Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Neurobiol Aging ; 95: 154-160, 2020 11.
Article in English | MEDLINE | ID: mdl-32810756

ABSTRACT

The normal aging process is commonly associated with mild cognitive deficits including memory decline. Previous studies indicate a role of dysregulated messenger ribonucleic acid translation capacity in cognitive defects associated with aging and aging-related diseases, including hyperphosphorylation of eukaryotic elongation factor 2 (eEF2). Phosphorylation of eEF2 by the kinase eEF2K inhibits its activity, hindering general protein synthesis. Here, we sought to determine whether cognitive deficits in aged mice can be improved by genetically deleting eEF2K (eEF2K KO) and consequently reduction of eEF2 phosphorylation. We found that suppression of eEF2K prevented aging-related deficits in novel object recognition memory. Interestingly, deletion of eEF2K did not alter overall protein synthesis in the hippocampus. Ultrastructural analysis revealed increase size and larger active zone lengths of postsynaptic densities in the hippocampus of aged eEF2K KO mice. Biochemical assays showed hippocampal eIF2α hyperphosphorylation in aged eEF2K KO mice, indicating inhibition of translation initiation. Our findings may provide insight into mechanistic understanding and thus development of novel therapeutic strategies for aging-related cognitive decline.


Subject(s)
Aging/psychology , Cognitive Aging , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Elongation Factor 2 Kinase/metabolism , Elongation Factor 2 Kinase/physiology , Memory , Recognition, Psychology , Aging/pathology , Animals , Cognitive Dysfunction/psychology , Disease Models, Animal , Female , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice, Knockout , Phosphorylation
2.
Curr Biol ; 30(18): 3507-3521.e7, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32707059

ABSTRACT

Levels of adult neurogenesis in the dentate gyrus (DG) of the hippocampus are correlated with unique cognitive functions. However, the molecular pathways controlling it are poorly understood. Here, we found that the known physiological ways to enhance neurogenesis converged on the eEF2/eEF2K pathway via AMPK in the DG. Enhancing the elongation phase of mRNA translation in eEF2K-knockout (eEF2K-KO) mice induced the expression of neurogenesis-related proteins in the hippocampus. We thus tested the hypothesis that inducing eEF2K-KO in mature neurons of the DG controls neurogenesis. Indeed, both general eEF2K-KO and targeted KO in DG excitatory mature neurons resulted in enhanced neurogenesis levels and upregulation of neurogenesis-related proteins. Increased neurogenesis was correlated with enhanced performance in DG-dependent learning. Moreover, general and local eEF2K-KO in old mice rejuvenated the DG, paving the way for better mechanistic understanding of how neurogenesis is controlled in the mature DG and possible treatments for incurable aging-associated diseases.


Subject(s)
Cognition/physiology , Dentate Gyrus/metabolism , Elongation Factor 2 Kinase/physiology , Hippocampus/metabolism , Neurogenesis , Neurons/cytology , Animals , Male , Mice , Mice, Knockout , Neurons/metabolism , Phosphorylation , Signal Transduction
3.
J Pharmacol Sci ; 137(1): 86-92, 2018 May.
Article in English | MEDLINE | ID: mdl-29778449

ABSTRACT

Eukaryotic elongation factor 2 kinase (eEF2K) is a calmodulin-related protein kinase which regulates protein translation. A484954 is an inhibitor of eEF2K. In the present study, we investigated the acute effects of A484954 on contractility of isolated blood vessels. Isometric contraction of rat isolated aorta and main branch of superior mesenteric artery (MA) was measured. Expression of an inward rectifier K+ (Kir) channel subtype mRNA and protein was examined. A484954 caused relaxation in endothelium-intact [E (+)] and -denuded [E (-)] aorta or MA precontracted with noradrenaline (NA). The relaxation was higher in MA than aorta. The relaxation was partially inhibited by a nitric oxide (NO) synthase inhibitor, NG-nitro-l-arginine methyl ester (300 µM) in E (+) MA. The relaxation was significantly smaller in MA precontracted with high K+ than NA. The A484954-induced relaxation was significantly inhibited by a Kir channel blocker, BaCl2 (1 mM) compared with vehicle control in E (-) MA. Expression of Kir2.2 mRNA and protein was significantly higher in MA than aorta. We for the first time revealed that A484954 induces relaxation through opening smooth muscle Kir (Kir2.2) channel and through endothelium-derived NO in MA.


Subject(s)
Cyclopropanes/pharmacology , Elongation Factor 2 Kinase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Isometric Contraction/drug effects , Mesenteric Artery, Superior/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Pyrimidines/pharmacology , Pyrrolidines/pharmacology , Animals , Elongation Factor 2 Kinase/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , In Vitro Techniques , Male , Muscle Contraction/drug effects , Nitric Oxide/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , Pyridines , Rats, Wistar
5.
Radiother Oncol ; 124(3): 439-447, 2017 09.
Article in English | MEDLINE | ID: mdl-28431753

ABSTRACT

OBJECTIVES: To investigate the biological function of eEF2K in esophageal squamous cell carcinoma (ESCC). MATERIALS AND METHODS: Tissue microarrays containing 100 pairs of ESCC tumor and adjacent normal tissues were completed. Overexpression and knockdown of eEF2K were constructed in ECA-109 and TE-13 ESCC cells. DNA damage, cell viability, migration and invasion, radioresistance, apoptosis and autophagy were determined by immunofluorescence, CCK-8, transwell assay, colony formation assay, flow cytometry and western blot, respectively. Tumor growth and radioresistance were also evaluated using xenograft models created in nude mice. RESULTS: eEF2K expression was higher in ESCC tissues compared with matched non-tumor tissues (P<0.05). Proliferation was increased in eEF2K overexpressing cells compared with controls (P<0.05), while silencing eEF2K reduced cell proliferation (P<0.05). Furthermore, lower levels of eEF2K expression correlated with slower migration and invasion rates (P<0.05), while higher levels of eEF2K expression with faster migration and invasion rates (P<0.05). eEF2K overexpression resulted in radioresistance and radiation-induced autophagy, and reduced radiation-induced apoptosis compared with controls, but silencing eEF2K promoted radiosensitivity and apoptosis, and reduced autophagy. In addition, eEF2K overexpression promoted the tumor growth in vivo (P<0.01). Combined treatment of NH125 (a pharmacological inhibitor of eEF2K) and radiation was more effective at delaying xenograft tumor growth than NH125 and radiation alone (P<0.05). CONCLUSION: eEF2K induced progression and radioresistance in ESCC, which may be a novel therapeutic target for ESCC to increase radiosensitivity.


Subject(s)
Carcinoma, Squamous Cell/pathology , Elongation Factor 2 Kinase/physiology , Esophageal Neoplasms/pathology , Animals , Apoptosis , Autophagy , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cell Proliferation , Disease Progression , Esophageal Neoplasms/radiotherapy , Esophageal Squamous Cell Carcinoma , Female , Humans , Mice , Mice, Inbred BALB C , Radiation Tolerance
6.
Dev Cell ; 28(5): 561-572, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24582807

ABSTRACT

The control of germline quality is critical to reproductive success and survival of a species; however, the mechanisms underlying this process remain unknown. Here, we demonstrate that elongation factor 2 kinase (eEF2K), an evolutionarily conserved regulator of protein synthesis, functions to maintain germline quality and eliminate defective oocytes. We show that disruption of eEF2K in mice reduces ovarian apoptosis and results in the accumulation of aberrant follicles and defective oocytes at advanced reproductive age. Furthermore, the loss of eEF2K in Caenorhabditis elegans results in a reduction of germ cell death and significant decline in oocyte quality and embryonic viability. Examination of the mechanisms by which eEF2K regulates apoptosis shows that eEF2K senses oxidative stress and quickly downregulates short-lived antiapoptotic proteins, XIAP and c-FLIPL by inhibiting global protein synthesis. These results suggest that eEF2K-mediated inhibition of protein synthesis renders cells susceptible to apoptosis and functions to eliminate suboptimal germ cells.


Subject(s)
Apoptosis , Caenorhabditis elegans/physiology , Elongation Factor 2 Kinase/physiology , Germ Cells/pathology , Oocytes/physiology , Quality Control , Animals , Blotting, Western , Caenorhabditis elegans/cytology , Caspases/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Immunoenzyme Techniques , In Situ Nick-End Labeling , Male , Mice , Mice, Knockout , NIH 3T3 Cells , Oocytes/cytology , Ovary/cytology , Ovary/physiology , Phosphorylation
7.
Encephale ; 40(1): 48-55, 2014 Feb.
Article in French | MEDLINE | ID: mdl-24434007

ABSTRACT

BACKGROUND: In recent years, discovery of ketamine's fast and powerful antidepressant effects for treatment-resistant depression (TRD) has led to rethinking of the pathophysiology of depression. Numerous studies in humans and animals have focused on mechanisms of action underlying this effect, producing a number of explanatory pathways. METHOD: The aim of this article is to summarize the various hypotheses underlying rapid antidepressant action of ketamine and therefore to better understand the mechanisms underlying depression and antidepressant action. RESULTS: Ketamine unique antidepressant properties have led to many studies on its neurobiological grounds. Intracellular signaling pathways such as mTOR, GSK3 or eEF2 seem to play a key role and are associated with an increased synaptic plasticity. Other hypotheses are discussed such as ketamine effects on neuro-inflammation, the role of anterior cingulate cortex in brain changes induced by ketamine, and the potential benefits of analgesic properties of ketamine in depressive disorders. CONCLUSION: Our review highlights the potential role of the glutamatergic system in the pathophysiology and treatment of mood disorders. Understanding which pathways underlie the fast antidepressant effect of ketamine paves the way for the development of new antidepressants.


Subject(s)
Antidepressive Agents/therapeutic use , Brain/drug effects , Depressive Disorder, Treatment-Resistant/drug therapy , Ketamine/therapeutic use , Animals , Antidepressive Agents/adverse effects , Brain/physiopathology , Depressive Disorder, Treatment-Resistant/diagnosis , Depressive Disorder, Treatment-Resistant/physiopathology , Depressive Disorder, Treatment-Resistant/psychology , Elongation Factor 2 Kinase/physiology , Glycogen Synthase Kinase 3/physiology , Gyrus Cinguli/drug effects , Gyrus Cinguli/physiopathology , Humans , Ketamine/adverse effects , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Receptors, Neurotransmitter/drug effects , Receptors, Neurotransmitter/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , TOR Serine-Threonine Kinases/physiology
9.
Am J Physiol Heart Circ Physiol ; 305(5): H756-68, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23812389

ABSTRACT

Eukaryotic elongation factor 2 kinase (eEF2K) is a Ca2+/calmodulin-dependent protein kinase. We recently demonstrated that eEF2K protein increases in mesenteric artery from spontaneously hypertensive rats (SHR). Pathogenesis of hypertension is regulated in part by vascular inflammation. We tested the hypothesis whether eEF2K mediates vascular inflammatory responses and development of hypertension. In vascular endothelial cells, small interfering RNA (siRNA) against eEF2K inhibited induction of VCAM-1 and endothelial-selectin as well as monocyte adhesion by TNF-α (10 ng/ml). eEF2K siRNA inhibited phosphorylation of JNK and NF-κB p65 as well as reactive oxygen species (ROS) production by TNF-α. In vascular smooth muscle cells, eEF2K siRNA also inhibited VCAM-1 induction and phosphorylation of JNK and NF-κB by TNF-α. In vivo, increased blood pressure in SHR and ROS production, induction of inflammatory molecules, and hypertrophy in SHR superior mesenteric artery were reduced by an eEF2K inhibitor NH125 (500 µg·kg(-1)·day(-1)). In SHR superior mesenteric artery, impairment of ACh-induced relaxation was normalized by NH125. The present results for the first time demonstrate that eEF2K mediates TNF-α-induced vascular inflammation via ROS-dependent mechanism, which is at least partly responsible for the development of hypertension in SHR.


Subject(s)
Elongation Factor 2 Kinase/physiology , Hypertension/physiopathology , Oxidative Stress/physiology , Tumor Necrosis Factor-alpha/adverse effects , Vasculitis/chemically induced , Vasculitis/physiopathology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cells, Cultured , Disease Models, Animal , Elongation Factor 2 Kinase/antagonists & inhibitors , Elongation Factor 2 Kinase/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Hypertension/metabolism , Imidazoles/pharmacology , MAP Kinase Kinase 4/metabolism , Male , NF-kappa B/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Wistar , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism , Vasculitis/metabolism
10.
PLoS One ; 5(3): e9715, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20300520

ABSTRACT

BACKGROUND: Autophagy is a highly conserved and regulated cellular process employed by living cells to degrade proteins and organelles as a response to metabolic stress. We have previously reported that eukaryotic elongation factor-2 kinase (eEF-2 kinase, also known as Ca(2+)/calmodulin-dependent protein kinase III) can positively modulate autophagy and negatively regulate protein synthesis. The purpose of the current study was to determine the role of the eEF-2 kinase-regulated autophagy in the response of breast cancer cells to inhibitors of growth factor signaling. METHODOLOGY/PRINCIPAL FINDINGS: We found that nutrient depletion or growth factor inhibitors activated autophagy in human breast cancer cells, and the increased activity of autophagy was associated with a decrease in cellular ATP and an increase in activities of AMP kinase and eEF-2 kinase. Silencing of eEF-2 kinase relieved the inhibition of protein synthesis, led to a greater reduction of cellular ATP, and blunted autophagic response. We further showed that suppression of eEF-2 kinase-regulated autophagy impeded cell growth in serum/nutrient-deprived cultures and handicapped cell survival, and enhanced the efficacy of the growth factor inhibitors such as trastuzumab, gefitinib, and lapatinib. CONCLUSION/SIGNIFICANCE: The results of this study provide new evidence that activation of eEF-2 kinase-mediated autophagy plays a protective role for cancer cells under metabolic stress conditions, and that targeting autophagic survival may represent a novel approach to enhancing the effectiveness of growth factor inhibitors.


Subject(s)
Autophagy , Breast Neoplasms/metabolism , Elongation Factor 2 Kinase/genetics , Gene Expression Regulation, Neoplastic , Adenylate Kinase/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Calcium/metabolism , Cell Line, Tumor , Elongation Factor 2 Kinase/physiology , Female , Gefitinib , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lapatinib , Quinazolines/pharmacology , Signal Transduction , Trastuzumab
SELECTION OF CITATIONS
SEARCH DETAIL
...