Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Stroke ; 51(5): 1570-1577, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32212900

RESUMEN

Background and Purpose- Ischemic stroke impairs endoplasmic reticulum (ER) function, causes ER stress, and activates the unfolded protein response. The unfolded protein response consists of 3 branches controlled by ER stress sensor proteins, which include PERK (protein kinase RNA-like ER kinase). Activated PERK phosphorylates eIF2α (eukaryotic initiation factor 2 alpha), resulting in inhibition of global protein synthesis. Here, we aimed to clarify the role of the PERK unfolded protein response branch in stroke. Methods- Neuron-specific and tamoxifen-inducible PERK conditional knockout (cKO) mice were generated by cross-breeding Camk2a-CreERT2 with Perkf/f mice. Transient middle cerebral artery occlusion was used to induce stroke. Short- and long-term stroke outcomes were evaluated. Protein synthesis in the brain was assessed using a surface-sensing-of-translation approach. Results- After tamoxifen-induced deletion of Perk in forebrain neurons was confirmed in PERK-cKO mice, PERK-cKO and control mice were subjected to transient middle cerebral artery occlusion and 3 days or 3 weeks recovery. PERK-cKO mice had larger infarcts and worse neurological outcomes compared with control mice, suggesting that PERK-induced eIF2α phosphorylation and subsequent suppression of translation protects neurons from ischemic stress. Indeed, better stroke outcomes were observed in PERK-cKO mice that received postischemic treatment with salubrinal, which can restore the ischemia-induced increase in phosphorylated eIF2α in these mice. Finally, our data showed that post-treatment with salubrinal improved functional recovery after stroke. Conclusions- Here, we presented the first evidence that postischemic suppression of translation induced by PERK activation promotes recovery of neurological function after stroke. This confirms and further extends our previous observations that recovery of ER function impaired by ischemic stress critically contributes to stroke outcome. Therefore, future research should include strategies to improve stroke outcome by targeting unfolded protein response branches to restore protein homeostasis in neurons.


Asunto(s)
Estrés del Retículo Endoplásmico/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Neuronas/metabolismo , Neuroprotección/genética , Respuesta de Proteína Desplegada/genética , eIF-2 Quinasa/genética , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Cinamatos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Infarto de la Arteria Cerebral Media/fisiopatología , Ratones , Ratones Noqueados , Fosforilación , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología , Tiourea/análogos & derivados , Tiourea/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
2.
Pancreatology ; 19(4): 548-556, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31040063

RESUMEN

BACKGROUND: Endoplasmic reticulum (ER) stress in the pancreas is closely associated with the development of acute pancreatitis. However, the role of the protein kinase RNA-like ER kinase (PERK) in this disease is not fully understood. We investigated whether an inhibitor of the dephosphorylation of eukaryotic initiation factor 2α, salubrinal, could improve murine experimental pancreatitis through the amelioration of ER stress. METHODS: Acute pancreatitis was induced by the intraperitoneal administration of cerulein (50 µg/kg) six times at 1-h intervals followed by lipopolysaccharide (10 mg/kg). Salubrinal was administered intraperitoneally immediately after lipopolysaccharide injection and 3 h later. Mice were sacrificed 24 h after the first injection of cerulein, and serum amylase and proinflammatory cytokines were measured. The severity of pancreatitis was evaluated histologically using a scoring system. The expression levels of ER stress-related proteins were evaluated by Western blotting. RESULTS: The administration of salubrinal significantly attenuated the increase in serum amylase levels and improved histologically assessed pancreatitis. The serum levels of proinflammatory cytokines were significantly suppressed in salubrinal-treated mice, as was the expression of glucose-regulated protein 78, CCAAT/enhancer-binding protein homologous protein, and cleaved caspase-3. CONCLUSIONS: The amelioration of ER stress through augmentation of the PERK-signaling pathway may be a therapeutic target for the treatment of acute pancreatitis.


Asunto(s)
Cinamatos/uso terapéutico , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Pancreatitis/tratamiento farmacológico , Tiourea/análogos & derivados , Enfermedad Aguda , Amilasas/sangre , Animales , Apoptosis/efectos de los fármacos , Ceruletida , Citocinas/sangre , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inyecciones Intraperitoneales , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Pancreatitis/inducido químicamente , Fosforilación/efectos de los fármacos , Tiourea/uso terapéutico
3.
Ren Fail ; 38(4): 622-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26923138

RESUMEN

PURPOSE: This study was performed to assess the effect of resveratrol on the expression of eukaryotic initiation factor 2α (eIF2α) and activating transcription factor 4 (ATF4) in renal tissues of rats with unilateral ureteral obstruction (UUO). METHODS: Using UUO animal model, after 14 days of surgery, pathological changes were detected by HE staining, renal tubular damage index, renal interstitial collagen deposition area were evaluated by Masson staining, in situ cell apoptosis in renal tissue was analyzed by TUNEL assay, and protein expression of eIF2α and ATF4 in renal tissue was analyzed using western blot detection. RESULTS: After comparison of the treatment groups with model group, we observed that the degree of renal tubular damage, relative area of renal interstitial collagen and eIF2α, ATF4 protein expression were also significantly reduced (p<0.05, p <0.01) in the high-dose resveratrol group. CONCLUSION: Resveratrol can reduce the level of eIF2α protein expression, which further reduces the ATF4 levels.


Asunto(s)
Factor 2 Eucariótico de Iniciación/biosíntesis , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Riñón/patología , Estilbenos/farmacología , Estilbenos/uso terapéutico , Animales , Fibrosis/tratamiento farmacológico , Fibrosis/etiología , Masculino , Ratas , Ratas Wistar , Resveratrol , Obstrucción Ureteral/complicaciones
4.
Digestion ; 90(3): 167-78, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25339182

RESUMEN

BACKGROUND/AIMS: Endoplasmic reticulum (ER) stress in the intestine is closely associated with the development of inflammatory bowel disease (IBD). However, the role of the protein kinase RNA-like ER kinase in this disease is not fully known. We studied whether an inhibitor of the dephosphorylation of eukaryotic initiation factor 2α, salubrinal, improves murine experimental colitis through the amelioration of ER stress. METHODS: Colitis was induced by the administration of 3% dextran sulfate sodium (DSS) for 5 days. Mice were injected salubrinal intraperitoneally from the commencement of DSS treatment and were sacrificed on day 10. The severity of colitis was evaluated histologically using a scoring system.Myeloperoxidase activity and the expression of proinflammatory cytokine genes in the colon were analyzed. The expression levels of ER stress-related proteins were evaluated by Western blotting. RESULTS: The administration of salubrinal significantly attenuated body weight loss and improved colitis, as assessed histologically. The elevation of myeloperoxidase activity and the expression of proinflammatory cytokine genes were suppressed in salubrinal-treated mice. The expression of glucose-regulated protein 78, activating translation factor 4, and heat-shock protein 70 was elevated in mice treated with salubrinal. CONCLUSION: The amelioration of ER stress may be a therapeutic target for the treatment of IBD.


Asunto(s)
Cinamatos/administración & dosificación , Colitis/tratamiento farmacológico , Proteínas de Unión al ADN/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Tiourea/análogos & derivados , Factores de Transcripción/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Sulfato de Dextran , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Inyecciones Intraperitoneales , Interleucinas/genética , Interleucinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , ARN Mensajero/metabolismo , Factores de Transcripción del Factor Regulador X , Tiourea/administración & dosificación , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Pérdida de Peso/efectos de los fármacos , eIF-2 Quinasa/efectos de los fármacos , eIF-2 Quinasa/genética
5.
J Periodontal Res ; 47(3): 299-308, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22489671

RESUMEN

BACKGROUND AND OBJECTIVE: Tobacco smoking is considered to be one of the major risk factors for periodontitis. For example, about half the risk of periodontitis can be attributable to smoking in the USA. It is evident that smokers have greater bone loss, greater attachment loss and deeper periodontal pockets than nonsmoking patients. It has recently been reported that endoplasmic reticulum (ER) stress markers are upregulated in periodontitis patients; however, the direct effects of nicotine on ER stress in regard to extracellular matrix (ECM) degradation are unclear. The purpose of this study was to examine the effects of nicotine on cytotoxicity and expression of ER stress markers, selected ECM molecules and MMPs, and to identify the underlying mechanisms in human periodontal ligament cells. We also examined whether ER stress was responsible for the nicotine-induced cytotoxicity and ECM degradation. MATERIAL AND METHODS: Cytotoxicity and cell death were measured by 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyltetrazolium bromide assay and flow cytometric annexin V and propidium iodide staining. The mRNA and protein expressions of MMPs and ER markers were examined by RT-PCR and western blot analysis. RESULTS: Treatment with nicotine reduced cell viability and increased the proportion of annexin V-negative, propidium iodide-positive cells, an indication of cell death. Nicotine induced ER stress, as evidenced by survival molecules, such as phosphorylated protein kinase-like ER-resident kinase, phosphorylated eukaryotic initiation factor-2α and glucose-regulated protein-78, and apoptotic molecules, such as CAAT/enhancer binding protein homologous protein (CHOP). Nicotine treatment led to the downregulation of ECM molecules, including collagen type I, elastin and fibronectin, and upregulation of MMPs (MMP-1, MMP-2, MMP-8 and MMP-9). Inhibition of ER stress by salubrinal and transfection of CHOP small interfering RNA attenuated the nicotine-induced cell death, ECM degradation and production of MMPs. Salubrinal and CHOP small interfering RNA inhibited the effects of nicotine on the activation of Akt, JNK and nuclear factor-κB. CONCLUSION: These results indicate that nicotine-induced cell death is mediated by the ER stress pathway, involving ECM degradation by MMPs, in human periodontal ligament cells.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Nicotina/toxicidad , Ligamento Periodontal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proteínas Potenciadoras de Unión a CCAAT/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cinamatos/farmacología , Colágeno Tipo I/efectos de los fármacos , Elastina/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/enzimología , Chaperón BiP del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación/antagonistas & inhibidores , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Matriz Extracelular/enzimología , Fibronectinas/efectos de los fármacos , Proteínas de Choque Térmico/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 1 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 8 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasas de la Matriz/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Nicotina/antagonistas & inhibidores , Ligamento Periodontal/citología , Ligamento Periodontal/enzimología , Proteínas Quinasas/análisis , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Tiourea/análogos & derivados , Tiourea/farmacología , Factor de Transcripción CHOP/efectos de los fármacos , Factor de Transcripción CHOP/genética
6.
Arch Toxicol ; 86(1): 37-44, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21809093

RESUMEN

Cadmium exposure is known to cause endoplasmic reticulum (ER) stress. In our current study, we examined the effects of salubrinal, a selective inhibitor of eukaryotic translation initiation factor 2 subunit α (eIF2α) dephosphorylation, on apoptotic cell death and ER stress-signaling events in HK-2 human renal proximal tubular cells exposed to cadmium chloride (CdCl(2)). Using phase-contrast microscopy and a cell viability assay, we observed that salubrinal suppressed CdCl(2)-induced cellular damage and cell death. Treatment with salubrinal reduced the number of TUNEL-positive cells and the cleavages of caspase-3 and poly(ADP-ribose) polymerase, but not the cleavage of light chain 3B, indicating protection from CdCl(2)-induced apoptosis but not autophagy. Although eIF2α remained phosphorylated after CdCl(2) exposure to salubrinal-treated HK-2 cells, the expression of activating transcription factor 4 (ATF4) and the 78 kDa glucose-regulated protein (GRP78) was not increased. On the other hand, CdCl(2)-induced expression of C/EBP homologous protein (CHOP) was reduced by salubrinal treatment. Expression of ATF4, an upstream regulator of GRP78 and CHOP, appeared to be a prerequisite for full protection by salubrinal against cadmium cytotoxicity, because CdCl(2)-induced cellular damage was not fully suppressed in ATF4-deficient cells. Phosphorylated forms of mitogen-activated protein kinases (MAPKs), including c-Jun NH(2)-terminal kinase (JNK), p38, and extracellular signal-regulated protein kinase (ERK), increased after CdCl(2) exposure, whereas salubrinal suppressed the phosphorylation of JNK and p38 but not ERK. These results suggest that salubrinal protects CdCl(2)-exposed HK-2 cells from apoptosis by suppressing cell death signal transduction pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Cloruro de Cadmio/toxicidad , Cinamatos/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Tiourea/análogos & derivados , Células Cultivadas , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Etiquetado Corte-Fin in Situ , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/patología , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tiourea/farmacología
7.
Anticancer Drugs ; 21(4): 389-400, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20110807

RESUMEN

The antiproliferative and apoptotic effects of cucurbitacin E, a natural product isolated from Cucurbitaceae, were determined in human leukemia HL-60 cells. Cucurbitacin E at low concentrations (3-50 nmol/l) inhibited the growth of HL-60 cells, which was associated with G2/M cell-cycle arrest, decrease in the levels of cyclin-dependent kinase1, and increase in the levels of p21. Cucurbitacin E at high concentrations (1-10 mol/l) induced apoptosis of HL-60 cells and activation of caspase-3, caspase-8, and caspase-9. Jurkat leukemia cells with or without caspase-8 expression were nearly equally sensitive to cucurbitacin E-induced apoptosis. Cucurbitacin E did not increase the levels of reactive oxygen species and antioxidants, N-acetylcysteine and catalase, did not block cucurbitacin E-induced apoptosis. Cucurbitacin E decreased the levels of the antiapoptotic proteins XIAP, survivin, and Mcl-1, but increased the level of the proapoptotic protein, Bax. The levels of phosphorylated eukaryotic translation initiation factor 2 subunit (eIF2) were induced in cells undergoing both apoptosis and cell-cycle arrest. As phosphorylated eIF2 is an inhibitor of protein translation initiation, our data suggest that cucurbitacin E induces cell growth arrest and apoptosis through the induction of eIF2 phosphorylation, which leads to the inhibition of cyclin-dependent kinase 1, Mcl-1, survivin, and/or XIAP protein synthesis and that cucurbitacin E induces apoptosis mainly through a mitochondrial-mediated pathway.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Leucemia/tratamiento farmacológico , Triterpenos/farmacología , Acetilcisteína/metabolismo , Antineoplásicos/uso terapéutico , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Catalasa/metabolismo , División Celular/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Fase G2/efectos de los fármacos , Inhibidores de Crecimiento/farmacología , Inhibidores de Crecimiento/uso terapéutico , Humanos , Proteínas Inhibidoras de la Apoptosis , Células Jurkat , Leucemia/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Survivin , Triterpenos/uso terapéutico , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
8.
Toxicol Appl Pharmacol ; 234(1): 14-24, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18951912

RESUMEN

Acrolein is a ubiquitous environmental pollutant and an endogenous product of lipid peroxidation. It is also generated during the metabolism of several drugs and amino acids. In this study, we examined the effects of acrolein on endothelial cells. Treatment of human umbilical vein endothelial cells (HUVECs) with 2 to 10 microM acrolein led to an increase in the phosphorylation of eIF-2alpha within 10 to 30 min of exposure. This was followed by alternate splicing of XBP-1 mRNA and an increase in the expression of the endoplasmic reticulum (ER) chaperone genes Grp78 and Herp. Within 2-4 h of treatment, acrolein also increased the abundance and the nuclear transport of the transcription factors ATF3, AFT4, and CHOP. Acrolein-induced increase in ATF3 was prevented by treating the cells with the chemical chaperone - phenylbutyric acid (PBA). Treatment with acrolein increased phosphorylation of ERK1/2, p38, and JNK. The increase in JNK phosphorylation was prevented by PBA. Acrolein treatment led to activation and nuclear translocation of the transcription factor NF-kappaB and an increase in TNF-alpha, IL-6 and IL-8, but not MCP-1, mRNA. Increased expression of cytokine genes and NF-kappaB activation were not observed in cells treated with PBA. These findings suggest that exposure to acrolein induces ER stress and triggers the unfolded protein response and that NF-kappaB activation and stimulation of cytokine production by acrolein could be attributed, in part, to ER stress. Chemical chaperones of protein-folding may be useful in treating toxicological and pathological states associated with excessive acrolein exposure or production.


Asunto(s)
Acroleína/toxicidad , Retículo Endoplásmico/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Estrés Oxidativo/efectos de los fármacos , Acroleína/administración & dosificación , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Células Endoteliales/metabolismo , Contaminantes Ambientales/administración & dosificación , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Chaperonas Moleculares/genética , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Empalme del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Factores de Transcripción del Factor Regulador X , Factores de Tiempo , Factores de Transcripción/metabolismo , Venas Umbilicales/citología , Venas Umbilicales/efectos de los fármacos , Proteína 1 de Unión a la X-Box
9.
Oxid Med Cell Longev ; 2019: 8574386, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31223428

RESUMEN

Oxidative stress plays a crucial role in the occurrence and development of osteoarthritis (OA) through the activation of endoplasmic reticulum (ER) stress. Curcumin is a polyphenolic compound with significant antioxidant and anti-inflammatory activity among various diseases. To elucidate the role of curcumin in oxidative stress-induced chondrocyte apoptosis, this study investigated the effect of curcumin on ER stress-related apoptosis and its potential mechanism in oxidative stress-induced rat chondrocytes. The results of flow cytometry and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining showed that curcumin can significantly attenuate ER stress-associated apoptosis. Curcumin inhibited the expression of cleaved caspase3, cleaved poly (ADP-ribose) polymerase (PARP), C/EBP homologous protein (CHOP), and glucose-regulated protein78 (GRP78) and upregulated the chondroprotective protein Bcl2 in TBHP-treated chondrocytes. In addition, curcumin promoted the expression of silent information regulator factor 2-related enzyme 1 (SIRT1) and suppressed the expression of activating transcription factor 4 (ATF4), the ratio of p-PERK/PERK, p-eIF2α/eIF2α. Our anterior cruciate ligament transection (ACLT) rat OA model research demonstrated that curcumin (50 mg/kg and 150 mg/kg) ameliorated the degeneration of articular cartilage and inhibited chondrocyte apoptosis in ACLT rats in a dose-dependent manner. By applying immunohistochemical analysis, we found that curcumin enhanced the expression of SIRT1 and inhibited the expression of CHOP and cleaved caspase3 in ACLT rats. Taken together, our present findings firstly indicate that curcumin could inhibit the PERK-eIF2α-CHOP axis of the ER stress response through the activation of SIRT1 in tert-Butyl hydroperoxide- (TBHP-) treated rat chondrocytes and ameliorated osteoarthritis development in vivo.


Asunto(s)
Condrocitos/metabolismo , Curcumina/uso terapéutico , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Osteoartritis/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Sirtuina 1/metabolismo , Animales , Curcumina/farmacología , Modelos Animales de Enfermedad , Ratas , Ratas Sprague-Dawley
10.
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
11.
Exp Neurol ; 313: 16-25, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30529503

RESUMEN

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.


Asunto(s)
Factor de Transcripción Activador 4/efectos de los fármacos , Antiinflamatorios/farmacología , Hemorragia Cerebral/complicaciones , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Inflamación/etiología , Inflamación/prevención & control , Proteínas Serina-Treonina Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción Activador 4/biosíntesis , Animales , Conducta Animal , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Hemorragia Cerebral/psicología , Citocinas/biosíntesis , Factor 2 Eucariótico de Iniciación/biosíntesis , Inflamasomas/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Infiltración Neutrófila/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
12.
Eur J Neurosci ; 28(10): 2003-16, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19046382

RESUMEN

Activation of the double-stranded RNA-dependent protein kinase (PKR) has been implicated in the pathogenesis of several neurodegenerative diseases. We find that a compound widely used as a pharmacological inhibitor of this enzyme, referred to as PKR inhibitor (PKRi), {8-(imidazol-4-ylmethylene)-6H-azolidino[5,4-g]benzothiazol-7-one}, protects against the death of cultured cerebellar granule and cortical neurons. PKRi also prevents striatal neurodegeneration and improves behavioral outcomes in a chemically induced mouse model of Huntington's disease. Surprisingly, PKRi fails to block the phosphorylation of eIF2alpha, a downstream target of PKR, and does not reduce the autophosphorylation of PKR enzyme immunoprecipitated from neurons. Furthermore, neurons lacking PKR are fully protected from apoptosis by PKRi, demonstrating that neuroprotection by this compound is not mediated by PKR inhibition. Using in vitro kinase assays we investigated whether PKRi affects any other protein kinase. These analyses demonstrated that PKRi has no major inhibitory effect on pro-apoptotic kinases such as the c-Jun N-terminal kinases, the p38 MAP kinases and the death-associated protein kinases, or on other kinases including c-Raf, MEK1, MKK6 and MKK7. PKRi does, however, inhibit the activity of certain cyclin-dependent kinases (CDKs), including CDK1, CDK2 and CDK5 both in vitro and in low potassium-treated neurons. Consistent with its inhibitory action on mitotic CDKs, the treatment of HT-22 and HEK293T cell lines with PKRi sharply reduces the rate of cell cycle progression. Taken together with the established role of CDK activation in the promotion of neurodegeneration, our results suggest that PKRi exerts its neuroprotective action by inhibiting CDK.


Asunto(s)
Benzotiazoles/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Degeneración Nerviosa/tratamiento farmacológico , Neuronas/efectos de los fármacos , eIF-2 Quinasa/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular , Quinasas Ciclina-Dependientes/metabolismo , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/fisiopatología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/enzimología , Ratas , Ratas Wistar , eIF-2 Quinasa/metabolismo
13.
Neurobiol Learn Mem ; 89(3): 247-59, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17919940

RESUMEN

There is ongoing debate concerning whether new protein synthesis is necessary for, or even contributes to, memory formation and storage. This review summarizes a contemporary model proposing a role for altered protein synthesis in memory formation and its subsequent stabilization. One defining aspect of the model is that altered protein synthesis serves as a trigger for memory consolidation. Thus, we propose that specific alterations in the pattern of neuronal protein translation serve as an initial event in long-term memory formation. These specific alterations in protein readout result in the formation of a protein complex that then serves as a nidus for subsequent perpetuating reinforcement by a positive feedback mechanism. The model proposes this scenario as a minimal but requisite component for long-term memory formation. Our description specifies three aspects of prevailing scenarios for the role of altered protein synthesis in memory that we feel will help clarify what, precisely, is typically proposed as the role for protein translation in memory formation. First, that a relatively short initial time window exists wherein specific alterations in the pattern of proteins translated (not overall protein synthesis) is involved in initializing the engram. Second, that a self-perpetuating positive feedback mechanism maintains the altered pattern of protein expression (synthesis or recruitment) locally. Third, that other than the formation and subsequent perpetuation of the unique initializing proteins, ongoing constitutive protein synthesis is all that is minimally necessary for formation and maintenance of the engram. We feel that a clear delineation of these three principles will assist in interpreting the available experimental data, and propose that the available data are consistent with a role for protein synthesis in memory.


Asunto(s)
Memoria/fisiología , Biosíntesis de Proteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Anisomicina/farmacología , Biorretroalimentación Psicológica/efectos de los fármacos , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/genética , Factores Eucarióticos de Iniciación , Memoria/efectos de los fármacos , Ratones , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/genética , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Inhibidores de la Síntesis de la Proteína/farmacología , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/fisiología , Refuerzo en Psicología , Proteínas Quinasas S6 Ribosómicas 70-kDa/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR
14.
Int Arch Allergy Immunol ; 146 Suppl 1: 7-10, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18504399

RESUMEN

BACKGROUND: Acetylation and deacetylation of proteins occur in cells in response to various stimuli, and are reversibly catalyzed by histone acetyltransferase and histone deacetylase (HDAC), respectively. EoL-1 cells have an FIP1L1-PDGFRA fusion gene that causes transformation of eosinophilic precursor cells into leukemia cells. The HDAC inhibitors apicidin and n-butyrate suppress the proliferation of EoL-1 cells and induce differentiation into eosinophils by a decrease in the protein level of FIP1L1-PDGFRalpha without affecting the mRNA level for FIP1L1-PDGFRA. In this study, we analyzed the mechanism by which the protein level of FIP1L1-PDGFRalpha is decreased by apicidin and n-butyrate. METHODS: EoL-1 cells were incubated in the presence of the HDAC inhibitors apicidin, trichostatin A or n-butyrate. The protein levels of FIP1L1-PDGFRalpha and phosphorylated eIF-2alpha were determined by Western blotting. Actinomycin D and cycloheximide were used to block RNA synthesis and protein synthesis, respectively, in the chasing experiment of the amount of FIP1L1-PDGFRalpha protein. RESULTS: When apicidin- and n-butyrate-treated EoL-1 cells were incubated in the presence of actinomycin D, the decrease in the protein level of FIP1L1-PDGFRalpha was significantly enhanced when compared with controls. In contrast, the protein levels were not changed by cycloheximide among these groups. Apicidin and n-butyrate induced the continuous phosphorylation of eIF-2alpha for up to 8 days. CONCLUSIONS: The decrease in the level of FIP1L1-PDGFRalpha protein by continuous inhibition of HDAC may be due to the decrease in the translation rate of FIP1L1-PDGFRA.


Asunto(s)
Butiratos/farmacología , Inhibidores Enzimáticos/farmacología , Eosinófilos/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Inhibidores de Histona Desacetilasas , Síndrome Hipereosinofílico/tratamiento farmacológico , Proteínas de Fusión Oncogénica/análisis , Péptidos Cíclicos/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/análisis , Factores de Escisión y Poliadenilación de ARNm/análisis , Acetilación/efectos de los fármacos , Western Blotting , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Eosinófilos/metabolismo , Factor 2 Eucariótico de Iniciación/análisis , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Histona Desacetilasas/metabolismo , Humanos , Síndrome Hipereosinofílico/metabolismo , Proteínas de Fusión Oncogénica/efectos de los fármacos , Proteínas de Fusión Oncogénica/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Escisión y Poliadenilación de ARNm/efectos de los fármacos , Factores de Escisión y Poliadenilación de ARNm/metabolismo
15.
Neurochem Int ; 52(6): 1167-75, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18222016

RESUMEN

Glutamate, the main excitatory amino acid transmitter regulates protein biosynthesis at the transcriptional and translational levels. It is critically involved in the continuous change of the protein repertoire that is inherent to synaptic plasticity. Activity-dependent differential gene expression occurs both in neurons and glial cells. In fact, besides a membrane to nuclei signaling that leads to transcriptional control, a biphasic effect in overall protein synthesis takes place after glutamate receptors stimulation in cultured chick cerebellar Bergmann glia. Therefore, the effect of glutamate receptors activation on translation elongation was characterized. A time- and dose-dependent increase in eukaryotic elongation factor-2 phosphorylation was found. Pharmacological tools established that alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate and Kainate, but not N-methyl-d-aspartate trigger this phosphorylation. The removal of external Ca2+ or the pre-treatment with a calmodulin antagonist prevented the glutamate effect. Accordingly, glutamate receptors regulate eukaryotic elongation factor-2 kinase phosphorylation through the involvement of Ca2+/calmodulin/extracellular-regulated protein kinases 1/2. These results demonstrate that glutamate receptors regulate the elongation of peptide chains in glial cells.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Ácido Glutámico/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Neuroglía/metabolismo , Receptores de Glutamato/metabolismo , Transducción de Señal/fisiología , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Corteza Cerebelosa/metabolismo , Embrión de Pollo , Relación Dosis-Respuesta a Droga , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/farmacología , Proteínas del Tejido Nervioso/genética , Neuroglía/efectos de los fármacos , Péptidos/genética , Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Receptores de Glutamato/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
16.
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
17.
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
18.
Circ Res ; 90(7): 777-83, 2002 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-11964370

RESUMEN

The aim was to identify energy-consuming processes, other than contraction, downregulated during moderate hypoxia ( approximately 5 micromol/L, 0.5% O(2)) and severe hypoxia (<0.5 micromol/L, <0.05% O(2)) in isolated neonatal cardiomyocytes. The metabolic response of cardiomyocytes to moderate and severe hypoxia was assessed by measuring rates of energy consumption and energetic status of cells maintained under these conditions. We found that the rates of energy production were decreased during both forms of hypoxia. Decreased rates of energy production under moderate hypoxia were associated with reduced energy wastage through a downregulation of proton leak in the mitochondria. Cellular protein synthesis and RNA synthesis, major energy-consuming pathways, were downregulated only during severe hypoxia, when oxygen concentrations were low enough to induce energetic stress (quantitatively defined as being any situation in which phosphocreatine concentrations had fallen by > or = 40%). Our results suggest that energetic stress is the signal responsible for this downregulation.


Asunto(s)
Hipoxia de la Célula/fisiología , Regulación hacia Abajo/fisiología , Miocardio/metabolismo , Proteínas/metabolismo , ARN/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Antifúngicos/farmacología , Separación Celular , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Técnicas In Vitro , Metacrilatos , Mitocondrias/metabolismo , Contracción Miocárdica/fisiología , Miocardio/citología , Oxígeno/metabolismo , Consumo de Oxígeno/fisiología , Fosfocreatina/metabolismo , Fosforilación/efectos de los fármacos , Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Sodio/metabolismo , Tiazoles/farmacología
19.
J Leukoc Biol ; 78(1): 187-94, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15845644

RESUMEN

The unfolded protein response pathway (UPR) compensates for excessive protein accumulation in the endoplasmic reticulum (ER). As insulin induces global protein synthesis, it may cause accumulation of unfolded proteins in the ER, thus triggering UPR. We assessed UPR activation in insulin-treated murine peritoneal macrophages using a number of markers including 78 kDa glucose response protein (GRP78), X-box-binding protein (XBP)-1, pancreatic ER kinase (PERK), eukaryotic initiation factor 2 (eIF2)alpha, and growth arrest and DNA damage (GADD)34. Exposure of cells to insulin activated UPR, as evidenced by an increased expression of GRP78, XBP-1, phosphorylated PERK (p-PERK), and p-eIF2alpha. The insulin-induced, elevated expression of GRP78 was comparable with that observed with tunicamycin, a classical inducer of ER stress. Concomitantly, insulin also up-regulated prosurvival mechanisms by elevating GADD34 and elements of the antiapoptotic pathway including Bcl-2, X-linked inhibitor of apoptosis, and phosphorylated forkhead transcription factor. In conclusion, we show here that insulin treatment does cause ER stress in macrophages, but insulin-dependent mechanisms overcome this ER stress by up-regulating UPR and the antiapoptotic pathway to promote cell survival.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Insulina/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Chaperonas Moleculares/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Antígenos de Diferenciación , Antivirales/farmacología , Apoptosis/fisiología , Biomarcadores/metabolismo , Proteínas de Ciclo Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Proteínas de Choque Térmico/genética , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Chaperonas Moleculares/genética , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/metabolismo , Pliegue de Proteína , Proteína Fosfatasa 1 , Proteínas/efectos de los fármacos , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factores de Transcripción del Factor Regulador X , Transducción de Señal/fisiología , Factores de Transcripción , Tunicamicina/farmacología , Regulación hacia Arriba/fisiología , Proteína 1 de Unión a la X-Box , eIF-2 Quinasa/efectos de los fármacos , eIF-2 Quinasa/metabolismo
20.
Transl Psychiatry ; 6(7): e852, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27404284

RESUMEN

Recent studies implicate the arginine-decarboxylation product agmatine in mood regulation. Agmatine has antidepressant properties in rodent models of depression, and agmatinase (Agmat), the agmatine-degrading enzyme, is upregulated in the brains of mood disorder patients. We have previously shown that mice lacking CREB-regulated transcription coactivator 1 (CRTC1) associate behavioral and molecular depressive-like endophenotypes, as well as blunted responses to classical antidepressants. Here, the molecular basis of the behavioral phenotype of Crtc1(-/-) mice was further examined using microarray gene expression profiling that revealed an upregulation of Agmat in the cortex of Crtc1(-/-) mice. Quantitative polymerase chain reaction and western blot analyses confirmed Agmat upregulation in the Crtc1(-/-) prefrontal cortex (PFC) and hippocampus, which were further demonstrated by confocal immunofluorescence microscopy to comprise an increased number of Agmat-expressing cells, notably parvalbumin- and somatostatin-positive interneurons. Acute agmatine and ketamine treatments comparably improved the depressive-like behavior of male and female Crtc1(-/-) mice in the forced swim test, suggesting that exogenous agmatine has a rapid antidepressant effect through the compensation of agmatine deficit because of upregulated Agmat. Agmatine rapidly increased brain-derived neurotrophic factor (BDNF) levels only in the PFC of wild-type (WT) females, and decreased eukaryotic elongation factor 2 (eEF2) phosphorylation in the PFC of male and female WT mice, indicating that agmatine might be a fast-acting antidepressant with N-methyl-D-aspartate (NMDA) receptor antagonist properties. Collectively, these findings implicate Agmat in the depressive-like phenotype of Crtc1(-/-) mice, refine current understanding of the agmatinergic system in the brain and highlight its putative role in major depression.


Asunto(s)
Agmatina/metabolismo , Encéfalo/metabolismo , Trastorno Depresivo/genética , Factores de Transcripción/genética , Ureohidrolasas/genética , Agmatina/farmacología , Animales , Conducta Animal/efectos de los fármacos , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Trastorno Depresivo/metabolismo , Trastorno Depresivo/psicología , Factor 2 Eucariótico de Iniciación/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Interneuronas/metabolismo , Ketamina/farmacología , Masculino , Ratones , Ratones Noqueados , Análisis por Micromatrices , Fenotipo , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Corteza Prefrontal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA