Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Free Radic Biol Med ; 212: 493-504, 2024 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-38184120

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) has extremely poor prognosis, with a 5-year survival rate of approximately 11 %. Yes-associated protein (YAP) is a major downstream effector of the Hippo-YAP pathway and plays a pivotal role in regulation of cell proliferation and organ regeneration and tumorigenesis. Activation of YAP signaling has been associated with PDAC progression and drug resistance. Verteporfin (VP) is a photosensitizer used for photodynamic therapy and previous work showed that it can function as a YAP inhibitor. The efficacy of VP on human cancer are being tested in several trials. In this study, we examined the effect of VP on reactive oxygen species (ROS) and lipid peroxidation in pancreatic cancer cells, by using fluorescent molecular probes and by measuring the levels of malondialdehyde, a metabolic byproduct and marker of lipid peroxidation. We found that VP causes rapid increase of both overall ROS and lipid peroxide levels, independent of light activation. These effects were not dependent on YAP, as knockdown of YAP did not cause ROS or lipid peroxidation or enhance VP-induced ROS production. Temoporfin, another photodynamic drug, did not show similar activities. In addition, VP treatment led to loss of cell membrane integrity and reduction of viability. Notably, the activity of VP to induce lipid peroxidation was neutralized by ferroptosis inhibitors ferrostatin-1 or liproxstatin-1. VP treatment also reduced the levels of glutathione peroxidase 4 (GPX4), an enzyme that protects against lipid peroxidation. These results indicate that VP can induce lipid peroxidation and ferroptosis in the absence of light activation. Our findings reveal a novel mechanism by which VP inhibits tumor growth and provide insights into development of new therapeutic strategies for the treatment of pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático , Ferroptose , Neoplasias Pancreáticas , Humanos , Verteporfina/farmacologia , Verteporfina/uso terapêutico , Peroxidação de Lipídeos , Espécies Reativas de Oxigênio , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética
2.
J Biomol Struct Dyn ; 41(17): 8517-8534, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36271642

RESUMO

Pancreatic adenocarcinoma is a disease with no effective treatment. Chemo-resistance contributes to the dismal prognosis for patients diagnosed with the disease. This study aims to evaluate the toxicity and the effect of Caralluma europaea (C.E) extracts on cancer cell survival, apoptosis, chemo-resistance, and pro-cancer pathways, in pancreatic cancer. The acute and subacute toxicities of C.E extracts were evaluated. The cytotoxic effect on pancreatic cancer cell survival and apoptosis was determined by MTT assay and DNA fragmentation. The expression of cancer stemness markers was measured using Western blot. A molecular docking was used to test the possible effects of C.E compounds in inhibiting the Hedgehog and activating caspase-3. The hydroethanolic extract's DL50 was over 5000 mg/kg. During the subacute toxicity, only saponins extract showed some hepatic toxicity signs. Cells treated with C.E extracts combined with gemcitabine revealed an additive anti-survival activity. C.E extracts sensitized resistant MIA-PaCa-2 to gemcitabine treatment. Most of the C.E extracts downregulated the expression of cancer stemness-associated genes. Luteolin-7-O-glucoside presented the highest docking Gscore on human Smoothened. Isorhamnetin-3-O-rutinoside induced apoptosis via activation of caspase-3. C.E extracts can be considered safe in inhibiting pancreatic cancer cell survival, inducing apoptosis, and sensitizing cells to chemotherapy via Hedgehog inhibition and caspase-3 activation.Communicated by Ramaswamy H. Sarma.

3.
Adv Ther (Weinh) ; 5(10)2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36590644

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a disease with no effective therapeutics. We have developed a novel targeted therapy drug consisting of a tumor-targeting ligand, near-infrared (NIR) organic heptamethine carbocyanine dye (HMCD), and HMG-CoA inhibitor simvastatin (SIM), and assessed its efficacy in PDAC. PDAC cell specific targeting of DZ-SIM was measured by determining the fluorescence in cells and animals. Mitochondrial bioenergetics and functions were measured by Seahorse and flow cytometry, respectively. Apoptosis was assessed by DNA fragmentation, AnnexinV/Propidium Iodide staining, and TUNEL. Markers of cell invasion, epithelial-to-mesenchymal transition, and cancer stemness were measured. The effect of DZ-SIM on survival, tumor growth and metastasis was measured in the Krasþ/LSLG12D;Trp53þ/LSLR172H;Pdx-1-Cre (KPC) transgenic mice and in syngeneic and subcutaneous PDAC models. NIR fluorescence imaging showed specific localization of DZ-SIM to cancer, but not to normal cells and tissues. DZ-SIM significantly inhibited tumor growth and re-sensitized therapeutically resistant PDAC cells to conventional therapies. DZ-SIM killed cancer cells through unique pathways involving decreasing mitochondrial bioenergetics, including oxygen consumption and ATP production, and increasing ROS production. Mitochondrial depletion prevented the effect of DZ-SIM. Administration of DZ-SIM in 3 PDAC animal models resulted in a marked increase in survival and a decrease in tumor growth and metastasis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...