Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Theranostics ; 12(16): 7032-7050, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36276638

RESUMO

Rationale: Glioblastoma (GBM) displays a complex metabolic reprogramming in cancer cells. Adenosine triphosphate (ATP) is one of the central mediators of cell metabolism and signaling. GBM cells generate ATP by glycolysis and the tricarboxylic acid (TCA) cycle associated with oxidative phosphorylation (OXPHOS) through the breaking-down of pyruvate or fatty acids to meet the growing energy demand of cancer cells. Therefore, it's urgent to develop novel treatments targeting energy metabolism to hinder tumor cell proliferation in GBM. Methods: Non-targeted metabolomic profiling analysis was utilized to evaluate cell metabolic reprogramming using a small molecule inhibitor (SMI) EPIC-0412 treatment. Cellular oxygen consumption rate (OCR) and the total proton efflux rate (PER), as well as ATP concentration, were tracked to study metabolic responses to specifically targeted inhibitors, including EPIC-0412, arachidonyl trifluoromethyl ketone (AACOCF3), and 2 deoxy-D-glucose (2-DG). Cancer cell proliferation was assessed by CCK-8 measurements and colony formation assay. Additionally, flow cytometry, immunoblotting (IB), and immunofluorescence (IF) analyses were performed with GBM cells to understand their tumorigenic properties under treatments. Finally, the anticancer effects of this combination therapy were evaluated in the GBM mouse model by convection-enhanced delivery (CED). Results: We found that SMI EPIC-0412 could effectively perturb the TCA cycle, which participated in the combination therapy of cytosolic phospholipase A2 (cPLA2)-inhibitor AACOCF3, and hexokinase II (HK2)-inhibitor 2-DG to disrupt the GBM energy metabolism for targeted metabolic treatments. ATP production was significantly declined in glioma cells when treated with monotherapy (EPIC-0412 or AACOCF3), dual therapy (EPIC-0412 + AACOCF3), or triple therapy (EPIC-0412 + AACOCF3 +2-DG) regimen. Our experiments revealed that these therapies hindered glioma cell proliferation and growth, leading to the reduction in ATP production and G0/G1 cell cycle arrest. We demonstrated that the combination therapy effectively extended the survival of cerebral tumor-bearing mice. Conclusion: Our findings indicate that the TCA-phospholipid-glycolysis metabolism axis can be blocked by specific inhibitors that significantly disrupt the tumor energy metabolism and suppress tumor proliferation in vitro and in vivo, suggesting that targeting ATP synthesis inhibition in cancer cells might be an attractive therapeutic avenue in GBM management.


Assuntos
Glioblastoma , Glioma , Fosfolipídeos , Animais , Camundongos , Trifosfato de Adenosina/metabolismo , Ácidos Graxos , Glioblastoma/metabolismo , Glucose/metabolismo , Glicólise/fisiologia , Hexoquinase/antagonistas & inibidores , Fosfolipases A2/metabolismo , Fosfolipases A2 Citosólicas/metabolismo , Fosfolipídeos/metabolismo , Prótons , Piruvatos/metabolismo , Ácidos Tricarboxílicos/uso terapêutico
2.
Hum Exp Toxicol ; 41: 9603271221132140, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36198566

RESUMO

Herbacetin (HBN) is a glycosylated flavonoid, which possesses numerous pharmacological properties. Cyclophosphamide (CYC) is a chemotherapeutic drug that adversely affects the kidneys. The present investigation aimed to evaluate the curative potential of HBN against CYC-induced nephrotoxicity. Sprague Dawley rats (n = 48) were randomly divided into four groups: control (0.1% DMSO + food), CYC (150 mg/kg b.wt.), CYC+HBN (150 + 40 mg/kg b.wt.), and HBN (40mg/kg b.wt.). CYC treatment significantly decreased the activities of antioxidant enzymes such as catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GSR) while elevating the concentration of reactive oxygen species (ROS) and malondialdehyde (MDA). Treatment with HBN significantly recovered the activity of CAT, SOD, GPx, and GSR while reducing the concentrations of ROS and MDA. Moreover, an increase in the level of renal functional markers, including Urea, creatinine, kidney injury molecule-1 (KIM-1), and neutrophil gelatinase-associated lipocalin (NGAL), and a decrease in creatinine clearance after CYC administration was recovered to control values by HBN treatment. Furthermore, HBN treatment normalized the increased levels of inflammatory markers such as nuclear factor kappa-B (NF-κB), tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) after CYC administration. Besides, HBN administration increased the expression of anti-apoptotic markers (Bcl-2) while decreasing the apoptotic markers (Bax and Caspase-3). Furthermore, HBN decreased the activities of tricarboxylic acid (TCA) cycle enzymes (ICDH, αKGDH, SDH, and MDH) as well as renal mitochondrial respiratory-chain complexes (I-IV) and repolarized mitochondrial membrane potential (ΔΨm). Additionally, HBN administration significantly protected against renal histological damage induced by CYC. In conclusion, CYC-induced toxicity was effectively ameliorated by the HBN administration. These results indicate that HBN might be considered as a potential protective agent against nephrotoxicity. The observed protection may be due to its antioxidant, anti-inflammatory, and anti-apoptotic potential.


Assuntos
NF-kappa B , Fator de Necrose Tumoral alfa , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Apoptose , Caspase 3/metabolismo , Catalase/metabolismo , Creatinina/metabolismo , Ciclo-Oxigenase 2/metabolismo , Ciclofosfamida/uso terapêutico , Ciclofosfamida/toxicidade , Dimetil Sulfóxido/metabolismo , Dimetil Sulfóxido/farmacologia , Dimetil Sulfóxido/uso terapêutico , Flavonoides/farmacologia , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Rim , Lipocalina-2 , Malondialdeído/metabolismo , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Ácidos Tricarboxílicos/metabolismo , Ácidos Tricarboxílicos/farmacologia , Ácidos Tricarboxílicos/uso terapêutico , Fator de Necrose Tumoral alfa/metabolismo , Ureia , Proteína X Associada a bcl-2/metabolismo
3.
J Ethnopharmacol ; 295: 115381, 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35595220

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Marsdenia tenacissima injection (MTE), a traditional Chinese medical injection extracted from the rattan of Marsdenia tenacissima (Roxb.) Moon, has been approved for clinical use in China as an adjuvant therapeutic agent in multiple cancers, including esophageal cancer, gastric cancer, lung cancer, and liver cancer. However, the activity and mechanism of MTE on prostate cancer (PCa) remain to be defined. AIM OF THE STUDY: To investigate the activity and the underlying mechanism of MTE in the treatment of PCa. MATERIALS AND METHODS: The component characterization of MTE was analyzed by HPLC-CAD-QTOF-MS/MS technology. Cell Counting Kit-8 (CCK-8) assay was used to assess PCa cell proliferation. Colony formation assay was applied to detect the clonogenic ability of the cells. MetaboAnalyst5.0 database was employed to analyze the altered metabolites of PC3 cells treated with MTE obtained by UPLC-QTOF-MS/MS. Combined with metabolomics analysis and network pharmacology, we predicted the potential targets, which further were verified by Western Blot, RT-qPCR, and Immunohistochemistry assays. Finally, SeeSAR software was applied to predict the potential active components of MTE against PCa. RESULTS: A total of 21 components in MTE were confirmed by HPLC-CAD-QTOF-MS/MS analysis. MTE inhibited the proliferation and colony formation of PCa cells. A total of 20 metabolites closely related to glycerophospholipid metabolism, glycolysis/gluconeogenesis, and tricarboxylic acid (TCA) cycle were significantly changed in PC3 cells treated with MTE. The network pharmacology analysis revealed that MTE suppressed the growth of PC3 cells might by regulating the ErbB2-GSK3ß-HIF1α signaling axis. Furthermore, we also confirmed that stimulation of MTE significantly inhibited the phosphorylation of ErbB2 at Tyr877 and the activities of its downstream signal transducers (GSK3ß and HIF1α) in PCa, as well as the mRNA levels of critical factors (IDH2, LDHA, and HIF1A) in the tricarboxylic acid (TCA) cycle. Molecular docking further suggested that Tenacissimoside E, cryptochlorogenic acid, and scopoletin might be the active ingredients of MTE for PCa treatment. CONCLUSION: This study proposed that MTE exerts a potential anti-tumor effect in PCa through inhibiting ErbB2-GSK3ß-HIF1α signaling axis, which may be related to the TCA cycle.


Assuntos
Neoplasias Pulmonares , Marsdenia , Neoplasias da Próstata , Glicogênio Sintase Quinase 3 beta , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Marsdenia/química , Simulação de Acoplamento Molecular , Extratos Vegetais/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Receptor ErbB-2 , Espectrometria de Massas em Tandem , Ácidos Tricarboxílicos/uso terapêutico
4.
Int J Pharm ; 587: 119667, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32702448

RESUMO

Curcumin (CUR) is a Biopharmaceutics Classification System (BCS) class IV drug with poor aqueous solubility and low permeability. The dissolution of CUR can be enhanced through the cocrystallization approach. In this work, we report a new cocrystal phase of CUR with trimesic acid (TMA) with the enhanced dissolution of CUR. Cytotoxicity and cell invasion assays were conducted on (2D) monolayers and three-dimensional (3D) tumor models of triple-negative breast cancer (TNBC) cells, MDA-MB-231 using the new CUR-TMA cocrystal phase along with different CUR solid forms prepared in our previous works. The cytotoxicity and internalization assays conducted on 2D monolayers indicated that all CUR multicomponent solid forms except Curcumin-Folic Acid Dihydrate (CUR-FAD) (1:1) coamorphous solid exhibited enhanced bioavailability than unprocessed CUR. Cell invasion assay conducted on 3D tumor spheroid models showed that Curcumin-Hydroxyquinol (CUR-HXQ) cocrystal completely inhibited cell invasion whereas CUR-FAD (1:1) coamorphous solid induced enhanced invasion of cells from spheroid models.


Assuntos
Antineoplásicos , Curcumina , Preparações Farmacêuticas , Neoplasias de Mama Triplo Negativas , Antineoplásicos/uso terapêutico , Humanos , Solubilidade , Ácidos Tricarboxílicos/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
5.
Cancer Immunol Immunother ; 65(11): 1303-1315, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27520505

RESUMO

Tumor-derived metabolites dampen tumor-infiltrating immune cells and antitumor immune responses. Among the various metabolites produced by tumors, we recently showed that cholesterol oxidized products, namely oxysterols, favor tumor growth through the inhibition of DC migration toward lymphoid organs and by promoting the recruitment of pro-tumor neutrophils within the tumor microenvironment. Here, we tested different drugs capable of blocking cholesterol/oxysterol formation. In particular, we tested efficacy and safety of different administration schedules, and of immunotherapy-based combination of a class of compounds, namely zaragozic acids, which inhibit cholesterol pathway downstream of mevalonate formation, thus leaving intact the formation of the isoprenoids, which are required for the maturation of proteins involved in the immune cell function. We show that zaragozic acids inhibit the in vivo growth of the RMA lymphoma and the Lewis lung carcinoma (LLC) without inducing side effects. Tumor growth inhibition requires an intact immune system, as immunodeficient tumor-bearing mice do not respond to zaragozic acid treatment. Of note, the effect of zaragozic acids is accompanied by a marked reduction in the LXR target genes Abcg1, Mertk, Scd1 and Srebp-1c in the tumor microenvironment. On the other hand, zoledronate, which blocks also isoprenoid formation, did not control the LLC tumor growth. Finally, we show that zaragozic acids potentiate the antitumor effects of active and adoptive immunotherapy, significantly prolonging the overall survival of tumor-bearing mice treated with the combo zaragozic acids and TAA-loaded DCs. This study identifies zaragozic acids as new antitumor compounds exploitable for the treatment of cancer patients.


Assuntos
Antineoplásicos/uso terapêutico , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Carcinoma Pulmonar de Lewis/terapia , Células Dendríticas/imunologia , Imunoterapia Adotiva/métodos , Linfoma de Células T/terapia , Ácidos Tricarboxílicos/uso terapêutico , Animais , Carcinoma Pulmonar de Lewis/imunologia , Colesterol/metabolismo , Terapia Combinada , Células Dendríticas/transplante , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Linfoma de Células T/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Oxisteróis/metabolismo , Evasão Tumoral , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA