Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurosci Lett ; 778: 136603, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35364125

RESUMEN

During neurodevelopment, differentiation of neural stem/progenitor cells (NSPCs) into neurons are regulated by many factors including Notch signaling pathway. Herein, we report the effect of a Notch signaling blocker, i.e. γ -secretase inhibitor (GSI), on this differentiating process, especially on the morphological development. NSPCs were cultured and induced to differentiate with or without GSI. The neurite outgrowth was impeded by GSI application and the expression of a Notch signaling downstream effector miR-342-5p increased with the downregulated expression of Notch effectors Hes1 and Hes5. Upregulated expression of miR-342-5p in differentiating NSPCs could shorten the neurite length of progeny neurons, which was similar to the effect of GSI. To avoid the possible influence from astrocytes into neurons, we directly applied cultured neurons, on which GSI could shorten the processes and RBP-J knockdown could also reduce the neurite length. Similarly, transfection of miR-342-5p mimics or inhibitors into PC12 cells led to shorter or longer processes of cells compared with control ones. Furthermore, in differentiating NSPCs, GSI-induced shorter neurites could be partially rescued by miR-342-5p inhibitors, and STAT3 was one of the possible targets of miR-342-5p during this differentiating process as indicated by results of Western Blot test, luciferase reporter assay and GFP reporter assay. To further demonstrate the role of STAT3, it was introduced into GSI-treated neurons and the GSI-affected neurites could also be partially rescued. In conclusion, GSI could influence the morphological development of neurons and the possible mechanism involved Notch/miR-342-5p and STAT3. These results would be informative for future therapeutic research.


Asunto(s)
Inhibidores y Moduladores de Gamma Secretasa , MicroARNs , Células-Madre Neurales , Receptores Notch , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Inhibidores y Moduladores de Gamma Secretasa/farmacología , MicroARNs/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Ratas , Receptores Notch/metabolismo , Transducción de Señal
2.
Scand J Immunol ; 96(2): e13169, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35384009

RESUMEN

Rheumatoid arthritis (RA) is a chronic immune disease involving the small joints, which often causes irreversible damage. In recent years, elevated interleukin 20 (IL-20) has been observed in synovial fluid, while IL-20 receptor overexpression has been observed in synovial cells. IL-20 is a pleiotropic cytokine that participates in various immune diseases. Further understanding of the relationship between IL-20 and RA can help to identify a potential clinical treatment for RA. This study demonstrated that IL-20 can regulate osteoclast differentiation and function in a dose-dependent manner, while influencing the expression of Notch signalling. Quantitative reverse transcription polymerase chain reaction and western blotting showed that γ-secretase-inhibiting drugs can reverse the effects of IL-20. The effects of Notch2 on IL-20-induced osteoclastogenesis were investigated by immunofluorescence and Notch2 gene silencing via transfection of small interfering RNA; the results showed that Notch2 obviously affected the expression levels of the key protein NFATc1 and downstream osteoclastic proteins. In conclusion, we found that IL-20 regulated the osteoclastogenesis in a dose-dependent manner via Notch signalling, primarily by means of Notch2 activity. This study may help to find new targets for RA treatment.


Asunto(s)
Artritis Reumatoide , Inhibidores y Moduladores de Gamma Secretasa , Interleucinas , Osteogénesis , Receptor Notch2 , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Células Cultivadas , Inhibidores y Moduladores de Gamma Secretasa/farmacología , Humanos , Interleucinas/metabolismo , Osteoclastos/metabolismo , Receptor Notch2/genética , Receptor Notch2/metabolismo , Membrana Sinovial/metabolismo
3.
Microvasc Res ; 140: 104308, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34995552

RESUMEN

Intrauterine growth restriction (IUGR) is associated with increased perinatal mortality and morbidity, and plays an important role in the development of adult cardiovascular diseases. This study brings forward a hypothesis that Human umbilical vein endothelial cells (HUVECs) from IUGR newborns present dysfunctions and varying changes of signaling pathways as compared to the Control group. Similar pathways may also be present in pulmonary or systemic vasculatures. HUVECs were derived from newborns. There were three groups according to the different fetal origins: normal newborns (Control), IUGR from poor maternal nutrition (IUGR1), and pregnancy-induced hypertension (IUGR2). We found that IUGR-derived HUVECs showed a proliferative phenotype compared to those from normal subjects. Interestingly, two types IUGR could cause varying degrees of cellular dysfunction. Meanwhile, the Notch1 signaling pathway showed enhanced activation in the two IUGR-induced HUVECs, with subsequent activation of Akt or extracellular signal regulated protein kinases1/2 (ERK1/2). Pharmacological inhibition or gene silencing of Notch1 impeded the proliferative phenotype of IUGR-induced HUVECs and reduced the activation of ERK1/2 and AKT. In summary, elevated Notch1 levels might play a crucial role in IUGR-induced HUVECs disorders through the activation of ERK1/2 and AKT. These pathways could be potential therapeutic targets for prevention of the progression of IUGR associated diseases later in life.


Asunto(s)
Retardo del Crecimiento Fetal/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Patológica , Receptor Notch1/metabolismo , Adulto , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Diaminas/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Retardo del Crecimiento Fetal/patología , Inhibidores y Moduladores de Gamma Secretasa/farmacología , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Recién Nacido , Fenotipo , Fosforilación , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Transducción de Señal , Tiazoles/farmacología
4.
Hepatology ; 75(3): 584-599, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34687050

RESUMEN

BACKGROUND AND AIMS: The mechanisms involved in liver regeneration after partial hepatectomy (pHx) are complicated. Cellular senescence, once linked to aging, plays a pivotal role in wound repair. However, the regulatory effects of cellular senescence on liver regeneration have not been fully elucidated. APPROACH AND RESULTS: Mice subjected to pHx were analyzed 14 days after surgery. The incomplete remodeling of liver sinusoids affected shear stress-induced endothelial nitric oxide synthase (eNOS) signaling on day 14, resulting in the accumulation of senescent LSECs. Removing macrophages to augment LSEC senescence led to a malfunction of the regenerating liver. A dynamic fluctuation in Notch activity accompanied senescent LSEC accumulation during liver regeneration. Endothelial Notch activation by using Cdh5-CreERT NICeCA mice triggered LSEC senescence and senescence-associated secretory phenotype, which disrupted liver regeneration. Blocking the Notch by γ-secretase inhibitor (GSI) diminished senescence and promoted LSEC expansion. Mechanically, Notch-hairy and enhancer of split 1 signaling inhibited sirtuin 1 (Sirt1) transcription by binding to its promoter region. Activation of Sirt1 by SRT1720 neutralized the up-regulation of P53, P21, and P16 caused by Notch activation and eliminated Notch-driven LSEC senescence. Finally, Sirt1 activator promoted liver regeneration by abrogating LSEC senescence and improving sinusoid remodeling. CONCLUSIONS: Shear stress-induced LSEC senescence driven by Notch interferes with liver regeneration after pHx. Sirt1 inhibition accelerates liver regeneration by abrogating Notch-driven senescence, providing a potential opportunity to target senescent cells and facilitate liver repair after injury.


Asunto(s)
Senescencia Celular , Regeneración Hepática , Receptores Notch , Transducción de Señal/efectos de los fármacos , Sirtuina 1/metabolismo , Animales , Senescencia Celular/efectos de los fármacos , Senescencia Celular/fisiología , Inhibidores y Moduladores de Gamma Secretasa/farmacología , Hepatectomía/métodos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Regeneración Hepática/efectos de los fármacos , Regeneración Hepática/fisiología , Ratones , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptores Notch/antagonistas & inhibidores , Receptores Notch/metabolismo , Fenotipo Secretor Asociado a la Senescencia/genética
5.
J Med Chem ; 64(19): 14426-14447, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34550687

RESUMEN

The recent approval of aducanumab for Alzheimer's disease has heightened the interest in therapies targeting the amyloid hypothesis. Our research has focused on identification of novel compounds to improve amyloid processing by modulating gamma secretase activity, thereby addressing a significant biological deficit known to plague the familial form of the disease. Herein, we describe the design, synthesis, and optimization of new gamma secretase modulators (GSMs) based on previously reported oxadiazine 1. Potency improvements with a focus on predicted and measured properties afforded high-quality compounds further differentiated via robust Aß42 reductions in both rodents and nonhuman primates. Extensive preclinical profiling, efficacy studies, and safety studies resulted in the nomination of FRM-024, (+)-cis-5-(4-chlorophenyl)-6-cyclopropyl-3-(6-methoxy-5-(4-methyl-1H-imidazole-1-yl)pyridin-2-yl)-5,6-dihydro-4H-1,2,4-oxadiazine, as a GSM preclinical candidate for familial Alzheimer's disease.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Encéfalo/metabolismo , Descubrimiento de Drogas , Inhibidores y Moduladores de Gamma Secretasa/farmacología , Péptidos beta-Amiloides/metabolismo , Animales , Área Bajo la Curva , Perros , Inhibidores y Moduladores de Gamma Secretasa/farmacocinética , Semivida , Haplorrinos , Humanos , Ratones , Fragmentos de Péptidos/metabolismo , Ratas
6.
Int J Med Sci ; 18(12): 2551-2560, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34104086

RESUMEN

Malignant gliomas are a type of central nervous system cancer with extremely high mortality rates in humans. γ-secretase has been becoming a potential target for cancer therapy, including glioma, because of the involvement of its enzymatic activity in regulating the proliferation and metastasis of cancer cells. In this study, we attempted to determine whether γ-secretase activity regulates E-cadherin to affect glioma cell migration. The human glioma cell lines, including LN18 and LN229, and the γ-secretase inhibitors, including N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) and RO4929097, were used in this study. It was shown that γ-secretase activity inhibition by DAPT and RO4929097 could promote LN18 and LN229 glioma cell migration via downregulating E-cadherin mRNA and protein expressions, but not via affecting E-cadherin protein processing. In addition, γ-secretase activity inhibition was regulated by bone morphogenetic proteins-independent Smad5 activation in glioma cells. Moreover, endogenous Smad1 in glioma cells was found to play an important role in regulating E-cadherin expression and subsequent cell migration but did not affect DAPT-stimulated effects. These results help further elucidate the molecular mechanisms of γ-secretase activity regulation involved in controlling glioma cell malignancy. Information about a potential role for Smad1/5 activity upregulation and subsequent E-cadherin downregulation during inhibition of γ-secretase activity in the development of gliomas is therefore relevant for future research.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Inhibidores y Moduladores de Gamma Secretasa/farmacología , Glioma/tratamiento farmacológico , Antígenos CD/genética , Benzazepinas/farmacología , Benzazepinas/uso terapéutico , Neoplasias Encefálicas/patología , Cadherinas/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Diaminas/farmacología , Diaminas/uso terapéutico , Regulación hacia Abajo/efectos de los fármacos , Inhibidores y Moduladores de Gamma Secretasa/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/patología , Humanos , Proteína Smad5/metabolismo , Tiazoles/farmacología , Tiazoles/uso terapéutico
7.
Molecules ; 27(1)2021 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-35011410

RESUMEN

γ-Secretase is an intramembrane aspartyl protease that is important in regulating normal cell physiology via cleavage of over 100 transmembrane proteins, including Amyloid Precursor Protein (APP) and Notch family receptors. However, aberrant proteolysis of substrates has implications in the progression of disease pathologies, including Alzheimer's disease (AD), cancers, and skin disorders. While several γ-secretase inhibitors have been identified, there has been toxicity observed in clinical trials associated with non-selective enzyme inhibition. To address this, γ-secretase modulators have been identified and pursued as more selective agents. Recent structural evidence has provided an insight into how γ-secretase inhibitors and modulators are recognized by γ-secretase, providing a platform for rational drug design targeting this protease. In this study, docking- and pharmacophore-based screening approaches were evaluated for their ability to identify, from libraries of known inhibitors and modulators with decoys with similar physicochemical properties, γ-secretase inhibitors and modulators. Using these libraries, we defined strategies for identifying both γ-secretase inhibitors and modulators incorporating an initial pharmacophore-based screen followed by a docking-based screen, with each strategy employing distinct γ-secretase structures. Furthermore, known γ-secretase inhibitors and modulators were able to be identified from an external set of bioactive molecules following application of the derived screening strategies. The approaches described herein will inform the discovery of novel small molecules targeting γ-secretase.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/química , Descubrimiento de Drogas/métodos , Inhibidores y Moduladores de Gamma Secretasa/química , Modelos Moleculares , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Inhibidores y Moduladores de Gamma Secretasa/farmacología , Humanos , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Reproducibilidad de los Resultados , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA