Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Ecotoxicol Environ Saf ; 274: 116193, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38460407

ABSTRACT

Chlorocholine chloride (CCC) is a plant growth regulator used worldwide that is detectable in cereals, fruits and animal products. The health effects of CCC exposure have raised public concern. Our previous research showed that CCC exposure decreased testosterone synthesis in pubertal rats. However, little is known about whether and how pubertal CCC exposure impacts spermatogenesis. In this study, we used BALB/c mice and spermatogonia-derived GC-1 cells to examine CCC-induced spermatogenic dysfunction. In vivo, pubertal CCC exposure led to decreased testicular weight, decreased testicular germ cells and poor sperm quality. This effect worsened after cessation of CCC exposure for the next 30 days. RNA-seq and western blot analysis revealed that CCC induced aryl hydrocarbon receptor (AhR) signaling, endoplasmic reticulum stress (ERS) and ferritinophagy. Increased iron content and lipid peroxidation levels were also observed in CCC-treated testes. In vitro, it was identified that iron overload mediated by enhanced ferritinophagy occurred in CCC-treated GC-1 cells, which might be attributed to the PERK pathway in ERS. Further, for the first time, our study elucidated the involvement of AhR in CCC-induced iron overload, which aggravated testicular oxidative damage via lipid peroxidation. Considering the adverse impact of CCC exposure on rodents, supportive evidence from GC-1 cells, and the critical importance of spermatogenesis on male development, the effects of CCC on the male reproduction warrant increased attention.


Subject(s)
Acetates , Chlormequat , Iron Overload , Phenols , Spermatogenesis , Animals , Male , Mice , Rats , Chlormequat/metabolism , Chlormequat/toxicity , Iron Overload/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Seeds , Spermatogenesis/drug effects , Testis , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism
2.
Dis Markers ; 2022: 9340353, 2022.
Article in English | MEDLINE | ID: mdl-36523813

ABSTRACT

Mycoplasma pneumoniae pneumonia (MPP) is usually found in school-aged children and relapses easily because of antibiotic resistance. The Qingfei Tongluo formula (QTF) is a clinically used traditional Chinese medicine to treat MPP. Our previous study demonstrated that QTF exhibited ameliorative effects on the experimental MPP mice model. In this study, the function and underlying QTF mechanism in MPP was attempted to be further explored. Mycoplasma pneumoniae (MP) was applied to infect A549 cells and BALB/c mice to mimic MPP in vitro and in vivo. Cytokine release and reactive oxygen species (ROS) production were analyzed using enzyme-linked immunosorbent assay (ELISA) assay and flow cytometry. Western blot analysis was used to detect the protein involved in ER stress. MP infection was found to enhance cytokine release and ER stress in vitro and in vivo, and this effect could be alleviated by QTF. Moreover, protein kinase RNA-like endoplasmic reticulum kinase (PERK) knockdown alleviated MP infection-induced cytokine release, ROS production, and ER stress in A549 cells while the PERK overexpression exhibited the opposite effects. In conclusion, QTF alleviated MP infection-induced cytokine release, ROS production, and ER stress via PERK signaling pathway inhibition.


Subject(s)
Drugs, Chinese Herbal , Pneumonia, Mycoplasma , eIF-2 Kinase , Animals , Mice , Cytokines , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism , Endoplasmic Reticulum/metabolism , Mice, Inbred BALB C , Pneumonia, Mycoplasma/drug therapy , Pneumonia, Mycoplasma/metabolism , Protein Kinases , Reactive Oxygen Species , Signal Transduction
3.
Acta Pharmacol Sin ; 43(3): 712-723, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33980998

ABSTRACT

Previous reports suggested that cinnamaldehyde (CA), the bioactive ingredient in Cinnamomum cassia, can suppress tumor growth, migratory, and invasive abilities. However, the role and molecular mechanisms of CA in GC are not completely understood. In the present study, we found that CA-induced ER stress and cell death via the PERK-CHOP axis and Ca2+ release in GC cells. Inhibition of ER stress using specific-siRNA blocked CA-induced cell death. Interestingly, CA treatment resulted in autophagic cell death by inducing Beclin-1, ATG5, and LC3B expression and by inhibiting p62 expression whereas autophagy inhibition suppressed CA-induced cell death. We showed that CA induces the inhibition of G9a and the activation of LC3B. Moreover, CA inhibited G9a binding on Beclin-1 and LC3B promoter. Overall, these results suggested that CA regulates the PERK-CHOP signaling, and G9a inhibition activates autophagic cell death via ER stress in GC cells.


Subject(s)
Acrolein/analogs & derivatives , Autophagic Cell Death/drug effects , Endoplasmic Reticulum Stress/drug effects , Epigenesis, Genetic/drug effects , Stomach Neoplasms/pathology , Acrolein/pharmacology , Autophagy-Related Protein 5/drug effects , Beclin-1/drug effects , Calcium/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Microtubule-Associated Proteins/drug effects , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Transcription Factor CHOP/drug effects , eIF-2 Kinase/drug effects
4.
Aging (Albany NY) ; 13(3): 3405-3427, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33495405

ABSTRACT

Cerebral ischemia/reperfusion (IR) after ischemic stroke causes deleterious microglial activation. Protein tyrosine phosphatase 1B (PTP1B) exacerbates neuroinflammation, yet the effect of the inhibition on microglial activation and cerebral IR injury is unknown. A cerebral IR rat model was induced by middle cerebral artery occlusion (MCAO) and reperfusion. The PTP1B inhibitor, sc-222227, was administered intracerebroventricularly. Neurologic deficits, infarct volume, and brain water content were examined. An in vitro oxygen glucose deprivation/reoxygenation (OGD/R) model was established in primary microglia and BV-2 cells. Microglial activation/polarization, endoplasmic reticulum (ER) stress, autophagy, and apoptosis were detected using western blot, immunohistology, ELISA, and real-time PCR. Protein interaction was assessed by a proximity ligation assay. The results showed a significant increase in microglial PTP1B expression after IR injury. Sc-222227 attenuated IR-induced microglial activation, ER stress, and autophagy and promoted M2 polarization. Upon OGD/R, sc-222227 mitigated microglial activation by inhibiting ER stress-dependent autophagy, the effect of which was abolished by PERK activation, and PERK inhibition attenuated microglial activation. The PTP1B-phosphorylated PERK protein interaction was significantly increased after OGD/R, but decreased upon sc-222227 treatment. Finally, sc-222227 mitigated neuronal damage and neurologic deficits after IR injury. Treatment targeting microglial PTP1B might be a potential therapeutic strategy for ischemic stroke treatment.


Subject(s)
Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/pharmacology , Ischemic Stroke/metabolism , Microglia/drug effects , Neurons/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Reperfusion Injury/metabolism , eIF-2 Kinase/drug effects , Animals , Apoptosis/drug effects , Cytokines/drug effects , Cytokines/genetics , In Vitro Techniques , Inflammation , Injections, Intraventricular , Ischemic Stroke/immunology , Mice , Microglia/immunology , Neurons/metabolism , Primary Cell Culture , RNA, Messenger/metabolism , Rats , Reperfusion Injury/immunology , eIF-2 Kinase/metabolism
5.
Addict Biol ; 26(2): e12905, 2021 03.
Article in English | MEDLINE | ID: mdl-32293776

ABSTRACT

Micro-RNAs (miRNAs) are highly evolutionarily conserved short-length/noncoding RNA molecules that modulate a wide range of cellular functions in many cell types by regulating the expression of a variety of targeted genes. miRNAs have also recently emerged as key regulators of neuronal genes mediating the effects of psychostimulant drugs and memory-related neuroplasticity processes. Smoking is a predominant addictive behaviour associated with millions of deaths worldwide, and nicotine is a potent natural psychoactive agonist of cholinergic receptors, highly abundant in cigarettes. The influence of miRNAs modulation on cholinergic signalling in the nervous system remains however poorly explored. Using miRNA knockout mice and biochemical, electrophysiological and pharmacological approaches, we examined the effects of miR-132/212 gene disruption on the levels of hippocampal nicotinic acetylcholine receptors, total ERK and phosphorylated ERK (pERK) and MeCP2 protein levels, and studied the impact of nicotine stimulation on hippocampal synaptic transmission and synaptic depression and strengthening. miR-132/212 deletion significantly altered α7-nAChR and pERK protein levels, but not total ERK or MeCP2, and resulted in both exacerbated synaptic depression and virtually abolished memory-related synaptic strengthening upon nicotine stimulation. These observations reveal a functional miRNAs/nicotinergic signalling interplay critical for nicotinic-receptor expression and neuroplasticity in brain structures relevant for drug addiction and learning and memory functions.


Subject(s)
Dentate Gyrus/drug effects , MicroRNAs/drug effects , Neuronal Plasticity/drug effects , Nicotine/pharmacology , Animals , Extracellular Signal-Regulated MAP Kinases/drug effects , Hippocampus/drug effects , Male , Methyl-CpG-Binding Protein 2/drug effects , Mice , Mice, Knockout , Receptors, Nicotinic/drug effects , Synaptic Transmission/drug effects , eIF-2 Kinase/drug effects
6.
Mol Med Rep ; 22(4): 2887-2895, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32700746

ABSTRACT

Prostaglandin E2 (PGE2) is involved in numerous physiological and pathological processes of the kidney via its four receptors. A previous study has suggested that a defect in the PGE2 receptor 1 (EP1) gene markedly suppressed the transforming growth factor­ß1 (TGF­ß1)­induced mesangial cell (MC) proliferation and extracellular matrix aggregation. Therefore, the present study aimed to adopt a pharmacological method of specifically suppressing or activating the EP1 receptor to further verify and demonstrate these results. The EP1 receptor antagonist SC­19220 and EP1 receptor agonist 17­phenyl­trinor­PGE2 ethyl amide (17­pt­PGE2) were selectively used to treat five­sixths nephrectomy renal fibrosis model mice and TGF­ß1­stimulated MCs. An Alpha screen PGE2 assay kit, flow cytometry, western blotting and immunohistochemical techniques were adopted to perform in vivo and in vitro experiments. The present results suggested that compared with the control group, the selective EP1 receptor antagonist SC­19220 improved renal function, markedly reduced the plasma blood urea nitrogen and creatinine levels (P<0.05) and alleviated glomerulosclerosis (P<0.05). By contrast, the EP1 receptor agonist 17­pt­PGE2 aggravated renal dysfunction and glomerulosclerosis (P<0.05). To verify the renal protection mechanisms mediated by suppression of the EP1 receptor, the expression levels of endoplasmic reticulum stress (ERS)­related proteins, including chaperone glucose­regulated protein 78 (GRP78), transient receptor potential channel 1 (TRPC1) and protein kinase R­like endoplasmic reticulum kinase (PERK), were further evaluated histologically. The expression of GRP78, TRPC1 and PERK in the antagonist treatment group were markedly downregulated (P<0.05), whereas those in the agonist treatment group were upregulated (P<0.05). The present in vitro experiments demonstrated that, compared with the control group, the EP1 receptor antagonist suppressed the expression of GRP78, TRPC1 and PERK (P<0.05), reduced the production of PGE2 (P<0.05) and decreased the MC apoptosis rate (P<0.05), thus alleviating TGF­ß1­stimulated MC injury. Consequently, consistent with previous results, selectively antagonizing the EP1 receptor improved renal function and mitigated glomerulosclerosis, and its potential mechanism might be associated with the suppression of ERS.


Subject(s)
Dinoprostone/metabolism , Glomerulonephritis/drug therapy , Receptors, Prostaglandin E, EP1 Subtype/agonists , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cells, Cultured , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Glomerulonephritis/etiology , Glomerulonephritis/physiopathology , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , Mice, Inbred C57BL , Nephrectomy/adverse effects , Prostaglandin Antagonists/pharmacology , TRPC Cation Channels/drug effects , TRPC Cation Channels/metabolism , Transforming Growth Factor beta1/toxicity , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism
8.
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
9.
Endocr J ; 67(2): 185-200, 2020 Feb 28.
Article in English | MEDLINE | ID: mdl-31748431

ABSTRACT

Growth hormone (GH) has multiple physiological roles, acting on many organs. In order to investigate its roles in rat liver, we tried to identify novel genes whose transcription was regulated by GH. We identified X-box binding protein 1 (Xbp1) as a candidate gene. XBP1 is a key transcription factor activated in response to endoplasmic reticulum (ER) stress. The purpose of this study was to investigate the mode of action of GH on XBP1, including the relation with ER stress, sex-dependent expression of the mRNA, and the signaling pathway. Intravenous administration of GH rapidly and transiently increased Xbp1 mRNA in hypophysectomized rat livers. Neither phosphorylated inositol-requiring-1α (IRE1α) nor phosphorylated PKR-like ER kinase (PERK) increased, suggesting that Xbp1 expression is induced by an ER stress-independent mechanism. The active form of XBP1(S) protein was increased by GH administration and was followed by an increased ER-associated dnaJ protein 4 (ERdj4) mRNA level. XBP1(S) protein levels were predominantly identified in male rat livers with variations among individuals similar to those of phosphorylated signal transducer and activator of transcription 5B (STAT5B), suggesting that XBP1(S) protein levels are regulated by the sex-dependent secretary pattern of GH. The GH signaling pathway to induce Xbp1 mRNA was examined in rat hepatoma H4IIE cells. GH induced the phosphorylation of CCAAT/enhancer-binding protein ß (C/EBPß) following extracellular signal-regulated protein kinase (ERK) phosphorylation. Taken together, the results indicated that XBP1 is activated by GH in rat liver in a sexually dimorphic manner via ERK and C/EBPß pathway.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Growth Hormone/pharmacology , Liver/drug effects , MAP Kinase Signaling System , RNA, Messenger/drug effects , X-Box Binding Protein 1/drug effects , Animals , Cell Line, Tumor , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/drug effects , Endoribonucleases/metabolism , HSP40 Heat-Shock Proteins/drug effects , HSP40 Heat-Shock Proteins/genetics , Hypophysectomy , Liver/metabolism , Multienzyme Complexes/drug effects , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Rats , STAT5 Transcription Factor/drug effects , STAT5 Transcription Factor/metabolism , Sex Characteristics , Signal Transduction , X-Box Binding Protein 1/genetics , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism
10.
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
11.
Neurochem Int ; 131: 104581, 2019 12.
Article in English | MEDLINE | ID: mdl-31639405

ABSTRACT

The present study was conducted to investigate the effect of salubrinal on nitric oxide mediated endoplasmic reticulum stress signaling and neuronal apoptosis. Rotenone treatment to neuro2a cells caused significantly decreased cell viability, increased cytotoxicity, augmented nitrite levels, increased nitrotyrosine level and augmented level of key ER stress markers (GRP-78, GADD153 and caspase-12). These augmented levels of ER stress markers could be attenuated with pretreatment of nitric oxide synthase inhibitor-aminoguanidine as well as with salubrinal. The rotenone treatment to neuro2a cells also triggered the ER stress induced up regulation of various signaling factors of unfolded protein response involving pPERK, ATF4, p-IRE1α, XBP-1 and ATF-6. Pretreatment of salubrinal significantly attenuated the activation of transmembrane kinases (PERK and IRE1) and ATF6 and restored the rotenone induced altered level of other UPR related signaling factors. Rotenone induced dephosphorylation of eIF2α was also inhibited with salubrinal treatment. Biochemically rotenone treatment to neuro2a cells caused the reactive oxygen species generation, depleted mitochondrial membrane potential and increased intra cellular calcium level which was attenuated with salubrinal treatment. Rotenone treatment to neuro2a cells also caused neuronal apoptosis, DNA fragmentation and chromatin condensation which were attenuated with salubrinal treatment. In conclusion, the findings suggested that rotenone causes the augmented level of nitric oxide which contributes in ER stress and could be inhibited by both aminoguanidine and/or salubrinal treatment. Further, salubrinal treatment attenuates the nitric oxide induced ER stress axis PERK:IRE1α:ATF-6 and inhibits the DNA damage and neuronal apoptosis.


Subject(s)
Activating Transcription Factor 6/drug effects , Cinnamates/pharmacology , DNA Damage/drug effects , Endoribonucleases/drug effects , Neurons/drug effects , Nitric Oxide/physiology , Protein Serine-Threonine Kinases/drug effects , Signal Transduction/drug effects , Thiourea/analogs & derivatives , eIF-2 Kinase/drug effects , Animals , Calcium Signaling/drug effects , Cell Line , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Rotenone/pharmacology , Thiourea/pharmacology , Uncoupling Agents/pharmacology
12.
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
13.
Biol Trace Elem Res ; 188(1): 2-10, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30196486

ABSTRACT

Boron is abundant in vegetables, nuts, legumes, and fruit and intake is associated with reduced risk of cancer and DNA damage and increased antioxidant status. Blood boric acid (BA) levels are approximately 10 µM BA in men at the mean US boron intake. Treatment of DU-145 human prostate cancer cells with 10 µM BA stimulates phosphorylation of elongation initiation factor 2α (eIF2α) at Ser51 leading to activation of the eIF2α/ATF4 pathway which activates the DNA damage-inducible protein GADD34. In the present study, we used MEF WT and MEF PERK (±) cells to test the hypothesis that BA-activated eIF2α phosphorylation requires protein kinase RNA-like endoplasmic reticulum kinase (PERK) and activates Nrf2 and the antioxidant response element (ARE). BA (10 µM) increased phosphorylation of eIF2α Ser51 in MEF WT cells at 1 h, but not in MEF Perk -/- cells exposed for as long as 6 h. GCN2 kinase-dependent phosphorylation of eIF2α Ser51 was activated in MEF PERK -/- cells by amino acid starvation. Nrf2 phosphorylation is PERK dependent and when activated is translocated from the cytoplasm to the nucleus where it acts as a transcription factor for ARE. DU-145 cells were treated with 10 µM BA and Nrf2 measured by immunofluorescence. Cytoplasmic Nrf2 was translocated to the nucleus at 1.5-2 h in DU-145 and MEF WT cells, but not MEF PERK -/- cells. Real-time PCR was used to measure mRNA levels of three ARE genes (HMOX-1, NQO1, and GCLC). Treatment with 10 µM BA increased the mRNA levels of all three genes at 1-4 h in DU-145 cells and HMOX1 and GCLC in MEF WT cells. These results extend the known boric acid signaling pathway to ARE-regulated genes. The BA signaling pathway can be expressed using the schematic [BA + cADPR → cADPR-BA → [[ER]i Ca2+↓] → 3 pathways: PERK/eIF2αP → pathways ATF4 and Nrf2; and [[ER]i Ca2+↓] → ER stress → ATF6 pathway. This signaling pathway provides a framework that links many of the molecular changes that underpin the biological effects of boron intake.


Subject(s)
Antioxidants/pharmacology , Boric Acids/pharmacology , Boron/pharmacology , DNA Damage/drug effects , Eukaryotic Initiation Factor-2/drug effects , NF-E2-Related Factor 2/drug effects , Trace Elements/pharmacology , eIF-2 Kinase/drug effects , Amino Acids/deficiency , Antioxidant Response Elements/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cytoplasm/drug effects , Eukaryotic Initiation Factor-1 , Gene Expression Regulation/drug effects , Humans , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Translocation, Genetic/drug effects
14.
Bosn J Basic Med Sci ; 18(1): 49-54, 2018 Feb 20.
Article in English | MEDLINE | ID: mdl-28686850

ABSTRACT

Status epilepticus (SE) is defined as continuous seizure activity lasting more than 5 minutes. It results in neuronal cell death, mediated by endoplasmic reticulum (ER) stress response. Previously, metformin demonstrated neuroprotective effects in primary cortical neurons. In this study, we analyzed the effect of metformin on ER stress via the pro-apoptotic protein kinase RNA-like endoplasmic reticulum kinase (PERK)-eukaryotic initiation factor 2α (eIF2α)-C/EBP homologous protein (CHOP) pathway. SE was induced in rats by pentylenetetrazole. Following SE, the rats were treated with salubrinal, GSK2656157, or metformin. In a control group (normal saline) SE was not induced. CHOP, eIF2α, and PERK expression was determined by Western blot; apoptosis was analyzed by TUNEL assay. CHOP expression was significantly increased at 6 and 24 hours following SE. At both time points, eIF2α and PERK levels were also increased. At 6 hours, CHOP expression was significantly reduced in salubrinal, GSK2656157 and metformin groups versus SE group. eIF2α and PERK levels were decreased in metformin compared to SE group. eIF2α expression was markedly decreased in salubrinal versus SE group, while PERK expression was markedly reduced in GSK2656157 versus SE group. At 6 and 24 hours, the apoptosis rate was significantly increased in SE versus control group, while it was significantly reduced in salubrinal, GSK2656157, and metformin groups compared to SE group. The apoptosis rate also decreased in salubrinal group at 24 hours, although not to the extent observed in metformin group. Overall, CHOP expression and apoptosis induced by SE in rats were reduced with metformin. Further studies are required to evaluate the clinical relevance of metformin for patients with SE.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/drug effects , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Signal Transduction/drug effects , Status Epilepticus/drug therapy , Transcription Factor CHOP/drug effects , eIF-2 Kinase/drug effects , Animals , Apoptosis/drug effects , Convulsants , Pentylenetetrazole , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology
15.
Expert Opin Ther Targets ; 21(12): 1171-1177, 2017 12.
Article in English | MEDLINE | ID: mdl-29063813

ABSTRACT

INTRODUCTION: The eIF2α kinase heme-regulated inhibitor (HRI) is one of four well-described kinases that phosphorylate eIF2α in response to various cell stressors, resulting in reduced ternary complex formation and attenuation of mRNA translation. Although HRI is well known for its role as a heme sensor in erythroid progenitors, pharmacologic activation of HRI has been demonstrated to have anti-cancer activity across a wide range of tumor sub-types. Here, the potential of HRI activators as novel cancer therapeutics is explored. Areas covered: We provide an introduction to eIF2 signaling pathways in general, and specifically review data on the eIF2α kinase HRI in erythroid and non-erythroid cells. We review aspects of targeting eIF2 signaling in cancer and highlight promising data using HRI activators against tumor cells. Expert opinion: Pharmacologic activation of HRI inhibits tumor growth as a single agent without appreciable toxicity in vivo. The ability of HRI activators to provide direct and sustained eIF2α phosphorylation without inducing oxidative stress or broad eIF2α kinase activation may be especially advantageous for tolerability. Combination therapy with established therapeutics may further augment anti-cancer activity to overcome disease resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , eIF-2 Kinase/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Drug Design , Erythrocytes/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/enzymology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects , eIF-2 Kinase/metabolism
16.
BMC Microbiol ; 17(1): 187, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28836948

ABSTRACT

BACKGROUND: The bronchial epithelium serves as the first defendant line of host against respiratory inhaled pathogens, mainly through releasing chemokines (e.g. interleukin-8 (IL-8), interferon-induced protein 10 (IP-10) etc.) responsible for neutrophil or lymphocyte recruitment to promote the clearance of inhaled pathogens including Streptococcus pneumoniae (S. pneumoniae). Previous studies have shown that IL-8 expression is induced by pneumococcal virulence factors (e.g. pneumolysin, peptidoglycan-polysaccharides, pneumococcal surface protein A (PspA) etc.), which contributes to the pathogenesis of pneumonia. Whether other pneumococcal virulence factors are involved in inducing chemokines expression in epithelium is still unknown. RESULTS: We studied the effect of PepO, a widely expressed and newly discovered pneumococcal virulence protein, on the release of proinflammatory cytokines, IL-8 and IP-10, from human bronchial epithelial cell line BEAS-2B and identified the relevant signaling pathways. Incubation of BEAS-2B with PepO resulted in increased synthesis and release of IL-8 and IP-10 in a dose and time independent manner. We also detected the increased and sustained expression of TLR2 and TLR4 transcripts in BEAS-2B stimulated by PepO. PepO activation leaded to the phosphorylation of MAPKs, Akt and p65. Pharmacologic inhibitors of MAPKs, PI3K and IκB-α phosphorylation attenuated IL-8 release, while IP-10 production was just suppressed by inhibitors of IκB-α phosphorylation, PI3K and P38 MAPK. CONCLUSION: These results suggest that PepO enhances IL-8 and IP-10 production in BEAS-2B in a MAPKs-PI3K/Akt-p65 dependent manner, which may play critical roles in the pathogenesis of pneumonia.


Subject(s)
Bacterial Proteins/pharmacology , Bronchi/metabolism , Chemokine CXCL10/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Interleukin-8/metabolism , Metalloendopeptidases/pharmacology , Streptococcus pneumoniae/metabolism , Bacterial Proteins/administration & dosage , Cell Line , Chemokine CXCL10/genetics , Cytokines/metabolism , Gene Expression Regulation, Enzymologic , Humans , Interleukin-8/genetics , Metalloendopeptidases/administration & dosage , Mitogen-Activated Protein Kinase Kinases/drug effects , NF-KappaB Inhibitor alpha/metabolism , Phosphatidylinositol 3-Kinases , Phosphorylation , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Streptococcus pneumoniae/pathogenicity , Time Factors , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Transcription, Genetic , Virulence Factors , eIF-2 Kinase/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Inflammation ; 40(6): 1903-1911, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28748322

ABSTRACT

Schisandrin B (Sch B), an active composition isolated from the fruit of Schisandra chinensis, has been proved to possess antiinflammatory, antioxidant and anti-endoplasmic reticulum (ER) stress effects in many rodent tissues. However, the exact mechanism of cardioprotective effect of Sch B still needs more study. Here, we detected the effects of Sch B on myocardial ischemia/reperfusion (I/R) injury rats. I/R injury model in this study was established by left anterior descending coronary artery ligation for 40 min followed by 1 h of reperfusion. Male healthy rats were randomly divided into five groups: the sham, I/R, Sch B (20 mg/kg) + I/R, and Sch B (40 mg/kg) + I/R, Sch B (80 mg/kg) + I/R, with 10 rats in each group. We showed that Sch B treatment significantly protected against myocardial I/R injury, as demonstrated by the decrease in the percentage of infarct formation assessed by 2,3,5-triphenyl tetrazolium chloride (TTC) staining in representative heart tissue slices, comparing with the I/R control group. The levels of creatine kinase (CK), lactate dehydrogenase (LDH), malondialdehyde (MDA), and total superoxide dismutase (T-SOD) were tested. The ER stress-related proteins such as C/EBP homologous protein (CHOP), activating transcription factor 6 (ATF6), and (PKR)-like ER kinase (PERK) were further measured by western blot, and their messenger RNA levels were measured by real-time PCR. The apoptosis of heart tissue cells was also tested through the expressions of caspase-9, caspase-3, Bcl-2, and Bax proteins. Collectively, these results revealed that Sch B exerts protection role on myocardial I/R injury through decreasing oxidative reaction, suppressing ATF6 and PERK pathway, and attenuating ER stress-induced apoptosis.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress , Lignans/pharmacology , Myocardial Reperfusion Injury/drug therapy , Polycyclic Compounds/pharmacology , Activating Transcription Factor 6/drug effects , Activating Transcription Factor 6/metabolism , Animals , Cyclooctanes/pharmacology , Male , Oxidative Stress/drug effects , Rats , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism
18.
Article in English | MEDLINE | ID: mdl-28642847

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) is a leading cause of diarrhea among children and travelers in developing countries, and heat-labile enterotoxin (LT) is one of the most important virulence factors. The pathogenesis of and virulence factors associated with ETEC have been well-characterized; however, the extent to which ETEC damages host cells remains unclear. In this study, we found that LT could induce decreases in intestinal epithelial cell viability and induce apoptosis in a dose- and time- dependent manner in both HCT-8 and Caco-2 cells. We analyzed the expression profiles of apoptosis-related proteins via protein array technology and found that Bax, p-p53(S46), cleaved caspase-3, and TNFRI/TNFRSF1A expression levels were significantly up-regulated in wild-type ETEC- but not in ΔLT ETEC-infected HCT-8 cells. Bax is essential for endoplasmic reticulum (ER) stress-triggered apoptosis, and our RNAi experiments showed that the PERK-eIF2-CHOP pathway and reactive oxygen species (ROS) are also main participants in this process. LT-induced ROS generation was decreased in CHOP-knockdown HCT-8 cells compared to that in control cells. Moreover, pretreatment with the ROS inhibitor NAC down-regulated GRP78, CHOP, Bim, and cleaved caspase-3 expression, resulting in a reduction in the apoptosis rate from 36.2 to 20.3% in LT-treated HCT-8 cells. Furthermore, ROS inhibition also attenuated LT-induced apoptosis in the small intestinal mucosa in the ETEC-inoculation mouse model.


Subject(s)
Apoptosis/drug effects , Enterotoxins/pharmacology , Epithelial Cells/drug effects , Intestinal Mucosa/metabolism , Transcription Factor CHOP/drug effects , Transcription Factor CHOP/metabolism , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11/drug effects , Caco-2 Cells , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Enterotoxigenic Escherichia coli/metabolism , Enterotoxigenic Escherichia coli/pathogenicity , Enterotoxins/administration & dosage , Escherichia coli Infections , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Gene Expression Regulation/drug effects , Gene Silencing , Heat-Shock Proteins/drug effects , Hot Temperature , Humans , Intestines/drug effects , Mice , Mice, Inbred ICR , RNA Interference , Reactive Oxygen Species/metabolism , Time Factors , Transcription Factor CHOP/genetics , bcl-2-Associated X Protein/metabolism , eIF-2 Kinase/genetics
19.
Mol Cells ; 40(4): 280-290, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28359145

ABSTRACT

Several lines of evidence suggest that endoplasmic reticulum (ER) stress plays a critical role in the pathogenesis of many neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Protein tyrosine phosphatase 1B (PTP1B) is known to regulate the ER stress signaling pathway, but its role in neuronal systems in terms of ER stress remains largely unknown. Here, we showed that rotenone-induced toxicity in human neuroblastoma cell lines and mouse primary cortical neurons was ameliorated by PTP1B inhibition. Moreover, the increase in the level of ER stress markers (eIF2α phosphorylation and PERK phosphorylation) induced by rotenone treatment was obviously suppressed by concomitant PTP1B inhibition. However, the rotenone-induced production of reactive oxygen species (ROS) was not affected by PTP1B inhibition, suggesting that the neuroprotective effect of the PTP1B inhibitor is not associated with ROS production. Moreover, we found that MG132-induced toxicity involving proteasome inhibition was also ameliorated by PTP1B inhibition in a human neuroblastoma cell line and mouse primary cortical neurons. Consistently, downregulation of the PTP1B homologue gene in Drosophila mitigated rotenone- and MG132-induced toxicity. Taken together, these findings indicate that PTP1B inhibition may represent a novel therapeutic approach for ER stress-mediated neurodegenerative diseases.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Neurodegenerative Diseases/enzymology , Neurons/drug effects , Neuroprotection , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Animals , Cell Death , Cerebral Cortex/enzymology , Down-Regulation , Drosophila/enzymology , Eukaryotic Initiation Factor-2/drug effects , Humans , Leupeptins/pharmacology , Mice , Neurons/enzymology , Phosphorylation , Reactive Oxygen Species/metabolism , Rotenone/pharmacology , Tumor Cells, Cultured , Uncoupling Agents/pharmacology , eIF-2 Kinase/drug effects
20.
Leuk Res ; 55: 23-32, 2017 04.
Article in English | MEDLINE | ID: mdl-28119225

ABSTRACT

Dexamethasone (dex) induces apoptosis in multiple myeloma (MM) cells and is a frontline treatment for this disease. However resistance to dex remains a major challenge and novel treatment approaches are needed. We hypothesized that dex utilizes translational pathways to promote apoptosis in MM and that specific targeting of these pathways could overcome dex-resistance. Global unbiased profiling of mRNA translational profiles in MM cells treated with or without dex revealed that dex significantly repressed eIF2 signaling, an important pathway for regulating ternary complex formation and protein synthesis. We demonstrate that dex induces the phosphorylation of eIF2α resulting in the translational upregulation of ATF4, a known eIF2 regulated mRNA. Pharmacologic induction of eIF2α phosphorylation via activation of the heme-regulated eIF2α kinase (HRI) induced apoptosis in MM cell lines and in primary MM cells from patients with dex-resistant disease. In addition, co-culture with marrow stroma failed to protect MM cells from apoptosis induced by targeting the eIF2 pathway. Combination therapy with rapamycin, an mTOR inhibitor, and BTdCPU, an activator of HRI, demonstrated additive effects on apoptosis in dex-resistant cells. Thus, specific activation of the eIF2α kinase HRI is a novel therapeutic target in MM that can augment current treatment strategies.


Subject(s)
Molecular Targeted Therapy/methods , Multiple Myeloma/drug therapy , eIF-2 Kinase/metabolism , Apoptosis/drug effects , Dexamethasone/pharmacology , Drug Resistance, Neoplasm , Humans , Phosphorylation , Protein Biosynthesis , Tumor Cells, Cultured , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...