Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Ethnopharmacol ; 290: 115100, 2022 May 23.
Article in English | MEDLINE | ID: mdl-35151835

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The natural extract glaucocalyxin A (GLA), purified from the aboveground sections of the Chinese traditional medicinal herb Rabdosia japonica (Burm. f.) Hara var. glaucocalyx (Maxim.) Hara, has various pharmacological benefits, such as anti-bacterial, anti-coagulative, anti-neoplastic, and anti-inflammatory activities. Although GLA has shown anti-tumor activity against various cancers, the therapeutic potential and biological mechanisms of GLA remain to be further explored in oral squamous cell carcinoma (OSCC). AIM OF THE STUDY: This study aimed to elucidate the therapeutic potential and regulatory mechanisms of GLA in OSCC. MATERIALS AND METHODS: The cell proliferation and apoptosis effects of GLA were analyzed by CCK-8, clone formation, Annexin V/PI staining, and apoptotic protein expression in vitro. An OSCC xenograft model was applied to confirm the anti-neoplastic effect in vivo. Furthermore, the changes of reactive oxygen species (ROS) were determined by DCFH-DA probe and GSH/GSSG assay, and inhibited by the pan-caspase inhibitor Z-VAD(OMe)-FMK and the ROS scavenger N-acetylcysteine (NAC). The modulation of GLA on mitochondria and ER-dependent apoptosis pathways was analyzed by JC-1 probe, quantitative real-time PCR, and Western blot. Finally, public databases, clinical samples, and transfection cells were analyzed to explore the importance of GLA's indirect targeting molecule CHAC1 in OSCC. RESULTS: GLA significantly inhibited cell proliferation and induced apoptosis in vitro and in vivo. GLA perturbed the redox homeostasis, and cell apoptosis was totally rescued by Z-VAD(OMe)-FMK and NAC. Furthermore, GLA activated the mitochondrial apoptosis pathway. Simultaneously, the overexpression and knockdown of CHAC1 dramatically affected GLA-mediated apoptosis. The endoplasmic reticulum stress-associated ATF4/CHOP signal was identified to participate in GLA-upregulated CHAC1 expression. Finally, we found that CHAC1 expression was lower in OSCC compared with normal tissues and positively correlated with 4-Hydroxynonenal (4-HNE) level. High CHAC1 expression also indicated better overall survival. Moreover, CHAC1 selectively regulated the viability of oral cancer cells. CONCLUSION: GLA is a promising therapeutic agent that activates the ROS-mediated ATF4/CHOP/CHAC1 axis in OSCC patients.


Subject(s)
Activating Transcription Factor 4/drug effects , Carcinoma, Squamous Cell/pathology , Diterpenes, Kaurane/pharmacology , Mouth Neoplasms/pathology , Transcription Factor CHOP/drug effects , gamma-Glutamylcyclotransferase/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , Humans , Isodon , Male , Mice , Mice, Inbred BALB C , Mitochondria/drug effects , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
2.
Pharmacology ; 106(11-12): 606-615, 2021.
Article in English | MEDLINE | ID: mdl-34515223

ABSTRACT

INTRODUCTION: Canagliflozin (CANA) is a sodium-glucose cotransporter 2 inhibitor that was recently approved for treating diabetes. However, its effects on liver function are not well understood. The function of asparagine synthetase (ASNS) has been studied in several cancers but not in liver injury. Therefore, we investigated the connection between CANA and ASNS in alleviating damage (i.e., their hepatoprotective effect) in a rat liver injury model. METHODS: The rat model of liver injury was established using carbon tetrachloride treatment. Rats with liver injury were administered CANA orally for 8 weeks daily. After week 8, peripheral blood was collected to measure serum alanine aminotransferase, aspartate aminotransferase, and lactate dehydrogenase levels. Liver histopathology was examined using hematoxylin and eosin staining to determine the degree of liver injury. Protein expression in the rat livers was examined using Western blotting. RESULTS: CANA treatment decreased serum alanine aminotransferase, aspartate aminotransferase, and lactate dehydrogenase levels compared with those of the untreated group, demonstrating diminished liver injury. Mechanistically, CANA treatment activated AMP-activated protein kinase (AMPK), leading to increased nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and activating transcription factor 4 (ATF4), which upregulated ASNS expression in liver-injured rats. CONCLUSION: CANA significantly alleviated liver injury by activating the AMPK/Nrf2/ATF4 axis and upregulating ASNS expression, indicating its potential for treating patients with type 2 diabetes mellitus with impaired liver function.


Subject(s)
Aspartate-Ammonia Ligase/pharmacology , Canagliflozin/pharmacology , Chemical and Drug Induced Liver Injury/pathology , Liver/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Activating Transcription Factor 4/drug effects , Adenylate Kinase/drug effects , Animals , Carbon Tetrachloride/pharmacology , Disease Models, Animal , Liver Function Tests , NF-E2-Related Factor 2/drug effects , Rats , Rats, Sprague-Dawley
3.
Sci Rep ; 11(1): 3765, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33580171

ABSTRACT

The initial sensing of dietary methionine restriction (MR) occurs in the liver where it activates an integrated stress response (ISR) that quickly reduces methionine utilization. The ISR program is regulated in part by ATF4, but ATF4's prototypical upstream regulator, eIF2α, is not acutely activated by MR. Bioinformatic analysis of RNAseq and metabolomics data from liver samples harvested 3 h and 6 h after initiating MR shows that general translation is inhibited at the level of ternary complex formation by an acute 50% reduction of hepatic methionine that limits formation of initiator methionine tRNA. The resulting ISR is induced by selective expression of ATF4 target genes that mediate adaptation to reduced methionine intake and return hepatic methionine to control levels within 4 days of starting the diet. Complementary in vitro experiments in HepG2 cells after knockdown of ATF4, or inhibition of mTOR or Erk1/2 support the conclusion that the early induction of genes by MR is partially dependent on ATF4 and regulated by both mTOR and Erk1/2. Taken together, these data show that initiation of dietary MR induces an mTOR- and Erk1/2-dependent stress response that is linked to ATF4 by the sharp, initial drop in hepatic methionine and resulting repression of translation pre-initiation.


Subject(s)
Activating Transcription Factor 4/metabolism , Gene Expression/drug effects , Methionine/metabolism , Activating Transcription Factor 4/drug effects , Animals , Diet Therapy/methods , Eukaryotic Initiation Factor-2/metabolism , Gene Expression/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hep G2 Cells , Humans , Liver/metabolism , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Stress, Physiological/physiology , TOR Serine-Threonine Kinases/metabolism , eIF-2 Kinase/metabolism
4.
Acta Cir Bras ; 35(1): e202000104, 2020.
Article in English | MEDLINE | ID: mdl-32159588

ABSTRACT

PURPOSE: Glutamine, as an essential part of enteral nutrition and parenteral nutrition agent, has been widely recognized to be a kind of important intestinal mucosa protectant in clinical practice and experimental research. However, the mechanisms of its protective effects are still not fully understand. Consequently, this study aimed to explore the potential mechanism of glutamine on ischemia-reperfusion (I/R) injury induced endoplasmic reticulum (ER) stress in intestine. METHODS: An experimental model of intestinal I/R in rats was established by 1 hour occlusion of the superior mesenteric artery followed by 3 hours of reperfusion. Morphologic changes of intestinal mucosa, apoptosis of epithelial cells, and expression of intestinal Grp78, Gadd153, Caspase-12, ATF4, PERK phosphorylation (P-PERK) and elF2αphosphorylation(P-elF2α) were determined. RESULTS: After I/R, the apoptotic index of intestinal mucosa epithelial cells observably increased with notable necrosis of intestinal mucosa, and the expressions of Grp78, Gadd153, Caspase-12, ATF4, P-PERK and P-elF2αall were increased. However, treatment with glutamine could significantly relieve intestinal I/R injury and apoptosis index. Moreover, glutamine could clearly up-regulate the expression of Grp78, restrain P-PERK and P-elF2α, and reduce ATF4, Gadd153 and Caspase-12 expressions. CONCLUSION: Glutamine may be involved in alleviating ER stress induced intestinal mucosa cells apoptosis.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Glutamine/pharmacology , Intestinal Mucosa/drug effects , Protective Agents/pharmacology , Reperfusion Injury/prevention & control , Activating Transcription Factor 4/drug effects , Animals , Caspase 12/drug effects , Heat-Shock Proteins/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Male , Mesenteric Artery, Superior/injuries , Models, Animal , RNA, Messenger/drug effects , Rats, Sprague-Dawley , Transcription Factor CHOP/drug effects , eIF-2 Kinase/drug effects
5.
Acta cir. bras ; 35(1): e202000104, 2020. graf
Article in English | LILACS | ID: biblio-1088525

ABSTRACT

Abstract Purpose Glutamine, as an essential part of enteral nutrition and parenteral nutrition agent, has been widely recognized to be a kind of important intestinal mucosa protectant in clinical practice and experimental research. However, the mechanisms of its protective effects are still not fully understand. Consequently, this study aimed to explore the potential mechanism of glutamine on ischemia-reperfusion (I/R) injury induced endoplasmic reticulum (ER) stress in intestine. Methods An experimental model of intestinal I/R in rats was established by 1 hour occlusion of the superior mesenteric artery followed by 3 hours of reperfusion. Morphologic changes of intestinal mucosa, apoptosis of epithelial cells, and expression of intestinal Grp78, Gadd153, Caspase-12, ATF4, PERK phosphorylation (P-PERK) and elF2αphosphorylation(P-elF2α) were determined. Results After I/R, the apoptotic index of intestinal mucosa epithelial cells observably increased with notable necrosis of intestinal mucosa, and the expressions of Grp78, Gadd153, Caspase-12, ATF4, P-PERK and P-elF2αall were increased. However, treatment with glutamine could significantly relieve intestinal I/R injury and apoptosis index. Moreover, glutamine could clearly up-regulate the expression of Grp78, restrain P-PERK and P-elF2α, and reduce ATF4, Gadd153 and Caspase-12 expressions. Conclusion Glutamine may be involved in alleviating ER stress induced intestinal mucosa cells apoptosis.


Subject(s)
Animals , Male , Reperfusion Injury/prevention & control , Apoptosis/drug effects , Protective Agents/pharmacology , Endoplasmic Reticulum Stress/drug effects , Glutamine/pharmacology , Intestinal Mucosa/drug effects , RNA, Messenger/drug effects , Rats, Sprague-Dawley , Mesenteric Artery, Superior/injuries , eIF-2 Kinase/drug effects , Models, Animal , Activating Transcription Factor 4/drug effects , Transcription Factor CHOP/drug effects , Caspase 12/drug effects , Heat-Shock Proteins/drug effects , Intestinal Mucosa , Intestinal Mucosa/ultrastructure
6.
Apoptosis ; 24(9-10): 798-811, 2019 10.
Article in English | MEDLINE | ID: mdl-31321634

ABSTRACT

Sirtuins have emerged as a promising novel class of anti-cancer drug targets. Inhibition of SIRT1 and SIRT2 induces apoptosis in cancer cells and they play multifaceted roles in regulating autophagy. In the present study, we found that salermide, a SIRT1/2-specific inhibitor or small interfering RNAs (siRNAs) to block SIRT1/2 expression could induce autophagy in human NSCLC cells. Moreover, SIRT1/2 inhibition increased the expression levels of ATF4 and DDIT4 and downregulated p-RPS6KB1 and p-EIF4EBP1, two downstream molecules of mTORC1. Moreover, ATF4 or DDIT4 knockdown attenuated salermide-induced autophagy, suggesting that SIRT1/2 inhibition induced autophagy through the ATF4-DDIT4-mTORC1 axis. Mechanistically, SIRT1/2 inhibition led to HSPA5 acetylation and dissociation from EIF2AK3, leading to ER stress response and followed by upregulation of ATF4 and DDIT4, triggering autophagy. Silencing of the autophagic gene ATG5 in lung cancer cells resulted in increased apoptotic cell death induced by SIRT1/2 inhibition. Our data show that inhibition of SIRT1/2 induces pro-survival autophagy via acetylation of HSPA5 and subsequent activation of ATF4 and DDIT4 to inhibit the mTOR signaling pathway in NSCLC cells. These findings suggest that combinatorial treatment with SIRT1/2 inhibitors and pharmacological autophagy inhibitors is an effective therapeutic strategy for cancer therapy.


Subject(s)
Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Mechanistic Target of Rapamycin Complex 1/drug effects , Naphthols/pharmacology , Phenylpropionates/pharmacology , Sirtuins/genetics , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Gene Expression/drug effects , HEK293 Cells , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , RNA Interference , Signal Transduction , Sirtuin 1/drug effects , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuin 2/drug effects , Sirtuin 2/genetics , Sirtuin 2/metabolism , Sirtuins/drug effects , Sirtuins/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism
7.
Dig Dis Sci ; 64(12): 3630-3641, 2019 12.
Article in English | MEDLINE | ID: mdl-31280390

ABSTRACT

AIM: Renal toxicity of adefovir disoproxil (ADV) and tenofovir disoproxil fumarate (TDF) is a significant concern in chronic hepatitis B (CHB) patients. Early observational clinical data suggested that telbivudine (LdT) might have renoprotective effects. METHODS: In this prospective study, consecutive CHB patients on combined lamivudine (LAM) + ADV/TDF were switched to LdT + ADV/TDF at recruitment and were followed up for 24 months. Estimated glomerular filtration rate (eGFR) was calculated with the modification of diet in renal disease equation. The effects of LdT on cell viability and expression of kidney injury or apoptotic biomarkers were investigated in cultured renal tubular epithelial cell line HK-2. RESULTS: Thirty-one patients (median age 55 years, 90.3% male) were recruited (54.8% TDF: 45.2% ADV). Serum HBV DNA was undetectable at all time points. Median eGFR was 70.2 (IQR 62.6-77.9) and 81.5 (IQR 63.6-99.1) mL/min/1.73 m2 at baseline and 24 months, respectively (p < 0.001). Downstaging of chronic kidney disease was observed in eight (25.8%) patients and was more common in ADV-treated compared to TDF-treated patients (7/8 vs. 1/17, p = 0.011; OR 16, 95% CI 1.643-155.766, p = 0.017). In vitro data showed that adding LdT to ADV or TDF was associated with improved cell viability and lower expression of injury and apoptotic biomarkers compared with ADV or TDF alone. Treatment was prematurely discontinued in four(12.9%) patients due to myalgia. CONCLUSIONS: Clinical and in vitro data suggest that LdT has renoprotective effects in patients on long-term ADV/TDF treatment. LdT may be considered as an adjuvant therapy in this special group of patients with renal impairment (NCT03778567).


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/therapeutic use , Glomerular Filtration Rate , Hepatitis B, Chronic/drug therapy , Organophosphonates/adverse effects , Renal Insufficiency, Chronic/metabolism , Telbivudine/therapeutic use , Tenofovir/adverse effects , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/genetics , Adenine/adverse effects , Adenine/pharmacology , Antiviral Agents/pharmacology , Apoptosis/drug effects , Caspase 12/drug effects , Caspase 12/genetics , Cell Line , Cell Survival/drug effects , Endoplasmic Reticulum Chaperone BiP , Epithelial Cells , Female , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/genetics , Hepatitis A Virus Cellular Receptor 1/drug effects , Hepatitis A Virus Cellular Receptor 1/genetics , Hepatitis B, Chronic/complications , Humans , In Vitro Techniques , Interleukin-18/genetics , Kidney Tubules , Lamivudine/pharmacology , Lipocalin-2/drug effects , Lipocalin-2/genetics , Male , Middle Aged , Organophosphonates/pharmacology , Prospective Studies , Protective Agents , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/complications , Severity of Illness Index , Tenofovir/pharmacology
8.
Am J Physiol Endocrinol Metab ; 316(6): E1081-E1092, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30964708

ABSTRACT

Musclin is a muscle-secreted cytokine that disrupts glucose uptake and glycogen synthesis in type 2 diabetes. The purpose of this study was to investigate the mechanisms responsible for the regulation of musclin gene expression in response to treatment with palmitate. RNA sequencing results showed that biological processes activated by palmitate are mainly enriched in endoplasmic reticulum (ER) stress. The protein kinase RNA-like ER kinase (PERK) signaling pathway is involved in the regulation of musclin expression induced by palmitate. Chromatin immunoprecipitation data showed that activating transcription factor 4 (ATF4)-downstream of PERK-bound to the promoter of the C/EBPß gene. Notably, C/EBPß also contains a binding site in the region -94~-52 of the musclin gene promoter. Knockdown or knockout of PERK and ATF4 using short hairpin RNA or CRISPR-Cas9 decreased the expression of C/EBPß and musclin induced by palmitate. Furthermore, knockdown and knockout of C/EBPß alleviated the high expression of musclin in response to treatment with palmitate. Moreover, CRISPR-Cas9 knockout of the region -94~-52 in which C/EBPß binds to the promoter of musclin abrogated the induction of high musclin expression caused by palmitate. Collectively, these findings suggest that treatment with palmitate activates the PERK/ATF4 signaling pathway, which in turn increases the expression of C/EBPß. C/EBPß binds directly to the promoter of the musclin gene and upregulates its expression.


Subject(s)
Activating Transcription Factor 4/drug effects , CCAAT-Enhancer-Binding Protein-beta/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/drug effects , Palmitates/pharmacology , Transcription Factors/drug effects , eIF-2 Kinase/drug effects , Activating Transcription Factor 4/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Gene Knockdown Techniques , Gene Knockout Techniques , Mice , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Promoter Regions, Genetic , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , eIF-2 Kinase/metabolism
9.
Exp Neurol ; 313: 16-25, 2019 03.
Article in English | MEDLINE | ID: mdl-30529503

ABSTRACT

Intracerebral hemorrhage (ICH) is a common and severe neurological disorder, which is associated with high rates of mortality and morbidity. This study aimed to evaluate whether general control non-derepressible-2 (GCN2) stimulation ameliorated neuroinflammation after ICH. Male CD-1 mice were subjected to experimental ICH by infusion of bacterial collagenase. Post-ictus assessment included neurobehavioral tests, brain edema measurement, quantification of neutrophil infiltration and microglia activation, and measurement of TNF-α and IL-1ß expression at 24h after ICH. Furthermore, we tested the long-term neurological improvement by GCN2 at 21 days after ICH. Our results showed that GCN2 improved neurological function and reduced brain edema at 24 and 72 h following experimental ICH in CD-1 mice in contrast to the vehicle administration alone. GCN2 was also found to decrease levels of IL-1ß and TNF-α, and inhibit neutrophil infiltration activation. In addititon, GCN2 also alleviated long-term neurological impairment after ICH. However, inhibition of eIF2α or ATF4 abolished the protective effects of GCN2, indicating eIF2α/ATF4 signaling pathway as the downstream mediator of GCN2.


Subject(s)
Activating Transcription Factor 4/drug effects , Anti-Inflammatory Agents/pharmacology , Cerebral Hemorrhage/complications , Eukaryotic Initiation Factor-2/drug effects , Inflammation/etiology , Inflammation/prevention & control , Protein Serine-Threonine Kinases/pharmacology , Signal Transduction/drug effects , Activating Transcription Factor 4/biosynthesis , Animals , Behavior, Animal , Brain Edema/etiology , Brain Edema/prevention & control , Cerebral Hemorrhage/psychology , Cytokines/biosynthesis , Eukaryotic Initiation Factor-2/biosynthesis , Inflammasomes/drug effects , Male , Maze Learning/drug effects , Mice , Neutrophil Infiltration/drug effects , Up-Regulation/drug effects
10.
J Nutr Sci Vitaminol (Tokyo) ; 64(3): 200-208, 2018.
Article in English | MEDLINE | ID: mdl-29962431

ABSTRACT

Fibroblast growth factor 21 (FGF21), mainly synthesized and secreted from the liver, is an endocrine FGF that regulates glucose and fatty acid metabolism to maintain whole body energy homeostasis. Gene expression of FGF21 was previously reported to be induced by endoplasmic reticulum (ER) stress through activating transcription factor 4 (ATF4). It has been reported that drug-induced ER stress is reduced by overexpression of FGF21. However, the function of endogenous FGF21 under physiological conditions such as the postprandial state remains unknown. Here, we examined the effects of both endogenous and exogenous FGF21 on postprandial hepatic ER stress. In mice, postprandial and tunicamycin-induced ER stress was significantly reduced by overexpression of FGF21 using a recombinant adenovirus. FGF21-deficient mice exhibited a more considerable increase in drug-induced ER stress target gene expression than wild-type mice. Following refeeding after fasting, FGF21 deficiency caused severe ER stress in the liver. The postprandial ER stress response was significantly reduced when hepatic FGF21 gene expression was increased by feeding a diet containing the soy protein ß-conglycinin which activates ATF4. Together, these results demonstrate that FGF21 reduces the increased expression of a subset of genes in the liver in response to ER stress and may correct metabolic disorders caused by ER stress.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Fibroblast Growth Factors/pharmacology , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/physiology , Animals , Antigens, Plant/pharmacology , Endoplasmic Reticulum Stress/genetics , Fasting , Fibroblast Growth Factors/deficiency , Fibroblast Growth Factors/genetics , Gene Expression/drug effects , Globulins/pharmacology , Liver/metabolism , Liver/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Postprandial Period , Seed Storage Proteins/pharmacology , Soybean Proteins/pharmacology , Tunicamycin/pharmacology
11.
Pharmazie ; 73(3): 156-160, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29544563

ABSTRACT

Previous studies have shown sorafenib to function as a multitargeted tyrosine kinase inhibitor in different tumors. However, whether sorafenib improves renal cell carcinoma (RCC) through activating transcription factor 4 (ATF4) has never been explored. In the current study, we showed that sorafenib could suppress RCC cell viability in a time- and dose-dependent manner. Furthermore, sorafenib is demonstrated to enhance the mRNA and protein levels of ATF4. Meanwhile, overexpression of ATF4 was demonstrated to induce ACHN cell cycle arrest and cell apoptosis. Moreover, treatment with sorafenib could enhance the expression of CCAAT/enhancer-binding protein-homologous protein (CHOP) and p53 upregulated modulator of apoptosis (PUMA), thereby leading to ACHN cell apoptosis. More importantly, silencing of ATF4 could largely abolish sorafenib-induced upregulation of CHOP and PUMA in ACHN cells. Meanwhile, sorafenib-induced cell apoptosis may be dependent on the activation of ATF4 since knockdown of ATF4 partially reversed sorafenib-induced ACHN cell apoptosis. In summary, the present study demonstrates that sorafenib activates ATF4-CHOP-PUMA pathway in RCC cells, resulting in enhanced ER stress-related cell apoptosis.


Subject(s)
Activating Transcription Factor 4/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Sorafenib/pharmacology , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Humans , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/drug effects , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/drug effects , Transfection , Up-Regulation/drug effects
12.
J Asthma ; 54(2): 125-133, 2017 03.
Article in English | MEDLINE | ID: mdl-27383524

ABSTRACT

INTRODUCTION: Endoplasmic reticulum (ER) stress has been considered to be an important regulator of airway inflammation in the pathogenesis of bronchial asthma, but the mechanism of ER stress involved in neutrophilic asthma remain not fully understood. METHODS: Tunicamycin is a mixture of homologous nucleoside antibiotics, which is used to induce ER stress. In the present study, Tunicamycin was administered to mouse bronchial epithelial cells and a neutrophilic asthma model (OVALPS-OVA mice), and ER stress indicators and inflammatory cytokines were measured by Western blotting and Elisa. RESULTS: Tunicamycin not only induced ER stress in mouse bronchial epithelial cells, but also increased expression of inflammation indicators such as IL-6, IL-8, and TNF-α via PERK-ATF4-CHOP signaling. Additionally, the phosphorylation of PERK and the expression levels of ATF4 and CHOP proteins and inflammatory cytokines (IL-6, IL-8 and TNF-α) were elevated in the lung tissue of OVALPS-OVA mice. Administering tunicamycin further increased protein expression levels of ER stress indicators and inflammatory cytokines, and resulted in more severe asthma phenotypes in OVALPS-OVA mice, suggesting that PERK-ATF4-CHOP signaling is associated with airway inflammation in neutrophil-dominant asthma. CONCLUSIONS: These data support the emerging notion that regulation of ER stress could be strongly associated with the development of neutrophilic asthma.


Subject(s)
Asthma/physiopathology , Endoplasmic Reticulum Stress/drug effects , Inflammation Mediators/metabolism , Tunicamycin/pharmacology , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/metabolism , Animals , Bronchi/drug effects , Bronchi/metabolism , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Mice , Signal Transduction/drug effects , Transcription Factor CHOP/drug effects , Transcription Factor CHOP/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism
13.
Am J Physiol Heart Circ Physiol ; 311(4): H871-H880, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27496879

ABSTRACT

We previously reported that endoplasmic reticulum (ER) stress is induced in the subfornical organ (SFO) and the hypothalamic paraventricular nucleus (PVN) of heart failure (HF) rats and is reduced by inhibition of mitogen-activated protein kinase (MAPK) signaling. The present study further examined the relationship between brain MAPK signaling, ER stress, and sympathetic excitation in HF. Sham-operated (Sham) and HF rats received a 4-wk intracerebroventricular (ICV) infusion of vehicle (Veh) or the ER stress inhibitor tauroursodeoxycholic acid (TUDCA, 10 µg/day). Lower mRNA levels of the ER stress biomarkers GRP78, ATF6, ATF4, and XBP-1s in the SFO and PVN of TUDCA-treated HF rats validated the efficacy of the TUDCA dose. The elevated levels of phosphorylated p44/42 and p38 MAPK in SFO and PVN of Veh-treated HF rats, compared with Sham rats, were significantly reduced in TUDCA-treated HF rats as shown by Western blot and immunofluorescent staining. Plasma norepinephrine levels were higher in Veh-treated HF rats, compared with Veh-treated Sham rats, and were significantly lower in the TUDCA-treated HF rats. TUDCA-treated HF rats also had lower mRNA levels for angiotensin converting enzyme, angiotensin II type 1 receptor, tumor necrosis factor-α, interleukin-1ß, cyclooxygenase-2, and NF-κB p65, and a higher mRNA level of IκB-α, in the SFO and PVN than Veh-treated HF rats. These data suggest that ER stress contributes to the augmented sympathetic activity in HF by inducing MAPK signaling, thereby promoting inflammation and renin-angiotensin system activity in key cardiovascular regulatory regions of the brain.


Subject(s)
Brain/metabolism , Endoplasmic Reticulum Stress , Heart Failure/metabolism , Inflammation/metabolism , Mitogen-Activated Protein Kinases/metabolism , Renin-Angiotensin System , Sympathetic Nervous System/metabolism , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/genetics , Activating Transcription Factor 6/drug effects , Activating Transcription Factor 6/genetics , Animals , Blotting, Western , Brain/drug effects , Cholagogues and Choleretics/pharmacology , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Echocardiography , Heart Failure/physiopathology , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/genetics , Infusions, Intraventricular , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Male , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/drug effects , NF-KappaB Inhibitor alpha/drug effects , NF-KappaB Inhibitor alpha/genetics , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Peptidyl-Dipeptidase A/drug effects , Peptidyl-Dipeptidase A/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/genetics , Signal Transduction , Subfornical Organ/drug effects , Subfornical Organ/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology , Taurochenodeoxycholic Acid/pharmacology , Transcription Factor RelA/drug effects , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , X-Box Binding Protein 1/drug effects , X-Box Binding Protein 1/genetics , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
14.
BMC Gastroenterol ; 16: 25, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26917416

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of most common and aggressive human malignancies in the world, especially, in eastern Asia, and its mortality is very high at any phase. We want to investigate mechanism of niclosamide inducing cell apoptosis in HCC. METHODS: Two hepatoma cell lines were used to evaluate activity of niclosamide inducing cell apoptosis and study its mechanism. Quantitative real-time PCR and western blotting were used in analysis of genes expression or protein active regulated by niclosamide. RESULTS: Niclosamide remarkably induced cell apoptosis in hepatoma cells. Furthermore, our study revealed that RNA-dependent protein kinase-like kinase (PERK) is activated and its expression is up-regulated in HCC cells which are exposed to niclosamide. niclosamide also significantly increase activating transcription factor 3 (ATF3), activating transcription factor 4 (ATF4) and CCAAT/enhancer-binding protein-homologous protein (CHOP) expression in HCC cells. It's suggested that the function of niclosamide was abrogated by PERK inhibitor or absent ATF3. Expression of PERK and CHOP is correlated with ATF3 level in the cells. CONCLUSION: Taken together, our results indicate that ATF3 plays an integral role in ER stress activated and cell apoptosis induced by niclosamide in HCC cells. In this study, the new mechanism of niclosamide as anti-cancer we investigated, too.


Subject(s)
Activating Transcription Factor 3/drug effects , Anthelmintics/pharmacology , Apoptosis/drug effects , Niclosamide/pharmacology , RNA, Messenger/drug effects , eIF-2 Kinase/drug effects , Activating Transcription Factor 3/genetics , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/genetics , Carcinoma, Hepatocellular , Cell Line, Tumor , Cell Survival/drug effects , Flow Cytometry , Gene Knockout Techniques , Hep G2 Cells , Humans , In Situ Nick-End Labeling , Liver Neoplasms , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Transcription Factor CHOP/drug effects , Transcription Factor CHOP/genetics , Transcriptional Activation/drug effects , Up-Regulation/drug effects , eIF-2 Kinase/metabolism
15.
Chin J Nat Med ; 13(4): 250-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25908621

ABSTRACT

Serum palmitic acid (PA), a type of saturated fatty acid, causes lipid accumulation and induces toxicity in hepatocytes. Ethanol (EtOH) is metabolized by the liver and induces hepatic injury and inflammation. Herein, we analyzed the effects of EtOH on PA-induced lipotoxicity in the liver. Our results indicated that EtOH aggravated PA-induced apoptosis and lipid accumulation in primary rat hepatocytes in dose-dependent manner. EtOH intensified PA-caused endoplasmic reticulum (ER) stress response in vitro and in vivo, and the expressions of CHOP, ATF4, and XBP-1 in nucleus were significantly increased. EtOH also increased PA-caused cleaved caspase-3 in cytoplasm. In wild type and CHOP(-/-) mice treated with EtOH and high fat diet (HFD), EtOH worsened the HFD-induced liver injury and dyslipidemia, while CHOP knockout blocked toxic effects of EtOH and PA. Our study suggested that targeting UPR-signaling pathways is a promising, novel approach to reducing EtOH and saturated fatty acid-induced metabolic complications.


Subject(s)
Chemical and Drug Induced Liver Injury , Endoplasmic Reticulum Stress/drug effects , Ethanol/toxicity , Fatty Liver/metabolism , Palmitic Acid/toxicity , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/metabolism , Animals , Apoptosis/drug effects , Caspase 3/drug effects , Chemical and Drug Induced Liver Injury/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Diet, High-Fat/adverse effects , Dose-Response Relationship, Drug , Dyslipidemias/chemically induced , Dyslipidemias/metabolism , Ethanol/metabolism , Fatty Liver/chemically induced , Gene Knockout Techniques , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Metabolism/drug effects , Liver/metabolism , Male , Mice , Rats , Rats, Sprague-Dawley , Regulatory Factor X Transcription Factors , Signal Transduction/drug effects , Transcription Factor CHOP/drug effects , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism , Unfolded Protein Response/drug effects , X-Box Binding Protein 1
16.
Auris Nasus Larynx ; 41(1): 81-3, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23880367

ABSTRACT

Arsenic trioxide (As2O3) has been used in the treatment of acute promyelocytic leukemia (APL) and many malignant solid tumors. Recently, endoplasmic reticulum (ER) stress plays an important role in As2O3-treated laryngeal squamous cell line Hep-2 cells. In the present work, the expression of ER stress-related proteins was investigated in As2O3-treated Hep-2 cells. The results showed that As2O3 increased the expression of GRP78, CHOP, phosphorylated eIF2α and ATF4, all of which are the molecule of ER stress. Therefore, As2O3 induced ER stress in Hep-2 cells.


Subject(s)
Activating Transcription Factor 4/drug effects , Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/drug effects , Heat-Shock Proteins/drug effects , Oxides/pharmacology , Transcription Factor CHOP/drug effects , Activating Transcription Factor 4/metabolism , Arsenic Trioxide , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/physiology , Eukaryotic Initiation Factor-2/metabolism , Head and Neck Neoplasms/metabolism , Heat-Shock Proteins/metabolism , Humans , Laryngeal Neoplasms/metabolism , Squamous Cell Carcinoma of Head and Neck
17.
J Natl Cancer Inst ; 105(19): 1463-73, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24052624

ABSTRACT

BACKGROUND: L-type amino acid transporters (LATs) uptake neutral amino acids including L-leucine into cells, stimulating mammalian target of rapamycin complex 1 signaling and protein synthesis. LAT1 and LAT3 are overexpressed at different stages of prostate cancer, and they are responsible for increasing nutrients and stimulating cell growth. METHODS: We examined LAT3 protein expression in human prostate cancer tissue microarrays. LAT function was inhibited using a leucine analog (BCH) in androgen-dependent and -independent environments, with gene expression analyzed by microarray. A PC-3 xenograft mouse model was used to study the effects of inhibiting LAT1 and LAT3 expression. Results were analyzed with the Mann-Whitney U or Fisher exact tests. All statistical tests were two-sided. RESULTS: LAT3 protein was expressed at all stages of prostate cancer, with a statistically significant decrease in expression after 4-7 months of neoadjuvant hormone therapy (4-7 month mean = 1.571; 95% confidence interval = 1.155 to 1.987 vs 0 month = 2.098; 95% confidence interval = 1.962 to 2.235; P = .0187). Inhibition of LAT function led to activating transcription factor 4-mediated upregulation of amino acid transporters including ASCT1, ASCT2, and 4F2hc, all of which were also regulated via the androgen receptor. LAT inhibition suppressed M-phase cell cycle genes regulated by E2F family transcription factors including critical castration-resistant prostate cancer regulatory genes UBE2C, CDC20, and CDK1. In silico analysis of BCH-downregulated genes showed that 90.9% are statistically significantly upregulated in metastatic castration-resistant prostate cancer. Finally, LAT1 or LAT3 knockdown in xenografts inhibited tumor growth, cell cycle progression, and spontaneous metastasis in vivo. CONCLUSION: Inhibition of LAT transporters may provide a novel therapeutic target in metastatic castration-resistant prostate cancer, via suppression of mammalian target of rapamycin complex 1 activity and M-phase cell cycle genes.


Subject(s)
Activating Transcription Factor 4/metabolism , Amino Acid Transport Systems, Basic/antagonists & inhibitors , Amino Acid Transport Systems, Basic/metabolism , Amino Acids/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Leucine/antagonists & inhibitors , Neoadjuvant Therapy/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/physiopathology , Receptors, Androgen/metabolism , Activating Transcription Factor 4/drug effects , Amino Acid Transport Systems, Basic/genetics , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Biological Transport/drug effects , Cell Cycle/drug effects , Computer Simulation , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Leucine/metabolism , Luminescent Measurements , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/physiopathology , Orchiectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Array Analysis , Receptors, Androgen/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
18.
Planta Med ; 77(2): 141-5, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20717870

ABSTRACT

The involvement of unfolded protein response (UPR) activation in tumor survival and resistance to chemotherapies suggests a new anticancer strategy targeting UPR pathway. Arctigenin, a natural product, has been recently identified for its antitumor activity with selective toxicity against cancer cells under glucose starvation with unknown mechanism. Here we found that arctigenin specifically blocks the transcriptional induction of two potential anticancer targets, namely glucose-regulated protein-78 (GRP78) and its analog GRP94, under glucose deprivation, but not by tunicamycin. The activation of other UPR pathways, e.g., XBP-1 and ATF4, by glucose deprivation was also suppressed by arctigenin. A further transgene experiment showed that ectopic expression of GRP78 at least partially rescued arctigenin/glucose starvation-mediated cell growth inhibition, suggesting the causal role of UPR suppression in arctigenin-mediated cytotoxicity under glucose starvation. These observations bring a new insight into the mechanism of action of arctigenin and may lead to the design of new anticancer therapeutics.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Furans/pharmacology , Glucose/metabolism , Lignans/pharmacology , Transcriptional Activation/drug effects , Unfolded Protein Response/drug effects , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/genetics , Animals , Cell Line, Tumor , Cell Survival/drug effects , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Neoplastic/drug effects , Glucose/pharmacology , HSP70 Heat-Shock Proteins/drug effects , HSP70 Heat-Shock Proteins/genetics , Humans , Membrane Proteins/drug effects , Membrane Proteins/genetics , Regulatory Factor X Transcription Factors , Time Factors , Transcription Factors/drug effects , Transcription Factors/genetics , Unfolded Protein Response/genetics , X-Box Binding Protein 1
19.
Cancer Res ; 69(10): 4415-23, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19417138

ABSTRACT

The ubiquitin-proteasome system plays a key regulatory role in cellular homeostasis. The inhibition of the 26S proteasome by Bortezomib leads to the accumulation of misfolded proteins, resulting in endoplasmic reticulum stress followed by a coordinated cellular response called unfolded protein response (UPR). Endoplasmic reticulum stress is also a potent inducer of macroautophagy. Bortezomib is a selective and potent inhibitor of the 26S proteasome and is approved for the treatment of multiple myeloma. Clinical trials with Bortezomib have shown promising results for some types of cancers, but not for some others, including those of the breast. In this study, we show that Bortezomib induces the UPR and autophagy in MCF7 breast cancer cells. Surprisingly, Bortezomib did not induce phosphorylation of PERK, a key initial step of the UPR. We show that induction of autophagy by Bortezomib is dependent on the proteasomal stabilisation of ATF4 and up-regulation of LC3B by ATF4. We show that ATF4 and LC3B play a critical role in activating autophagy and protecting cells from Bortezomib-induced cell death. Our experiments also reveal that HDAC6 knockdown results in decreased LC3B protein and reduced autophagy. Our work shows that the induction of autophagy through ATF4 may be an important resistance mechanism to Bortezomib treatment in breast cancer, and targeting autophagy may represent a novel approach to sensitize breast cancers to Bortezomib.


Subject(s)
Activating Transcription Factor 4/metabolism , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Boronic Acids/therapeutic use , Pyrazines/therapeutic use , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/genetics , Bortezomib , Breast Neoplasms/drug therapy , Cell Division/drug effects , Cell Line, Tumor , Female , Flow Cytometry , Humans , Immunohistochemistry , Neoplasm Proteins/drug effects , Neoplasm Proteins/metabolism , Polymerase Chain Reaction , RNA, Neoplasm/drug effects , RNA, Neoplasm/genetics , RNA, Small Interfering/drug effects , RNA, Small Interfering/genetics
20.
Cancer Res ; 69(10): 4225-34, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19435925

ABSTRACT

Glucose deprivation, a cell condition that occurs in solid tumors, activates the unfolded protein response (UPR). A key feature of the UPR is the transcription program activation, which allows the cell to survive under stress conditions. Here, we show that the UPR transcription program is disrupted by the antidiabetic biguanides metformin, buformin, and phenformin depending on cellular glucose availability. These drugs inhibit production of the UPR transcription activators XBP1 and ATF4 and induce massive cell death during glucose deprivation as did the antitumor macrocyclic compound versipelostatin. Gene expression profiling shows remarkable similarity in the modes of action of biguanides and versipelostatin determined by the broad range of glucose deprivation-inducible genes. Importantly, during glucose deprivation, most of the biguanide suppression genes overlap with the genes induced by tunicamycin, a chemical UPR inducer. Gene expression profiling also identifies drug-driven signatures as a tool for discovering pharmacologic UPR modulators. Our findings show that disrupting the UPR during glucose deprivation could be an attractive approach for selective cancer cell killing and could provide a chemical genomic basis for developing UPR-targeting drugs against solid tumors.


Subject(s)
Activating Transcription Factor 4/drug effects , DNA-Binding Proteins/drug effects , Gene Expression Profiling , Genomics , Glucose/deficiency , Hypoglycemic Agents/pharmacology , Macrolides/pharmacology , Neoplasms/genetics , Oligosaccharides/pharmacology , Protein Denaturation/genetics , Transcription Factors/drug effects , Cell Death , Cell Survival/drug effects , Genes, Reporter , Humans , Neoplasms/drug therapy , Phenformin/pharmacology , Plasmids , Protein Folding/drug effects , Regulatory Factor X Transcription Factors , Transfection , X-Box Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...