Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Cell Death Dis ; 13(9): 754, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36050295

ABSTRACT

Ivermectin is a widely used antiparasitic drug and shows promising anticancer activity in various cancer types. Although multiple signaling pathways modulated by ivermectin have been identified in tumor cells, few studies have focused on the exact target of ivermectin. Herein, we report the pharmacological effects and targets of ivermectin in prostate cancer. Ivermectin caused G0/G1 cell cycle arrest, induced cell apoptosis and DNA damage, and decreased androgen receptor (AR) signaling in prostate cancer cells. Further in vivo analysis showed ivermectin could suppress 22RV1 xenograft progression. Using integrated omics profiling, including RNA-seq and thermal proteome profiling, the forkhead box protein A1 (FOXA1) and non-homologous end joining (NHEJ) repair executer Ku70/Ku80 were strongly suggested as direct targets of ivermectin in prostate cancer. The interaction of ivermectin and FOXA1 reduced the chromatin accessibility of AR signaling and the G0/G1 cell cycle regulator E2F1, leading to cell proliferation inhibition. The interaction of ivermectin and Ku70/Ku80 impaired the NHEJ repair ability. Cooperating with the downregulation of homologous recombination repair ability after AR signaling inhibition, ivermectin increased intracellular DNA double-strand breaks and finally triggered cell death. Our findings demonstrate the anticancer effect of ivermectin in prostate cancer, indicating that its use may be a new therapeutic approach for prostate cancer.


Subject(s)
Hepatocyte Nuclear Factor 3-alpha , Ivermectin , Ku Autoantigen , Prostatic Neoplasms , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Repair , Hepatocyte Nuclear Factor 3-alpha/drug effects , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Ivermectin/pharmacology , Ivermectin/therapeutic use , Ku Autoantigen/drug effects , Ku Autoantigen/metabolism , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism
2.
Biomed Res Int ; 2020: 1283214, 2020.
Article in English | MEDLINE | ID: mdl-32076604

ABSTRACT

Bupivacaine, a common local anesthetic, causes serious nerve injury, especially in diabetic patients, as high glucose has been reported to enhance bupivacaine-induced neurotoxicity. However, the key regulator for synergism remains unknown. To our surprise, the expression of repair protein Ku70 is suppressed, while the high-glucose environment induces DNA oxidative damage in neurons. Here, we aim to investigate whether the inhibition of Ku70 by high-glucose conditions aggrandized bupivacaine-induced DNA damage. Consistent with previous results, bupivacaine induced reactive oxygen species production and upregulated Ku70 and cleaved caspase-3 expressions at both transcript and protein levels and ultimately caused nucleic acid damage and apoptosis in human neuroblastoma (SH-SY5Y) cells. High-glucose treatment inhibited the expression of Ku70 and enhanced bupivacaine-induced neurotoxicity. In contrast, the overexpression of Ku70 mitigated DNA damage and apoptosis triggered by bupivacaine and high glucose. In conclusion, our data indicated that local anesthetics may aggravate nerve toxicity in a high-glucose environment.


Subject(s)
Bupivacaine/toxicity , DNA Damage/drug effects , DNA Repair/drug effects , Glucose/metabolism , Ku Autoantigen/drug effects , Ku Autoantigen/metabolism , Neurotoxicity Syndromes/metabolism , Apoptosis , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Ku Autoantigen/genetics , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
3.
Pathol Res Pract ; 215(11): 152677, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31591052

ABSTRACT

Sanguinarine (SAG), a benzophenanthridine alkaloid extracted from Sanguinaria canadensis, exerts antioxidant, anti-inflammatory and antiproliferative activities in a variety of malignancies. However, the underlying mechanisms by which SAG affects the tumorigenesis of gastric cancer (GC) are unclear. The common targets of SAG and GC were identified by network pharmacology, and the association of thymocyte selection-associated high mobility group box (TOX) with the clinicopathological characteristics and prognosis of patients with GC was analyzed by using datasets from The Cancer Genome Atlas (TCGA). 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assays, colony formation assays, flow cytometry analysis, and a xenograft tumor model were conducted to assess the effects of SAG on the growth of GC cells, and Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis were used to determine the effects of SAG on the TOX/DNA-PKcs/KU70/80 signaling pathway. We identified 9 collective targets of SAG and GC, of which TOX expression levels were dramatically downregulated in GC tissues compared with adjacent normal tissues, and a low expression of TOX served as an independent prognostic factor of poor survival in patients with GC. SAG suppressed cell viability, colony formation and in vivo tumorigenesis and induced cell apoptosis and cell cycle arrest. Furthermore, SAG increased the expression levels of TOX but decreased those of DNA-PKcs and KU70/80 in GC cells. Our findings indicate that SAG inhibits the tumorigenesis of GC cells by regulating TOX/DNA-PKcs/KU70/80 signaling and may provide therapeutic strategies for the treatment of GC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzophenanthridines/pharmacology , Carcinogenesis/drug effects , Isoquinolines/pharmacology , Signal Transduction/drug effects , Stomach Neoplasms/metabolism , Animals , Cell Proliferation/drug effects , High Mobility Group Proteins/drug effects , High Mobility Group Proteins/metabolism , Humans , Ku Autoantigen/drug effects , Ku Autoantigen/metabolism , Mice, Inbred BALB C , Mice, Nude , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL