Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Heliyon ; 10(13): e33936, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39071636

RESUMEN

Introduction and Objectives: Ferroptosis is a novel form of cell death driven by iron dependence and lipid peroxidation, presenting a promising potential as an innovative strategy for cancer treatment. Celastrol (Cel) is particularly effective in inducing ferroptosis, but its molecular mechanism remains unclear. The study aims to elucidate the potential mechanism through both in vitro and in vivo experiments. Materials and methods: CCK-8 assay, Western blot analysis and measurements of reactive oxygen species (ROS), malondialdehyde (MDA), and glutathione (GSH) were performed to investigate how Cel inhibits the proliferation of hepatocellular carcinoma (HCC) cells via the ferroptosis mechanism. Bioinformatics analysis based on the TCGA-LIHC and FerrDb databases was performed to identify the target gene RRM2, and molecular docking-simulated binding between RRM2 and Cel. The role of RRM2 in the effects of Cel was determined through lentiviral transfection, Transwell assays, and in vivo experiments. Results: Cel inhibited HCC cell proliferation via the ferroptosis pathway. Inhibition RRM2 significantly reduces mTOR protein phosphorylation, while overexpressing RRM2 can attenuate theeffects of Cel on the proliferation, migration, invasion, and ferroptosis induction of HCC cells. The result of in vivo experiments in nude mice demonstrated that Cel inhibited tumor growth without adversely affecting liver and kidney function indicators. Immunohistochemistry and Western blot analyses revealed that Cel activated the key proteins in the ferroptosis pathway and affected crucial indicators such as malondialdehyde (MDA) and glutathione (GSH). Conclusion: In this study, we clarifiy the molecular mechanism of Cel, thus broadening its clinical applications for treating various cancer types, including liver cancer.

2.
Drug Des Devel Ther ; 18: 3121-3141, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39071814

RESUMEN

Background: As a traditional Chinese medicine monomer derived from Tripterygium wilfordii Hook.f. with potential anticancer activity, celastrol can induce ferroptosis in hepatic stellate cells and inhibit their activation to alleviate liver fibrosis. Activation of ferroptosis can effectively inhibit Hepatocellular carcinoma (HCC). Whether celastrol inhibits HCC by inducing ferroptosis remains to be studied. Purpose: To explore the potential targets of celastrol against HCC through ferroptosis based on network pharmacology and to verify the anticancer effect of celastrol on HepG2 cells. Methods: We collected celastrol targets, HCC, and ferroptosis-related genes through online databases, and got their intersection targets. Subsequently, we obtained a protein-protein interaction (PPI) network, and performed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis to gain key genes for further study. They were verified in vitro and were performed molecular docking. The changes in cell proliferation and ferroptosis characteristics of HepG2 cells after celastrol treatment were detected. Results: 31 core target genes were screened for PPI network and enrichment analysis. The most significantly related KEGG pathway was chemical carcinogenesis-reactive oxygen species. The mRNA and protein levels of GSTM1 were significantly decreased after celastrol treatment. Molecular docking demonstrated the interaction between celastrol and GSTM1. Ferroptosis was induced and cell proliferation was inhibited by celastrol in HCC cells. Conclusion: Celastrol induces ferroptosis in HCC via regulating GSTM1 expression and may serve as a novel therapeutic compound with clinical potential in HCC treatment.


Asunto(s)
Carcinoma Hepatocelular , Proliferación Celular , Ferroptosis , Neoplasias Hepáticas , Farmacología en Red , Triterpenos Pentacíclicos , Triterpenos Pentacíclicos/farmacología , Triterpenos Pentacíclicos/química , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Ferroptosis/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Proliferación Celular/efectos de los fármacos , Células Hep G2 , Simulación del Acoplamiento Molecular , Triterpenos/farmacología , Triterpenos/química , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/química , Ensayos de Selección de Medicamentos Antitumorales , Mapas de Interacción de Proteínas/efectos de los fármacos , Relación Dosis-Respuesta a Droga
3.
J Integr Med ; 22(3): 286-294, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38565435

RESUMEN

OBJECTIVE: Research has shown that celastrol can effectively treat a variety of diseases, yet when passing a certain dosage threshold, celastrol becomes toxic, causing complications such as liver and kidney damage and erythrocytopenia, among others. With this dichotomy in mind, it is extremely important to find ways to preserve celastrol's efficacy while reducing or preventing its toxicity. METHODS: In this study, insulin-resistant HepG2 (IR-HepG2) cells were prepared using palmitic acid and used for in vitro experiments. IR-HepG2 cells were treated with celastrol alone or in combination with N-acetylcysteine (NAC) or ferrostatin-1 (Fer-1) for 12, 24 or 48 h, at a range of doses. Cell counting kit-8 assay, Western blotting, quantitative reverse transcription-polymerase chain reaction, glucose consumption assessment, and flow cytometry were performed to measure celastrol's cytotoxicity and whether the cell death was linked to ferroptosis. RESULTS: Celastrol treatment increased lipid oxidation and decreased expression of anti-ferroptosis proteins in IR-HepG2 cells. Celastrol downregulated glutathione peroxidase 4 (GPX4) mRNA. Molecular docking models predicted that solute carrier family 7 member 11 (SLC7A11) and GPX4 were covalently bound by celastrol. Importantly, we found for the first time that the application of ferroptosis inhibitors (especially NAC) was able to reduce celastrol's toxicity while preserving its ability to improve insulin sensitivity in IR-HepG2 cells. CONCLUSION: One potential mechanism of celastrol's cytotoxicity is the induction of ferroptosis, which can be alleviated by treatment with ferroptosis inhibitors. These findings provide a new strategy to block celastrol's toxicity while preserving its therapeutic effects. Please cite this article as: Liu JJ, Zhang X, Qi MM, Chi YB, Cai BL, Peng B, Zhang DH. Ferroptosis inhibitors reduce celastrol toxicity and preserve its insulin sensitizing effects in insulin resistant HepG2 cells. J Integr Med. 2024; 22(3): 286-294.


Asunto(s)
Ferroptosis , Resistencia a la Insulina , Triterpenos Pentacíclicos , Humanos , Células Hep G2 , Triterpenos Pentacíclicos/farmacología , Ferroptosis/efectos de los fármacos , Triterpenos/farmacología , Ciclohexilaminas/farmacología , Acetilcisteína/farmacología , Fenilendiaminas/farmacología , Simulación del Acoplamiento Molecular , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo
4.
Neurosci Lett ; 821: 137629, 2024 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-38191089

RESUMEN

Hyperglycemia exacerbates ischemic brain injury by up-regulating autophagy. However, the underlying mechanisms are unknown. This study aims to determine whether hyperglycemia activates autophagy through the p53-Sesn2-AMPK signaling pathway. Rats were subjected to 30-min middle cerebral artery occlusion (MCAO) with reperfusion for 1- and 3-day under normo- and hyperglycemic conditions; and HT22 cells were exposed to oxygen deprivation (OG) or oxygen-glucose deprivation and re-oxygenation (OGD/R) with high glucose. Autophagy inhibitors, 3-MA and ARI, were used both in vivo and in vitro. The results showed that, compared with the normoglycemia group (NG), hyperglycemia (HG) increased infarct volume and apoptosis in penumbra area, worsened neurological deficit, and augmented autophagy. after MCAO followed by 1-day reperfusion. Further, HG promoted the conversion of LC-3I to LC-3II, decreased p62, increased protein levels of aldose reductase, p53, P-p53ser15, Sesn2, AMPK and numbers of autophagosomes and autolysosomes, detected by transmission electron microscopy and mRFP-GFP-LC3 molecular probe, in the cerebral cortex after ischemia and reperfusion injury in animals or in cultured HT22 cells exposed to hypoxia with high glucose content. Finally, experiments with autophagy inhibitors 3-MA and aldose reductase inhibitor (ARI) revealed that while both inhibitors reduced the number of TUNEL positive neurons and reversed the effects of hyperglycemic ischemia on LC3 and p62, only ARI decreased the levels of p53, P-p53ser15. These results suggested that hyperglycemia might induce excessive autophagy to aggravate the brain injury resulted from I/R and that hyperglycemia might activate the p53-Sesn2-AMPK signaling pathway, in addition to the classical PI3K/AKT/mTOR autophagy pathway.


Asunto(s)
Isquemia Encefálica , Hiperglucemia , Daño por Reperfusión , Animales , Ratas , Aldehído Reductasa/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Glucosa/farmacología , Infarto de la Arteria Cerebral Media , Oxígeno/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
5.
PLoS One ; 18(9): e0291192, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37682882

RESUMEN

Hyperglycemia can exacerbate cerebral ischemia/reperfusion (I/R) injury, and the mechanism involves oxidative stress, apoptosis, autophagy and mitochondrial function. Our previous research showed that selenium (Se) could alleviate this injury. The aim of this study was to examine how selenium alleviates hyperglycemia-mediated exacerbation of cerebral I/R injury by regulating ferroptosis. Middle cerebral artery occlusion (MCAO) and reperfusion models were established in rats under hyperglycemic conditions. An in vitro model of hyperglycemic cerebral I/R injury was created with oxygen-glucose deprivation and reoxygenation (OGD/R) and high glucose was employed. The results showed that hyperglycemia exacerbated cerebral I/R injury, and sodium selenite pretreatment decreased infarct volume, edema and neuronal damage in the cortical penumbra. Moreover, sodium selenite pretreatment increased the survival rate of HT22 cells under OGD/R and high glucose conditions. Pretreatment with sodium selenite reduced the hyperglycemia mediated enhancement of ferroptosis. Furthermore, we observed that pretreatment with sodium selenite increased YAP and TAZ levels in the cytoplasm while decreasing YAP and TAZ levels in the nucleus. The Hippo pathway inhibitor XMU-MP-1 eliminated the inhibitory effect of sodium selenite on ferroptosis. The findings suggest that pretreatment with sodium selenite can regulate ferroptosis by activating the Hippo pathway, and minimize hyperglycemia-mediated exacerbation of cerebral I/R injury.


Asunto(s)
Isquemia Encefálica , Ferroptosis , Hiperglucemia , Daño por Reperfusión , Selenio , Animales , Ratas , Vía de Señalización Hippo , Selenito de Sodio , Daño por Reperfusión/tratamiento farmacológico , Glucosa , Hiperglucemia/complicaciones , Hiperglucemia/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico
6.
Biomed Res Int ; 2023: 1076522, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37082194

RESUMEN

Background: Stroke is the third main reason of mortality, which is the leading reason for adult disability in the globe. Poststroke inflammation is well known to cause acute ischemic stroke- (AIS-) induced brain injury (BI) exacerbation. Celastrol (CL) has exhibited anti-inflammatory activities in various inflammatory traits though underlying mechanisms remain unknown. So, the present investigation is aimed at studying CL protective mechanism against AIS-induced BI. Methods: A mouse model regarding middle cerebral artery occlusion and an oxygen-glucose deprivation (OGD) cell model with or not CL treatment were constructed to study CL protective effects. NF-E2-related factor 2 (Nrf2) was then silenced in BV2 microglia cells (BV2) to study Nrf2 role regarding CL-mediated neuroprotection. Results: The results showed that CL treatment suppressed AIS-induced BI by inhibiting NLRP3/caspase-1 pathway activations and induction of apoptosis and pyroptosis in vivo and in vitro. NLRP3/caspase-1 pathway blocking activation suppressed OGD-induced cell pyroptosis and apoptosis. Also, CL treatment reversed OGD-induced microglial injury by promoting Nrf2/heme oxygenase-1 (HO-1) pathway activations. Nrf2 downregulation reversed CL protective effects against OGD-induced microglial injury, pyroptosis, and apoptosis. Conclusion: The findings advise that CL treatment ameliorated AIS-induced BI by inhibiting microglial injury and activating the Nrf2/HO-1 pathway.


Asunto(s)
Lesiones Encefálicas , Accidente Cerebrovascular Isquémico , Daño por Reperfusión , Ratones , Animales , Hemo-Oxigenasa 1/metabolismo , Microglía/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal , Lesiones Encefálicas/tratamiento farmacológico , Oxígeno/metabolismo , Caspasas/metabolismo , Daño por Reperfusión/metabolismo
7.
Neurochem Res ; 47(5): 1369-1382, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35099719

RESUMEN

Hyperglycemia aggravates cerebral ischemia/reperfusion (I/R) injury via vascular injury. There is still a lack of effective pharmaceutical preparations for cerebral I/R injury under hyperglycemia. This study aimed to investigate the effects of oxymatrine (OMT) on hyperglycemia-exacerbated cerebral I/R injury in vitro and in vivo. The middle cerebral artery occlusion (MCAO) and reperfusion was established in the rats under hyperglycemia. Meanwhile, oxygen-glucose deprivation and reoxygenation (OGD/R) with high glucose was used as an in vitro model of hyperglycemic cerebral I/R injury. The results showed that the neurological deficit score, mortality, infarct volume and penumbra apoptosis in hyperglycemia group were significantly higher than those in normal glucose group. OMT pre-treated obviously reduced the degree of neurological deficit, mortality, infarct volume, improve cerebral blood flow after I/R in rats with hyperglycemia, and increase the survival rate of human brain microvascular endothelial cells (HBMECs) in high glucose and OGD/R group. OMT significantly improved the ultrastructure changes of endothelial cells, and maintain the migration and angiogenesis potency of HBMECs in high glucose and OGD/R group. OMT obviously alleviated the down-regulating CD31 and CD105 expression in cerebral microvessels caused by hyperglycemia. It is concluded that OMT treatment might alleviate cerebral I/R injury under hyperglycemia via protecting microvessels.


Asunto(s)
Alcaloides , Isquemia Encefálica , Quinolizinas , Daño por Reperfusión , Alcaloides/uso terapéutico , Animales , Apoptosis , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Células Endoteliales/metabolismo , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Microvasos/metabolismo , Quinolizinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo
8.
Commun Biol ; 3(1): 466, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811894

RESUMEN

Chinese herbal formulas including the lung-cleaning and toxicity-excluding (LCTE) soup have played an important role in treating the ongoing COVID-19 pandemic (caused by SARS-CoV-2) in China. Applying LCTE outside of China may prove challenging due to the unfamiliar rationale behind its application in terms of Traditional Chinese Medicine. To overcome this barrier, a biochemical understanding of the clinical effects of LCTE is needed. Here, we explore the chemical compounds present in the reported LCTE ingredients and the proteins targeted by these compounds via a network pharmacology analysis. Our results indicate that LCTE contains compounds with the potential to directly inhibit SARS-CoV-2 and inflammation, and that the compound targets proteins highly related to COVID-19's main symptoms. We predict the general effect of LCTE is to affect the pathways involved in viral and other microbial infections, inflammation/cytokine response, and lung diseases. Our work provides a biochemical basis for using LCTE to treat COVID-19 and its main symptoms.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Medicina Tradicional China , Pandemias , Neumonía Viral/tratamiento farmacológico , Antiinflamatorios/análisis , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antivirales/química , Antivirales/uso terapéutico , COVID-19 , Sulfato de Calcio , China/epidemiología , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/metabolismo , Sistemas de Liberación de Medicamentos , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/uso terapéutico , Tracto Gastrointestinal/efectos de los fármacos , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Fitoterapia , Plantas Medicinales/química , Neumonía Viral/epidemiología , Neumonía Viral/metabolismo , Sistema Respiratorio/efectos de los fármacos , SARS-CoV-2 , Proteínas Virales/antagonistas & inhibidores , Tratamiento Farmacológico de COVID-19
9.
J Integr Med ; 18(2): 152-158, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32113846

RESUMEN

OBJECTIVE: In this study we execute a rational screen to identify Chinese medical herbs that are commonly used in treating viral respiratory infections and also contain compounds that might directly inhibit 2019 novel coronavirus (2019-nCoV), an ongoing novel coronavirus that causes pneumonia. METHODS: There were two main steps in the screening process. In the first step we conducted a literature search for natural compounds that had been biologically confirmed as against sever acute respiratory syndrome coronavirus or Middle East respiratory syndrome coronavirus. Resulting compounds were cross-checked for listing in the Traditional Chinese Medicine Systems Pharmacology Database. Compounds meeting both requirements were subjected to absorption, distribution, metabolism and excretion (ADME) evaluation to verify that oral administration would be effective. Next, a docking analysis was used to test whether the compound had the potential for direct 2019-nCoV protein interaction. In the second step we searched Chinese herbal databases to identify plants containing the selected compounds. Plants containing 2 or more of the compounds identified in our screen were then checked against the catalogue for classic herbal usage. Finally, network pharmacology analysis was used to predict the general in vivo effects of each selected herb. RESULTS: Of the natural compounds screened, 13 that exist in traditional Chinese medicines were also found to have potential anti-2019-nCoV activity. Further, 125 Chinese herbs were found to contain 2 or more of these 13 compounds. Of these 125 herbs, 26 are classically catalogued as treating viral respiratory infections. Network pharmacology analysis predicted that the general in vivo roles of these 26 herbal plants were related to regulating viral infection, immune/inflammation reactions and hypoxia response. CONCLUSION: Chinese herbal treatments classically used for treating viral respiratory infection might contain direct anti-2019-nCoV compounds.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Medicamentos Herbarios Chinos/uso terapéutico , Neumonía Viral/tratamiento farmacológico , COVID-19 , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Medicamentos Herbarios Chinos/farmacología , Humanos , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Virosis/tratamiento farmacológico
10.
Int J Biochem Cell Biol ; 114: 105564, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31276786

RESUMEN

Exosomes derived from mesenchymal stem cells (MSCs) are known to participate in myocardial repair after myocardial infarction (MI), but the mechanism remains unclear. Here, we isolated exosomes from adipose-derived MSCs (ADSCs) and examined their effect on MI-induced cardiac damage. To examine the underlying mechanism, H9c2 cells, cardiac fibroblasts, and HAPI cells were used to study the effect of ADSC-exosomes on hypoxia-induced H9c2 apoptosis, TGF-ß1-induced fibrosis of cardiac fibroblasts, and hypoxia-induced macrophage M1 polarization using qRT-PCR, western blot, ELISA, immunohistochemistry, immunofluorescence and flow cytometry. ADSC-exosome treatment mitigated MI-induced cardiac damage by suppressing cardiac dysfunction, cardiac apoptosis, cardiac fibrosis, and inflammatory responses in vitro and in vivo. In addition, ADSC-exosome treatment promoted macrophage M2 polarization. Further experiments found that S1P/SK1/S1PR1 signaling was involved in the ADSC-exosome-mediated myocardial repair. Silencing of S1PR1 reversed the inhibitory effect of ADSC-exosomes on MI-induced cardiac apoptosis and fibrosis in vitro. ADSC-exosome-induced macrophage M2 polarization was also reversed after downregulation of S1PR1 under hypoxia conditions, which promoted NFκB and TGF-ß1 expression, and suppressed the MI-induced cardiac fibrosis and inflammatory response. In sum, these results indicate that ADSC-derived exosomes ameliorate cardiac damage after MI by activating S1P/SK1/S1PR1 signaling and promoting macrophage M2 polarization.


Asunto(s)
Tejido Adiposo/metabolismo , Exosomas/trasplante , Lisofosfolípidos/metabolismo , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Miocardio/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Esfingosina/análogos & derivados , Tejido Adiposo/patología , Animales , Línea Celular , Macrófagos/patología , Masculino , Células Madre Mesenquimatosas/patología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/patología , Ratas , Ratas Sprague-Dawley , Esfingosina/metabolismo
11.
J Cell Biochem ; 120(11): 19019-19030, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31241217

RESUMEN

Circular RNAs (circRNAs) have an important function in human diseases, especially in cancer. circRNA hsa_circ_0014130 (circPIP5K1A), a particularly abundant circRNA, participates in the tumorigenesis of non-small cell lung cancer (NSCLC), although the underlying regulatory mechanism remains unclear. Here, we investigated the circPIP5K1A role in NSCLC. Expression of circPIP5K1A in NSCLC cell lines was explored with quantitative real-time PCR. The effect of circPIP5K1A on NSCLC was evaluated with circPIP5K1A silencing, miR-600 mimic transfection, and hypoxia-inducible factor (HIF)-1α overexpression, followed by assessment of cell proliferation, metastasis, and tumorigenesis in nude mice. The subcellular localization of circPIP5K1A was evaluated via fluorescence in situ hybridization (FISH), and correlation between circPIP5K1A, miR-600, and HIF-1α was assessed by luciferase assay. The data demonstrated that circPIP5K1A expression was increased in NSCLC cells. FISH showed that circPIP5K1A localized to the cytoplasm. The circPIP5K1A knockdown suppressed NSCLC cell metastasis and proliferation by promoting expression of miR-600. Overexpression of miR-600 inhibited HIF-1α-mediated metastasis and proliferation of NSCLC cell by downregulating the endothelial mesenchymal transition-related proteins, Snail and vimentin, and upregulating E-cadherin. In vivo experiments illustrated that circPIP5K1A silence suppressed tumor growth and pulmonary metastasis. The circPIP5K1A may function as an miR-600 sponge to facilitate NSCLC proliferation and metastasis by promoting HIF-1α. A bifluorescein reporter experiment confirmed that miR-600 was the circPIP5K1A target, and miR-600 interacted with the 3' untranslated region of HIF-1α. These results show that circPIP5K1A acted as a tumor promoter through a novel circPIP5K1A/miR-600/HIF-1α axis, which provides candidate markers and therapeutic targets for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proliferación Celular , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Pulmonares/metabolismo , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Circular/metabolismo , ARN Neoplásico/metabolismo , Células A549 , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , ARN Circular/genética , ARN Neoplásico/genética
12.
Can J Diabetes ; 43(3): 165-172, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30287053

RESUMEN

OBJECTIVES: The natural triterpenoid compound celastrol ameliorates insulin resistance (IR) in animal models, but the underlying molecular mechanism is unclear. In this study, we investigated how celastrol regulates IR. METHODS: The HepG2 cellular IR model was initially established with palmitic acid (PA). The expression and activity of glucose transporter 4 (GLUT4), insulin receptor substrate-1 (IRS1) and 9 microRNAs (miRNAs) (miR-7, -34a, -96, -113, -126, -145, -150, -223 and -370) were detected before and after celastrol treatment using the PA-induced HepG2 IR model. RESULTS: The results showed that 250 µM PA for ≥2 days was optimal for inducing IR in HepG2 cells; 600 nM celastrol significantly attenuated the PA-induced IR in HepG2 cells. The PA-induced GLUT4 and IRS1 downregulation and Ser307 phosphorylation on IRS1 was reversed by subsequent treatment with 600 nM celastrol for 6 h. We next investigated which IR-related miRNAs were possible upstream regulators of celastrol-mediated reversal of PA-induced HepG2 IR. Two miRNAs, miR-150 and -223, were significantly downregulated by PA and were re-raised by subsequent celastrol treatment; and miR-223 was upstream of miR-150. Moreover, knocking down miR-223 abolished celastrol's anti-IR effects in the PA-induced model. CONCLUSIONS: Collectively, our results demonstrated that celastrol reverses PA-induced IR-related alterations, in part via miR-223 in HepG2 cells. Further investigation is warranted for establishing the clinical potential of celastrol in treating IR-related disorders.


Asunto(s)
Transportador de Glucosa de Tipo 4/metabolismo , Resistencia a la Insulina , Ácido Palmítico/metabolismo , Triterpenos/farmacología , Animales , Regulación hacia Abajo , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4/genética , Células Hep G2 , Humanos , Resistencia a la Insulina/fisiología , MicroARNs/metabolismo , Triterpenos Pentacíclicos , Fosforilación , Transducción de Señal
13.
Anticancer Drugs ; 29(8): 748-755, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29927769

RESUMEN

The development of resistance to therapy continues to be a serious clinical problem in lung cancer management. Epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) is one of the most common chemotherapy drugs to treat non-small-cell lung cancer. However, almost all treatments fail after ∼1 year of treatment because of drug tolerance, probably occurring from the threonine 790 mutation (T790M) of the EGFR, resulting in overactivation of the EGFR. Celastrol is a natural compound that exhibits antiproliferative activity. In this study, we showed that celastrol combined with EGFR-TKIs significantly suppressed cell invasion of lung cancer cells with a T790M mutation by suppressing the EGFR pathway. Combined therapy with celastrol and EGFR-TKIs inhibited tumor growth in vivo. Together, these results suggested that combined therapy with EGFR-TKIs and celastrol may be a more effective treatment of patients with non-small-cell lung cancer with T790M mutations of the EGFR.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Triterpenos/farmacología , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Triterpenos Pentacíclicos , Inhibidores de Proteínas Quinasas/administración & dosificación , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Triterpenos/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Cell Physiol ; 233(10): 6814-6824, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29667734

RESUMEN

Elevated plasma statured fatty acids (FFAs) cause TLR4/MD2 activation-dependent inflammation and insulin tolerance, which account for the occurrence and development of obesity. It has been confirmed that statured palmitic acid (PA) (the most abundant FFA) could bind MD2 to cause cellular inflammation. The natural compound celastrol could improve obesity, which is suggested via inhibiting inflammation, yet the detailed mechanism for celastrol is still unclear. As celastrol is reported to directly target MD2, we thought disrupting the binding between FFAs and MD2 might be one of the ways for celastrol to inhibit FFAs-caused inflammation and insulin resistance. In this study, we found evidence to support our hypothesis: celastrol could reverse PA-caused TLR4/MD2 activation-dependent insulin resistance, as determined by glucose-lowering ability, cellular glucose uptake, insulin action-related proteins and TLR4/MD2/NF-κB activation. Bioinformatics and cellular experiments showed that both celastrol and PA could bind MD2, and that celastrol could expel PA from cells. Finally, celastrol could reverse high fat diet caused hyperglycemia and obesity, and liver NF-kB activations. Taking together, we proved that celastrol could reverses PA-caused TLR4-MD2 activation-dependent insulin resistance via disrupting PA binding to MD2.


Asunto(s)
Resistencia a la Insulina/fisiología , Ácido Palmítico/metabolismo , Receptor Toll-Like 4/efectos de los fármacos , Triterpenos/farmacología , Animales , Dieta Alta en Grasa , Regulación de la Expresión Génica , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ratones Endogámicos C57BL , Ácido Palmítico/farmacología , Triterpenos Pentacíclicos , Transducción de Señal , Receptor Toll-Like 4/metabolismo
15.
J Neuroinflammation ; 15(1): 78, 2018 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-29540209

RESUMEN

BACKGROUND: Acute ischemic stroke (AIS) is the most common type of cerebrovascular disease and is a leading cause of disability and death worldwide. Recently, a study suggested that transformation of microglia from the pro-inflammatory M1 state to the anti-inflammatory and tissue-reparative M2 phenotype may be an effective therapeutic strategy for ischemic stroke. Celastrol, a traditional oriental medicine, may have anti-inflammatory and neuroprotective effects. However, the underlying mechanisms remain unknown. METHODS: We first determined the expression levels of inflammatory factors in patients and rodent models associated with AIS; we then determined the anti-inflammatory effects of celastrol in AIS, both in vivo and in vitro, using animal models of middle cerebral artery occlusion (MCAO) and cell models of oxygen-glucose deprivation (OGD) treatment with or without celastrol, respectively. RESULTS: The results indicated that expression of both inflammatory (interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α) cytokines, as well as the anti-inflammatory cytokine, IL-33, and IL-10, were increased following AIS in patients and in animal models. Furthermore, in vitro experiments confirmed that celastrol treatment decreased inflammatory cytokine expression induced by OGD through an IL-33/ST2 axis-mediated M2 microglia/macrophage polarization. Finally, celastrol is protected against ischemic-induced nerve injury, both in vivo and in vitro. CONCLUSIONS: Taken together, these data suggest that celastrol post-treatment reduces ischemic stroke-induced brain damage, suggesting celastrol may represent a novel potent pharmacological therapy.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Polaridad Celular/efectos de los fármacos , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-3/metabolismo , Triterpenos/uso terapéutico , Anciano , Animales , Apoptosis/efectos de los fármacos , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/etiología , Lesiones Encefálicas/etiología , Isquemia Encefálica/complicaciones , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/genética , Macrófagos/efectos de los fármacos , Masculino , Microglía/efectos de los fármacos , Persona de Mediana Edad , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Triterpenos Pentacíclicos , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Filtrado Sensorial/efectos de los fármacos , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/etiología
16.
Oncol Lett ; 15(1): 324-330, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29391882

RESUMEN

The present study aimed to investigate the role of endothelial progenitor cells (EPCs) and endothelial cells (ECs) in the peripheral blood of patients with gastric cancer (GC), and to investigate vascular endothelial growth factor (VEGF) expression and microvessel density (MVD) in GC tissues. First, 6 ml peripheral blood with added anticoagulant was collected from each of the 42 patients with GC, followed by determination of the number of EPCs and ECs by flow cytometry using the surface markers cluster of differentiation (CD)34brightCD133+CD31+CD45dim and CD34dimCD133-CD31brightCD45-, respectively. VEGF expression in patients with GC was detected by the streptomycin avidin-peroxidase immunohistochemical method, and MVD was calculated using the marker CD34. EPC and EC levels were positively associated with VEGF expression level, as well as with MVD. VEGF expression was positive in 66.67% GC cases, and its level was significantly associated with tumor-node-metastasis (TNM) stage, invasion depth and lymph-node metastasis (P<0.05). VEGF expression level was also positively associated with MVD. MVD in GC was significantly larger than that in normal tissue (P<0.01), and it was significantly associated with TNM stage (P<0.05), invasion depth (P<0.01) and lymph-node metastasis (P<0.01). EPCs in the peripheral blood have an important role in GC development, and may be a promising indicator of GC diagnosis and prognosis.

17.
Biotechnol Appl Biochem ; 65(3): 390-396, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29274099

RESUMEN

To observe the effects of celastrol on Tau hyperphosphorylation induced by amyloid-ß peptides (Aß) in SH-SY5Y neuroblastoma cells, the changes of Tau hyperphosphorylation and the expression of heat shock protein 90 (HSP90), HSP70, and heat shock factor 1 (HSF-1) in SH-SY5Y cells treated with Aß1-42 and celastrol were measured. Tau hyperphosphorylation and HSP90 expression induced by Aß1-42 was also measured by Western blotting after HSP70 or HSF-1 knockdown by siRNA. The interaction between HSP70 and Tau or HSP70 and carboxyl terminus of HSP70 interacting protein (CHIP) was measured by co-immunoprecipitation. Compared with the control group, the expressions of HSP70 and HSF-1 were markedly decreased after the induction of Aß1-42 , whereas the expressions of HSP90, Tau phospho S199/202, and Tau phospho S396 were markedly increased. Meanwhile, both celastrol treatment and knockdown of HSP70 or HSF-1 in SH-SY5Y cells significantly inhibited the Tau hyperphosphorylation and HSP90 expression induced by Aß1-42 . Moreover, celastrol treatment had no effects on Aß1-42 -induced decreased expression of HSP70 and HSF-1, Tau ubiquitination, and the interaction of HSP70/Tau and HSP70/CHIP. These results suggest that celastrol- inhibited Tau hyperphosphorylation may not be dependent on the cause of HSF-1/HSP70/CHIP-mediated ubiquitination of Tau.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/biosíntesis , Factores de Transcripción del Choque Térmico/biosíntesis , Fragmentos de Péptidos/antagonistas & inhibidores , Triterpenos/farmacología , Proteínas tau/metabolismo , Péptidos beta-Amiloides/farmacología , Humanos , Triterpenos Pentacíclicos , Fragmentos de Péptidos/farmacología , Fosforilación/efectos de los fármacos , Células Tumorales Cultivadas
18.
An Bras Dermatol ; 92(4): 556-558, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28954112

RESUMEN

We report an 80-year-old male patient with severe rheumatoid arthritis who was treated with tripterygium glycoside, an immunosuppressive agent made from the extract of a Chinese medicinal herb called Tripterygium wilfordii Hook F. The patient had no apparent skin lesions before the treatment, but he developed aggressive hyperkeratotic lesions with rapid progression after using tripterygium glycoside. He was repeatedly diagnosed with eczema, but treatment failed to achieve efficacy. Interestingly, a microscopic examination of the lesions revealed numerous scabies mites and eggs. Thus, we confirmed the diagnosis of Norwegian scabies infection. Treated with crotamiton 10% cream and 10% sulfur ointment for one month, the patient's clinical symptoms disappeared.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Glicósidos/efectos adversos , Escabiosis/inducido químicamente , Tripterygium/química , Anciano de 80 o más Años , Animales , Humanos , Masculino , Extractos Vegetales/efectos adversos , Sarcoptes scabiei , Escabiosis/diagnóstico
19.
Exp Ther Med ; 14(2): 1659-1664, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28810633

RESUMEN

The aims of the present study were to establish a single-platform flow cytometry method using 5-(and 6)-carboxyfluorescein diacetate succinimidyl ester (CFSE)-labeled microspheres as the reference for determining endothelial progenitor cell (EPC) number and to evaluate the efficacy of this detection method. Single-platform flow cytometry was used to count cell numbers using CFSE-stained fluorescent microspheres as the internal reference and the EPC numbers in specimens using this novel method were compared with an in vitro clonogenic counting assay. The results of the two counting methods were consistent and compared with the in vitro clonogenic counting assay, the time and cost of the novel method was markedly reduced, as were the corresponding technical requirements. The present findings indicated that single-platform flow cytometry, with CFSE-labeled microspheres as the reference, provides faster and improved detection of EPCs in human peripheral blood specimens, with reduced time and cost, making it more suitable for routine clinical application.

20.
An. bras. dermatol ; 92(4): 556-558, July-Aug. 2017. graf
Artículo en Inglés | LILACS | ID: biblio-887008

RESUMEN

Abstract: We report an 80-year-old male patient with severe rheumatoid arthritis who was treated with tripterygium glycoside, an immunosuppressive agent made from the extract of a Chinese medicinal herb called Tripterygium wilfordii Hook F. The patient had no apparent skin lesions before the treatment, but he developed aggressive hyperkeratotic lesions with rapid progression after using tripterygium glycoside. He was repeatedly diagnosed with eczema, but treatment failed to achieve efficacy. Interestingly, a microscopic examination of the lesions revealed numerous scabies mites and eggs. Thus, we confirmed the diagnosis of Norwegian scabies infection. Treated with crotamiton 10% cream and 10% sulfur ointment for one month, the patient's clinical symptoms disappeared.


Asunto(s)
Humanos , Animales , Masculino , Anciano de 80 o más Años , Artritis Reumatoide/tratamiento farmacológico , Escabiosis/inducido químicamente , Tripterygium/química , Glicósidos/efectos adversos , Sarcoptes scabiei , Escabiosis/diagnóstico , Extractos Vegetales/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA