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

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Phytomedicine ; 129: 155563, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38552377

RESUMEN

BACKGROUND: Mitotic clonal expansion (MCE) is a prerequisite for preadipocyte differentiation and adipogenesis. Epigallocatechin gallate (EGCG) has been shown to inhibit preadipocyte differentiation. However, the exact molecular mechanisms are still elusive. PURPOSE: This study investigated whether EGCG could inhibit adipogenesis and lipid accumulation by regulating the cell cycle in the MCE phase of adipogenesis and its underlying molecular mechanisms. METHOD: 3T3-L1 preadipocytes were induced to differentiate by a differentiation cocktail (DMI) and were treated with EGCG (25-100 µM) for 9, 18, and 24 h to examine the effect on MCE, or eight days to examine the effect on terminal differentiation. C57BL/6 mice were fed a high-fat diet (HFD) for three months to induce obesity and were given EGCG (50 or 100 mg/kg) daily by gavage. RESULTS: We showed that EGCG significantly inhibited terminal adipogenesis and lipid accumulation in 3T3-L1 cells and decreased expressions of PPARγ, C/EBPα, and FASN. Notably, at the MCE phase, EGCG regulated the cell cycle in sequential order, induced G0/G1 arrest at 18 h, and inhibited the G2/M phase at 24 h upon DMI treatment. Meanwhile, EGCG regulated the expressions of cell cycle regulators (cyclin D1, cyclin E1, CDK4, CDK6, cyclin B1, cyclin B2, p16, and p27), and decreased C/EBPß, PPARγ, and C/EBPα expressions at MCE. Mechanistic studies using STAT3 agonist Colivelin and antagonist C188-9 revealed that EGCG-induced cell cycle arrest in the MCE phase and terminal adipocyte differentiation was mediated by the inhibition of JAK2/STAT3 signaling cascades and STAT3 (Tyr705) nuclear translocation. Furthermore, EGCG significantly protected mice from HFD-induced obesity, reduced body weight and lipid accumulations in adipose tissues, reduced hyperlipidemia and leptin levels, and improved glucose intolerance and insulin sensitivity. Moreover, RNA sequencing (RNA-seq) analysis showed that the cell cycle changes in epididymal white adipose tissue (eWAT) were significantly enriched upon EGCG treatment. We further verified that EGCG treatment significantly reduced expressions of adipogenic factors, cell cycle regulators, and p-STAT3 in eWAT. CONCLUSION: EGCG inhibits MCE, resulting in the inhibition of early and terminal adipocyte differentiation and lipid accumulation, which were mediated by inhibiting p-STAT3 nucleus translocation and activation.


Asunto(s)
Células 3T3-L1 , Adipocitos , Adipogénesis , Catequina , Dieta Alta en Grasa , Janus Quinasa 2 , Ratones Endogámicos C57BL , Factor de Transcripción STAT3 , Animales , Catequina/farmacología , Catequina/análogos & derivados , Ratones , Factor de Transcripción STAT3/metabolismo , Adipogénesis/efectos de los fármacos , Janus Quinasa 2/metabolismo , Adipocitos/efectos de los fármacos , Masculino , Mitosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Obesidad/tratamiento farmacológico , PPAR gamma/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
2.
Phytomedicine ; 110: 154626, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36603342

RESUMEN

BACKGROUND: Ganoderma lucidum polysaccharide (GLP) has many biological properties, however, the anti-fibrosis effect of GLP is unknown at present. PURPOSE: This study aimed to examine the anti-fibrogenic effect of GLP and its underlying molecular mechanisms in vivo and in vitro. STUDY DESIGN: Both CCl4-induced mouse and TGF-ß1-induced HSC-T6 cellular models of fibrosis were established to examine the anti-fibrogenic effect of a water-soluble GLP (25 kDa) extracted from the sporoderm-removed spores of G. lucidum.. METHOD: Serum markers of liver injury, histology and fibrosis of liver tissues, and collagen formation were examined using an automatic biochemical analyzer, H&E staining, Sirius red staining, immunohistochemistry, immunofluorescence, ELISA, Western blotting, and qRT-PCR. RNA-sequencing, enrichment pathway analysis, Western blotting, qRT-PCR, and flow cytometry were employed to identify the potential molecular targets and signaling pathways that are responsible for the anti-fibrotic effect of GLP. RESULTS: We showed that GLP (150 and 300 mg/kg) significantly inhibited hepatic fibrogenesis and inflammation in CCl4-treated mice as mediated by the TLR4/NF-κB/MyD88 signaling pathway. We further demonstrated that GLP significantly inhibited hepatic stellate cell (HSCs) activation in mice and in TGF-ß1-induced HSC-T6 cells as manifested by reduced collagen I and a-SMA expressions. RNA-sequencing uncovered inflammation, apoptosis, cell cycle, ECM-receptor interaction, TLR4/NF-κB, and TGF-ß/Smad signalings as major pathways suppressed by GLP administration. Further studies demonstrated that GLP elicits anti-fibrotic actions that are associated with a novel dual effect on apoptosis in vivo (inhibit) or in vitro (promote), suppression of cell cycle in vivo, induction of S phase arrest in vitro, and attenuation of ECM-receptor interaction-associated molecule expressions including integrins ITGA6 and ITGA8. Furthermore, GLP significantly inhibited the TGF-ß/Smad signaling in mice, and reduced TGF-ß1 or its agonist SRI-011381-induced Smad2 and Smad3 phosphorylations, but increased Samd7 expression in HSC-T6 cells. CONCLUSION: This study provides the first evidence that GLP could be a promising dietary strategy for treating liver fibrosis, which protects against liver fibrosis and HSC activation through targeting inflammation, apoptosis, cell cycle, and ECM-receptor interactions that are mediated by TGF-ß/Smad signaling.


Asunto(s)
Reishi , Factor de Crecimiento Transformador beta1 , Ratones , Animales , Factor de Crecimiento Transformador beta1/metabolismo , FN-kappa B/metabolismo , Receptor Toll-Like 4/metabolismo , Proteínas Smad/metabolismo , Células Estrelladas Hepáticas , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Colágeno Tipo I/metabolismo , Ciclo Celular , Inflamación/metabolismo , Apoptosis , ARN/metabolismo
3.
Phytomedicine ; 103: 154225, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35689899

RESUMEN

BACKGROUND: Koumine is the most abundant alkaloid extracted from Gelsemium elegans Benth.. Preliminary studies by our research group have shown that koumine has significant anxiolytic effect, but this needs to be further confirmed. HYPOTHESIS/PURPOSE: To investigate the potential anxiolytic effect of koumine on predatory sound (PS) stress-induced anxiety models and preliminarily explore its therapeutic targets and molecular mechanisms. STUDY DESIGN AND METHODS: The anxiolytic effect of koumine in an animal model of acute PS stress-induced anxiety were determined. Then, neurosteroids levels in the main brain regions involved in anxiety disorders, as well as plasma adrenocorticotropic hormone (ACTH) and corticosterone (CORT) levels, were determinated. Finally, to clarify the effect of koumine on translocator protein 18 kDa (TSPO), the affinity between koumine and TSPO was evaluated by surface plasmon resonance (SPR) technology. RESULTS: Koumine treatment mitigated anxiety-like behavior following acute PS stress in the open field test and elevated plus maze test. PS exposure significantly decreased progesterone and allopregnanolone levels in the PFC, Hip, and Amy and increased ACTH and CORT levels in plasma, and koumine administration significantly reversed these effects. Finally, the reliable SPR results showed that the KD of koumine with TSPO was 155.33 ± 11.0 µM, indicating that koumine is a human TSPO high-affinity ligand that has an affinity comparable to typical TSPO ligands. CONCLUSION: Our results show that koumine has obvious anxiolytic effect in the PS-induced anxiety model. Targeting TSPO-neurosteroids-HPA axis may be an important mechanism by which koumine exerts its anxiolytic effect.


Asunto(s)
Ansiolíticos , Neuroesteroides , Hormona Adrenocorticotrópica , Animales , Ansiolíticos/farmacología , Ansiolíticos/uso terapéutico , Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/tratamiento farmacológico , Corticosterona , Sistema Hipotálamo-Hipofisario , Alcaloides Indólicos , Ligandos , Sistema Hipófiso-Suprarrenal
4.
J Chem Neuroanat ; 111: 101888, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33212191

RESUMEN

Koumine (KM) is a major alkaloid monomer in the traditional Chinese medicine herb Gelsemium elegans Benth that has exhibited therapeutic potential in clinical applications. However, the pharmacological toxicological mechanism of this drug has not been fully explored. The purpose of this study was to evaluate the impacts of KM administration at a therapeutic dose in offspring. On gestational day 0, mice were injected with KM once daily for 4 consecutive days. Male and female offspring were subjected to behavioral tests and neuropathological analyses from postnatal day 60. Prenatal KM exposure resulted in cognitive and memory impairments in the Morris water maze, Y-maze test, and novel object recognition test. The open field test and elevated plus maze test indicated that prenatal KM exposure induced anxiety-like behavior in offspring. Electrophysiological experiments demonstrated that KM exposure inhibited hippocampal long-term potentiation. Immunostaining for neurogenesis markers DCX and BrdU demonstrated that KM suppressed adult neurogenesis in the subgranular zone of the dentate gyrus. In addition, prenatal KM exposure induced a significant reduction in dendritic spine density in hippocampal neurons. Synaptic formation-related proteins were decreased in the KM group based on western blot. No sex differences in the effects of KM were observed. Collectively, our results indicate that prenatal KA exposure has detrimental neural effects on offspring. This study provides a preliminary preclinical toxicological assessment of the safety of KM use during pregnancy.


Asunto(s)
Ansiedad/fisiopatología , Conducta Animal/efectos de los fármacos , Disfunción Cognitiva/fisiopatología , Alcaloides Indólicos/farmacología , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Espinas Dendríticas/efectos de los fármacos , Proteína Doblecortina , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Masculino , Ratones , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Embarazo
5.
J Nat Med ; 71(2): 397-408, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28105568

RESUMEN

Koumine, an active alkaloid of neurotoxic plant Gelsemium, has been focused on its therapeutic uses, especially in central nervous system. Nevertheless, less is known about the neurological effects of koumine, which hampers its potential therapeutic exploitation. Moreover, as the anxiolytic potential of Gelsemium has raised many critical issues, its active principles on the anxiolytic and other neurological effects need to be further investigated. Here, we used functional observation battery (FOB) of mice to systematically measure the neurological effects of koumine at the effective doses, and then further confirmed its anxiolytic properties in open-field test (OFT) of mice and Vogel conflict test (VCT) of rats. Koumine exhibited anxiolytic-like activities but did not affect other autonomic, neurological and physical functions in FOB. Furthermore, koumine released anxiolytic responses and anti-punishment action in a manner similar to diazepam in OFT and VCT, respectively. The results constitutes solid set of fundamental data further demonstrating anxiolytic properties of koumine at the therapeutic doses without inducing adverse neurological effects, which supports the perspectives for the development of safe and effective koumine medicine against pathological anxiety.


Asunto(s)
Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Gelsemium/química , Alcaloides Indólicos/uso terapéutico , Plantas/química , Animales , Alcaloides Indólicos/farmacología , Masculino , Ratones , Ratas , Ratas Wistar
6.
Zhongguo Zhong Yao Za Zhi ; 39(14): 2716-20, 2014 Jul.
Artículo en Chino | MEDLINE | ID: mdl-25272502

RESUMEN

OBJECTIVE: To investigate the protective effect of extracts from Cichorium endivia (CEE) in H2O2-induced HepG2 cell oxidative stress injury, and explore the antioxidant mechanism of CEE in HepG2 cells. METHOD: The viability of H2O2-induced HepG2 cells and the intracellular ROS level were measured by MTT assay and DCFH-DA fluorescence staining assay. The antioxidant-response element (ARE)-Luciferase activity was tested in HepG2 cells stably transected by ARE reporter gene. The fluorescence quantitative RT-PCR was adopted to determine the mRNA expressions of genes containing ARE sequence in HepG2 cells. RESULT: The cell viability reduced, while the ROS level increased after HepG2 cells were treated by H2O2. Different concentrations of CEE could be added to significantly improve the above results. After HepG2 cells transected by ARE reporter gene were treated with different concentrations of CEE, the intracellular ARE activity could increase in a concentration-dependent manner. In addition, the mRNA expressions of regulatory genesGCLC, GCLM and HMOX-1 containing ARE sequence in HepG2 cells were up-regulated in a concentration-dependent manner by CEE. CONCLUSION: CEE inhibited the H2O2-injured HepG2 cells by reducing the ROS level. CEE's antioxidant mechanism for HepG2 cells may be closely related to the antioxidant defense system associated with its effect of activating Nrf2-ARE pathway in HepG2 cells.


Asunto(s)
Antioxidantes/aislamiento & purificación , Antioxidantes/farmacología , Asteraceae/química , Medicamentos Herbarios Chinos/aislamiento & purificación , Medicamentos Herbarios Chinos/farmacología , Supervivencia Celular/efectos de los fármacos , Células Hep G2 , Humanos , Peróxido de Hidrógeno/farmacología , Especies Reactivas de Oxígeno/metabolismo , Elementos de Respuesta/genética
7.
Molecules ; 16(11): 9049-66, 2011 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-22033140

RESUMEN

The objective of the present study was to investigate the in vitro and in vivo hepatoprotective properties of Cichorium endivia L. extract (CEE), and to identify its chemical constituents. CEE significantly blocked the oxidative stress and cytotoxicity induced by tert-butyl hydroperoxide (t-BHP) in HepG2 cells. Meanwhile, oral administration of CEE to mice before the treatment of t-BHP exhibited a markedly protective effect by lowering serum levels of ALT and AST, inhibiting the changes in liver biochemistry including MDA, SOD, GSH and GST, as well as ameliorating the liver injuries according to the histopathological observations. According to the acute oral toxicity test, the LD(50) of CEE was greater than 5,000 mg/kg, which demonstrates that the CEE can be considered practically non-toxic. Phytochemical analysis of CEE showed the presence of five compounds identified as 2-furanmethanol-(5'→11)-1,3-cyclopentadiene-[5,4-c]-1H-cinnoline, which is a new cinnoline derivative derived from a natural source but not synthesis, 2-phenylethyl-ß-D-glucopyranoside, kaempferol-3-O-ß-D-glucoside, kaempferol, and adenosine. In the ORAC assay, CEE and its constituents kaempferol and kaempferol-3-O-ß-D-glucoside had considerable antioxidant potency. Taken together, CEE protects hepatic tissue from oxidative damage in vitro and in vivo, potentially due to its phenolic substances, and does not cause acute oral toxicity, which suggests that CEE may be a valid and safe remedy to cure liver disease.


Asunto(s)
Asteraceae/química , Hígado/efectos de los fármacos , Extractos Vegetales/química , Extractos Vegetales/farmacología , Sustancias Protectoras/química , Sustancias Protectoras/farmacología , Animales , Antioxidantes/química , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Femenino , Células Hep G2 , Humanos , Quempferoles/química , Quempferoles/farmacología , Quempferoles/uso terapéutico , Hígado/citología , Hígado/patología , Hepatopatías/tratamiento farmacológico , Hepatopatías/patología , Masculino , Ratones , Monosacáridos/química , Monosacáridos/farmacología , Monosacáridos/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Sustancias Protectoras/uso terapéutico , Distribución Aleatoria , Especies Reactivas de Oxígeno/metabolismo , terc-Butilhidroperóxido/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA