Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Redox Biol ; 73: 103176, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38705094

ABSTRACT

Excitotoxicity is a prevalent pathological event in neurodegenerative diseases. The involvement of ferroptosis in the pathogenesis of excitotoxicity remains elusive. Transcriptome analysis has revealed that cytoplasmic reduced nicotinamide adenine dinucleotide phosphate (NADPH) levels are associated with susceptibility to ferroptosis-inducing compounds. Here we show that exogenous NADPH, besides being reductant, interacts with N-myristoyltransferase 2 (NMT2) and upregulates the N-myristoylated ferroptosis suppressor protein 1 (FSP1). NADPH increases membrane-localized FSP1 and strengthens resistance to ferroptosis. Arg-291 of NMT2 is critical for the NADPH-NMT2-FSP1 axis-mediated suppression of ferroptosis. This study suggests that NMT2 plays a pivotal role by bridging NADPH levels and neuronal susceptibility to ferroptosis. We propose a mechanism by which the NADPH regulates N-myristoylation, which has important implications for ferroptosis and disease treatment.

2.
Neurochem Int ; 152: 105244, 2022 01.
Article in English | MEDLINE | ID: mdl-34826530

ABSTRACT

Excitotoxicity refers to the ability of excessive extracellular excitatory amino acids to damage neurons via receptor activation. It is a crucial pathogenetic process in neurodegenerative diseases. TP53 is confirmed to be involved in excitotoxicity. It is demonstrated that TP53 induced glycolysis and apoptotic regulator (TIGAR)-regulated metabolic pathway can protect against neuronal injury. However, the role of TIGAR in excitotoxicity and specific mechanisms is still unknown. In this study, an in vivo excitotoxicity model was constructed via stereotypical kainic acid (KA) injection into the striatum of mice. KA reduced TIGAR expression levels, neuroinflammatory responses and mitochondrial dysfunction. TIGAR overexpression could reverse KA-induced neuronal injury by reducing neuroinflammation and improving mitochondrial function, thereby exerting neuroprotective effects. Therefore, this study could provide a potential therapeutic target for neurodegenerative diseases.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Mitochondria/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroprotection/drug effects , Phosphoric Monoester Hydrolases/metabolism , Animals , Apoptosis Regulatory Proteins/drug effects , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , Mice, Transgenic , Mitochondria/metabolism , Neuroinflammatory Diseases/chemically induced , Neuroprotective Agents/pharmacology
3.
J Pharmacol Exp Ther ; 378(3): 276-286, 2021 09.
Article in English | MEDLINE | ID: mdl-34253647

ABSTRACT

Our previous studies have shown that cathepsin L (CTSL) is involved in the ability of tumors to resist ionizing radiation (IR), but the specific mechanisms responsible for this remain unknown. We report here that mutant p53 (mut-p53) is involved in IR-induced transcription of CTSL. We found that irradiation caused activation of CTSL in mut-p53 cell lines, whereas there was almost no activation in p53 wild-type cell lines. Additionally, luciferase reporter gene assay results demonstrated that IR induced the p53 binding region on the CTSL promoter. We further demonstrated that the expression of p300 and early growth response factor-1 (Egr-1) was upregulated in mut-p53 cell lines after IR treatment. Accordingly, the expression of Ac-H3, Ac-H4, AcH3K9 was upregulated after IR treatment in mut-p53 cell lines, whereas histone deacetylase (HDAC) 4 and HDAC6 were reciprocally decreased. Moreover, knockdown of either Egr-1 or p300 abolished the binding of mut-p53 to the promoter of CTSL. Chromatin immunoprecipitation assay results showed that the IR-activated transcription of CTSL was dependent on p300. To further delineate the clinical relevance of interactions between Egr-1/p300, mut-p53, and CTSL, we accessed primary tumor samples to evaluate the relationships between mut-p53, CTSL, and Egr-1/p300 ex vivo. The results support the notion that mut-p53 is correlated with CTSL transcription involving the Egr-1/p300 pathway. Taken together, the results of our study revealed that p300 is an important target in the process of IR-induced transcription of CTSL, which confirms that CTSL participates in mut-p53 gain-of-function. SIGNIFICANCE STATEMENT: Transcriptional activation of cathepsin L by ionizing radiation required the involvement of mutated p53 and Egr-1/p300. Interference with Egr-1 or p300 could inhibit the expression of cathepsin L induced by ionizing radiation. The transcriptional activation of cathepsin L by p300 may be mediated by p53 binding sites on the cathepsin L promoter.


Subject(s)
Cathepsin L , Tumor Suppressor Protein p53 , Histone Deacetylases , Repressor Proteins
4.
Front Cell Dev Biol ; 9: 612554, 2021.
Article in English | MEDLINE | ID: mdl-33644049

ABSTRACT

AIM: Previous research recognizes that NADPH can produce reduced glutathione (GSH) as a coenzyme and produce ROS as a substrate of NADPH oxidase (NOX). Besides, excessive activation of glutamate receptors results in mitochondrial impairment. The study aims at spelling out the effects of NADPH and Mito-apocynin, a NOX inhibitor which specifically targets the mitochondria, on the excitotoxicity induced by Kainic acid (KA) and its mechanism. METHODS: The in vivo neuronal excitotoxicity model was constructed by stereotypically injecting KA into the unilateral striatum of mice. Administrated NADPH (i.v, intravenous) 30 min prior and Mito-apocynin (i.g, intragastric) 1 day prior, respectively, then kept administrating daily until mice were sacrificed 14 days later. Nissl staining measured the lesion of striatum and survival status of neurons. Cylinder test of forelimb asymmetry and the adhesive removal test reflected the behavioral deficit caused by neural dysfunction. Determined Total superoxide dismutase (T-SOD), malondialdehyde (MDA), and GSH indicated oxidative stress. Western blot presented the expression levels of LC3-II/LC3-I, SQSTM1/p62, TIGAR, and NOX4. Assessed oxygen consumption rate using High-Resolution Respirometry. In vitro, the MitoSOX Indicator reflected superoxide released by neuron mitochondria. JC-1 and ATP assay Kit were used to detect mitochondrial membrane potential (MMP) and energy metabolism, respectively. RESULTS: In this study, we have successfully established excitotoxic model by KA in vivo and in vitro. KA induced decreased SOD activity and increased MDA concentration. KA cause the change of LC3-II/LC3-I, SQSTM1/p62, and TIGAR expression, indicating the autophagy activation. NADPH plays a protective role in vivo and in vitro. It reversed the KA-mediated changes in LC3, SQSTM1/p62, TIGAR, and NOX4 protein expression. Mito-apocynin inhibited KA-induced increases in mitochondrial NOX4 expression and activity. Compared with NADPH, the combination showed more significant neuroprotective effects, presenting more neurons survive and better motor function recovery. The combination also better inhibited the over-activated autophagy. In vitro, combination of NADPH and Mito-apocynin performed better in restoring mitochondria membrane potential. CONCLUSION: In summary, combined administration of NADPH and NOX inhibitors offers better neuroprotection by reducing NADPH as a NOX substrate to generate ROS. The combined use of NADPH and Mito-apocynin can better restore neurons and mitochondrial function through autophagy pathway.

5.
Acta Pharmacol Sin ; 41(4): 508-515, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32123301

ABSTRACT

Olanzapine is an antipsychotic drug used to treat patients with schizophrenia due to its lower incidence of extrapyramidal symptoms. Previous studies have shown that olanzapine activates AMP-activated protein kinase (AMPK), and induce autophagy in SH-SY5Y cell line. In this study, we investigated whether olanzapine protected against rotenone-induced neurotoxicity in PC12 cells. We showed that treatment with olanzapine increased the phosphorylation of AMPK in both dose- and time-dependent manners in PC12 cells. In addition, olanzapine activated autophagy and increased autophagic vacuoles. Furthermore, olanzapine pretreatment could protect PC12 cells from rotenone-induced apoptosis. Besides, olanzapine pretreatment could suppress the rotenone-induced depolarization of mitochondrial potential and thus protect the cells. Moreover, pretreatment with specific AMPK inhibitor compound C or with autophagy inhibitor 3-methyladenine impaired the protective effect of olanzapine on rotenone-treated PC12 cells. In summary, our results show for the first time that olanzapine ameliorates rotenone-induced injury by activating autophagy through AMPK pathway.


Subject(s)
Neuroprotective Agents/pharmacology , Olanzapine/pharmacology , Rotenone/antagonists & inhibitors , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/drug effects , Cell Survival/drug effects , PC12 Cells , Rats , Rotenone/toxicity , Tumor Cells, Cultured
6.
Toxicology ; 435: 152408, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32057834

ABSTRACT

PURPOSE: To investigate the effects and mechanisms of NADPH on Kainic acid (KA)-induced excitotoxicity. METHODS: KA, a non-N-methyl-d-aspartate glutamate receptor agonist, was exposed to adult SD rats via intrastriatal injection and rat primary cortical neurons to establish excitotoxic models in vivo and in vitro, respectively. To determine the effects of NADPH on KA-induced excitotoxicity, neuronal survival, neurologically behavioral score and oxidative stress were evaluated. To explore the mechanisms of neuroprotective effects of NADPH, the autophagy-lysosome pathway related proteins were detected. RESULTS: In vivo, NADPH (1 mg/kg or 2 mg/kg) diminished KA (2.5 nmol)-induced enlargement of lesion size in striatum, improved KA-induced dyskinesia and reversed KA-induced activation of glial cells. Nevertheless, the neuroprotective effect of NADPH was not significant under the condition of autophagy activation. NADPH (2 mg/kg) inhibited KA (2.5 nmol)-induced down-regulation of TP-53 induced glycolysis and apoptosis regulator (TIGAR) and p62, and up-regulation of the protein levels of LC3-II/LC3-I, Beclin-1 and Atg5. In vitro, the excitotoxic neuronal injury was induced after KA (50 µM, 100 µM or 200 µM) treatment as demonstrated by decreased cell viability. Moreover, KA (100 µM) increased the intracellular levels of calcium and reactive oxygen species (ROS) and declined the levels of the reduced form of glutathione (GSH). Pretreatment of NADPH (10 µM) effectively reversed these changes. Meanwhile NADPH (10 µM) inhibited KA (100 µM)-induced down-regulation of TIGAR and p62, and up-regulation of the ratio of LC3-II/LC3-I, Beclin-1, Atg5, active-cathepsin B and active-cathepsin D. CONCLUSIONS: Our data provide a possible mechanism that NADPH ameliorates KA-induced excitotoxicity by blocking the autophagy-lysosome pathway and up-regulating TIGAR along with its antioxidant properties.


Subject(s)
Autophagy/drug effects , Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Excitatory Amino Acid Agonists/toxicity , Kainic Acid/toxicity , Lysosomes/drug effects , NADP/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5/metabolism , Beclin-1/metabolism , Behavior, Animal/drug effects , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Lysosomes/metabolism , Lysosomes/pathology , Male , Microtubule-Associated Proteins/metabolism , Motor Activity/drug effects , Neurons/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Phosphoric Monoester Hydrolases/metabolism , Rats, Sprague-Dawley , Sequestosome-1 Protein/metabolism
7.
Acta Pharmacol Sin ; 40(11): 1394-1403, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31444477

ABSTRACT

Cathepsin L (CTSL), a cysteine protease, is responsible for the degradation of a variety of proteins. It is known to participate in neuronal apoptosis associated with abnormal cell cycle. However, the mechanisms underlying CTSL-induced cell apoptosis remain largely unclear. We reported here that rotenone caused an activation of CTSL expression in PC-12 cells, while knockdown of CTSL by small interfering RNAs or its inhibitor reduced the rotenone-induced cell cycle arrest and apoptosis. Moreover, elevation of CTSL and increased-apoptosis were accompanied by induction of B-Myb, a crucial cell cycle regulator. We found that B-Myb was increased in rotenone-treated PC-12 cells and knockdown of B-Myb ameliorated rotenone-stimulated cell apoptosis. Further analysis demonstrated that CTSL influenced the expression of B-Myb as suppression of CTSL activity led to a decreased B-Myb expression, whereas overexpression of CTSL resulted in B-Myb induction. Reduction of B-Myb in CTSL-overexpressing cells revealed that regulation of cell cycle-related proteins, including cyclin A and cyclin B1, through CTSL was mediated by the transcription factor B-Myb. In addition, we demonstrated that the B-Myb target, Bim, and its regulator, Egr-1, which was also associated with CTSL closely, were both involved in rotenone-induced apoptosis in PC-12 cells. Our data not only revealed the role of CTSL in rotenone-induced neuronal apoptosis, but also indicated the involvement of B-Myb in CTSL-related cell cycle regulation.


Subject(s)
Apoptosis/physiology , Cathepsin L/physiology , Cell Cycle Proteins/metabolism , Transcription Factors/metabolism , Animals , Apoptosis/drug effects , Bcl-2-Like Protein 11/metabolism , Cell Cycle/physiology , Cyclin A/metabolism , Cyclin B1/metabolism , Early Growth Response Protein 1/metabolism , PC12 Cells , Rats , Rotenone/pharmacology
8.
CNS Neurosci Ther ; 25(9): 911-921, 2019 09.
Article in English | MEDLINE | ID: mdl-30955240

ABSTRACT

AIMS: Our laboratory previously reported that olanzapine treatment inhibited growth of glioma cell lines and hypothesized that autophagy may be involved in the proliferation inhibitory effects of olanzapine. However, the mechanisms of olanzapine-contributed autophagy activation are unclear. METHODS: The inhibitory effects of olanzapine on glioma cells were evaluated by CCK8 assay, Hoechst 33258 staining and annexin V-FITC/PI staining. Western blotting, nuclear separation techniques, and immunofluorescence assays were used to investigate the relationship between the inhibition of NF-κB and autophagy activation by olanzapine. RESULTS: In this work, we verified that olanzapine increased autophagic flux and autophagic vesicles. In addition, we confirmed that autophagy was related to NF-κB inhibition in cancer progression, especially with the nuclear translocation of p65. Furthermore, we demonstrated that autophagy induced by olanzapine could be impaired with TNFα cotreatment. We also found that olanzapine had an inhibitory effect on T98 cells with positive MGMT protein expression, which may involve the inhibition of MGMT through effects on NF-κB. CONCLUSIONS: Our findings identify a pathway by which olanzapine induces autophagy by depressing NF-κB in a glioma cell line, providing evidence which supports the use of olanzapine as a potential anticancer drug.


Subject(s)
Autophagy/drug effects , Glioma/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Olanzapine/toxicity , Selective Serotonin Reuptake Inhibitors/toxicity , Antineoplastic Agents/toxicity , Autophagy/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Glioma/pathology , Humans
9.
Acta Pharmacol Sin ; 39(6): 1034-1047, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29219949

ABSTRACT

Cathepsin L (CTSL), a cysteine protease, is closely related to tumor occurrence, development, and metastasis, and possibly regulates cancer cell resistance to chemotherapy. miRNAs, especially the miR-200 family, have been implicated in drug-resistant tumors. In this study we explored the relationship of CTSL, miRNA-200c and drug resistance, and the potential regulatory mechanisms in human lung cancer A549 cells and A549/TAX cells in vitro. A549/TAX cells were paclitaxel-resistant A549 cells overexpressing CTSL and characterized by epithelial-mesenchymal transition (EMT). We showed that miRNA-200c and CTSL were reciprocally linked in a feedback loop in these cancer cells. Overexpression of miRNA-200c in A549/TAX cells decreased the expression of CTSL, and enhanced their sensitivity to paclitaxel and suppressed EMT, whereas knockdown of miRNA-200c in A549 cells significantly increased the expression of CTSL, and decreased their sensitivity to paclitaxel and induced EMT. Overexpression of CTSL in A549 cells significantly decreased the expression of miRNA-200c, and reduced their sensitivity to paclitaxel and induced EMT, but these effects were reversed by miRNA-200c, whereas knockdown of CTSL in A549/TAX cells attenuated paclitaxel resistance and remarkably inhibited EMT, but the inhibition of miRNA-200c could reverse these effects. Therefore, miRNA-200c may be involved in regulating paclitaxel resistance through CTSL-mediated EMT in A549 cells, and CTSL and miRNA-200c are reciprocally linked in a feedback loop.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cathepsin L/metabolism , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/drug therapy , MicroRNAs/metabolism , Paclitaxel/pharmacology , A549 Cells , Cathepsin L/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/genetics , Neoplasm Invasiveness , Signal Transduction/drug effects
10.
Acta Pharmacol Sin ; 37(12): 1606-1622, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27840408

ABSTRACT

AIM: Cathepsin L (CTSL), a lysosomal acid cysteine protease, is known to play important roles in tumor metastasis and chemotherapy resistance. In this study we investigated the molecular mechanisms underlying the regulation of chemoresistance by CTSL in human lung cancer cells. METHODS: Human lung cancer A549 cells, A549/PTX (paclitaxel-resistant) cells and A549/DDP (cisplatin-resistant) cells were tested. The resistance to cisplatin or paclitaxel was detected using MTT and the colony-formation assays. Actin remodeling was observed with FITC-Phalloidin fluorescent staining or immunofluorescence. A wound-healing assay or Transwell assay was used to assess the migration or invasion ability. The expression of CTSL and epithelial and mesenchymal markers was analyzed with Western blotting and immunofluorescence. The expression of EMT-associated transcription factors was measured with Western blotting or q-PCR. BALB/c nude mice were implanted subcutaneously with A549 cells overexpressing CTSL, and the mice were administered paclitaxel (10, 15 mg/kg, ip) every 3 d for 5 times. RESULTS: Cisplatin or paclitaxel treatment (10-80 ng/mL) induced CTSL expression in A549 cells. CTSL levels were much higher in A549/PTX and A549/DDP cells than in A549 cells. Silencing of CTSL reversed the chemoresistance in A549/DDP and A549/TAX cells, whereas overexpression of CTSL attenuated the sensitivity of A549 cells to cisplatin or paclitaxel. Furthermore, A549/DDP and A549/TAX cells underwent morphological and cytoskeletal changes with increased cell invasion and migration abilities, accompanied by decreased expression of epithelial markers (E-cadherin and cytokeratin-18) and increased expression of mesenchymal markers (N-cadherin and vimentin), as well as upregulation of EMT-associated transcription factors Snail, Slug, ZEB1 and ZEB2. Silencing of CTSL reversed EMT in A549/DDP and A549/TAX cells; In contrast, overexpression of CTSL induced EMT in A549 cells. In xenograft nude mouse model, the mice implanted with A549 cells overexpressing CTSL exhibited significantly reduced sensitivity to paclitaxel treatment, and increased expression of EMT-associated proteins and transcription factors in tumor tissues. CONCLUSION: Cisplatin and paclitaxel resistance is associated with CTSL upregulation-induced EMT in A549 cells. Thus, CTSL-mediated EMT may be exploited as a target to enhance the efficacy of cisplatin or paclitaxel against lung cancer and other types of malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Cathepsin L/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Paclitaxel/pharmacology , A549 Cells , Animals , Humans , Male , Mice , Mice, Inbred BALB C , Up-Regulation/drug effects
11.
Acta Pharmacol Sin ; 36(9): 1113-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26095040

ABSTRACT

AIM: Cathepsin L is a lysosomal cysteine protease that plays important roles in cancer tumorigenesis, proliferation and chemotherapy resistance. The aim of this study was to determine how cathepsin L regulated the radiosensitivity of human glioma cells in vitro. METHODS: Human glioma U251 cells (harboring the mutant type p53 gene) and U87 cells (harboring the wide type p53 gene) were irradiated with X-rays. The expression of cathepsin L was analyzed using Western blot and immunofluorescence assays. Cell survival and DNA damage were evaluated using clonogenic and comet assays, respectively. Flow cytometry was used to detect the cell cycle distribution. Apoptotic cells were observed using Hoechst 33258 staining and fluorescence microscopy. RESULTS: Irradiation significantly increased the cytoplasmic and nuclear levels of cathepsin L in U251 cells but not in U87 cells. Treatment with the specific cathepsin L inhibitor Z-FY-CHO (10 µmol/L) or transfection with cathepsin L shRNA significantly increased the radiosensitivity of U251 cells. Both suppression and knockdown of cathepsin L in U251 cells increased irradiation-induced DNA damage and G2/M phase cell cycle arrest. Both suppression and knockdown of cathepsin L in U251 cells also increased irradiation-induced apoptosis, as shown by the increased levels of Bax and decreased levels of Bcl-2. CONCLUSION: Cathepsin L is involved in modulation of radiosensitivity in human glioma U251 cells (harboring the mutant type p53 gene) in vitro.


Subject(s)
Brain Neoplasms/radiotherapy , Cathepsin L/antagonists & inhibitors , DNA Damage/radiation effects , G2 Phase Cell Cycle Checkpoints/radiation effects , Glioma/radiotherapy , Apoptosis/radiation effects , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cathepsin L/analysis , Cathepsin L/genetics , Cathepsin L/metabolism , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , RNA Interference , RNA, Small Interfering/genetics , Radiation Tolerance
12.
Acta Pharmacol Sin ; 36(3): 400-10, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25661319

ABSTRACT

AIM: Cathepsin L, a lysosomal cysteine proteinase, is exclusively elevated in a variety of malignancies, including gliomas. In this study we investigated the relationship between cathepsin L and NF-κB, two radiation-responsive elements, in regulating the sensitivity of human glioma cells ionizing radiation (IR) in vitro. METHODS: Human glioma U251 cells were exposed to IR (10 Gy), and the expression of cathepsin L and NF-κB was measured using Western blotting. The nuclear translocation of NF-κB p65 and p50 was analyzed with immunofluorescence assays. Cell apoptosis was examined with clonogenic assays. NF-κB transcription and NF-κB-dependent cyclin D1 and ATM transactivation were monitored using luciferase reporter and ChIP assays, respectively. DNA damage repair was investigated using the comet assay. RESULTS: IR significantly increased expression of cathepsin L and NF-κB p65 and p50 in the cells. Furthermore, IR significantly increased the nuclear translocation of NF-κB, and NF-κB-dependent cyclin D1 and ATM transactivation in the cells. Knockdown of p65 did not change the expression of cathepsin L in IR-treated cells. Pretreatment with Z-FY-CHO (a selective cathepsin L inhibitor), or knockdown of cathepsin L significantly attenuated IR-induced nuclear translocation of NF-κB and cyclin D1 and ATM transactivation, and sensitized the cells to IR. Pretreatment with Z-FY-CHO, or knockdown of p65 also decreased IR-induced DNA damage repair and clonogenic cell survival, and sensitized the cells to IR. CONCLUSION: Cathepsin L acts as an upstream regulator of NF-κB activation in human glioma cells and contributes to their sensitivity to IR in vitro. Inhibition of cathepsin L can sensitize the cells to IR.


Subject(s)
Brain Neoplasms/radiotherapy , Cathepsin L/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Glioma/radiotherapy , NF-kappa B/metabolism , Neurons/drug effects , Neurons/radiation effects , Radiation-Sensitizing Agents/pharmacology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Active Transport, Cell Nucleus , Ataxia Telangiectasia Mutated Proteins/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cathepsin L/genetics , Cathepsin L/metabolism , Cell Line, Tumor , Cyclin D1/metabolism , DNA Damage , Dose-Response Relationship, Radiation , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Humans , NF-kappa B p50 Subunit/metabolism , Neurons/enzymology , Neurons/pathology , RNA Interference , Time Factors , Transcription Factor RelA/metabolism , Transfection
13.
Asian Pac J Cancer Prev ; 15(19): 8107-13, 2014.
Article in English | MEDLINE | ID: mdl-25338992

ABSTRACT

The aim of this study was to investigate the effects of olanzapine on growth inhibition as well as autophagy in glioma cells in vitro and in vivo. The proliferation of both LN229 and T98 glioma cells, measured by MTT assay, was suppressed in a concentration-dependent and time-dependent manner. Moreover, apoptosis of both cells was significantly increased with the treatment of olanzapine as evidenced by increased Bcl-2 expression, Hoechst 33258 staining and annexinV-FITC/PI staining. Olanzapine treatment also enhanced activation of autophagy with increased expression of LC3-II, expression of protein p62, a substrate of autophagy, being decreased. The growth inhibition by olanzapine in both glioma cell lines could be blocked by co-treatment with 3-MA, an autophagy inhibitor. Furthermore, olanzapine effectively blocked the growth of subcutaneous xenografts of LN229 glioma cells in vivo. The increased level of protein LC3-II and decreased level of p62 followed by a decreased level of Bcl-2, suggesting that autophagy may contribute to apoptosis. In addition, reduced proliferation of glioma cells was shown by a decrease of Ki-67 staining and increased caspase-3 staining indicative of apoptosis in mouse xenografts. These results indicated that olanzapine inhibited the growth of glioma cells accompanied by induction of autophagy and apoptosis both in vitro and in vivo. Olanzapine-induced autophagy plays a tumor-suppressing role in glioma cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Benzodiazepines/pharmacology , Glioma/pathology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Biomarkers, Tumor/metabolism , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/prevention & control , Cell Proliferation/drug effects , Flow Cytometry , Glioma/metabolism , Glioma/prevention & control , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Olanzapine , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Pharm Biol ; 52(10): 1302-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24992201

ABSTRACT

CONTEXT: The principal bioactive lignan of Schisandra chinensis fructus, commonly used for traditional Chinese medicine, is schisandrin A. Schisandrin A has been widely reported as being very effective for the treatment of liver disease. However, the mechanisms of its protective effects in liver remain unclear. OBJECTIVE: To explore the hepatoprotective mechanisms of schisandrin A. MATERIALS AND METHODS: d-Galactosamine (d-GalN)-induced liver injury in mice was used as a model. Schisandrin A was examined for its protective mechanisms using hematoxylin-eosin (HE) staining, enzyme-linked immunosorbent assay (ELISA), western blotting and real-time PCR (RT-PCR). RESULTS: Aspartate amino-transferase (AST) and alanine transaminase (ALT) levels in the schisandrin A group were significantly decreased (p < 0.01) compared with those in the d-GalN-treated group. HE results showed that the pathological changes in hepatic tissue seen in the d-GalN-treated were reduced in the schisandrin A/d-GalN-treated group, with the morphological characteristics being close to those of the control (untreated) group. Western blotting results showed that schisandrin A can activate autophagy flux and inhibit progression of apoptosis. The immune function of the schisandrin A-pretreated group was assayed by flow cytometry. It was found that the mechanism may involve activated autophagy flux, inhibited apoptosis, and improved immunity in response to liver damage. CONCLUSION: Our results show that the hepatoprotective mechanisms of schisandrin A may include activation of autophagy flux and inhibition of apoptosis. These results provide pharmacological evidence supporting its future clinical application.


Subject(s)
Autophagy/drug effects , Chemical and Drug Induced Liver Injury/prevention & control , Cyclooctanes/therapeutic use , Galactosamine/toxicity , Lignans/therapeutic use , Polycyclic Compounds/therapeutic use , Schisandra , Animals , Autophagy/physiology , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Cyclooctanes/pharmacology , Lignans/pharmacology , Male , Mice , Mice, Inbred ICR , Polycyclic Compounds/pharmacology , Random Allocation
15.
PLoS One ; 8(9): e75702, 2013.
Article in English | MEDLINE | ID: mdl-24073275

ABSTRACT

The present study seeks to investigate the role of cathepsin L in glutamate receptor-induced transcription factor nuclear factor-kappa B (NF-κB) activation and excitotoxicity in rats striatal neurons. Stereotaxic administration of the N-methyl-d-aspartate (NMDA) receptor agonist Quinolinic acid (QA) into the unilateral striatum was used to produce the in vivo excitotoxic model. Co-administration of QA and the cathepsin L inhibitor Z-FF-FMK or 1-Naphthalenesulfonyl-IW-CHO (NaphthaCHO) was used to assess the contribution of cathepsin L to QA-induced striatal neuron death. Western blot analysis and cathepsin L activity assay were used to assess the changes in the levels of cathepsin L after QA treatment. Western blot analysis was used to assess the changes in the protein levels of inhibitor of NF-κB alpha isoform (IκB-α) and phospho-IκB alpha (p-IκBα) after QA treatment. Immunohistochemical analysis was used to detect the effects of Z-FF-FMK or NaphthaCHO on QA-induced NF-κB. Western blot analysis was used to detect the effects of Z-FF-FMK or NaphthaCHO on QA-induced IκB-α phosphorylation and degradation, changes in the levels of IKKα, p-IKKα, TP53, caspase-3, beclin1, p62, and LC3II/LC3I. The results show that QA-induced loss of striatal neurons were strongly inhibited by Z-FF-FMK or NaphthaCHO. QA-induced degradation of IκB-α, NF-κB nuclear translocation, up-regulation of NF-κB responsive gene TP53, and activation of caspase-3 was strongly inhibited by Z-FF-FMK or NaphthaCHO. QA-induced increases in beclin 1, LC3II/LC3I, and down-regulation of p62 were reduced by Z-FF-FMK or NaphthaCHO. These results suggest that cathepsin L is involved in glutamate receptor-induced NF-κB activation. Cathepsin L inhibitors have neuroprotective effects by inhibiting glutamate receptor-induced IκB-α degradation and NF-κB activation.


Subject(s)
Apoptosis/drug effects , Cathepsin L/physiology , Corpus Striatum/drug effects , NF-kappa B/metabolism , Neurons/drug effects , Quinolinic Acid/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Corpus Striatum/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Fluorescent Antibody Technique , Glutamic Acid/metabolism , Ketones/pharmacology , Male , Neurons/metabolism , Proteolysis , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
16.
PLoS One ; 8(5): e63232, 2013.
Article in English | MEDLINE | ID: mdl-23658815

ABSTRACT

Autophagy can be induced under nutrition stress conditions. Bcl-2 is a pro-survival protein which inhibits apoptosis and autophagy. However, the role of Bcl-2 in autophagy regulation and cell survival under nutrition deprivation has not been fully understood. This study sought to investigate if Bcl-2 upregulation is essential in limiting autophagic activity and prevent cell death under nutrition deprivation conditions. Autophagic activity was monitored by the changes in GFP-LC3 localization and protein levels of Beclin1, LC3-II, cathepsin D and p62 in neuroblastoma SH-SY5Y cells underwent serum deprivation. Manipulation of Bcl-2 function was achieved with siRNAs and small molecular inhibitors. The cell viability and apoptosis were assessed with MTT assay and Annexin V/PI staining. The results showed that serum starvation increased protein levels of LC3-II and Beclin1 but decreased autophagy substrate p62. Autophagy activation induced by serum deprivation and rapamycin was accompanied by an upregulation of Bcl-2 protein levels. When Bcl-2 was knocked down with siRNA or inhibited with HA 14-1 or ABT-737, serum starvation induced profound cell death and enhanced autophagic flux under nutrition deprivation conditions, while knockdown of autophagic gene Beclin1 or autophagy inhibitors (bafilomycin A1 and E64D), rescued cell death. In contrast, overexpression of Bcl-2 inhibited autophagy and blocked cell death in response to serum deprivation. These data suggest that Bcl-2 plays an essential role in limiting autophagy activation and preventing initiation of programmed cell death. Thus Bcl-2 may be an important mechanism for balancing beneficial and detrimental impacts of autophagy on cell survival.


Subject(s)
Autophagy/genetics , Neurons/metabolism , Stress, Physiological/genetics , bcl-2-Associated X Protein/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Benzopyrans/pharmacology , Biphenyl Compounds/pharmacology , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Line, Tumor , Cell Survival/genetics , Culture Media/chemistry , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Nitriles/pharmacology , Nitrophenols/pharmacology , Piperazines/pharmacology , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , Sulfonamides/pharmacology , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/metabolism
17.
Neurochem Int ; 63(3): 133-40, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23714208

ABSTRACT

Rotenone is an environmental neurotoxin that induces degeneration of dopaminergic (DA) neurons in substantia nigra pars compacta (SNpc), which ultimately results in parkinsonism, but the molecular mechanisms of selective degeneration of nigral DA neurons are not fully understood. In the present study, we investigated the induction of p38(MAPK)/p53 and Bax in SNpc of Lewis rats after chronic treatment with rotenone and the contribution of Bax to rotenone-induced apoptotic commitment of differentiated PC12 cells. Lewis rats were subcutaneously treated with rotenone (1.5mg/kg) twice a day for 50days and the loss of tyrosine hydroxylase (THase), motor function impairment, and expression of p38(MAPK), P-p38(MAPK), p53, and Bax were assessed. After differentiated PC cells were treated with rotenone (500nM) for 6-36h, protein levels of p38(MAPK) and P-p38(MAPK), p53 nuclear translocation, Bax induction and cell death were measured. The results showed that rotenone administration significantly reduced motor activity and caused a loss of THase immunoreactivity in SNpc of Lewis rats. The degeneration of nigral DA neurons was accompanied by the increases in p38(MAPK), P-p38(MAPK), p53, and Bax protein levels. In cultured PC12 cells, rotenone also induced an upregulation of p38(MAPK), P-p38(MAPK), p53 and Bax. Pharmacological inhibition of p38(MAPK) with SB203580 (25µM) blunted rotenone-induced cell apoptosis. Treatment with SB203580 prevented the p53 nuclear translocation and upregulation of Bax. Inhibition of p53 with pifthrin-alpha or Bax with siRNAs significantly reduced rotenone-induced Bax induction and apoptotic cell death. These results suggest that the p38(MAPK)/p53-dependent induction of Bax contributes to rotenone's neurotoxicity in PD models.


Subject(s)
Disease Models, Animal , Neurons/pathology , Parkinson Disease/pathology , Rotenone/toxicity , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Male , Neurons/enzymology , Neurons/metabolism , PC12 Cells , Parkinson Disease/enzymology , Parkinson Disease/metabolism , Rats , Rats, Inbred Lew , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
18.
Acta Pharmacol Sin ; 34(5): 681-90, 2013 May.
Article in English | MEDLINE | ID: mdl-23603977

ABSTRACT

AIM: NVP-BEZ235 is a novel dual PI3K/mTOR inhibitor and shows dramatic effects on gliomas. The aim of this study was to investigate the effects of NVP-BEZ235 on the radiosensitivity and autophagy of glioma stem cells (GSCs) in vitro. METHODS: Human GSCs (SU-2) were tested. The cell viability and survival from ionizing radiation (IR) were evaluated using MTT and clonogenic survival assay, respectively. Immunofluorescence assays were used to identify the formation of autophagosomes. The apoptotic cells were quantified with annexin V-FITC/PI staining and flow cytometry, and observed using Hoechst 33258 staining and fluorescence microscope. Western blot analysis was used to analyze the expression levels of proteins. Cell cycle status was determined by measuring DNA content after staining with PI. DNA repair in the cells was assessed using a comet assay. RESULTS: Treatment of SU-2 cells with NVP-BEZ235 (10-320 nmol/L) alone suppressed the cell growth in a concentration-dependent manner. A low concentration of NVP-BEZ235 (10 nmol/L) significantly increased the radiation sensitivity of SU-2 cells, which could be blocked by co-treatment with 3-MA (50 µmol/L). In NVP-BEZ235-treated SU-2 cells, more punctate patterns of microtubule-associated protein LC3 immunoreactivity was observed, and the level of membrane-bound LC3-II was significantly increased. A combination of IR with NVP-BEZ235 significantly increased the apoptosis of SU-2 cells, as shown in the increased levels of BID, Bax, and active caspase-3, and decreased level of Bcl-2. Furthermore, the combination of IR with NVP-BEZ235 led to G1 cell cycle arrest. Moreover, NVP-BEZ235 significantly attenuated the repair of IR-induced DNA damage as reflected by the tail length of the comet. CONCLUSION: NVP-BEZ235 increases the radiosensitivity of GSCs in vitro by activating autophagy that is associated with synergistic increase of apoptosis and cell-cycle arrest and decrease of DNA repair capacity.


Subject(s)
Brain Neoplasms/radiotherapy , Glioma/radiotherapy , Imidazoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Radiation-Sensitizing Agents/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Autophagy/drug effects , Autophagy/radiation effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/radiation effects , Glioma/drug therapy , Glioma/pathology , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Radiation Tolerance/drug effects
19.
Chin J Cancer ; 30(10): 712-20, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21959048

ABSTRACT

Glioma stem/progenitor cells (GSPCs) are considered to be responsible for the initiation, propagation, and recurrence of gliomas. The factors determining their differentiation remain poorly defined. Accumulating evidences indicate that alterations in autophagy may influence cell fate during mammalian development and differentiation. Here, we investigated the role of autophagy in GSPC differentiation. SU-2 cells were treated with rapamycin, 3-methyladenine (3-MA) plus rapamycin, E64d plus rapamycin, or untreated as control. SU-2 cell xenografts in nude mice were treated with rapamycin or 3-MA plus rapamycin, or untreated as control. Western blotting and immunocytochemistry showed up-regulation of microtubule-associated protein light chain-3 (LC3)-II in rapamycin-treated cells. The neurosphere formation rate and the number of cells in each neurosphere were significantly lower in the rapamycin treatment group than in other groups. Real-time PCR and immunocytochemistry showed down-regulation of stem/progenitor cell markers and up-regulation of differentiation markers in rapamycin-treated cells. Transmission electron microscopy revealed autophagy activation in rapamycin-treated tumor cells in mice. Immunohistochemistry revealed decreased Nestin-positive cells and increased GFAP-positive cells in rapamycin-treated tumor sections. These results indicate that rapamycin induces differentiation of GSPCs by activating autophagy.


Subject(s)
Autophagy/drug effects , Brain Neoplasms/pathology , Cell Differentiation/drug effects , Glioma/pathology , Neoplastic Stem Cells/pathology , Sirolimus/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Brain Neoplasms/metabolism , Cell Line, Tumor , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glioma/metabolism , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/metabolism , RNA, Messenger/metabolism , Xenograft Model Antitumor Assays
20.
Brain Res ; 1371: 7-15, 2011 Jan 31.
Article in English | MEDLINE | ID: mdl-21108935

ABSTRACT

Glioblastoma (GBM) is a highly aggressive brain tumor characterized by increased proliferation and resistance to chemotherapy and radiotherapy. A growing body of evidence suggests that only a small subpopulation of malignant glioma cells, called glioma stem cells or glioma-initiating cells (GICs), have true tumorigenic potential and confer glioma radioresistance. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) plays a major role in the repair of DNA double-strand breaks induced by ionizing radiation (IR). Suppression of one of these components of the DNA-PK complex can inhibit the DNA double-strand break repair and radiosensitize the cells. In general, the cell death induced by IR is considered to be apoptotic. Recently, autophagy, an alternative form of programmed cell death, has been shown to contribute significantly to anti-neoplastic effects of radiation therapy. Autophagy is independent of phagocytes and differs from apoptosis by the presence of autophagosomes, autolysosomes, and an intact nucleus in the cell. Little is known, however, regarding the relationship between DNA-PKcs and IR-induced autophagy in GICs. In the present study, we constructed plasmids encoding short hairpin RNA (shRNA) targeting DNA-PKcs, which were then transfected into GICs. Then, we used GICs and DNA-PKcs-RNAi transfected cells to investigate the role of DNA-PKcs in IR-induced apoptotic and autophagic cell death. IR induced massive autophagic cell death in DNA-PKcs-RNAi transfected cells, but only occasional apoptotic cells were detected among GICs. Specific inhibition of DNA-PKcs in GICs induced autophagy and radiosensitized the cells. Our results suggest that such radiation-induced autophagy may enhance the effect of glioma therapies.


Subject(s)
Autophagy/radiation effects , Brain Neoplasms/pathology , DNA-Activated Protein Kinase/physiology , Gene Knockdown Techniques , Glioblastoma/pathology , Neoplastic Stem Cells/enzymology , RNA, Small Interfering/pharmacology , Radiation Tolerance/physiology , Apoptosis , Autophagy/drug effects , Autophagy/physiology , Brain Neoplasms/radiotherapy , Catalytic Domain/genetics , DNA Breaks, Double-Stranded , DNA Repair , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/genetics , Glioblastoma/radiotherapy , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , RNA Interference , RNA, Small Interfering/genetics , Radiation Tolerance/drug effects , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...