Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Neural Plast ; 2021: 4090441, 2021.
Article in English | MEDLINE | ID: mdl-34471408

ABSTRACT

Our previous study showed that growth arrest- and DNA damage-inducible gene 153 (GAD153/CHOP) plays an important role in intermittent hypoxia- (IH-) induced apoptosis and impaired synaptic plasticity. This study is aimed at determining which signaling pathway is activated to induce CHOP and the role of this protein in mitochondria-dependent apoptosis induced by IH. In the in vivo study, mice were placed in IH chambers for 8 h daily over a period of 2 weeks; the IH chambers had oxygen (O2) concentrations that oscillated between 10% and 21%, cycling every 90 s. In the in vitro study, PC12 cells were exposed to 21% O2 (normoxia) or 8 IH cycles (25 min at 21% O2 and 35 min at 0.1% O2 for each cycle). After 2 weeks of IH treatment, we observed that the expression levels of phosphorylated protein kinase-like endoplasmic reticulum kinase (p-PERK), activating transcription factor 4 (ATF-4) and phosphorylated eukaryotic initiation factor 2 alpha (p-elf2α), were increased, but the levels of activating transcription factor 6 (ATF-6) and inositol-requiring enzyme 1 (IRE-1) were not increased. GSK2606414, a specific chemical inhibitor of the PERK pathway, reduced the expression of p-PERK, ATF-4, p-elf2α, and CHOP and rescued ER structure. In addition, Bax and Bak accumulated in the mitochondria after IH treatment, which induced cytochrome c release and initiated apoptosis. These effects were prevented by GSK2606414 and CHOP shRNA. Finally, the impaired long-term potentiation and long-term spatial memory in the IH group were rescued by GSK2606414. Together, the data from the in vitro and in vivo experiments indicate that IH-induced apoptosis and impaired synaptic plasticity were mediated by the PERK-ATF-4-CHOP pathway. Suppressing PERK-ATF-4-CHOP signaling pathway attenuated mitochondria-dependent apoptosis by reducing the expression of Bax and Bak in mitochondria, which may serve as novel adjunct therapeutic strategy for ameliorating obstructive sleep apnea- (OSA-) induced neurocognitive impairment.


Subject(s)
Cognitive Dysfunction/metabolism , Hypoxia/metabolism , Neurons/metabolism , Transcription Factor CHOP/biosynthesis , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cognitive Dysfunction/drug therapy , Hypoxia/drug therapy , Indoles/pharmacology , Indoles/therapeutic use , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , PC12 Cells , Rats , Transcription Factor CHOP/antagonists & inhibitors
2.
PLoS One ; 16(4): e0249591, 2021.
Article in English | MEDLINE | ID: mdl-33819286

ABSTRACT

Traumatic spinal cord injury (SCI) is a devastating neurological condition that involves both primary and secondary tissue loss. Various cytotoxic events including hypoxia, hemorrhage and blood lysis, bioenergetic failure, oxidative stress, endoplasmic reticulum (ER) stress, and neuroinflammation contribute to secondary injury. The HIF prolyl hydroxylase domain (PHD/EGLN) family of proteins are iron-dependent, oxygen-sensing enzymes that regulate the stability of hypoxia inducible factor-1α (HIF-1α) and also mediate oxidative stress caused by free iron liberated from the lysis of blood. PHD inhibition improves outcome after experimental intracerebral hemorrhage (ICH) by reducing activating transcription factor 4 (ATF4)-driven neuronal death. As the ATF4-CHOP (CCAAT-enhancer-binding protein homologous protein) pathway plays a role in the pathogenesis of contusive SCI, we examined the effects of PHD inhibition in a mouse model of moderate T9 contusive SCI in which white matter damage is the primary driver of locomotor dysfunction. Pharmacological inhibition of PHDs using adaptaquin (AQ) moderately lowers acute induction of Atf4 and Chop mRNAs and prevents the acute decline of oligodendrocyte (OL) lineage mRNAs, but does not improve long-term recovery of hindlimb locomotion or increase chronic white matter sparing. Conditional genetic ablation of all three PHD isoenzymes in OLs did not affect Atf4, Chop or OL mRNAs expression levels, locomotor recovery, and white matter sparing after SCI. Hence, PHDs may not be suitable targets to improve outcomes in traumatic CNS pathologies that involve acute white matter injury.


Subject(s)
Activating Transcription Factor 4/antagonists & inhibitors , Endoplasmic Reticulum Stress , Locomotion , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Recovery of Function , Spinal Cord Injuries/physiopathology , Transcription Factor CHOP/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Spinal Cord Injuries/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
3.
Cell Mol Life Sci ; 78(6): 3021-3044, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33230565

ABSTRACT

Arginine deprivation therapy (ADT) is a new metabolic targeting approach with high therapeutic potential for various solid cancers. Combination of ADT with low doses of the natural arginine analog canavanine effectively sensitizes malignant cells to irradiation. However, the molecular mechanisms determining the sensitivity of intrinsically non-auxotrophic cancers to arginine deficiency are still poorly understood. We here show for the first time that arginine deficiency is accompanied by global metabolic changes and protein/membrane breakdown, and results in the induction of specific, more or less pronounced (severe vs. mild) ER stress responses in head and neck squamous cell carcinoma (HNSCC) cells that differ in their intrinsic ADT sensitivity. Combination of ADT with canavanine triggered catastrophic ER stress via the eIF2α-ATF4(GADD34)-CHOP pathway, thereby inducing apoptosis; the same signaling arm was irrelevant in ADT-related radiosensitization. The particular strong supra-additive effect of ADT, canavanine and irradiation in both intrinsically more and less sensitive cancer cells supports the rational of ER stress pathways as novel target for improving multi-modal metabolic anti-cancer therapy.


Subject(s)
Canavanine/pharmacology , Endoplasmic Reticulum Stress/drug effects , Radiation Tolerance/drug effects , X-Rays , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Apoptosis/drug effects , Arginine/deficiency , Arginine/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Culture Media/chemistry , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Endoribonucleases/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
4.
Acta Biochim Biophys Sin (Shanghai) ; 53(1): 46-53, 2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33242093

ABSTRACT

Endoplasmic reticulum (ER) stress plays a critical role in pancreatic ß cell destruction which leads to the pathogenesis of type 1 diabetes mellitus (T1DM). Vitamin D (VD) has been reported to reduce the risk of T1DM; however, it remains unknown whether VD affects ER stress in pancreatic ß cells. In this study, we investigated the role of the active form of VD, 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3], in ER stress-induced ß cell apoptosis and explored its potential mechanism in mouse insulinoma cell line mouse insulinoma 6 (MIN6). The results of cell counting kit-8 (CCK8) and flow cytometric analyses showed that 1,25-(OH)2D3 caused a significant increase in the viability of MIN6 cells injured by H2O2. The protein kinase like ER kinase (PERK) signal pathway, one of the most conserved branches of ER stress, was found to be involved in this process. H2O2 activated the phosphorylation of PERK, upregulated the activating transcription factor 4 (ATF4) and C/EBP homologous protein (CHOP) expression, and subsequently initiated cell apoptosis, which were significantly reversed by 1,25-(OH)2D3 pretreatment. In addition, GSK2606414, a specific inhibitor of PERK, suppressed PERK phosphorylation and reduced the expressions of ATF4 and CHOP, leading to a significant decrease in ß cell apoptosis induced by H2O2. Taken together, the present findings firstly demonstrated that 1,25-(OH)2D3 could prevent MIN6 cells against ER stress-associated apoptosis by inhibiting the PERK-ATF4-CHOP pathway. Therefore, our results suggested that 1,25-(OH)2D3 might serve as a potential therapeutic target for preventing pancreatic ß cell destruction in T1DM.


Subject(s)
Activating Transcription Factor 4/antagonists & inhibitors , Calcitriol/pharmacology , Insulin-Secreting Cells/drug effects , Protective Agents/pharmacology , Signal Transduction/drug effects , Transcription Factor CHOP/antagonists & inhibitors , eIF-2 Kinase/antagonists & inhibitors , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Endoplasmic Reticulum Stress/drug effects , Hydrogen Peroxide/toxicity , Indoles/pharmacology , Insulin-Secreting Cells/cytology , Mice
5.
Chin J Nat Med ; 18(9): 704-713, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32928514

ABSTRACT

Chinese herbal compound Nao-Fu-Cong (NFC) has been mainly used to treat cognitive disorders in Traditional Chinese Medicine (TCM). The present study aimed to investigate whether its neuroprotective effects might be related to the inhibition of JNK/CHOP/Bcl2-mediated apoptosis pathway or not. We randomly assigned STZ (60 mg·kg-1)-induced diabetic rats into control group, diabetic model group and NFC groups (low-dose, medium-dose and high-dose). The primary culture of hippocampal neurons were transferred into different culture media on the third day. The cells were then divided into control group, high-glucose group, NFC (low-dose, medium-dose and high-dose) groups, CHOP si-RNA intervention group, JNK pathway inhibitor SP600125 group and oxidative stress inhibitor N-acetylcysteine (NAC) group. NFC significantly improved the cognitive function of diabetic rats, and had neuroprotective effect on hippocampal neurons cultured in high glucose. Further research results showed that NFC could reduce the apoptosis of hippocampal neurons in rats with diabetic cognitive dysfunction. NFC had inhibitory effects on CHOP/JNK apoptosis pathway induced by high glucose, and also decreased the levels of ROS and increased the mitochondrial membrane potential. These suggested that the neuroprotective effect of NFC might be related to the inhibition of CHOP and JNK apoptotic signaling pathways, and the cross pathway between oxidative stress and mitochondrial damage pathway.


Subject(s)
Apoptosis/drug effects , Cognitive Dysfunction/drug therapy , Drugs, Chinese Herbal/pharmacology , Neuroprotective Agents/pharmacology , Acetylcysteine/pharmacology , Animals , Anthracenes/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Dose-Response Relationship, Drug , Hippocampus/drug effects , MAP Kinase Signaling System/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Neurons/drug effects , Proto-Oncogene Proteins c-bcl-2/drug effects , Random Allocation , Rats , Transcription Factor CHOP/antagonists & inhibitors
6.
Chem Res Toxicol ; 33(5): 1237-1249, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32285662

ABSTRACT

To assess the toxicity of nanomaterials, most in vitro studies have been performed under submerged conditions, which do not reflect physiological conditions upon inhalation. An air-liquid interface (ALI) exposure may provide more reliable data on dosimetry and prevent interactions with cell culture media components. Therefore, an ALI exposure was combined with a high-throughput RT-qPCR approach to evaluate the toxicological potential of CuO and TiO2 nanoparticles (NP) in A549 cells. While TiO2 NP did not show any cytotoxicity or other effects compromising genomic stability up to 25.8 µg/cm2, CuO NP revealed a dose-dependent cytotoxicity, starting at 4.9 µg/cm2. Furthermore, CuO NP altered distinct gene expression patterns indicative for disturbed metal homeostasis, stress response, and DNA damage induction. Thus, induction of metal homeostasis associated genes (MT1X, MT2A) at 0.4 µg/cm2 and higher suggested uptake and intracellular dissolution of CuO NP, which was verified by a dose-dependent increase in intracellular copper concentration. Starting at 4.9 µg/cm2, oxidative stress markers (HMOX1, HSPA1A) were induced dose-dependently, supported by elevated ROS levels. Furthermore, a dose-dependent induction of genes associated with DNA damage response (DDIT3, GADD45A) was observed, in concordance with an increase in DNA strand breaks. Finally, transcriptional data suggested the induction of apoptosis at high doses, while flow cytometric analysis revealed increased numbers of either late apoptotic or necrotic cells and clearly necrotic cells at the highest concentrations. Thus, an ALI cell culture system was successfully combined with a comprehensive high-throughput RT-qPCR system, allowing the quantification of NP deposition and their impact on genomic stability. For CuO NP, in principle the data confirm observations made under submerged conditions with respect to intracellular copper ion release, as well as oxidative and genotoxic stress response. However, the results derived from ALI exposure allow the assessment of dose-response-relationships as well as the comparison of relative toxic potencies of different NP.


Subject(s)
Copper/toxicity , Gene Expression Profiling , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Titanium/toxicity , A549 Cells , Air , Apoptosis/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Copper/chemistry , DNA Damage , Dose-Response Relationship, Drug , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/genetics , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Humans , Metallothionein/antagonists & inhibitors , Metallothionein/genetics , Oxidative Stress/drug effects , Particle Size , Real-Time Polymerase Chain Reaction , Titanium/chemistry , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Tumor Cells, Cultured , Water/chemistry
7.
Neurobiol Dis ; 136: 104725, 2020 03.
Article in English | MEDLINE | ID: mdl-31911115

ABSTRACT

Identifying disease-causing pathways and drugs that target them in Parkinson's disease (PD) has remained challenging. We uncovered a PD-relevant pathway in which the stress-regulated heterodimeric transcription complex CHOP/ATF4 induces the neuron prodeath protein Trib3 that in turn depletes the neuronal survival protein Parkin. Here we sought to determine whether the drug adaptaquin, which inhibits ATF4-dependent transcription, could suppress Trib3 induction and neuronal death in cellular and animal models of PD. Neuronal PC12 cells and ventral midbrain dopaminergic neurons were assessed in vitro for survival, transcription factor levels and Trib3 or Parkin expression after exposure to 6-hydroxydopamine or 1-methyl-4-phenylpyridinium with or without adaptaquin co-treatment. 6-hydroxydopamine injection into the medial forebrain bundle was used to examine the effects of systemic adaptaquin on signaling, substantia nigra dopaminergic neuron survival and striatal projections as well as motor behavior. In both culture and animal models, adaptaquin suppressed elevation of ATF4 and/or CHOP and induction of Trib3 in response to 1-methyl-4-phenylpyridinium and/or 6-hydroxydopamine. In culture, adaptaquin preserved Parkin levels, provided neuroprotection and preserved morphology. In the mouse model, adaptaquin treatment enhanced survival of dopaminergic neurons and substantially protected their striatal projections. It also significantly enhanced retention of nigrostriatal function. These findings define a novel pharmacological approach involving the drug adaptaquin, a selective modulator of hypoxic adaptation, for suppressing Parkin loss and neurodegeneration in toxin models of PD. As adaptaquin possesses an oxyquinoline backbone with known safety in humans, these findings provide a firm rationale for advancing it towards clinical evaluation in PD.


Subject(s)
Activating Transcription Factor 4/metabolism , Cell Cycle Proteins/biosynthesis , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/prevention & control , Pyridines/pharmacology , Quinolines/pharmacology , Transcription Factor CHOP/metabolism , Activating Transcription Factor 4/antagonists & inhibitors , Animals , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Oxidopamine/toxicity , PC12 Cells , Parkinsonian Disorders/chemically induced , Pyridines/therapeutic use , Quinolines/therapeutic use , Rats , Transcription Factor CHOP/antagonists & inhibitors
8.
Nat Commun ; 10(1): 1280, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894532

ABSTRACT

Understanding the intrinsic mediators that render CD8+ T cells dysfunctional in the tumor microenvironment is a requirement to develop more effective cancer immunotherapies. Here, we report that C/EBP homologous protein (Chop), a downstream sensor of severe endoplasmic reticulum (ER) stress, is a major negative regulator of the effector function of tumor-reactive CD8+ T cells. Chop expression is increased in tumor-infiltrating CD8+ T cells, which correlates with poor clinical outcome in ovarian cancer patients. Deletion of Chop in T cells improves spontaneous antitumor CD8+ T cell immunity and boosts the efficacy of T cell-based immunotherapy. Mechanistically, Chop in CD8+ T cells is elevated primarily through the ER stress-associated kinase Perk and a subsequent induction of Atf4; and directly represses the expression of T-bet, a master regulator of effector T cell function. These findings demonstrate the primary role of Chop in tumor-induced CD8+ T cell dysfunction and the therapeutic potential of blocking Chop or ER stress to unleash T cell-mediated antitumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Ovarian Epithelial/genetics , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Neoplastic/immunology , Ovarian Neoplasms/genetics , T-Box Domain Proteins/genetics , Transcription Factor CHOP/genetics , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/therapy , Cell Line, Tumor , Female , Humans , Immunity, Cellular , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Knockout , Ovarian Neoplasms/immunology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Survival Analysis , T-Box Domain Proteins/immunology , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology
9.
Cell Oncol (Dordr) ; 42(3): 357-368, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30919222

ABSTRACT

PURPOSE: ABT-263 is a potent BH3 mimetic that possesses anticancer potential against various types of cancer. In general, this potential is due to its high binding affinity to anti-apoptotic proteins in the Bcl-2 family that disrupt sequestration of pro-apoptotic proteins. In the present study, we sought to identify an alternative regulatory mechanism responsible for ABT-263-mediated anticancer activity in human oral cancer. METHODS: We investigated the in vitro anti-cancer effects of ABT-263 using a trypan blue exclusion assay, Western blotting, DAPI staining, immunofluorescence staining, a live/dead assay, microarray-based expression profiling, and quantitative real-time PCR. In vivo anti-tumorigenic effects of ABT-263 were examined using a nude mouse tumor xenograft model, a TUNEL assay, and immunohistochemistry. RESULTS: We found that ABT-263 suppressed viability and induced apoptosis in human oral cancer-derived cell lines HSC-3 and HSC-4. Subsequent microarray-based gene expression profiling revealed 55 differentially expressed genes in the ABT-263-treatead group, including 12 genes associated with "endoplasmic reticulum stress and apoptosis." Consistent with the microarray results, the mRNA expression levels of the top four genes (CHOP, TRB3, ASNS, and STC2) were found to be significantly increased. In addition, we found that ABT-263 considerably enhanced the expression levels of the C/EBP-homologous protein (CHOP) and its mRNA, resulting in apoptosis induction in four other human oral cancer-derived cell lines (MC-3, YD-15, HN22, and Ca9.22). Extending our in vitro findings, we found that ABT-263 reduced the growth of HSC-4 cells in vivo at a dosage of 100 mg/kg/day without any change in body weight. TUNEL-positive cells were also found to be increased in tumors of ABT-263-treated mice without any apparent histopathological changes in liver or kidney tissues. CONCLUSIONS: These results provide evidence that ABT-263 may serve as an effective therapeutic agent for the treatment of human oral cancer.


Subject(s)
Aniline Compounds/pharmacology , Apoptosis/drug effects , Mouth Neoplasms/drug therapy , Sulfonamides/pharmacology , Transcription Factor CHOP/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Apoptosis/genetics , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Nude , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
10.
J Cell Mol Med ; 23(4): 2954-2969, 2019 04.
Article in English | MEDLINE | ID: mdl-30746902

ABSTRACT

This study was designed to explore the inductive effect of glycated high-density lipoprotein (gly-HDL) on endoplasmic reticulum (ER) stress-C/EBP homologous protein (CHOP)-mediated macrophage apoptosis and its relationship with autophagy. Our results showed that gly-HDL caused macrophage apoptosis with concomitant activation of ER stress pathway, including nuclear translocation of activating transcription factor 6, phosphorylation of protein kinase-like ER kinase (PERK) and eukaryotic translation initiation factor 2α, and CHOP up-regulation, which were inhibited by 4-phenylbutyric acid (PBA, an ER stress inhibitor) and the gene silencing of PERK and CHOP. Similar data were obtained from macrophages treated by HDL isolated from diabetic patients. Gly-HDL induced macrophage autophagy as assessed by up-regulation of beclin-1, autophagy-related gene 5 and microtubule-associated protein one light chain 3-II, which were depressed by PBA and PERK siRNA. Gly-HDL-induced apoptosis, PERK phosphorylation and CHOP up-regulation were suppressed by rapamycin (an autophagy inducer), whereas aggravated by 3-methyladenine (an autophagy inhibitor) and beclin-1 siRNA. Administration of diabetic apoE-/- mice with rapamycin attenuated MOMA-2 and CHOP up-regulation and apoptosis in atherosclerotic lesions. These data indicate that gly-HDL may induce macrophage apoptosis through activating ER stress-CHOP pathway and ER stress mediates gly-HDL-induced autophagy, which in turn protects macrophages against apoptosis by alleviating CHOP pathway.


Subject(s)
Apoptosis , Atherosclerosis/pathology , Autophagy , Endoplasmic Reticulum Stress , Lipoproteins, HDL/chemistry , Macrophages/pathology , Transcription Factor CHOP/antagonists & inhibitors , Animals , Atherosclerosis/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Glycolysis , Lipoproteins, HDL/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Signal Transduction
11.
Biomed Pharmacother ; 111: 468-475, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30594786

ABSTRACT

Kaempferol is a flavonoid compound that has many functions, such as anti-inflammation and antioxidation. Acute liver failure (ALF) is a life-threatening illness accompanied by serious inflammation and extensive hepatocyte apoptosis. The aim of this study was to examine the therapeutic potential of kaempferol and its mechanism in ALF. In a murine ALF model induced by d-galactosamine (d-GalN, 700 mg/kg) / lipopolysaccharide (LPS, 10 µg/kg), mice were pretreated with kaempferol at 2 h before d-GalN/LPS administration and then sacrificed 6 h after d-GalN/LPS injection. Lethality, liver damage, endoplasmic reticulum(ER) stress, hepatocyte viability and apoptosis were evaluated. Whether pretreatment of kaempferol protected hepatocytes from ER stress-induced apoptosis was detected in vitro. Pretreatment of kaempferol decreased lethality, prolonged the survival time and significantly protected against liver injury, which was indicated by decreased transaminase levels and the well-preserved liver structure. The protective effect of kaempferol on the ALF mouse model was achieved by inhibiting hepatocyte apoptosis. Moreover, pretreatment of kaempferol increased the expression of glucose-regulated/binding immunoglobulin protein 78 (Grp78), decreased the expression of C/EBP-homologous protein (CHOP), and protected hepatocytes from ER stress-induced apoptosis in vitro. Our results showed that pretreatment of Grp78 siRNA partially negated the hepatic protection from kaempferol and reversed the inhibition of CHOP protein expression in d-GalN/LPS-induced ALF mice. In conclusion, kaempferol inhibits hepatocyte apoptosis to protect mice from liver failure by regulating the ER stress-Grp78-CHOP signaling pathway. Therefore, kaempferol may be used to treat ALF.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Galactosamine/toxicity , Heat-Shock Proteins/biosynthesis , Kaempferols/therapeutic use , Liver Failure, Acute/metabolism , Transcription Factor CHOP/biosynthesis , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/agonists , Hepatocytes/drug effects , Hepatocytes/metabolism , Kaempferols/pharmacology , Lipopolysaccharides/toxicity , Liver Failure, Acute/chemically induced , Liver Failure, Acute/prevention & control , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factor CHOP/antagonists & inhibitors
12.
Bioorg Med Chem ; 26(18): 5151-5158, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30243792

ABSTRACT

Cadmium (Cd) is a potent toxic heavy metal, some studies showed that Cd-induced apoptosis is through ER stress pathway. Compounds of pyrrolo[2,1-c][1,4]benzodiazepine (PBD)-3,11-diones were discovered as potent neuroprotective agents against Cd-induced toxicity in SH-SY5Y cells for the first time. In this study, twenty-six PBD-3,11-dione derivatives were synthesized and evaluated for their neuroprotective activity against Cd-induced toxicity by CCK-8 assay. Their preliminary SARs studies indicated that various substituents were tolerated on the benzene ring, and alkyl heterocycles groups at the N10-position of the PBD-3,11-dione scaffold were important for the activities. Among them, compound 13c exhibited the best activity (cell viability = 68.6%, 25 µM). Furthermore, we found that the compound 13c could inhibit cadmium-induced cell apoptosis with the downregulation of the ER stress markers GRP78, CHOP, cleaved-caspase12 and cleaved-caspase3 through western blotting. The results of in silico evaluation of ADME/T properties showed that 13c exhibited medium BBB penetration level and promising toxicity profiles. These results proved the potential of 13c as a promising lead compound against Cd-induced neurotoxicity.


Subject(s)
Apoptosis/drug effects , Benzodiazepines/pharmacology , Cadmium/pharmacology , Endoplasmic Reticulum Stress/drug effects , Pyrroles/pharmacology , Benzodiazepines/chemistry , Caspase 12/metabolism , Caspase 3/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Humans , Molecular Structure , Pyrroles/chemistry , Structure-Activity Relationship , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/metabolism , Tumor Cells, Cultured
13.
Cell Physiol Biochem ; 48(5): 1995-2010, 2018.
Article in English | MEDLINE | ID: mdl-30092598

ABSTRACT

BACKGROUND/AIMS: Endoplasmic reticulum (ER) stress is an important event in atherosclerosis. Recent studies have shown that ER stress deregulates cholesterol metabolism via multiple pathways. This study aimed to determine the relationship between ER stress and lipid metabolism and to verify that upregulation of miR-33 is involved in this process. METHODS: An atherosclerosis model was established in apolipoprotein E-deficient (ApoE-/-) mice fed a Western diet, and THP-1 derived macrophages were used in this study. Hematoxylin-eosin and Oil Red O staining were used to quantify the atherosclerotic plaques. 1,1'-Dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate labeled oxidized low-density lipoprotein binding assay and a Cholesterol Efflux Fluorometric Assay Kit were used to observe cholesterol uptake and efflux. The mRNA and protein levels of biomarkers associated with ER stress and cholesterol metabolism in atherosclerotic plaques and macrophages were evaluated by real-time PCR and western blotting, respectively. Immunofluorescence was used to observe alterations of ABCA1 localization. Small interfering RNAs were used to knock down CHOP and miR-33 in macrophages to alter CHOP and miR-33 expression. RESULTS: Atherosclerotic lesions and systemic lipid levels were ameliorated after inhibition of ER stress (tauroursodeoxycholic acid) in vivo. In vitro studies confirmed that ER stress regulated the lipid catabolism of macrophages by promoting cholesterol uptake, inhibiting cholesterol efflux, and modulating the expression of related transporters. CHOP contributed to lipid metabolism disorder following ER stress. Furthermore, over-expression of miR-33 was involved in ER stress that induced lipid metabolism disorder in macrophages. These findings support a model of ER stress induction by oxidized low-density lipoprotein that affects macrophage lipid catabolism disorder. CONCLUSION: Our data shed new light on the relationship between ER stress and lipid metabolism in vivo and in vitro, and confirm that upregulation of miR-33 is involved in this process. The relationship between ER stress and miR-33 represents a novel target for the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/pathology , Endoplasmic Reticulum Stress , Lipid Metabolism , MicroRNAs/metabolism , Transcription Factor CHOP/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Antagomirs/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Atherosclerosis/veterinary , Cell Line , Cholesterol/metabolism , Endoplasmic Reticulum Stress/drug effects , Humans , Lipid Metabolism/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , RNA Interference , RNA, Small Interfering/metabolism , Taurochenodeoxycholic Acid/pharmacology , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics
14.
J Biochem ; 164(6): 415-426, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30165670

ABSTRACT

A small nuclear protein, C1D, has roles in various cellular processes, transcription regulation, genome stability surveillance, DNA repair and RNA processing, all of which are required to maintain the host life cycles. In the previous report, C1D directly interacts with XPB, a component of the nucleotide excision repair complex, and C1D knockdown reduced cell survival of 27-1 cells, CHO derivative cells, after UV irradiation. To find out the role of C1D in UV-damaged cells, we used human cell lines with siRNA or shRNA to knockdown C1D. C1D knockdown reduced cell survival rates of LU99 and 786-O after UV irradiation, although C1D knockdown did not affect the efficiency of the nucleotide excision repair. Immunostaining data support that C1D is not directly involved in the DNA repair process in UV-damaged cells. However, H2O2 treatment reduced cell viability in LU99 and 786-O cells. We also found that C1D knockdown upregulated DDIT3 expression in LU99 cells and downregulated APEX1 in 786-O cells, suggesting that C1D functions as a co-repressor/activator. The data accounts for the reduction of cell survival rates upon UV irradiation.


Subject(s)
Co-Repressor Proteins/metabolism , DNA Repair/radiation effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA/metabolism , Gene Expression Regulation, Enzymologic/radiation effects , Oxidative Stress/radiation effects , Transcription Factor CHOP/metabolism , Animals , Biomarkers/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , DNA Damage , DNA Repair/drug effects , DNA, Neoplasm/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Hydrogen Peroxide/toxicity , Oxidants/toxicity , Oxidative Stress/drug effects , Pyrimidine Dimers/metabolism , RNA Interference , Radiation Injuries, Experimental/enzymology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Transcription Factor CHOP/agonists , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics
15.
Int J Mol Sci ; 19(7)2018 06 21.
Article in English | MEDLINE | ID: mdl-29933620

ABSTRACT

Unfolded protein response (UPR) is a cytoprotective mechanism that alleviates the protein-folding burden in eukaryotic organisms. Moderate activation of UPR is required for maintaining endoplasmic reticulum (ER) homeostasis and profoundly contributes to tumorigenesis. Defects in UPR signaling are implicated in the attenuation of various malignant phenotypes including cell proliferation, migration, and invasion, as well as angiogenesis. This suggests UPR as a promising target in cancer therapy. The pharmacological effects of the plant Scindapsus cf. hederaceus on human cancer cell lines is not understood. In this study, we identified an ethyl acetate extract from Scindapsus cf. hederaceus (SH-EAE), which markedly altered the protein expression of UPR-related genes in human non-small cell lung cancer (NSCLC) cells. Treatment with the SH-EAE led to the dose-dependent suppression of colony forming ability of both H1299 and H460 cells, but not markedly in normal bronchial epithelial BEAS-2B cells. SH-EAE treatment also attenuated the migration and invasion ability of H1299 and H460 cells. Moreover, SH-EAE strikingly suppressed the protein expression of two ER stress sensors, including inositol requiring enzyme-1α (IRE-1α) and protein kinase R-like ER kinase (PERK), and antagonized the induction of C/EBP homologous protein (CHOP) expression by thapsigargin, an ER stress inducer. SH-EAE induced the formation of massive vacuoles which are probably derived from ER. Importantly, SH-EAE impaired the formation of intersegmental vessels (ISV) in zebrafish larvae, an index of angiogenesis, but had no apparent effect on the rate of larval development. Together, our findings demonstrate, for the first time, that the ability of SH-EAE specifically targets the two sensors of UPR, with significant anti-proliferation and anti-migration activities as a crude extract in human NSCLC cells. Our finding also indicates potential applications of SH-EAE in preventing UPR activation in response to Tg-induced ER stress. We suggest that SH-EAE attenuates UPR adaptive pathways for rendering the NSCLC cells intolerant to ER stress.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Araceae/chemistry , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic , Neovascularization, Physiologic/drug effects , Plant Extracts/pharmacology , Acetates/chemistry , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Bronchi/drug effects , Bronchi/metabolism , Bronchi/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Endoribonucleases/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Larva/drug effects , Neovascularization, Physiologic/genetics , Plant Extracts/isolation & purification , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Solvents/chemistry , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tumor Stem Cell Assay , Unfolded Protein Response/drug effects , Zebrafish , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
16.
Cell Physiol Biochem ; 46(1): 322-334, 2018.
Article in English | MEDLINE | ID: mdl-29590661

ABSTRACT

BACKGROUND/AIMS: Protodioscin (PD) is a steroidal saponin with anti-cancer effects on a number of cancer cells, but the anti-tumor effects and mechanism of action of PD on human cervical cancer cells is unclear. METHODS: We determined cell viability using the MTT assay. Cell death, mitochondrial membrane potential (MMP), intracellular reactive oxygen species (ROS) generation, and endoplasmic reticulum (ER) stress were measured on a flow cytometry. Caspase activation, ER stress, and MMP-dependent apoptosis proteins in cervical cancer cells in response to PD were determined by Western blot analysis. The ability of ATF4 binding to ChIP promoter was measured using the ChIP assay. RESULTS: We demonstrated that PD inhibits cell viability, causes a loss of mitochondrial function, and induces apoptosis, as evidenced by up-regulation of caspase-8, -3, -9, -PARP, and Bax activation, and down-regulation of Bcl-2 expression. PD was shown to induce ROS and the ER stress pathway, including GRP78, p-eIF-2α, ATF4, and CHOP. Pre-treatment with NAC, a ROS production inhibitor, significantly reduced ER stress and apoptosis-related proteins induced by PD. Transfection of GRP78/CHOP-siRNA effectively inhibited PD-induced ER stress-dependent apoptosis. Moreover, treatment with PD significantly increased p38 and JNK activation. Co-administration of a JNK inhibitor (SP600125) or p38 inhibitor (SB203580) abolished cell death and ER stress effects during PD treatment. In addition, PD induced the expression of nuclear ATF4 and CHOP, as well as the binding ability of ATF4 to the CHOP promoter. CONCLUSION: Our results suggest that PD is a promising therapeutic agent for the treatment of human cervical cancer.


Subject(s)
Diosgenin/analogs & derivatives , Endoplasmic Reticulum Stress/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Reactive Oxygen Species/metabolism , Saponins/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Diosgenin/chemistry , Diosgenin/pharmacology , Down-Regulation/drug effects , Drugs, Chinese Herbal/pharmacology , Endoplasmic Reticulum Chaperone BiP , Female , HeLa Cells , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Membrane Potential, Mitochondrial/drug effects , RNA Interference , Saponins/chemistry , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Up-Regulation/drug effects , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
17.
Mol Nutr Food Res ; 62(8): e1700890, 2018 04.
Article in English | MEDLINE | ID: mdl-29446867

ABSTRACT

SCOPE: We investigated the role of endoplasmic reticulum (ER) stress in the protective effects of EGCG against the neuronal apoptosis in Aß1-42 -induced SH-SY5Y cells and APP/PS1 transgenic mice. METHODS AND RESULTS: Cell viability (CCK8 assay), flow cytometry, Hoechst 33258 staining, immunohistochemistry, transmission electron microscopy (TEM), and western blotting were used. EGCG prevented Aß1-42-induced toxicity in SH-SY5Y cells, increased cell viability, and decreased apoptosis in a dose-dependent manner. In a subsequent mechanism study, it was found that this effect contributed to the down-regulation of GRP78, CHOP, cleaved-caspase-12 and -3. Moreover, EGCG also reduced the cytotoxicity induced by tunicamycin (TM) and thapsigargin (TG), two ER stress activators. Consistent with the in vitro study, EGCG inhibited neuronal apoptosis in the cortex of APP/PS1 transgenic mice, with the mitigation of ER abnormal ultrastructural swelling and the downregulation of ER-stress-associated proteins. CONCLUSION: These results indicate that EGCG attenuates the neurotoxicity in Alzheimer's disease (AD) via a novel mechanism that involves inhibition of ER-stress-associated neuronal apoptosis in vitro and in vivo, suggesting the tremendous potential of EGCG for use in a nutritional preventive strategy against AD.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Apoptosis , Catechin/analogs & derivatives , Dietary Supplements , Endoplasmic Reticulum Stress , Neurons/metabolism , Neuroprotective Agents/metabolism , Peptide Fragments/antagonists & inhibitors , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/metabolism , Animals , Caspase 12/chemistry , Caspase 12/genetics , Caspase 12/metabolism , Caspase 3/chemistry , Caspase 3/genetics , Caspase 3/metabolism , Catechin/metabolism , Catechin/therapeutic use , Cell Line, Tumor , Cell Survival , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/ultrastructure , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/agonists , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Mice, Transgenic , Microscopy, Electron, Transmission , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/pathology , Neurons/ultrastructure , Neuroprotective Agents/therapeutic use , Nootropic Agents/metabolism , Nootropic Agents/therapeutic use , Peptide Fragments/metabolism , Random Allocation , Transcription Factor CHOP/agonists , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
18.
J Biol Chem ; 293(8): 2801-2814, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29305424

ABSTRACT

The stress-induced unfolded protein response (UPR) in the endoplasmic reticulum (ER) involves various signaling cross-talks and controls cell fate. B-cell receptor (BCR) signaling, which can trigger UPR, induces gammaherpesvirus lytic replication and serves as a physiological mechanism for gammaherpesvirus reactivation in vivo However, how the UPR regulates BCR-mediated gammaherpesvirus infection is unknown. Here, we demonstrate that the ER stressors tunicamycin and thapsigargin inhibit BCR-mediated murine gammaherpesvirus 68 (MHV68) lytic replication by inducing expression of the UPR mediator Bip and blocking activation of Akt, ERK, and JNK. Both Bip and the downstream transcription factor ATF4 inhibited BCR-mediated MHV68 lytic gene expression, whereas UPR-induced C/EBP homologous protein (CHOP) was required for and promoted BCR-mediated MHV68 lytic replication by suppressing upstream Bip and ATF4 expression. Bip knockout was sufficient to rescue BCR-mediated MHV68 lytic gene expression in CHOP knockout cells, and this rescue was blocked by ectopic ATF4 expression. Furthermore, ATF4 directly inhibited promoter activity of the MHV68 lytic switch transactivator RTA. Altogether, we show that ER stress-induced CHOP inhibits Bip and ATF4 expression and that ATF4, in turn, plays a critical role in CHOP-mediated regulation of BCR-controlled MHV68 lytic replication. We conclude that ER stress-mediated UPR and BCR signaling pathways are interconnected and form a complex network to regulate the gammaherpesvirus infection cycle.


Subject(s)
Activating Transcription Factor 4/metabolism , B-Lymphocytes/virology , Endoplasmic Reticulum Stress , Gammaherpesvirinae/physiology , Heat-Shock Proteins/metabolism , Receptors, Antigen, B-Cell/agonists , Transcription Factor CHOP/metabolism , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Animals , Antiviral Agents/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line, Transformed , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Gammaherpesvirinae/drug effects , Gammaherpesvirinae/growth & development , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Lysogeny/drug effects , Mice , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Promoter Regions, Genetic/drug effects , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/drug effects , Thapsigargin/pharmacology , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Tunicamycin/pharmacology , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Activation/drug effects , Virus Replication/drug effects
19.
Cell Physiol Biochem ; 45(1): 267-280, 2018.
Article in English | MEDLINE | ID: mdl-29357323

ABSTRACT

BACKGROUND/AIMS: Prostate cancer (PCa) is the second most frequently diagnosed cancer in men worldwide. Currently available therapies for hormone-refractory PCa are only marginally effective. Plumbagin (PLB), a natural naphthoquinone isolated from the traditional folk medicine Plumbago zeylanica, is known to selectively kill tumor cells. Nevertheless, antitumor mechanisms initiated by PLB in cancer cells have not been fully defined. METHODS: MTT assay was used to evaluate the effect of PLB on the viability of cancer cells. Cell apoptosis and reactive oxygen species (ROS) production were determined by flow cytometry. Protein expression was detected by western blotting. In vivo anti-tumor effect was measured by using tumor xenoqraft model in nude mice. RESULTS: In the present study, we found that PLB decreases cancer cell growth and induces apoptosis in DU145 and PC-3 cells. In addition, by increasing intracellular ROS levels, PLB induced a lethal endoplasmic reticulum stress response in PCa cells. Importantly, blockage of ROS production significantly reversed PLB-induced ER stress activation and cell apoptosis. In vivo, we found that PLB inhibits the growth of PCa xenografts without exhibiting toxicity Treatment of mice bearing human PCa xenografts with PLB was also associated with induction of ER stress activation. CONCLUSION: Inducing ER stress by PLB thus discloses a previously unrecognized mechanism underlying the biological activity of PLB and provides an in-depth insight into the action of PLB in the treatment of hormone-refractory PCa.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Naphthoquinones/pharmacology , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Activating Transcription Factor 4/metabolism , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Naphthoquinones/chemistry , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transplantation, Heterologous , eIF-2 Kinase/metabolism
20.
Arch Physiol Biochem ; 124(2): 131-138, 2018 May.
Article in English | MEDLINE | ID: mdl-28857622

ABSTRACT

CONTEXT: Molecular pathogenesis of chronic alcoholism is linked to increased endoplasmic reticulum stress. Ethanol is a competitive inhibitor of vitamin A metabolism and vitamin A supplementation aggravates existing liver problems. Hence, we probed into the impact of supplementation of all trans retinoic acid (ATRA), the active metabolite of vitamin A on ethanol-induced endoplasmic reticulcum stress. METHODS: Male Sprague-Dawley rats were divided into four groups - I: Control; II: Ethanol; III: ATRA; IV: ATRA + Ethanol. After 90 days the animals were sacrificed to study markers of lipid peroxidation in hepatic microsomal fraction and expression of ER stress proteins and apoptosis in liver. RESULTS AND CONCLUSION: Ethanol caused hepatic hyperlipidemia, enhanced microsomal lipid peroxidation, upregulated expression of unfolded protein response associated proteins and that of apoptosis. Ethanol also led to downregulation of retinoid receptors. ATRA supplementation reversed all these alterations indicating the decrease in ethanol-induced endoplasmic reticulum stress.


Subject(s)
Dietary Supplements , Endoplasmic Reticulum Stress , Fatty Liver, Alcoholic/prevention & control , Liver/metabolism , Protective Agents/therapeutic use , Receptors, Retinoic Acid/agonists , Tretinoin/therapeutic use , Activating Transcription Factor 4/agonists , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cytochrome P-450 CYP2E1/chemistry , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Endoplasmic Reticulum Stress/drug effects , Ethanol/toxicity , Fatty Liver, Alcoholic/enzymology , Fatty Liver, Alcoholic/metabolism , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/enzymology , Male , Rats, Sprague-Dawley , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/agonists , Retinoid X Receptors/antagonists & inhibitors , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Transcription Factor CHOP/agonists , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tretinoin/antagonists & inhibitors , Unfolded Protein Response/drug effects , X-Box Binding Protein 1/agonists , X-Box Binding Protein 1/antagonists & inhibitors , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL