Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Pharmacol Sci ; 150(2): 110-122, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36055749

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases, and there are no effective drugs available so far. Lactucin and Lactucopicrin belong to sesquiterpene lactones and are extracted from Cichorium glandulosum Boiss. et Huet (CG) possesses multiple biopharmacological activities. However, the therapeutic effects of both Lactucin and Lactucopicrin on many diseases and their underlying mechanisms remain largely unknown. Here, we analyzed the both natural compounds hypolipidemic effects on FFA-induced HepG2 cells and their potential mechanisms based on transcriptomics and experimental tests. Our results indicated that Lactucin (10 µM) and Lactucopicrin (20 µM) remarkably reduced TG accumulation. Transcriptomics analysis identified 1960, 1645, and 1791 differentially expressed genes (DEGs) and obtained 611 and 635 specific genes in different comparisons, respectively. The enrichment analysis and experimental validations (RT-qPCR and Western Blot) showed that their hypolipidemic activities were most probably exerted via regulating numerous key DEGs involved in lipid metabolism. Taken together, both Lactucin and Lactucopicrin may represent potent hepatoprotective agents. Both of them exhibited therapeutic effects against liver diseases such as NAFLD by regulating multi-gene and proteins like HADHA, ADAM17, SQSTM1, and GBA and modulating multi-pathways like fatty acid oxidation metabolic signaling.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Sesquiterpenos , Células Hep G2 , Humanos , Lactonas/farmacología , Lactonas/uso terapéutico , Metabolismo de los Lípidos/genética , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/genética , Forboles , Sesquiterpenos/farmacología , Sesquiterpenos/uso terapéutico
2.
Mol Biol Rep ; 48(11): 7261-7272, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34626309

RESUMEN

BACKGROUND: The tumor suppressor protein p53 is a most promising target for the development of anticancer drugs. Allicin (diallylthiosulfinate) is one of the most active components of garlic (Alliium sativum L.) and possesses a variety of health-promoting properties with pharmacological applications. However, whether allicin plays an anti-cancer role against breast cancer cells through the induction of p53-mediated apoptosis remains unknown. METHODS AND RESULTS: In this study, we investigate the anti-breast cancer effect of allicin in vitro by using MCF-7 and MD-MBA-231 cells. We found that allicin reduces cell viability, induces apoptosis and cell cycle arrest in both cells. Allicin activated p53 and caspase 3 expressions in both cells but produced different effects on the expression of p53-related biomarkers. In MDA-MB-231 cells, allicin up-regulated the mRNA and protein expression of A1BG and THBS1 while down-regulated the expression of TPM4. Conversely, the mRNA and protein expression of A1BG, THBS1 and TPM4 were all reduced in MCF-7 cells. Hence, allicin induces cell cycle arrest and apoptosis in breast cancer cells through p53 activation but it effects on the expression of p53-related biomarkers were dependent upon the specific type of breast cancer involved. CONCLUSIONS: These findings suggest that allicin induces apoptosis and regulates biomarker expression in breast cancer cell lines through modulating the p53 signaling pathway. Furthermore, our results promote the utility of allicin as compound for further studies as an anticancer drug targeting p53.


Asunto(s)
Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Puntos de Control del Ciclo Celular , Disulfuros/farmacología , Transducción de Señal , Ácidos Sulfínicos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/fisiopatología , Caspasa 3/genética , Línea Celular Tumoral , Disulfuros/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Ácidos Sulfínicos/uso terapéutico , Proteína p53 Supresora de Tumor/genética
3.
BMC Complement Altern Med ; 17(1): 201, 2017 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-28388901

RESUMEN

BACKGROUND: Abnormal Savda Munziq (ASMq) is a traditional prescription in Uyghur Medicine, and its treatment of complex diseases such as tumors and asthma has been proven to be effective in Uyghur medical clinical practice. The efficacy-enhancing and toxicity-reducing properties of ASMq were studied on mice with transplanted cervical cancer (U27) tumors, which were treated with 5-fluorouracil (5-FU) in this work. METHODS: To investigate the synergistic effect of ASMq and 5-FU on U27 cells, inhibitory effects on cell proliferation were determined through a MTT assay. 48 Kunming mice which were randomly divided in to 6 groups: control group, model group, 5-FU group, 5-FU combine with ASMq low-dose group, 5-FU combine with ASMq medium-dose group, and 5-FU combine with ASMq high- dose group, the inhibition rate of the tumor, the viscera indexes, and the content of serum tumor necrosis factor-α (TNF-α), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were determined. The expression levels of transforming growth factor-ß1 (TGF-ß1) and human papillomavirus type 16 E2 (HPV16 E2) protein were assessed by Western blot. Pathological changes in the liver were observed. RESULT: The inhibition rates of tumors, the 5-FU + ASMq.H group(80.64%), 5-FU + ASMq.M group (90.67%), 5-FU + ASMq.L group (72.03%) and 5-FU group (66.89%), clearly indicated that the effects of tumor inhibition. The thymus index and spleen index were increased, and the serum concentration of TNF-α increased while ALT and AST concentrations were decreased, and TNF-α protein expression were increased while TGF-ß1 and HPV16 E2 were decreased. ASMq might can improve livers central vein hyperemia and interstitial edema, and preserve the radial structure of the hepatic cords. CONCLUSIONS: The results suggested that ASMq might reduce toxicity and enhance the efficacy of the chemotherapeutic drug 5-fluorouracil in the treatment of cervical carcinoma.


Asunto(s)
Antineoplásicos Fitogénicos/toxicidad , Fluorouracilo/toxicidad , Extractos Vegetales/administración & dosificación , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Fluorouracilo/uso terapéutico , Humanos , Medicinas Tradicionales Africanas , Ratones , Factor de Crecimiento Transformador beta1/sangre , Factor de Necrosis Tumoral alfa/sangre , Neoplasias del Cuello Uterino/sangre
4.
Food Sci Nutr ; 10(12): 4371-4379, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36514773

RESUMEN

Harmol hydrochloride dihydrate (HHD) is a novel alkaloid salt of the natural ß-carboline harmol, which is isolated from Peganum harmala L. Here, we studied whether HHD could induce autophagy in neuro cells and investigated the underlying molecular mechanism. After incubation with HHD, the number of GFP-LC3 puncta in cells was measured using confocal microscopy. The distribution and colocalization of autophagosomes and autolysosomes in the cells were also detected. LC3 was gathered and cultured in a medium containing HHD. Compared with control cells and cells starved for 2 h, the number of GFP-LC3 puncta and the LC3-II expression level were significantly increased in HHD-treated cells (p < .05). The number of autophagosome (red) was increased and most of them were colocalized with lysosomes (green). Moreover, HHD induced the formation of puncta with Lysotracker Red positive in the L3 fat bodies (p < .05). When treated HEK cells with HHD, the protein expression level of LC3-II was markedly increased, and the protein expression level of α-Syn was significantly decreased (p < .05). HHD could induce the increased autophagosome in neuro cells by induction of autophagy. Moreover, HHD may promote the degradation of α-Syn protein to protect neuro cells by inducing autophagy.

5.
Artículo en Inglés | MEDLINE | ID: mdl-35432563

RESUMEN

This paper aims to study the potential biological mechanism of Üstikuddus Sherbiti (ÜS) in the treatment of ischemic cerebrovascular diseases (ICVD) by the network pharmacology method. Traditional Chinese Medicine Systems Pharmacology (TCMSP) database was used to obtain effective constituents of ÜS by screening eligible oral utilization, drug similarity, and blood-brain barrier permeability threshold. By drug target prediction and stroke treatment target mining, 2 target data sets were analyzed to find intersection targets and the corresponding constituents were used as active constituents. An active constituent target network and an effective constituent target network were constructed by using Cytoscape 3.7.2 software. Degree parameters of the effective constituent target network were analyzed to find important effective constituents and targets. Through protein-protein interaction (PPI) analysis/Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, potential signaling pathways of ÜS in ischemic stroke were found out. AutoDock was used for molecular docking verification. A total of 90 active constituents of ÜS were screened out. There were 10 active constituents against ICVD, including quercetin, luteolin, kaempferol, and naringenin, and 10 important targets for anticerebral ischemia, namely, PIK3CA, APP, PIK3R1, MAPK1, MAPK3, AKT1, PRKCD, Fyn, RAC1, and NF-κB1. Based on the protein interaction network, the important targets of ÜS were significantly enriched in PI3K-Akt signaling pathway, neuroactive ligand-receptor interaction pathway, Ras signaling pathway, etc. ÜS in ICVD has characteristics like multiple targets, multiple approaches, and multiple pathways. Results of molecular docking showed that the active components in ICVD had a good binding ability with the key targets. Its main biological mechanism may be related to the PI3K-Akt and Ras-MAPK centered signaling pathway. Our study demonstrated that ÜS exerted the effect of treating ICVD by regulating multiple targets and multiple channels with multiple components through the method of network pharmacology and molecular docking.

6.
Biomed Pharmacother ; 141: 111802, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34147903

RESUMEN

OBJECTIVE: To study the effects of irbesartan on pulmonary artery lesions in a rat model with chronic mountain sickness (CMS) and identify the biomarkers involved. METHODS: In this study, we used a rat model of CMS to evaluate the therapeutic effect of irbesartan by measuring pulmonary artery pressure and evaluating the histopathology of the pulmonary artery. We also used proteomics technology to identify differentially expressed proteins (DEPs) in the serum and performed bioinformatics analysis. Results were then verified by enzyme linked immunosorbent assay (ELISA) and immunohistochemistry (IHC). RESULTS: Irbesartan treatment induced a significant decrease (P < 0.05) in the pulmonary artery pressure of CMS rats. Histopathological and electron microscope further confirmed that high altitude hypoxia induced changes in the structure of the pulmonary artery tissue and caused ultrastructural lesions. Proteomics analysis identified 40 DEPs; bioinformatics analysis further revealed that the cholesterol metabolism pathway plays a crucial role in the occurrence of CMS. ELISA and IHC verified that several DEPs (Apo-A1, Apo-C1, Apo-E, IGF-1, Profilin1, and Col1a1) represent critical biological markers in pulmonary artery disease caused by CMS. CONCLUSIONS: Irbesartan significantly improved pulmonary artery damage in a rat model of CMS possibly by impacting on the cholesterol metabolism pathway and by reducing damage to vascular endothelial cells. Irbesartan also inhibited the expression levels of IGF-1, Profilin1 and Col1a1 to relieve pulmonary artery pressure and improve lung function by inhibiting vascular remodeling. Several proteins were identified as potential biomarkers of CMS, including Apo-A1, Apo-C1, Apo-E, IGF-1, Profilin1, and Col1a1.


Asunto(s)
Mal de Altura/tratamiento farmacológico , Mal de Altura/metabolismo , Colesterol/metabolismo , Irbesartán/uso terapéutico , Mapas de Interacción de Proteínas/efectos de los fármacos , Proteómica/métodos , Animales , Antihipertensivos/farmacología , Antihipertensivos/uso terapéutico , Enfermedad Crónica/tratamiento farmacológico , Irbesartán/farmacología , Mapas de Interacción de Proteínas/fisiología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Circulación Pulmonar/efectos de los fármacos , Circulación Pulmonar/fisiología , Ratas , Ratas Sprague-Dawley , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/fisiología
7.
J Biophotonics ; 14(11): e202100117, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34331509

RESUMEN

Photothermal therapy (PTT) has attracted wide attention due to its noninvasiveness and its thermal ablation ability. As photothermal agents are crucial factor in PTT, those with the characteristics of biocompatibility, non-toxicity and high photothermal stability have attracted great interest. In this work, new indocyanine green (IR-820) was utilized as a photothermal agent and near-infrared (NIR) fluorescence imaging nanoprobe. To improve the biocompatibility, poly(styrene-co-maleic anhydride) (PSMA) was utilized to encapsulate the IR-820 molecules to form novel IR-820@PSMA nanoparticles (NPs). Then, the optical and thermal properties of IR-820@PSMA NPs were studied in detail. The IR-820@PSMA NPs showed excellent photothermal stability and biocompatibility. The cellular uptaking ability of the IR-820@PSMA NPs was further confirmed in HeLa cells by the NIR fluorescent confocal microscopic imaging technique. The IR-820@PSMA NPs assisted PTT of living HeLa cells was conducted under 793 nm laser excitation, and a high PTT efficiency of 73.3% was obtained.


Asunto(s)
Nanopartículas , Neoplasias del Cuello Uterino , Femenino , Células HeLa , Humanos , Verde de Indocianina/análogos & derivados , Fototerapia , Terapia Fototérmica , Polímeros , Neoplasias del Cuello Uterino/diagnóstico por imagen , Neoplasias del Cuello Uterino/terapia
8.
Eur J Pharmacol ; 903: 174136, 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-33940032

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease. At present, the incidence rate of PD is increasing worldwide, there is no effective cure available so far, and currently using drugs are still limited in efficacy due to serious side effects. Acteoside (ACT) is an active ingredient of many valuable medicinal plants, possesses potential therapeutic effects on many pathological conditions. In this study, we dissected the neuroprotection effects of ACT on PD and its potential molecular mechanism in our PD model pathology based on network pharmacology prediction and experimental assays. Network pharmacology and bioinformatics analysis demonstrated that ACT has 381 potential targets; among them 78 putative targets associated with PD were closely related to cellular autophagy and apoptotic processes. Our experimental results showed that ACT exerted significant neuroprotection effects on Rotenone (ROT) -induced injury of neuronal cells and Drosophila melanogaster (D. melanogaster). Meanwhile, ACT treatment induced autophagy in both neuronal cell lines and fat bodies of D. melanogaster. Furthermore, ACT treatment decreased ROT induced apoptotic rate and reactive oxygen species production, increased mitochondrial membrane potentials in neuronal cells, and promoted clearance of α-synuclein (SNCA) aggregations in SNCA overexpressed cell model through the autophagy-lysosome pathway. Interestingly, ACT treatment significantly enhanced mitophagy and protected cell injury in neuronal cells. Taken together, ACT may represent a potent stimulator of mitophagy pathway, thereby exerts preventive and therapeutic effects against neurodegenerative diseases such as PD by clearing pathogenic proteins and impaired cellular organelles like damaged mitochondria in neurons.


Asunto(s)
Autofagia/efectos de los fármacos , Glucósidos/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/prevención & control , Fenoles/farmacología , Adenilato Quinasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Biología Computacional , Drosophila melanogaster , Glucósidos/uso terapéutico , Células HEK293 , Humanos , Longevidad/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Modelos Biológicos , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Fenoles/uso terapéutico , Ratas , Especies Reactivas de Oxígeno/metabolismo , Rotenona/toxicidad , alfa-Sinucleína/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA