) suppressed the TR4 transactivation with increased DTX chemo-sensitivity. Mechanism dissection studies revealed that TR4 might alter the DTX chemo-sensitivity via modulating the TR4/lincRNA-p21/HIF-1α/VEGF-A signaling. Together, these results suggest that targeting this newly identified TR4/lincRNA-p21/HIF-1α/VEGF-A signaling with Bex, an FDA-approved drug, may increase the DTX chemo-sensitivity to better suppress the mCRPC progression.


Asunto(s)
Bexaroteno/farmacología , Docetaxel/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Hormona Tiroidea/antagonistas & inhibidores , Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/secundario , Células Tumorales Cultivadas
9.
Bioorg Chem ; 94: 103471, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31813476

RESUMEN

Schweinfurthin A (Sch A) is a natural product with a selective and strong anti-cancer effect. Although it is known to target oxysterol binding proteins, the detailed mode of action is not well understood. Here, we provide strong evidence that yeast cells can be used as a eukaryotic model system to decipher the molecular modes of Sch A. We show that Sch A (100 µM) targets Osh2 (a yeast oxysterol binding protein homolog) genetically and taking advantage of computational chemistry indicate that the tetrahydro-2H-xanthene portion of Sch A forms H-bonds with residues Ser105, Val113, and Lys201, while its isoprenoid side chain is placed in a hydrophobic pocket lined by the side chains of Leu41, Leu45, Leu58, Met56, and Phe174 in Osh2. This model suggests that Sch A occupies the same binding pocket in Osh2 which is occupied by its natural substrate, ergosterol. Osh proteins transport sterol and PI(4)P in a cyclic manner between two membranes. Therefore, we suggest that Sch A interferes with this function of Osh2. In support of this hypothesis, we show that Sch A toxicity rate changes upon manipulating the enzymes that modify the levels of sterol and PI(4)P. This approach also informs how Sch A exerts its toxic effect in yeast cells. These enzymes include Coq1, Sac1, Plc1, Stt4, Pik1, and Mss4. We demonstrate that Coq1 an enzyme required for coenzyme Q synthesis (also involved in sterol metabolism indirectly), Sac1, and Stt4 the enzymes governing PI(4)P level modify Sch A toxicity and finally propose Sch A disrupts sterol/PI(4)P exchange between membranes by occupying the sterol/PI(4)P binding pocket in Osh2.


Asunto(s)
Antineoplásicos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Modelos Biológicos , Receptores de Esteroides/antagonistas & inhibidores , Saccharomyces cerevisiae/efectos de los fármacos , Estilbenos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Receptores de Esteroides/metabolismo , Saccharomyces cerevisiae/metabolismo , Estilbenos/síntesis química , Estilbenos/química , Relación Estructura-Actividad
10.
Nat Cell Biol ; 21(10): 1234-1247, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31570833

RESUMEN

Phosphoinositides have a pivotal role in the maturation of nascent phagosomes into microbicidal phagolysosomes. Following degradation of their contents, mature phagolysosomes undergo resolution, a process that remains largely uninvestigated. Here we studied the role of phosphoinositides in phagolysosome resolution. Phosphatidylinositol-4-phosphate (PtdIns(4)P), which is abundant in maturing phagolysosomes, was depleted as they tubulated and resorbed. Depletion was caused, in part, by transfer of phagolysosomal PtdIns(4)P to the endoplasmic reticulum, a process mediated by oxysterol-binding protein-related protein 1L (ORP1L), a RAB7 effector. ORP1L formed discrete tethers between the phagolysosome and the endoplasmic reticulum, resulting in distinct regions with alternating PtdIns(4)P depletion and enrichment. Tubules emerged from PtdIns(4)P-rich regions, where ADP-ribosylation factor-like protein 8B (ARL8B) and SifA- and kinesin-interacting protein/pleckstrin homology domain-containing family M member 2 (SKIP/PLEKHM2) accumulated. SKIP binds preferentially to monophosphorylated phosphoinositides, of which PtdIns(4)P is most abundant in phagolysosomes, contributing to their tubulation. Accordingly, premature hydrolysis of PtdIns(4)P impaired SKIP recruitment and phagosome resolution. Thus, resolution involves phosphoinositides and tethering of phagolysosomes to the endoplasmic reticulum.


Asunto(s)
Retículo Endoplásmico/metabolismo , Monocitos/metabolismo , Fagosomas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Receptores de Esteroides/genética , Transducción de Señal , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Animales , Sistemas CRISPR-Cas , Retículo Endoplásmico/ultraestructura , Edición Génica , Regulación de la Expresión Génica , Humanos , Ratones , Monocitos/ultraestructura , Fagocitosis , Fagosomas/ultraestructura , Cultivo Primario de Células , Proteolisis , Células RAW 264.7 , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
11.
PLoS Biol ; 17(10): e3000145, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31589603

RESUMEN

Male reproductive glands like the mammalian prostate and the paired Drosophila melanogaster accessory glands secrete seminal fluid components that enhance fecundity. In humans, the prostate, stimulated by environmentally regulated endocrine and local androgens, grows throughout adult life. We previously showed that in fly accessory glands, secondary cells (SCs) and their nuclei also grow in adults, a process enhanced by mating and controlled by bone morphogenetic protein (BMP) signalling. Here, we demonstrate that BMP-mediated SC growth is dependent on the receptor for the developmental steroid ecdysone, whose concentration is reported to reflect sociosexual experience in adults. BMP signalling appears to regulate ecdysone receptor (EcR) levels via one or more mechanisms involving the EcR's N terminus or the RNA sequence that encodes it. Nuclear growth in virgin males is dependent on ecdysone, some of which is synthesised in SCs. However, mating induces additional BMP-mediated nuclear growth via a cell type-specific form of hormone-independent EcR signalling, which drives genome endoreplication in a subset of adult SCs. Switching to hormone-independent endoreplication after mating allows growth and secretion to be hyperactivated independently of ecdysone levels in SCs, permitting more rapid replenishment of the accessory gland luminal contents. Our data suggest mechanistic parallels between this physiological, behaviour-induced signalling switch and altered pathological signalling associated with prostate cancer progression.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Ecdisona/metabolismo , Genoma de los Insectos , Proteínas de Insectos/genética , Receptores de Esteroides/genética , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Copulación/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Insectos/antagonistas & inhibidores , Proteínas de Insectos/metabolismo , Masculino , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/metabolismo , Transducción de Señal
12.
Chemosphere ; 237: 124551, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31549662

RESUMEN

To characterize the potential endocrine-disrupting chemicals (EDCs) in the environment that interact with the crustacean ecdysone receptor (EcR), we established a method involving in silico modeling/molecular docking and in vitro reporter gene assay. Cherry shrimp (Neocaridina davidi) EcR (NdEcR) and retinoid X receptor (NdRxR) were identified and cloned for use in this method. A theoretical 3D model of NdEcR ligand-binding domain (LBD) was built in silico based on sequence homology with the established X-ray structure of insect EcR. The interaction of the NdEcR LBD with ecdysteroids, diacylhydrazine (DAH) pesticides, and other potential EDCs was evaluated using molecular docking programs. The results revealed that the ligand-binding pocket in the NdEcR LBD was flexible and adaptive for accommodating ligands of different shapes. The agonistic and antagonistic activities of the candidate compounds were further assessed by in vitro reporter gene assay using human cell lines transiently transfected with NdEcR and NdRxR expression plasmids and a reporter plasmid containing synthesized ecdysone response element. The assay was validated by the dose-dependent responses of EcR-mediated gene transcription after treating the transfected cell lines with ecdysteroids, 20-hydroxyecdysone, and ponasterone A. Examination of the candidate compounds using the reporter gene assay revealed restricted functional specificity to ecdysteroids and DAHs. Three of the tested DAH pesticides originally targeting the insect EcR were found to be weak agonists and strong antagonists of NdEcR. These results suggest that DAHs are potential EDCs for crustaceans that disrupt their ecdysteroid signals by functioning as EcR agonists or antagonists.


Asunto(s)
Crustáceos/efectos de los fármacos , Ecdisteroides/farmacología , Plaguicidas/toxicidad , Receptores de Esteroides/metabolismo , Animales , Sitios de Unión , Línea Celular , Simulación por Computador , Crustáceos/metabolismo , Decápodos/genética , Ecdisona/metabolismo , Ecdisona/farmacología , Ecdisteroides/toxicidad , Ecdisterona/análogos & derivados , Ecdisterona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Simulación del Acoplamiento Molecular , Plaguicidas/química , Plaguicidas/metabolismo , Filogenia , Receptores de Esteroides/agonistas , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/genética , Receptores X Retinoide/química , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo
13.
Inorg Chem ; 58(17): 11782-11792, 2019 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-31433630

RESUMEN

Reproduction of the dominant vector of Zika and dengue diseases, Aedes aegypti mosquito, is controlled by an active heterodimer complex composed of the 20-hydroxyecdysone receptor (EcR) and ultraspiracle protein. Although A. aegypti EcR shares the structural and functional organization with other nuclear receptors, its C-terminus has an additional long F domain (AaFEcR). Recently, we showed that the full length AaFEcR is intrinsically disordered with the ability to specifically bind divalent metal ions. Here, we describe the details of the exhaustive structural and thermodynamic properties of Zn2+- and Cu2+-complexes with the AaFEcR domain, based on peptide models of its two putative metal binding sites (Ac-HGPHPHPHG-NH2 and Ac-QQLTPNQQQHQQQHSQLQQVHANGS-NH2). Unexpectedly, only in the presence of increasing concentrations of Cu2+ ions, the Ac-HGPHPHPHG-NH2 peptide gained a metal ion-induced poly-l-proline type II helical structure, which is unique for members of the family of nuclear receptors.


Asunto(s)
Aedes/efectos de los fármacos , Antivirales/farmacología , Cobre/farmacología , Compuestos Organometálicos/farmacología , Péptidos/farmacología , Receptores de Esteroides/antagonistas & inhibidores , Animales , Antivirales/química , Sitios de Unión/efectos de los fármacos , Cobre/química , Dengue/tratamiento farmacológico , Dengue/metabolismo , Estructura Molecular , Compuestos Organometálicos/química , Péptidos/química , Receptores de Esteroides/metabolismo , Termodinámica , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/metabolismo
14.
Life Sci Alliance ; 2(5)2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31451509

RESUMEN

The small GTPase KRAS, which is frequently mutated in human cancers, must be localized to the plasma membrane (PM) for biological activity. We recently showed that the KRAS C-terminal membrane anchor exhibits exquisite lipid-binding specificity for select species of phosphatidylserine (PtdSer). We, therefore, investigated whether reducing PM PtdSer content is sufficient to abrogate KRAS oncogenesis. Oxysterol-related binding proteins ORP5 and ORP8 exchange PtdSer synthesized in the ER for phosphatidyl-4-phosphate synthesized in the PM. We show that depletion of ORP5 or ORP8 reduced PM PtdSer levels, resulting in extensive mislocalization of KRAS from the PM. Concordantly, ORP5 or ORP8 depletion significantly reduced proliferation and anchorage-independent growth of multiple KRAS-dependent cancer cell lines, and attenuated KRAS signaling in vivo. Similarly, functionally inhibiting ORP5 and ORP8 by inhibiting PI4KIIIα-mediated synthesis of phosphatidyl-4-phosphate at the PM selectively inhibited the growth of KRAS-dependent cancer cell lines over normal cells. Inhibiting KRAS function through regulating PM lipid PtdSer content may represent a viable strategy for KRAS-driven cancers.


Asunto(s)
Membrana Celular/metabolismo , Fosfatidilserinas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/química , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Interferente Pequeño/farmacología , Receptores de Esteroides/antagonistas & inhibidores , Animales , Sitios de Unión/efectos de los fármacos , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Perros , Retículo Endoplásmico/metabolismo , Células HCT116 , Humanos , Células de Riñón Canino Madin Darby , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptores de Esteroides/metabolismo , Transducción de Señal
15.
Antiviral Res ; 170: 104548, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31271764

RESUMEN

Oxysterol-binding Protein (OSBP) is a human lipid-transport protein required for the cellular replication of many types of viruses, including several human pathogens. The structurally-diverse small molecule compounds OSW-1, itraconazole (ITZ), T-00127-HEV2 (THEV) and TTP-8307 (TTP) inhibit viral replication through interaction with the OSBP protein. The OSW-1 compound reduces intracellular OSBP, and the reduction of OSBP protein levels persists multiple days after the OSW-1-compound treatment is stopped. The OSW-1-induced reduction of OSBP levels inhibited Enterovirus replication prophylactically in cells. In this report, the OSBP-interacting compounds ITZ, THEV, and TTP are shown not to reduce OSBP levels in cells, unlike the OSW-1-compound, and the OSW-1 compound is determined to be the only compound capable of providing prophylactic antiviral activity in cells. Furthermore, OSW-1 and THEV inhibit the binding of 25-hydroxycholesterol (25-OHC) to OSBP indicating that these compounds bind at the conserved sterol ligand binding site. The ITZ and TTP compounds do not inhibit 25-hydroxycholesterol binding to OSBP, and therefore ITZ and TTP interact with OSBP through other, unidentified binding sites. Co-administration of the THEV compound partially blocks the cellular activity of OSW-1, including the reduction of cellular OSBP protein levels; co-administration of the ITZ and TTP compounds have minimal effect on OSW-1 cellular activity further supporting different modes of interaction with these compounds to OSBP. OSW-1, ITZ, THEV, and TTP treatment alter OSBP cellular localization and levels, but in four distinct ways. Co-administration of OSW-1 and ITZ induced OSBP cellular localization patterns with features similar to the effects of ITZ and OSW-1 treatment alone. Based on these results, OSBP is capable of interacting with multiple structural classes of antiviral small molecule compounds at different binding sites, and the different compounds have distinct effects on OSBP cellular activity.


Asunto(s)
Antivirales/farmacología , Enterovirus/efectos de los fármacos , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/metabolismo , Replicación Viral/efectos de los fármacos , Línea Celular , Células HEK293 , Células HeLa , Humanos , Hidroxicolesteroles/metabolismo , Unión Proteica
16.
Theriogenology ; 134: 42-52, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31132720

RESUMEN

The objective of the present study was to examine the effects of neonatal exposure to either agonists or antagonists of androgen and estrogen receptors on the expression of growth and differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) and their cognate receptors (TGFBR1, BMPR1B, and BMPR2) in ovarian follicles of adult pigs. Piglets were injected subcutaneously with testosterone propionate (TP, an androgen, at 20 mg/kg bw), flutamide (FLU, an antiandrogen, at 50 mg/kg bw), 4-tert-octylphenol (OP, an estrogenic compound, 100 mg/kg bw), ICI 182,780 (ICI, an antiestrogen, 400 µg/kg bw), or corn oil (control) between postnatal Days 1 and 10 (n = 5/group). Ovarian follicles were excised from adult pigs on Days 8-11 of the estrous cycle. The expression of GDF9, BMP15, TGFBR1, BMPR1B and BMPR2 were examined in the population of preantral and small antral ovarian follicles using real-time PCR, Western blot and immunohistochemistry. In preantral follicles, the upregulation of GDF9 mRNA and protein expression was found in pigs that were neonatally exposed to TP or FLU, while administration of TP or ICI resulted in upregulation of BMP15. TGFBR1 and BMPR2 mRNA and protein expression were upregulated in preantral follicles of adult pigs that were neonatally exposed to TP or FLU, while administration of TP or ICI resulted in upregulation of BMPR1B. In small antral follicles, the mRNA and protein for TGFBR1 and BMPR2 were upregulated, while BMPR1B was downregulated in response to neonatal OP treatment. In addition, treatment with FLU upregulated BMPR1B and BMPR2 mRNA and protein expression, while downregulated the expression of TGFBR1. Moreover, GDF9 and BMP15 were immunolocalized in oocytes and granulosa cells of preantral follicles obtained from both control and treated ovaries. TGFBR1, BMPR1B and BMPR2 receptors were observed in the oocytes and granulosa cells of preantral follicles as well as in granulosa and theca cells of small antral follicles. In conclusion, the present study demonstrated neonatal exposure to either agonists or antagonists of androgen and estrogen receptors affected GDF9 and BMP15 signalling in ovaries of adult pigs. It seems that neonatal androgen excess or deficiency may lead to the acceleration of initial follicle recruitment, while neonatal exposure to compounds with antiandrogenic and estrogenic activity may disturb small antral follicles fate. Therefore, it confirms that neonatal window is critical for programming of ovarian function in pigs.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Folículo Ovárico/metabolismo , Porcinos/fisiología , Animales , Proteína Morfogenética Ósea 15/genética , Proteína Morfogenética Ósea 15/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Femenino , Regulación de la Expresión Génica , Células de la Granulosa/metabolismo , Factor 9 de Diferenciación de Crecimiento/genética , Factor 9 de Diferenciación de Crecimiento/metabolismo , Oocitos/metabolismo , Folículo Ovárico/efectos de los fármacos , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Receptores de Esteroides/agonistas , Receptores de Esteroides/antagonistas & inhibidores , Transducción de Señal
17.
ACS Infect Dis ; 5(6): 962-973, 2019 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-30919621

RESUMEN

Phosphatidylinositol-4 kinase III ß (PI4KB) and oxysterol-binding protein (OSBP) family I provide a conserved host pathway required for enterovirus replication. Here, we analyze the role and essentiality of this pathway in enterovirus replication. Phosphatidylinositol 4-phosphate (PI4P) production and cholesterol accumulation in the replication organelle (RO) are severely suppressed in cells infected with a poliovirus (PV) mutant isolated from a PI4KB-knockout cell line (RD[Δ PI4KB]). Major determinants of the mutant for infectivity in RD(Δ PI4KB) cells map to the A5270U(3A-R54W) and U3881C(2B-F17L) mutations. The 3A mutation is required for PI4KB-independent development of RO. The 2B mutation rather sensitizes PV to PI4KB/OSBP inhibitors by itself but confers substantially complete resistance to the inhibitors with the 3A mutation. The 2B mutation also confers hypersensitivity to interferon alpha treatment on PV. These suggest that the PI4KB/OSBP pathway is not necessarily essential for enterovirus replication in vitro. This work supports a two-step resistance model of enterovirus to PI4KB/OSBP inhibitors involving unique recessive epistasis of 3A and 2B and offers insights into a potential evolutionary pathway of enterovirus toward independence from the PI4KB/OSBP pathway.


Asunto(s)
Evolución Molecular , Mutación , Fosfatidilinositol-4-Fosfato 3-Quinasa/genética , Poliovirus/genética , Receptores de Esteroides/genética , Antivirales/farmacología , Línea Celular Tumoral , Epistasis Genética , Técnicas de Inactivación de Genes , Humanos , Proteínas de la Membrana/genética , Redes y Vías Metabólicas , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilinositol-4-Fosfato 3-Quinasa/antagonistas & inhibidores , Fosfatidilinositol-4-Fosfato 3-Quinasa/metabolismo , Poliovirus/fisiología , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/metabolismo , Proteínas del Núcleo Viral/genética , Proteínas no Estructurales Virales/genética , Replicación Viral
18.
Endocr Rev ; 40(5): 1207-1249, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30869771

RESUMEN

Nuclear receptors (NRs) are transcription factors actively involved in many aspects of human physiology and pathology, serving as sensors of stimuli, master regulators of downstream molecular events, and hubs governing complex gene regulatory networks. The importance of various members of the NR superfamily in cancer has led to substantial efforts to target them therapeutically. Notably, drugs that block the action of estrogen receptor (ER)α in patients with ERα+ breast cancer or the androgen receptor (AR) in patients with prostate cancer have provided remarkable improvements in survival. However, there is continuing need for novel drugs that target ERα or the AR owing to resistance to established drugs, and there are also promising opportunities for targeting other NRs in cancer. In this review, we provide an overview of NR-based drug discovery in cancer and related resistance mechanisms, focusing on novel strategies for targeting well-established NR targets, including ERα, the AR, the glucocorticoid receptor, and the progesterone receptor, as well as opportunities to target other NRs that are attracting interest in immuno-oncology, such as liver X receptors, retinoic acid-related orphan receptors, and farnesoid X receptors.


Asunto(s)
Núcleo Celular/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Receptores de Esteroides/metabolismo , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Descubrimiento de Drogas , Femenino , Humanos , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Progesterona/metabolismo , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/efectos de los fármacos
19.
Cell Rep ; 26(8): 2166-2177.e9, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30784597

RESUMEN

Leukemia stem cells (LSCs) are a rare subpopulation of abnormal hematopoietic stem cells (HSCs) that propagates leukemia and are responsible for the high frequency of relapse in therapies. Detailed insights into LSCs' survival will facilitate the identification of targets for therapeutic approaches. Here, we develop an inhibitor, LYZ-81, which targets ORP4L with high affinity and specificity and selectively eradicates LCSs in vitro and in vivo. ORP4L is expressed in LSCs but not in normal HSCs and is essential for LSC bioenergetics and survival. It extracts PIP2 from the plasma membrane and presents it to PLCß3, enabling IP3 generation and subsequent Ca2+-dependent bioenergetics. LYZ-81 binds ORP4L competitively with PIP2 and blocks PIP2 hydrolysis, resulting in defective Ca2+ signaling. The results provide evidence that LSCs can be eradicated through the inhibition of ORP4L by LYZ-81, which may serve as a starting point of drug development for the elimination of LSCs to eventually cure leukemia.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Leucemia/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Receptores de Esteroides/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Membrana Celular/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia/sangre , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Fosfolipasa C beta/metabolismo , Receptores de Esteroides/antagonistas & inhibidores
20.
Bioorg Med Chem ; 27(6): 1065-1075, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30770256

RESUMEN

The ecdysone receptor (EcR) is an insect nuclear receptor that is activated by the molting hormone, 20-hydroxyecdysone. Because synthetic EcR ligands disrupt the normal growth of insects, they are attractive candidates for new insecticides. In this study, the Molecular Mechanics/Poisson-Boltzmann Surface Area (MM/PBSA) method was used to predict the binding activity of EcR ligands. Validity analyses using 40 known EcR ligands showed that the binding activity was satisfactorily predicted when the ligand conformational free energy term was introduced. Subsequently, this MM/PBSA method was applied to structure-based hierarchical virtual screening, and 12 candidate compounds were selected from a database of 3.8 million compounds. Five of these compounds were active in a cell-based competitive binding assay. The most potent compound is a simple proline derivative with low micromolar binding activity, representing a valuable lead compound for further structural optimization.


Asunto(s)
Proteínas de Insectos/antagonistas & inhibidores , Insecticidas/química , Receptores de Esteroides/antagonistas & inhibidores , Animales , Bases de Datos Farmacéuticas , Diseño de Fármacos , Proteínas de Insectos/metabolismo , Insectos/efectos de los fármacos , Insectos/metabolismo , Insecticidas/metabolismo , Insecticidas/toxicidad , Ligandos , Simulación de Dinámica Molecular , Receptores de Esteroides/metabolismo , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...