Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters











Database
Language
Publication year range
1.
Environ Toxicol ; 38(1): 90-100, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36176197

ABSTRACT

Temozolomide (TMZ) can cross the blood-brain barrier (BBB) and deliver methyl groups to the purine (guanine) bases of DNA, leading to mispairing during DNA replication and subsequent cell death. However, increased expression of the repair enzyme methyl guanine methyltransferase (MGMT), which removes methyl groups from purine bases, counteracts methylation by TMZ. We evaluated the anticancer potential of thymoquinone (TQ), a hydrophobic flavonoid that inhibits resistance and induces apoptosis in various cancer cells, both in vitro and in vivo. In vitro experiments showed that compared with the Hs683 and M059J cell lines, U251 cells were more sensitive to TMZ. Compared to U251 cells, U251R cells, a TMZ drug-resistant strain established in this study, are characterized by increased expression of phosphorylated extracellular signal-regulated kinase (p-ERK) and MGMT. TQ treatments induced apoptosis in all cell lines. The p38 mitogen-activated protein kinase signal pathway was mainly activated in U251 and U251R cells; however, p-ERK and MGMT upregulation could not suppress TQ effects. Furthermore, si-p38 pretreatment of U251R cells in TQ treatments inhibited cell apoptosis. We speculate that TQ contributed to the phosphorylation and activation of p38, but not of ERK-induced apoptosis (irrespective of TMZ resistance). In vivo, U251R-derived tumors subcutaneously inoculated in nude mice exhibited significant tumor volume reduction after TQ or TQ + TMZ cotreatments. High-performance liquid chromatography assay confirmed the presence of TQ in murine brain tissues. Our findings demonstrate that TQ can effectively cross the BBB and function alone or in combination with TMZ to treat glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Mice , Animals , Temozolomide/pharmacology , Temozolomide/therapeutic use , Glioblastoma/pathology , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Mice, Nude , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Apoptosis , Signal Transduction , Purines/pharmacology , Purines/therapeutic use , Guanine/pharmacology , Guanine/therapeutic use , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/metabolism
2.
Environ Toxicol ; 36(10): 1990-1998, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34173702

ABSTRACT

Although enormous achievements have been made in targeted molecular therapies against hepatocellular carcinoma (HCC), the treatments can only prolong the life of patients with extrahepatic metastases. We evaluated thymoquinone (TQ), a compound from Nigella sativa Linn., for its anti-cancer effect on SK-Hep1 cells and HCC-xenograft nude mice. TQ effectively triggered cell death    and activated p38 and extracellular signal-regulated kinases (Erk) pathways up to 24 h after treatment in cells. TQ-induced cell death was reversed by p38 inhibitor; however, it was enhanced by si-Erk. The caspase3 activation and TUNEL assay revealed a stronger toxic effect upon co-treatment with TQ and si-Erk. Our study suggested that phosphorylation of p38 in SK-Hep1 cells constituted the major factor leading to cell apoptosis, whereas phosphorylation of Erk led to drug resistance. Furthermore, TQ therapeutic effect was improved upon Erk inhibition in HCC-xenograft nude mice. TQ could present excellent anti-HCC potential under suitable p-Erk inhibiting conditions.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Apoptosis , Benzoquinones , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Liver Neoplasms/drug therapy , Mice , Mice, Nude , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL