Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros












Intervalo de año de publicación
1.
Ecotoxicol Environ Saf ; 274: 116193, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38460407

RESUMEN

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.


Asunto(s)
Acetatos , Clormequat , Sobrecarga de Hierro , Fenoles , Espermatogénesis , Animales , Masculino , Ratones , Ratas , Clormequat/metabolismo , Clormequat/toxicidad , Sobrecarga de Hierro/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Semillas , Espermatogénesis/efectos de los fármacos , Testículo , eIF-2 Quinasa/efectos de los fármacos , eIF-2 Quinasa/metabolismo
2.
Dis Markers ; 2022: 9340353, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36523813

RESUMEN

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.


Asunto(s)
Medicamentos Herbarios Chinos , Neumonía por Mycoplasma , eIF-2 Quinasa , Animales , Ratones , Citocinas , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , eIF-2 Quinasa/efectos de los fármacos , eIF-2 Quinasa/metabolismo , Retículo Endoplásmico/metabolismo , Ratones Endogámicos BALB C , Neumonía por Mycoplasma/tratamiento farmacológico , Neumonía por Mycoplasma/metabolismo , Proteínas Quinasas , Especies Reactivas de Oxígeno , Transducción de Señal
3.
Acta Pharmacol Sin ; 43(3): 712-723, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33980998

RESUMEN

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.


Asunto(s)
Acroleína/análogos & derivados , Muerte Celular Autofágica/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Neoplasias Gástricas/patología , Acroleína/farmacología , Proteína 5 Relacionada con la Autofagia/efectos de los fármacos , Beclina-1/efectos de los fármacos , Calcio/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción CHOP/efectos de los fármacos , eIF-2 Quinasa/efectos de los fármacos
4.
Aging (Albany NY) ; 13(3): 3405-3427, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33495405

RESUMEN

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.


Asunto(s)
Autofagia/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Accidente Cerebrovascular Isquémico/metabolismo , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Daño por Reperfusión/metabolismo , eIF-2 Quinasa/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Citocinas/efectos de los fármacos , Citocinas/genética , Técnicas In Vitro , Inflamación , Inyecciones Intraventriculares , Accidente Cerebrovascular Isquémico/inmunología , Ratones , Microglía/inmunología , Neuronas/metabolismo , Cultivo Primario de Células , ARN Mensajero/metabolismo , Ratas , Daño por Reperfusión/inmunología , eIF-2 Quinasa/metabolismo
5.
Addict Biol ; 26(2): e12905, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32293776

RESUMEN

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.


Asunto(s)
Giro Dentado/efectos de los fármacos , MicroARNs/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Nicotina/farmacología , Animales , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Hipocampo/efectos de los fármacos , Masculino , Proteína 2 de Unión a Metil-CpG/efectos de los fármacos , Ratones , Ratones Noqueados , Receptores Nicotínicos/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , eIF-2 Quinasa/efectos de los fármacos
6.
Mol Med Rep ; 22(4): 2887-2895, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32700746

RESUMEN

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.


Asunto(s)
Dinoprostona/metabolismo , Glomerulonefritis/tratamiento farmacológico , Subtipo EP1 de Receptores de Prostaglandina E/agonistas , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Ácido Dibenzo(b,f)(1,4)oxazepina-10(11H)-carboxílico, 8-cloro-, 2-acetilhidrazida/farmacología , Dinoprostona/análogos & derivados , Dinoprostona/farmacología , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glomerulonefritis/etiología , Glomerulonefritis/fisiopatología , Proteínas de Choque Térmico/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Masculino , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Ratones , Ratones Endogámicos C57BL , Nefrectomía/efectos adversos , Antagonistas de Prostaglandina/farmacología , Canales Catiónicos TRPC/efectos de los fármacos , Canales Catiónicos TRPC/metabolismo , Factor de Crecimiento Transformador beta1/toxicidad , eIF-2 Quinasa/efectos de los fármacos , eIF-2 Quinasa/metabolismo
8.
Acta Cir Bras ; 35(1): e202000104, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32159588

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glutamina/farmacología , Mucosa Intestinal/efectos de los fármacos , Sustancias Protectoras/farmacología , Daño por Reperfusión/prevención & control , Factor de Transcripción Activador 4/efectos de los fármacos , Animales , Caspasa 12/efectos de los fármacos , Proteínas de Choque Térmico/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/ultraestructura , Masculino , Arteria Mesentérica Superior/lesiones , Modelos Animales , ARN Mensajero/efectos de los fármacos , Ratas Sprague-Dawley , Factor de Transcripción CHOP/efectos de los fármacos , eIF-2 Quinasa/efectos de los fármacos
9.
Endocr J ; 67(2): 185-200, 2020 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-31748431

RESUMEN

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.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Hormona del Crecimiento/farmacología , Hígado/efectos de los fármacos , Sistema de Señalización de MAP Quinasas , ARN Mensajero/efectos de los fármacos , Proteína 1 de Unión a la X-Box/efectos de los fármacos , Animales , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/genética , Endorribonucleasas/efectos de los fármacos , Endorribonucleasas/metabolismo , Proteínas del Choque Térmico HSP40/efectos de los fármacos , Proteínas del Choque Térmico HSP40/genética , Hipofisectomía , Hígado/metabolismo , Complejos Multienzimáticos/efectos de los fármacos , Complejos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/metabolismo , Ratas , Factor de Transcripción STAT5/efectos de los fármacos , Factor de Transcripción STAT5/metabolismo , Caracteres Sexuales , Transducción de Señal , Proteína 1 de Unión a la X-Box/genética , eIF-2 Quinasa/efectos de los fármacos , eIF-2 Quinasa/metabolismo
10.
Acta cir. bras ; 35(1): e202000104, 2020. graf
Artículo en Inglés | LILACS | ID: biblio-1088525

RESUMEN

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.


Asunto(s)
Animales , Masculino , Daño por Reperfusión/prevención & control , Apoptosis/efectos de los fármacos , Sustancias Protectoras/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glutamina/farmacología , Mucosa Intestinal/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Ratas Sprague-Dawley , Arteria Mesentérica Superior/lesiones , eIF-2 Quinasa/efectos de los fármacos , Modelos Animales , Factor de Transcripción Activador 4/efectos de los fármacos , Factor de Transcripción CHOP/efectos de los fármacos , Caspasa 12/efectos de los fármacos , Proteínas de Choque Térmico/efectos de los fármacos , Mucosa Intestinal , Mucosa Intestinal/ultraestructura
11.
Neurochem Int ; 131: 104581, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31639405

RESUMEN

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.


Asunto(s)
Factor de Transcripción Activador 6/efectos de los fármacos , Cinamatos/farmacología , Daño del ADN/efectos de los fármacos , Endorribonucleasas/efectos de los fármacos , Neuronas/efectos de los fármacos , Óxido Nítrico/fisiología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tiourea/análogos & derivados , eIF-2 Quinasa/efectos de los fármacos , Animales , Señalización del Calcio/efectos de los fármacos , Línea Celular , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Óxido Nítrico Sintasa/antagonistas & inhibidores , Rotenona/farmacología , Tiourea/farmacología , Desacopladores/farmacología
12.
Am J Physiol Endocrinol Metab ; 316(6): E1081-E1092, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30964708

RESUMEN

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.


Asunto(s)
Factor de Transcripción Activador 4/efectos de los fármacos , Proteína beta Potenciadora de Unión a CCAAT/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Proteínas Musculares/efectos de los fármacos , Palmitatos/farmacología , Factores de Transcripción/efectos de los fármacos , eIF-2 Quinasa/efectos de los fármacos , Factor de Transcripción Activador 4/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Línea Celular , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Ratones , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , eIF-2 Quinasa/metabolismo
13.
Biol Trace Elem Res ; 188(1): 2-10, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30196486

RESUMEN

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.


Asunto(s)
Antioxidantes/farmacología , Ácidos Bóricos/farmacología , Boro/farmacología , Daño del ADN/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Oligoelementos/farmacología , eIF-2 Quinasa/efectos de los fármacos , Aminoácidos/deficiencia , Elementos de Respuesta Antioxidante/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Citoplasma/efectos de los fármacos , Factor 1 Eucariótico de Iniciación , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Translocación Genética/efectos de los fármacos
14.
Bosn J Basic Med Sci ; 18(1): 49-54, 2018 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-28686850

RESUMEN

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.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Transducción de Señal/efectos de los fármacos , Estado Epiléptico/tratamiento farmacológico , Factor de Transcripción CHOP/efectos de los fármacos , eIF-2 Quinasa/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Convulsivantes , Pentilenotetrazol , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/fisiopatología
15.
Expert Opin Ther Targets ; 21(12): 1171-1177, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29063813

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , eIF-2 Quinasa/efectos de los fármacos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Diseño de Fármacos , Eritrocitos/metabolismo , Humanos , Terapia Molecular Dirigida , Neoplasias/enzimología , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , eIF-2 Quinasa/metabolismo
16.
BMC Microbiol ; 17(1): 187, 2017 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-28836948

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/farmacología , Bronquios/metabolismo , Quimiocina CXCL10/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Interleucina-8/metabolismo , Metaloendopeptidasas/farmacología , Streptococcus pneumoniae/metabolismo , Proteínas Bacterianas/administración & dosificación , Línea Celular , Quimiocina CXCL10/genética , Citocinas/metabolismo , Regulación Enzimológica de la Expresión Génica , Humanos , Interleucina-8/genética , Metaloendopeptidasas/administración & dosificación , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Inhibidor NF-kappaB alfa/metabolismo , Fosfatidilinositol 3-Quinasas , Fosforilación , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Streptococcus pneumoniae/patogenicidad , Factores de Tiempo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Transcripción Genética , Factores de Virulencia , eIF-2 Quinasa/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Inflammation ; 40(6): 1903-1911, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28748322

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico , Lignanos/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Compuestos Policíclicos/farmacología , Factor de Transcripción Activador 6/efectos de los fármacos , Factor de Transcripción Activador 6/metabolismo , Animales , Ciclooctanos/farmacología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , eIF-2 Quinasa/efectos de los fármacos , eIF-2 Quinasa/metabolismo
18.
Artículo en Inglés | MEDLINE | ID: mdl-28642847

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Enterotoxinas/farmacología , Células Epiteliales/efectos de los fármacos , Mucosa Intestinal/metabolismo , Factor de Transcripción CHOP/efectos de los fármacos , Factor de Transcripción CHOP/metabolismo , eIF-2 Quinasa/efectos de los fármacos , eIF-2 Quinasa/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2/efectos de los fármacos , Células CACO-2 , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Escherichia coli Enterotoxigénica/metabolismo , Escherichia coli Enterotoxigénica/patogenicidad , Enterotoxinas/administración & dosificación , Infecciones por Escherichia coli , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Proteínas de Choque Térmico/efectos de los fármacos , Calor , Humanos , Intestinos/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Factor de Transcripción CHOP/genética , Proteína X Asociada a bcl-2/metabolismo , eIF-2 Quinasa/genética
19.
Mol Cells ; 40(4): 280-290, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28359145

RESUMEN

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.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Enfermedades Neurodegenerativas/enzimología , Neuronas/efectos de los fármacos , Neuroprotección , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Animales , Muerte Celular , Corteza Cerebral/enzimología , Regulación hacia Abajo , Drosophila/enzimología , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Humanos , Leupeptinas/farmacología , Ratones , Neuronas/enzimología , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Rotenona/farmacología , Células Tumorales Cultivadas , Desacopladores/farmacología , eIF-2 Quinasa/efectos de los fármacos
20.
Leuk Res ; 55: 23-32, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28119225

RESUMEN

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.


Asunto(s)
Terapia Molecular Dirigida/métodos , Mieloma Múltiple/tratamiento farmacológico , eIF-2 Quinasa/metabolismo , Apoptosis/efectos de los fármacos , Dexametasona/farmacología , Resistencia a Antineoplásicos , Humanos , Fosforilación , Biosíntesis de Proteínas , Células Tumorales Cultivadas , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...