Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Front Biosci (Landmark Ed) ; 29(1): 26, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-38287818

RESUMEN

Vascular diseases are amongst the most serious diseases affecting human life and health globally. Energy metabolism plays a crucial role in multiple vascular diseases, and the imbalance of energy metabolism in cells from the blood vessel wall can cause various vascular diseases. Energy metabolism studies have often focused on atherosclerosis (AS) and pulmonary hypertension (PH). However, the roles of energy metabolism in the development of other vascular diseases is becoming increasingly appreciated as both dynamic and essential. This review summarizes the role of energy metabolism in various vascular diseases, including AS, hemangioma, aortic dissection, PH, vascular aging, and arterial embolism. It also discusses how energy metabolism participates in the pathophysiological processes of vascular diseases and potential drugs that may interfere with energy metabolism. This review presents suggestions for the clinical prevention and treatment of vascular diseases from the perspective of energy metabolism.


Asunto(s)
Hipertensión Pulmonar , Enfermedades Vasculares , Humanos , Metabolismo Energético , Enfermedades Vasculares/metabolismo , Hipertensión Pulmonar/metabolismo
2.
Yao Xue Xue Bao ; 48(1): 149-54, 2013 Jan.
Artículo en Zh | MEDLINE | ID: mdl-23600157

RESUMEN

The purpose of this study is to investigate the intracellular transporters effect and the cytotoxicity of carboxyl nanodiamond (CND) - podophyllotoxin (PPT). Nanodiamond (ND) was treated with mixed carboxylic acid and finally got 64 nm CND by centrifugation, and then it was reacted with PPT to form CND-PPT. UV spectrophotometry was used to calculate the content of PPT in CND-PPT, the particle size distribution and zeta potential were measured by Dynamic laser scattering instrument. CND, PPT, CND-PPT and CND + PPT (physical mixture of CND and PPT) were characterized by Fourier transform infrared spectroscopy, at the same time, thermal analysis and element analysis were used to estimate the content of the PPT in CND-PPT. The affect of CND, PPT, CND-PPT on HeLa cell was measured with MTT assay. The results showed that content of PPT combined with CND accounted for about 10%. MTT assay showed that CND has low cytotoxicity and CND-PPT can increase the water soluble of PPT. As a conclusion, CND as a hydrophilic pharmaceutical carrier combined with PPT is able to increase the water solubility of PPT, at low concentration, CND-PPT can enhance the antitumor activity in comparison with PPT, so CND can be used as a potential anticancer drug carrier.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Nanodiamantes/química , Podofilotoxina/administración & dosificación , Podofilotoxina/farmacología , Antineoplásicos Fitogénicos/química , Ácidos Carboxílicos/química , Portadores de Fármacos , Células HeLa/efectos de los fármacos , Humanos , Tamaño de la Partícula , Podofilotoxina/química , Solubilidad , Espectrofotometría Ultravioleta , Espectroscopía Infrarroja por Transformada de Fourier
3.
Phytomedicine ; 120: 155046, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37659297

RESUMEN

BACKGROUND: Doxorubicin (Dox), a chemotherapeutic agent known for its efficacy, has been associated with the development of severe cardiotoxicity, commonly referred to as doxorubicin-induced cardiotoxicity (DIC). The role and mechanism of action of phloretin (Phl) in cardiovascular diseases are well-established; however, its specific function and underlying mechanism in the context of DIC have yet to be fully elucidated. OBJECTIVE: This research aimed to uncover the protective effect of Phl against DIC in vivo and in vitro, while also providing a comprehensive understanding of the underlying mechanisms involved. METHODS: DIC cell and murine models were established. The action targets and mechanism of Phl against DIC were comprehensively examined by systematic network pharmacology, molecular docking, transcriptomics technologies, transcription factor (TF) prediction, and experimental validation. RESULTS: Phl relieved Dox-induced cell apoptosis in vitro and in vivo. Through network pharmacology analysis, a total of 554 co-targeted genes of Phl and Dox were identified. Enrichment analysis revealed several key pathways including the PI3K-Akt signaling pathway, Apoptosis, and the IL-17 signaling pathway. Protein-protein interaction (PPI) analysis identified 24 core co-targeted genes, such as Fos, Jun, Hif1a, which were predicted to bind well to Phl based on molecular docking. Transcriptomics analysis was performed to identify the top 20 differentially expressed genes (DEGs), and 202 transcription factors (TFs) were predicted for these DEGs. Among these TFs, 10 TFs (Fos, Jun, Hif1a, etc.) are also the co-targeted genes, and 3 TFs (Fos, Jun, Hif1a) are also the core co-targeted genes. Further experiments validated the finding that Phl reduced the elevated levels of Hif3a (one of the top 20 DEGs) and Fos (one of Hif3a's predicted TFs) induced by Dox. Moreover, the interaction between Fos protein and the Hif3a promoter was confirmed through luciferase reporter assays. CONCLUSION: Phl actively targeted and down-regulated the Fos protein to inhibit its binding to the promoter region of Hif3a, thereby providing protection against DIC.


Asunto(s)
Cardiotoxicidad , Floretina , Animales , Ratones , Simulación del Acoplamiento Molecular , Fosfatidilinositol 3-Quinasas , Doxorrubicina/efectos adversos , Proteínas Represoras , Proteínas Reguladoras de la Apoptosis
4.
Biochem Pharmacol ; 214: 115662, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37331637

RESUMEN

Trastuzumab (Tra), the first humanized monoclonal antibody that targets human epidermal growth factor receptor 2 (HER2), is commonly used alongside doxorubicin (Dox) as a combination therapy in HER2-positive breast cancer. Unfortunately, this leads to a more severe cardiotoxicity than Dox alone. NLRP3 inflammasome is known to be involved in Dox-induced cardiotoxicity and multiple cardiovascular diseases. However, whether the NLRP3 inflammasome contributes to the synergistic cardiotoxicity of Tra has not been elucidated. In this study, primary neonatal rat cardiomyocyte (PNRC), H9c2 cells and mice were treated with Dox (15 mg/kg in mice or 1 µM in cardiomyocyte) or Tra (15.75 mg/kg in mice or 1 µM in cardiomyocyte), or Dox combined Tra as cardiotoxicity models to investigate this question. Our results demonstrated that Tra significantly potentiated Dox-induced cardiomyocyte apoptosis and cardiac dysfunction. These were accompanied by the increased expressions of NLRP3 inflammasome components (NLRP3, ASC and cleaved caspase-1), the secretion of IL-ß and the pronounced production of ROS. Inhibiting the activation of NLRP3 inflammasome by NLRP3 silencing significantly reduced cell apoptosis and ROS production in Dox combined Tra-treated PNRC. Compared with the wild type mice, the systolic dysfunction, myocardial hypertrophy, cardiomyocyte apoptosis and oxidative stress induced by Dox combined Tra were alleviated in NLRP3 gene knockout mice. Our data revealed that the co-activation of NLRP3 inflammasome by Tra promoted the inflammation, oxidative stress and cardiomyocytes apoptosis in Dox combined Tra-induced cardiotoxicity model both in vivo and in vitro. Our results suggest that NLRP3 inhibition is a promising cardioprotective strategy in Dox/Tra combination therapy.


Asunto(s)
Cardiotoxicidad , Inflamasomas , Ratas , Ratones , Humanos , Animales , Inflamasomas/metabolismo , Cardiotoxicidad/etiología , Cardiotoxicidad/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Trastuzumab , Especies Reactivas de Oxígeno/metabolismo , Doxorrubicina/toxicidad , Doxorrubicina/metabolismo , Miocitos Cardíacos/metabolismo , Apoptosis , Estrés Oxidativo
5.
Front Pharmacol ; 14: 1292088, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38143497

RESUMEN

Doxorubicin is a widely used anticancer drug in clinical practice for the treatment of various human tumors. However, its administration is associated with cardiotoxicity. Administration of doxorubicin with low side effects for cancer treatment and prevention are, accordingly, urgently required. The human body harbors various endogenous metal ions that exert substantial influences. Consequently, extensive research has been conducted over several decades to investigate the potential of targeting endogenous metal ions to mitigate doxorubicin's side effects and impede tumor progression. In recent years, there has been a growing body of research indicating the potential efficacy of metal ion-associated therapeutic strategies in inhibiting doxorubicin-induced cardiotoxicity (DIC). These strategies offer a combination of favorable safety profiles and potential clinical utility. Alterations in intracellular levels of metal ions have been found to either facilitate or mitigate the development of DIC. For instance, ferroptosis, a cellular death mechanism, and metal ions such as copper, zinc, and calcium have been identified as significant contributors to DIC. This understanding can contribute to advancements in cancer treatment and provide valuable insights for mitigating the cardiotoxic effects of other therapeutic drugs. Furthermore, potential therapeutic strategies have been investigated to alleviate DIC in clinical settings. The ultimate goal is to improve the efficacy and safety of Dox and offer valuable insights for future research in this field.

6.
Drug Des Devel Ther ; 16: 1621-1645, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35669282

RESUMEN

Background: Huo Luo Xiao Ling Dan (HLXLD), a famous Traditional Chinese Medicine (TCM) classical formula, possesses anti-atherosclerosis (AS) activity. However, the underlying molecular mechanisms remain obscure. Aim: The network pharmacology approach, molecular docking strategy, and in vitro validation experiment were performed to explore the potential active compounds, key targets, main signaling pathways, and underlying molecular mechanisms of HLXLD in treating AS. Methods: Several public databases were used to search for active components and targets of HLXLD, as well as AS-related targets. Crucial bioactive ingredients, potential targets, and signaling pathways were acquired through bioinformatics analysis. Subsequently, the molecular docking strategy and molecular dynamics simulation were carried out to predict the affinity and stability of active compounds and key targets. In vitro cell experiment was performed to verify the findings from bioinformatics analysis. Results: A total of 108 candidate compounds and 321 predicted target genes were screened. Bioinformatics analysis suggested that quercetin, dihydrotanshinone I, pelargonidin, luteolin, guggulsterone, and ß-sitosterol may be the main ingredients. STAT3, HSP90AA1, TP53, and AKT1 could be the key targets. MAPK signaling pathway might play an important role in HLXLD against AS. Molecular docking and molecular dynamics simulation results suggested that the active compounds bound well and stably to their targets. Cell experiments showed that the intracellular accumulation of lipid and increased secretory of TNF-α, IL-1ß, and MCP-1 in ox-LDL treated RAW264.7 cells, which can be significantly suppressed by pretreating with dihydrotanshinone I. The up-regulation of STAT3, ERK, JNK, and p38 phosphorylation induced by ox-LDL can be inhibited by pretreating with dihydrotanshinone I. Conclusion: Our findings comprehensively demonstrated the active compounds, key targets, main signaling pathways, and underlying molecular mechanisms of HLXLD in treating AS. These findings would provide a scientific basis for the study of the complex mechanisms underlying disease and drug action.


Asunto(s)
Aterosclerosis , Medicamentos Herbarios Chinos , Aterosclerosis/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Furanos , Humanos , Medicina Tradicional China , Simulación del Acoplamiento Molecular , Farmacología en Red , Fenantrenos , Quinonas
7.
Comput Biol Med ; 151(Pt A): 106298, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36403355

RESUMEN

OBJECTIVES: Recently, it has been reported that cepharanthine (CEP) is highly likely to be an agent against Coronavirus disease 2019 (COVID-19). In the present study, a network pharmacology-based approach combined with RNA-sequencing (RNA-seq), molecular docking, and molecular dynamics (MD) simulation was performed to determine hub targets and potential pharmacological mechanism of CEP against COVID-19. METHODS: Targets of CEP were retrieved from public databases. COVID-19-related targets were acquired from databases and RNA-seq datasets GSE157103 and GSE155249. The potential targets of CEP and COVID-19 were then validated by GSE158050. Hub targets and signaling pathways were acquired through bioinformatics analysis, including protein-protein interaction (PPI) network analysis and enrichment analysis. Subsequently, molecular docking was carried out to predict the combination of CEP with hub targets. Lastly, MD simulation was conducted to further verify the findings. RESULTS: A total of 700 proteins were identified as CEP-COVID-19-related targets. After the validation by GSE158050, 97 validated targets were retained. Enrichment results indicated that CEP acts on COVID-19 through multiple pathways, multiple targets, and overall cooperation. Specifically, PI3K-Akt signaling pathway is the most important pathway. Based on PPI network analysis, 9 central hub genes were obtained (ACE2, STAT1, SRC, PIK3R1, HIF1A, ESR1, ERBB2, CDC42, and BCL2L1). Molecular docking suggested that the combination between CEP and 9 central hub genes is extremely strong. Noteworthy, ACE2, considered the most important gene in CEP against COVID-19, binds to CEP most stably, which was further validated by MD simulation. CONCLUSION: Our study comprehensively illustrated the potential targets and underlying molecular mechanism of CEP against COVID-19, which further provided the theoretical basis for exploring the potential protective mechanism of CEP against COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Simulación de Dinámica Molecular , Humanos , Simulación del Acoplamiento Molecular , Enzima Convertidora de Angiotensina 2 , Farmacología en Red , Fosfatidilinositol 3-Quinasas , ARN
8.
Eur J Pharmacol ; 891: 173690, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33127362

RESUMEN

Emerging evidence suggests the cardiovascular protective effects of Xanthohumol (Xn), a prenylated flavonoid isolated from the hops (Humulus lupulus L.). However, the cardioprotective effect of Xn remains unclear. Present study aimed to investigate the protective role of Xn against isoprenaline (ISO)-induced cardiac hypertrophy and fibrosis, and elucidate the underlying mechanism. The cardiac hypertrophy and fibrosis model were established via subcutaneously administration of ISO. ISO reduced the left ventricular contractile function and elevated myocardial enzyme levels, suggesting cardiac dysfunction. Moreover, the increased cardiac myocyte area, heart weight/body weight (HW/BW) ratio and ANP/BNP expressions indicated the ISO-induced hypertrophy, while the excessive collagen-deposition and up-regulation of fibrosis marker protein (α-SMA, Collagen-I/III) expression indicated the ISO-induced fibrosis. The ISO-induced cardiac dysfunction, hypertrophy and fibrosis were significantly attenuated by oral administrated with Xn. PTEN/AKT/mTOR pathway has been reported to involve in pathogenesis of cardiac hypertrophy and fibrosis. We found that Xn administration up-regulated PTEN expression and inhibited the phosphorylation of AKT/mTOR in ISO-treated mice. Moreover, treating with VO-ohpic, a specific PTEN inhibitor, abolished the cardioprotective effect of Xn. Collectively, these results suggested that Xn attenuated ISO-induced cardiac hypertrophy and fibrosis through regulating PTEN/AKT/mTOR pathway.


Asunto(s)
Flavonoides/farmacología , Hipertrofia Ventricular Izquierda/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Fosfohidrolasa PTEN/metabolismo , Propiofenonas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Fibrosis , Hipertrofia Ventricular Izquierda/inducido químicamente , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/fisiopatología , Isoproterenol , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/fisiopatología
9.
J Cancer ; 12(21): 6576-6587, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659548

RESUMEN

Aims: Bai-He-Gu-Jin-Tang (BHGJT) is a classic Chinese formula used to treat lung cancer, while the underlying molecular mechanism remains obscure. The aim of the study was to investigate the molecular mechanism of BHGJT on lung cancer and demonstrate the potential for synergistic treatment combining BHGJT with conventional therapy. Methods: Cell viability assay, colony formation assay and EdU assay were used to determine the in vitro effects of BHGJT, and a subcutaneous xenograft model was used to evaluate the in vivo effect. Cell cycle analysis, apoptosis rate analysis, immunohistochemical and immunofluorescent staining, Western blot assays and network pharmacology-based analysis were used to explore the underlying mechanisms. Results: We found that BHGJT inhibited cell proliferation via a dose-dependent pathway and obviously hindered tumor growth in vivo in lung cancer. Cell cycle arrest and apoptosis were pronouncedly induced by BHGJT via dysregulation of the cell cycle regulators CDK4 and Cyclin D1 and dysregulation of apoptosis-associated proteins, such as cleaved caspase 3/9 and the BCL-2 family. Based on a network pharmacology-based analysis and experimental evidence, we demonstrated that the AKT/GSK3ß/ß-catenin signaling pathways were responsible for BHGJT-induced apoptosis in lung cancer cells. Additionally, autophagy was induced by BHGJT via the AMPK/mTORC1/ULK1 signaling pathway, and blocking autophagy with either chloroquine or a ULK1 inhibitor increased the killing efficiency of BHGJT in lung cancer cells. Conclusion: Our findings indicate that the BHGJT formula efficiently inhibits lung cancer growth and represents a potential complementary and alternative treatment for lung cancer.

10.
Biomed Res Int ; 2021: 6688855, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33860051

RESUMEN

Polygonatum sibiricum polysaccharides (PSP), the extract of Polygonatum sibiricum, are demonstrated to exhibit a wide range of pharmacological activities. A recent study reported that PSP alleviated the aging of the kidney and meninges. However, the effect of PSP on heart aging remains unclear. The present study is aimed at investigating the protection of PSP on D-galactose- (D-gal-) induced heart aging. Results showed that irregularly arranged cardiac muscle fibers were observed in heart tissues of D-gal-treated mice, and the levels of cardiac troponin T (cTnT), creatine kinase (CK), p21, and p53 were increased after D-gal treatment. D-gal-induced heart aging and injury can be attenuated by oral administration of PSP. Moreover, PSP also decreased reactive oxygen species (ROS) and malondialdehyde (MDA) and increased the level of superoxide dismutase (SOD) in the hearts of D-gal-treated mice. DNA damages and lipid peroxidation induced by oxidative stress were also inhibited by PSP as indicated by reduced levels of 8-hydroxydeoxyguanosine (8-OHdG) and 4-hydroxy-2-nonenal (4-HNE). Collectively, PSP attenuated D-gal-induced heart aging via inhibiting oxidative stress, suggesting that PSP might serve as a potential effective Chinese herbal active constituent for antiaging therapy.


Asunto(s)
Envejecimiento/patología , Antioxidantes/farmacología , Miocardio/patología , Polygonatum/química , Polisacáridos/farmacología , Envejecimiento/efectos de los fármacos , Animales , Cardiotónicos/farmacología , Daño del ADN , Galactosa , Peroxidación de Lípido/efectos de los fármacos , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos
11.
Drug Des Devel Ther ; 15: 3255-3276, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34349502

RESUMEN

BACKGROUND: Huai Hua San (HHS), a famous Traditional Chinese Medicine (TCM) formula, has been widely applied in treating ulcerative colitis (UC). However, the interaction of bioactives from HHS with the targets involved in UC has not been elucidated yet. AIM: A network pharmacology-based approach combined with molecular docking and in vitro validation was performed to determine the bioactives, key targets, and potential pharmacological mechanism of HHS against UC. MATERIALS AND METHODS: Bioactives and potential targets of HHS, as well as UC-related targets, were retrieved from public databases. Crucial bioactive ingredients, potential targets, and signaling pathways were acquired through bioinformatics analysis, including protein-protein interaction (PPI), as well as the Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Subsequently, molecular docking was carried out to predict the combination of active compounds with core targets. Lastly, in vitro experiments were conducted to further verify the findings. RESULTS: A total of 28 bioactive ingredients of HHS and 421 HHS-UC-related targets were screened. Bioinformatics analysis revealed that quercetin, luteolin, and nobiletin may be potential candidate agents. JUN, TP53, and ESR1 could become potential therapeutic targets. PI3K-AKT signaling pathway might play an important role in HHS against UC. Moreover, molecular docking suggested that quercetin, luteolin, and nobiletin combined well with JUN, TP53, and ESR1, respectively. Cell experiments showed that the most important ingredient of HHS, quercetin, could inhibit the levels of inflammatory factors and phosphorylated c-Jun, as well as PI3K-Akt signaling pathway in LPS-induced RAW264.7 cells, which further confirmed the prediction by network pharmacology strategy and molecular docking. CONCLUSION: Our results comprehensively illustrated the bioactives, potential targets, and molecular mechanism of HHS against UC. It also provided a promising strategy to uncover the scientific basis and therapeutic mechanism of TCM formulae in treating diseases.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Medicina Tradicional China , Simulación del Acoplamiento Molecular , Farmacología en Red , Animales , Ratones , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Quercetina/farmacología , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
12.
Biomed Res Int ; 2021: 9806412, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34195284

RESUMEN

[This corrects the article DOI: 10.1155/2021/6688855.].

13.
Drug Des Devel Ther ; 14: 5431-5439, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324040

RESUMEN

BACKGROUND: Xanthohumol (Xn) is the most abundant prenylated flavonoid in Hops (Humulus lupulus L.), and exhibits a range of pharmacological activities. This study aimed to investigate the effect of Xn on TGF-ß1-induced cardiac fibroblasts activation and elucidate the underlying mechanism. MATERIALS AND METHODS: The cellTiter 96® AQueous one solution cell proliferation assay kit was adopted to determine the cell viability of cardiac fibroblasts, and the proliferation was detected through 5-ethynyl-2'-deoxyuridine (EdU) incorporation assay. The α-SMA protein expression was measured by using immunofluorescence and Western blotting. Western blotting was conducted to test the protein expressions of collagen I and III, PTEN, p-Akt, Akt, p-mTOR, mTOR, p-Smad3, Smad3 and GAPDH. The mRNA levels of α-SMA, collagen I and III were determined by quantitative real-time polymerase chain reaction (PCR). RESULTS: Xn inhibited the TGF-ß1-induced proliferation, differentiation and collagen overproduction of cardiac fibroblasts. TGF-ß1 induced the down-regulated PTEN expression, Akt and mTOR phosphorylation. These effects of TGF-ß1 were suppressed by Xn, while blocking of PTEN reduced Xn-mediated inhibitory effect on cardiac fibroblasts activation induced by TGF-ß1. CONCLUSION: Xn inhibits TGF-ß1-induced cardiac fibroblasts activation via mediating PTEN/Akt/mTOR signaling pathway.


Asunto(s)
Antineoplásicos/farmacología , Flavonoides/farmacología , Miofibroblastos/efectos de los fármacos , Fosfohidrolasa PTEN/antagonistas & inhibidores , Propiofenonas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Antineoplásicos/química , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Flavonoides/química , Humulus/química , Masculino , Estructura Molecular , Miofibroblastos/metabolismo , Fosfohidrolasa PTEN/metabolismo , Propiofenonas/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/farmacología
14.
Cardiovasc Toxicol ; 20(5): 507-519, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32607760

RESUMEN

Doxorubicin (Dox) is widely used in cancer therapy, but the clinical application is limited by its cardiotoxicity. The underlying mechanism of Dox-induced cardiotoxicity remains unclear. Present study aimed to evaluate the role of NLRP3 inflammasome in Dox-induced cardiotoxicity. The NLRP3 inflammasome was activated in the myocardium of Dox-treating (5 mg/kg, once every other day, cumulative dosage to 15 mg/kg and sacrificed after 2 days of last Dox injection) C57BL/6 mice as shown by the up-regulation of NLRP3 and Caspase-1 p20. Dox (1 µM for 48 h) induced the apoptosis of H9c2 cells and primary cardiomyocytes concomitantly with up-regulation of NLRP3, ASC and Caspase-1 p20 expressions, as well as the increased IL-1ß secretion, suggesting the activation of NLRP3 inflammasome. These effects of Dox on H9c2 cells and primary cardiomyocytes can be reversed by MCC950, a specific inhibitor of NLRP3. In view of the key role of ROS on the Dox-induced cardiotoxicity, the relationship between ROS and NLRP3 was further investigated. The ROS level was increased in myocardium, H9c2 cells and primary cardiomyocytes after treating with Dox. Decreasing ROS level by NAC can inhibit the NLRP3 inflammasome activation, secretion of IL-1ß and apoptosis in Dox-treating H9c2 cells and primary cardiomyocytes. Collectively, this study reveals a crucial role of ROS/NLRP3-associated inflammasome activation in Dox-induced cardiotoxicity, and NLRP3 inflammasome may represent a new therapeutic target for Dox-induced cardiotoxicity.


Asunto(s)
Doxorrubicina , Cardiopatías/metabolismo , Inflamasomas/metabolismo , Miocitos Cardíacos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis , Cardiotoxicidad , Línea Celular , Modelos Animales de Enfermedad , Cardiopatías/inducido químicamente , Cardiopatías/patología , Interleucina-1beta/metabolismo , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley , Transducción de Señal
15.
Anal Chim Acta ; 1087: 76-85, 2019 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-31585569

RESUMEN

Aluminum is a very important analyte, and developing biosensors for aluminum is an important analytical task. In this work, we report a novel mechanism to design colorimetric sensor based on gold nanoparticles (AuNPs). The AuNPs were prepared by reducing HAuCl4 using catechols, and the resulting AuNPs can be directly adapted for Al3+ detection without any post-modifications, showing high sensitivity and selectivity against other metal ions. Interestingly, our mechanistic studies revealed that Al3+-induced AuNPs aggregation was not due to the formation of interparticle crosslinks that refers to the design principle of most AuNPs-based colorimetric sensors reported before. But rather, Al3+ competitively coordinated with the capping ligands on AuNPs surface through the formation of stable Al-O bond, which dissociated these ligands from AuNPs surface. As a result, the AuNPs aggregated due to the loss of surface stabilizers. Based on this mechanism, several catechols, including pyrocatechol (PC), 3-(3,4-dihydroxyphenyl) propionic acid (DHCA), levodopa (LDA) and dopamine (DA), were used as reductant to prepare AuNPs for Al3+ sensing, and the AuNPs prepared by DA (AuNPs/DA) displayed the highest sensitivity, with detection limit of 0.81 µM. The sensor was then tested for Al content analysis in river water and food samples, and the results supported its practical applications. Importantly, this work expands the design principles for colorimetric sensors by using AuNPs.


Asunto(s)
Aluminio/análisis , Contaminación de Alimentos/análisis , Oro/química , Nanopartículas del Metal/química , Contaminantes Químicos del Agua/análisis , Técnicas Biosensibles/métodos , Pan/análisis , Catecoles/química , Quelantes/química , Colorimetría/métodos , Ligandos , Límite de Detección , Ríos/química
16.
Int J Nanomedicine ; 14: 8345-8360, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31695371

RESUMEN

BACKGROUND: The protective role of puerarin (PUE) against myocardial infarction is closely related to its regulation on mitochondria. However, free PUE can hardly reach the mitochondria of ischemic cardiomyocytes due to the lack of mitochondrial targeting of PUE. Here PUE was loaded into mitochondria-targeted micelles (PUE@TPP/PEG-PE) for precisely delivering PUE into mitochondria with the aim of enhancing the anti-apoptosis effect. METHODS: The mitochondriotropic polymer TPP-PEG-PE was synthesized for the preparation of PUE@TPP/PEG-PE micelles modified with triphenylphosphonium (TPP) cation. The physicochemical properties and anti-apoptosis effect of PUE@TPP/PEG-PE micelles were investigated. The coumarin 6 (C6)-labeled TPP/PEG-PE (C6@TPP/PEG-PE) micelles were used to observe the enhanced cellular uptake, mitochondrial targeting and lysosomes escape. Moreover, in vivo and ex vivo biodistribution of lipophilic near-infrared dye 1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide (DiR)-labeled PUE@TPP/PEG-PE (DiR@TPP/PEG-PE) micelles were detected through fluorescence imaging. RESULTS: The successful synthesis of TPP-PEG-PE conjugate was confirmed. PUE@TPP/PEG-PE micelles had a particle size of 17.1 nm, a zeta potential of -6.2 mV, and a sustained-release behavior. The in vitro results showed that the intracellular uptake of C6@TPP/PEG-PE micelles was significantly enhanced in H9c2 cells. C6@TPP/PEG-PE micelles could deliver C6 to mitochondria and reduce the capture of lysosomes. In addition, compared with the PUE@PEG-PE micelles and free PUE, the PUE@TPP/PEG-PE micelles exerted an enhanced protective effect against isoprenaline-induced H9c2 cell apoptosis, as evident by the decreased percentage of apoptotic cells, Caspase-3 activity, ROS level, Bax expression, and increased Bcl-2 expression. The in vivo detecting results of the targeting effect using DiR probe also indicated that TPP/PEG-PE micelles could accumulate and retain in the ischemic myocardium. CONCLUSION: The results of this study demonstrate the promising potential of applying PUE@TPP/PEG-PE micelles in mitochondria-targeted drug delivery to achieve maximum therapeutic effects of PUE.


Asunto(s)
Apoptosis/efectos de los fármacos , Cardiotónicos/farmacología , Isoflavonas/farmacología , Micelas , Mitocondrias/metabolismo , Miocitos Cardíacos/patología , Fosfinas/química , Animales , Cationes , Línea Celular , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Endocitosis/efectos de los fármacos , Femenino , Humanos , Isoproterenol , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Nanopartículas/química , Nanopartículas/ultraestructura , Tamaño de la Partícula , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Ratas , Electricidad Estática , Distribución Tisular/efectos de los fármacos
17.
Eur J Pharmacol ; 843: 96-103, 2019 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-30352200

RESUMEN

It has been documented cardiac fibroblasts as the predominant cell population undergoing senescence in heart. Calcitonin gene-related peptide (CGRP) exhibits a wide range of cardiovascular protective effects. Whether CGRP protects against cardiac fibroblasts senescence in cardiac fibrosis remains unknown. Here, we detected the down-regulation of CGRP concomitant with senescence in fibrotic myocardium, both hypertension- induced left ventricular fibrosis in SHR rats and hypoxia-induced right ventricular fibrosis in pulmonary artery hypertension rats. Exogenous CGRP inhibited the cardiac fibroblasts senescence and senescence-associated secretory phenotype (SASP) induced by TGF-ß1, which was abolished by CGRP8-37, a selective CGRP receptor antagonist. Moreover, the expression of klotho, an anti-senescence protein, was down-regulated in fibrotic myocardium, and CGRP up-regulated the klotho expression in TGF-ß1-treated cardiac fibroblasts. Klotho knockdown by siRNA reversed the inhibition of CGRP on senescence and SASP induced by TGF-ß1 in cardiac fibroblasts. These results suggested that CGRP inhibited the cardiac fibroblasts senescence and SASP in cardiac fibrosis via up-regulating klotho expression.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/farmacología , Senescencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Glucuronidasa/metabolismo , Animales , Animales Recién Nacidos , Fibroblastos/fisiología , Fibrosis , Glucuronidasa/genética , Proteínas Klotho , Masculino , Miocardio/citología , Miocardio/metabolismo , Miocardio/patología , ARN Interferente Pequeño , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba
18.
Oncol Rep ; 40(6): 3213-3222, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30272303

RESUMEN

Xanthohumol (Xn), a prenylated flavonoid isolated from Hops (Humulus lupulus L.), has demonstrated potent anticancer activity in multiple types of cancer. However, the effect of Xn on gastric cancer (GC) remains unknown. The aim of the present study was to investigate the effect of Xn on GC cell proliferation, apoptosis and metastasis. It was observed that Xn decreased the viability of GC cells, with very low or no toxicity to normal gastric epithelial cells GES­1 at a concentration of 1­100 µM. The proliferation of AGS cells was inhibited by Xn, as indicated by the decreased number of EdU­positive cells. Xn treatment increased the number of apoptotic cells, downregulated the expression of Bcl­2 and upregulated the expression of Bax, suggesting induction of apoptosis. The results from the wound healing and Transwell assays indicated that Xn suppressed AGS cell metastasis. Moreover, Xn induced reactive oxygen species (ROS) overproduction and inhibited nuclear factor (NF)­κB signaling in AGS cells, which was reversed by the ROS inhibitor N­acetylcysteine (NAC). NAC suppressed the effect of Xn on the proliferation, apoptosis and metastasis of AGS cells. Taken together, these results suggest that Xn exerts anticancer effects against GC via induction of ROS production and subsequent inhibition of NF­κB signaling. Therefore, Xn may be a promising candidate treatment against GC progression.


Asunto(s)
Antineoplásicos/farmacología , Flavonoides/farmacología , Propiofenonas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Neoplasias Gástricas/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/tratamiento farmacológico
19.
Front Pharmacol ; 9: 530, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29872398

RESUMEN

It has been observed that many phytochemicals, frequently present in foods or beverages, show potent chemopreventive or therapeutic properties that selectively affect cancer cells. Numerous studies have demonstrated the anticancer activity of xanthohumol (Xn), a prenylated flavonoid isolated from hops (Humulus lupulus L.), with a concentration up to 0.96 mg/L in beer. This review aims to summarize the existing studies focusing on the anticancer activity of Xn and its effects on key signaling molecules. Furthermore, the limitations of current studies and challenges for the clinical use of Xn are discussed.

20.
Food Chem Toxicol ; 93: 102-10, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27170490

RESUMEN

Chronic arsenic exposure in drinking water is associated with the abnormalities of cardiac tissue. Excessive generation of ROS induced by arsenic has a central role in arsenic-induced cardiotoxicity. (-)-Epigallocatechin-3-gallate (EGCG), the most abundant polyphenol in green tea, possesses a potent antioxidant capacity and exhibits extensive pharmacological activities. This study was aim to evaluate the effect of EGCG on arsenic-induced cardiotoxicity in vivo and in vitro. Treatment with NaAsO2 seriously affected the morphology and ultrastructure of myocardium, and induced cardiac injuries, oxidative stress, intracellular calcium accumulation and apoptosis in rats. In consistent with in vivo study, the injuries, oxidative stress and apoptosis were also observed in NaAsO2-treated H9c2 cells. All of these effects induced by NaAsO2 were attenuated by EGCG. These results suggest EGCG could attenuate NaAsO2-induced cardiotoxicity, and the mechanism may involve its potent antioxidant capacity.


Asunto(s)
Antioxidantes/farmacología , Arsénico/toxicidad , Cardiotoxicidad/prevención & control , Catequina/análogos & derivados , Corazón/efectos de los fármacos , Miocardio/patología , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Cardiotoxicidad/etiología , Catequina/farmacología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA