Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Arch Biochem Biophys ; 729: 109390, 2022 10 30.
Article in English | MEDLINE | ID: mdl-36067878

ABSTRACT

BACKGROUND: To investigate the protective effect of low-dose radiation (LDR) on brain injury in mice induced by doxorubicin (DOX). METHODS: Sixty female BALB/C mice were randomly divided into the control (CTR) group, low-dose radiation (LDR) group, doxorubicin treatment (DOX) group and low-dose radiation before doxorubicin treatment (COM) group. After 72 h of exposure to 75 mGy, the mice were intraperitoneally injected with 7.5 mg/kg of doxorubicin and sacrificed 5 days later. Neuron-specific enolase (NSE), lactate dehydrogenase (LDH), adenosine triphosphate (ATP), neurotransmitters, inflammatory mediators, apoptosis- and oxidative stress-related mediators as well as mitochondrial dysfunction were examined. RESULTS: Compared to the DOX group, the concentrations of DA, 5-HT, EPI and GABA in the COM group were significantly decreased, and the number of TUNEL-positive cells was decreased. In addition, the expression of proapoptotic proteins was downregulated in the COM group compared to the DOX group. Low-dose radiation in advance reduced reactive oxygen species and activated the SOD antioxidant defense system as indicated by significantly reduced GSH expression, increased GSSG expression, increased GPx expression and activation of the Nrf2 redox pathway. After low-dose radiation, the expression levels of ATP5f1, NDUFV1 and CYC1 were close to normal, and the mitochondrial respiratory control rate (RCR) and activity of respiratory chain complex enzymes also tended to be normal. Low-dose radiation upregulated the expression levels of IL-2 and IL-4 but downregulated the expression levels of IL-10 and TGF-ß. CONCLUSION: LDR has a protective effect on brain injury in mice treated with DOX. The mechanism is related to LDR alleviating mitochondrial dysfunction and oxidative stress, which promotes the production of antioxidant damage proteins, thus exerting an adaptive protective effect on cells.


Subject(s)
Brain Injuries , NF-E2-Related Factor 2 , Adenosine Triphosphate/metabolism , Animals , Antioxidants/pharmacology , Apoptosis , Brain Injuries/chemically induced , Brain Injuries/prevention & control , Doxorubicin/pharmacology , Female , Glutathione Disulfide/metabolism , Interleukin-10/metabolism , Interleukin-2/metabolism , Interleukin-2/pharmacology , Interleukin-4/metabolism , Lactate Dehydrogenases/metabolism , Mice , Mice, Inbred BALB C , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Phosphopyruvate Hydratase/metabolism , Phosphopyruvate Hydratase/pharmacology , Reactive Oxygen Species/metabolism , Serotonin/metabolism , Superoxide Dismutase/metabolism , Transforming Growth Factor beta/metabolism , gamma-Aminobutyric Acid/metabolism
2.
Med Sci Monit ; 26: e927106, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32804918

ABSTRACT

BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a life-threatening digestive tract malignancy with no known curative treatment. This study aimed to investigate the antineoplastic effects of omipalisib and its underlying molecular mechanisms in ESCC using a high throughput screen. MATERIAL AND METHODS MTT assay and clone formation were used to determine cell viability and proliferation. Flow cytometry was conducted to detect cell cycle distribution and apoptosis. Global gene expression and mRNA expression levels were determined by RNA sequencing and real-time PCR, respectively. Protein expression was evaluated in the 4 ESCC cell lines by Western blot analysis. Finally, a xenograft nude mouse model was used to evaluate the effect of omipalisib on tumor growth in vivo. RESULTS In the pilot screening of a 1404-compound library, we demonstrated that omipalisib markedly inhibited cell proliferation in a panel of ESCC cell lines. Mechanistically, omipalisib induced G0/G1 cell cycle arrest and apoptosis. RNA-seq, KEGG, and GSEA analyses revealed that the PI3K/AKT/mTOR pathway is the prominent target of omipalisib in ESCC cells. Treatment with omipalisib decreased expression of p-AKT, p-4EBP1, p-p70S6K, p-S6, and p-ERK, therefore disrupting the activation of PI3K/AKT/mTOR and ERK signaling. In the nude mouse xenograft model, omipalisib significantly suppressed the tumor growth in ESCC tumor-bearing mice without obvious adverse effects. CONCLUSIONS Omipalisib inhibited the proliferation and growth of ESCC by disrupting PI3K/AKT/mTOR and ERK signaling. The present study supports the rationale for using omipalisib as a therapeutic approach in ESCC patients. Further clinical studies are needed.


Subject(s)
Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Quinolines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/enzymology , Esophageal Squamous Cell Carcinoma/metabolism , Heterografts , Humans , Mice , Mice, Nude , Pyridazines
3.
Thorac Cancer ; 13(10): 1513-1524, 2022 05.
Article in English | MEDLINE | ID: mdl-35429141

ABSTRACT

BACKGROUND: Lung cancer is one of the most lethal cancers worldwide. Cisplatin, a widely used anti-lung cancer drug, has been limited in clinical application due to its drug resistance. Medicines targeting mitochondrial electron transport chain (ETC) complexes may be effective candidates for cisplatin-based chemotherapy. METHODS: In this study, the small molecule drug library from Food and Drug Administration FDA was used to screen for medicines targeting ETC. MTT and colony formation assays were used to investigate cell proliferation. Flow cytometry was employed to analyze cell cycle, apoptosis, reactive oxygen species (ROS), and mitochondrial membrane potential. Wound scratch and transwell assays were used to detect migration and invasion abilities. The activities of the ETC complex were tested using kits. Western blot analysis was used to investigate the expressions of related proteins. A mouse xenograft model was constructed to verify the antitumor effect in vivo. RESULTS: The results showed that mubritinib can reduce the activation of the PI3K/mTOR signal pathway, disrupt mitochondrial function, significantly increase ROS levels and induce oxidative stress, and ultimately exert its antitumor effect against non-small cell lung cancer (NSCLC) both in vivo and in vitro. In addition, the combination of cisplatin and mubritinib can improve the tumor-suppressive effect of cisplatin. CONCLUSION: Mubritinib can upregulate intracellular ROS concentration and cell apoptosis, inhibit the PI3K signaling pathway and interfere with the function of mitochondria, thus reducing cell proliferation and increasing ROS induced apoptosis by reducing the activation of Nrf2 by PI3K.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacology , Cisplatin/therapeutic use , Humans , Lung Neoplasms/pathology , Mice , Mitochondria/metabolism , Oxazoles , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Triazoles
SELECTION OF CITATIONS
SEARCH DETAIL