Significance of SGK1 in the regulation of neuronal function

新加坡元1 功能(生物学) 神经科学 生物 细胞生物学 磷酸化
作者
Florian Läng,Nathalie Strutz‐Seebohm,Guiscard Seebohm,Undine E. Lang
出处
期刊:The Journal of Physiology [Wiley]
卷期号:588 (18): 3349-3354 被引量:128
标识
DOI:10.1113/jphysiol.2010.190926
摘要

The Journal of PhysiologyVolume 588, Issue 18 p. 3349-3354 SYMPOSIUM REVIEWFree Access Significance of SGK1 in the regulation of neuronal function Florian Lang, Florian Lang Department of Physiology I, University of Tübingen, D-72076 Tübingen, GermanySearch for more papers by this authorNathalie Strutz-Seebohm, Nathalie Strutz-Seebohm Department of Biochemistry I, Cation Channel Group, Ruhr-University Bochum, D-44780 Bochum, GermanySearch for more papers by this authorGuiscard Seebohm, Guiscard Seebohm Department of Biochemistry I, Cation Channel Group, Ruhr-University Bochum, D-44780 Bochum, GermanySearch for more papers by this authorUndine E. Lang, Undine E. Lang Department of Psychiatry and Psychotherapy, Charité Campus Mitte, D-10117 Berlin, GermanySearch for more papers by this author Florian Lang, Florian Lang Department of Physiology I, University of Tübingen, D-72076 Tübingen, GermanySearch for more papers by this authorNathalie Strutz-Seebohm, Nathalie Strutz-Seebohm Department of Biochemistry I, Cation Channel Group, Ruhr-University Bochum, D-44780 Bochum, GermanySearch for more papers by this authorGuiscard Seebohm, Guiscard Seebohm Department of Biochemistry I, Cation Channel Group, Ruhr-University Bochum, D-44780 Bochum, GermanySearch for more papers by this authorUndine E. Lang, Undine E. Lang Department of Psychiatry and Psychotherapy, Charité Campus Mitte, D-10117 Berlin, GermanySearch for more papers by this author First published: 16 September 2010 https://doi.org/10.1113/jphysiol.2010.190926Citations: 100 Corresponding author F. Lang: Department of Physiology, University of Tübingen, Gmelinstrasse 5, D72076 Tübingen, Germany. Email: [email protected] This review was presented at The Journal of Physiology Symposium on Regulation of neuronal cell volume: From activation to inhibition to degeneration, which took place at Experimental Biology 2010, Anaheim, CA, USA, 26 April 2010. It was commissioned by the Editorial Board and reflects the views of the authors. AboutSectionsPDF ToolsRequest permissionExport citationAdd to favoritesTrack citation ShareShare Give accessShare full text accessShare full-text accessPlease review our Terms and Conditions of Use and check box below to share full-text version of article.I have read and accept the Wiley Online Library Terms and Conditions of UseShareable LinkUse the link below to share a full-text version of this article with your friends and colleagues. Learn more.Copy URL Abstract The present brief review highlights the putative role of the serum- and glucocorticoid-inducible-kinase-1 (SGK1) in the regulation of neuronal function. SGK1 is genomically upregulated by cell shrinkage and by a variety of hormones including mineralocorticoids and glucocorticoids. The kinase is activated by insulin and growth factors via phosphatidylinositide-3-kinase (PI3-kinase), phosphoinositide-dependent kinase PDK1 and mammalian target of rapamycin mTORC2. SGK1 upregulates ion channels (e.g. SCN5A, ENaC, ASIC1, TRPV5,6, ROMK, Kv1.1–5, KCNEx/KCNQ1–5, GluR6, VSOAC, ClC2, CFTR), carriers (e.g. NHE3, NKCC2, NCC, NaPiIIb, SMIT, GLUT1,4, SGLT1, NaDC, EAAT1–5, SN1, ASCT2, 4F2/LAT, PepT2), and the Na+/K+-ATPase. SGK1 regulates enzymes (e.g. glycogen-synthase-kinase-3, ubiquitin-ligase Nedd4-2, phosphomannose-mutase-2), and transcription factors (e.g. forkhead transcription factor Foxo3a, β-catenin, nuclear factor-kappa-B (NFκB)). SGK1 participates in the regulation of transport, hormone release, neuroexcitability, inflammation, coagulation, cell proliferation and apoptosis. SGK1 contributes to regulation of renal Na+ retention, renal K+ elimination, salt appetite, gastric acid secretion, intestinal Na+/H+ exchange and nutrient transport, insulin-dependent salt sensitivity of blood pressure, salt sensitivity of peripheral glucose uptake, cardiac repolarization and memory consolidation. Presumably, SGK1 contributes to the regulation of diverse cerebral functions (e.g. memory consolidation, fear retention) and the pathophysiology of several cerebral diseases (e.g. Parkinson's disease, schizophrenia, depression, Alzheimer's disease). Despite multiple SGK1 functions, the phenotype of the SGK1 knockout mouse is mild and becomes only apparent under challenging conditions. [ Florian Lang studied in Munich and Glasgow and received his MD in Munich. Prior to joining the department of physiology in Tübingen, he worked at the University of Munich, the University of Innsbruck (Austria), the Mayo Clinic (Rochester, MN, USA), the Max Planck Institute (Frankfurt, Germany), Yale University (New Haven, CT, USA) and the University of Naples (Italy). His research interests include properties, regulation and (patho)physiological significance of ion channels and transporters across the cell membrane. Functions in his focus include epithelial transport, blood pressure, metabolism, cell volume, cell proliferation, cell death, migration, inflammation and host–pathogen interaction.] Abbreviations ALS amyotrophic lateral sclerosis BDNF brain-derived neurotrophic factor mTOR mammalian target of rapamycin NFκB nuclear factor-kappa-B PDK1 phosphoinositide-dependent kinase 1 PI3-kinase phosphatidylinositide-3-kinase SGK1 serum- and glucocorticoid-inducible kinase-1 The serum- and glucocorticoid-inducible kinase was originally cloned as an early gene upregulated by the treatment of mammary tumour cells with glucocorticoids and serum (Firestone et al. 2003). Later the gene was discovered as a cell volume-regulated gene (Lang et al. 2006). Within some 20 min, osmotic and isotonic cell shrinkage profoundly upregulates the SGK1 expression (Lang et al. 2006). Presumably due to cell volume sensitivity of SGK1 transcription, cerebral SGK1 expression is further upregulated by dehydration (Lang et al. 2006). The hydration state of the brain is in turn a critical determinant of neuronal function (De Luca et al. 2010; Thornton, 2010). Hyperosmolar states decrease and hyposmolar states increase the susceptibility to epileptic seizures (Osehobo & Andrew, 1993; Ochoa, 2004), effects in part due to an osmotic gradient across the blood–brain barrier, leading to respective movements of water and changes of extracellular space (Cserr et al. 1991; Silver et al. 1996; Verkman, 2005). During osmotic cell swelling, excitability could be enhanced by the decrease of extracellular space (Osehobo & Andrew, 1993). Hydration may further modify cerebral function by influencing neuronal or glial cell volume (Lang et al. 1998). Dehydration alters the expression of a wide variety of genes (Mutsuga et al. 2005; Lee et al. 2006; Ramos et al. 2007; Hindmarch et al. 2008; Wang et al. 2009; Bourassa & Speth, 2010; Brocker et al. 2010; Pereira-Derderian et al. 2010; Scott & Brown, 2010) including SGK1 (Lang et al. 2006). SGK1-sensitive functions presumably contribute significantly to altered function of the dehydrated brain. Beyond that multiple additional physiological and pathophysiological regulators of SGK1 expression and function are known which may modify cerebral function in the absence of neuronal or glial cell volume changes. The present brief review amplifies on the role of SGK1 in the regulation of neuronal cell function. Due to N limitations of references, reviews are quoted covering earlier papers. Regulation and function of SGK1 SGK1 transcription is controlled by a wide variety of additional hormones and further regulators including increase of cytosolic Ca2+ activity and NO (Lang et al. 2006, 2009). SGK1 is ubiquitously expressed (Lang et al. 2006, 2009). The kinase is activated by insulin and growth factors via PI3-kinase, the 3-phosphoinositide-dependent kinase PDK1, and the mammalian target of rapamycin mTORC2 (Lang et al. 2006, 2009; Peterson et al. 2009). SGK1 increases the protein abundance and/or activity of a variety of ion channels (e.g. SCN5A, ENaC, ASIC1, TRPV5,6, ROMK, Kv1.1–5, KCNE1/KCNQ1, KCNQ2–5, GluR6, VSOAC, ClC2, CFTR), carriers (e.g. NHE3, NKCC2, NCC, NaPiIIb, SMIT, GLUT1,4, SGLT1, NaDC, EAAT1–5, SN1, ASCT2, 4F2/LAT, PepT2, CreaT), and the Na+/K+-ATPase (Lang et al. 2006, 2009; Boehmer et al. 2008a,b; Schuetz et al. 2008; Gehring et al. 2009; Krueger et al. 2009; Laufer et al. 2009). Several of the carriers could contribute to regulatory cell volume increase (RVI), such as NHE, NKCC or SMIT (Lang et al. 1998; Hoffmann et al. 2009). Thus, upregulation of SGK1 by cell shrinkage may support RVI. Stimulation of K+ channels and Cl− channels, however, would counteract RVI and rather support regulatory cell volume decrease (RVD) (Lang et al. 1998; Hoffmann et al. 2009). SGK1 presumably enhances the cellular capacity for both RVI and RVD but is presumably not involved in the rapid activation of cell volume regulatory mechanisms, which is accomplished by activation of other kinases. SGK1 regulates several enzymes (e.g. glycogensynthase-kinase-3, ubiquitin-ligase Nedd4-2, phosphomannose-mutase-2) and transcription factors (e.g. FKHRL1/Foxo3a, β-catenin, NFκB, p53) (Lang et al. 2006, 2009). SGK1 contributes to the regulation of transport, hormone release, neuroexcitability, inflammation, cell proliferation and apoptosis (Lang et al. 2006, 2009). SGK1 further participates in the regulation of renal Na+ retention and renal K+ elimination, mineralocorticoid stimulation of salt appetite, glucocorticoid stimulation of gastric acid secretion, intestinal mineral, electrolyte and nutrient transport, blood coagulation, insulin-dependent salt sensitivity of blood pressure and salt sensitivity of peripheral glucose uptake (Lang et al. 2006, 2009). A common (∼3–5% prevalence in Caucasians, ∼10% in Africans) SGK1 gene variant is associated with increased blood pressure, obesity, enhanced prevalence of diabetes and a shortening of the cardiac action potential (Lang et al. 2006, 2009). SGK1 is further involved in the pathophysiology of allergy, peptic ulcer, tumour growth, fibrosing disease, ischaemia and neurodegeneration (Lang et al. 2006, 2009). The phenotype of SGK1 knockout mice is surprisingly mild and becomes only apparent upon appropriate stress conditions. Putative role of SGK1 in neuronal function Cerebral SGK1 is upregulated by glucocorticoids (Sato et al. 2008; Sarabdjitsingh et al. 2010), corticotropin releasing hormone (Sheng et al. 2008), hyperactivity (Kalinichev et al. 2008), intracranial self-stimulation (Huguet et al. 2009), chronic escalating morphine regimen (Befort et al. 2008) and administration of the antipsychotic drug clozapine (Robbins et al. 2008). Hippocampal SGK1 expression is stimulated by fear conditioning, elevated plus maze exposure and after enrichment training (Lang et al. 2006). SGK1 transcript levels are further enhanced by the psychostimulant amphetamine, by the hallucinogenic drug lysergic acid dimethylamide (LSD), by neuronal injury, neuronal excitotoxicity and microgravity (Lang et al. 2006). Neuronal SGK1 expression is suppressed by Zif268 (Egr1/Krox24/NGF-IA), a transcription factor associated with neuronal plasticity (James et al. 2006). Hypothalamic SGK1 is upregulated by fasting or obesity with hyperphagia (Nonogaki et al. 2006). SGK1 is considered to play an important role in long-term memory formation (Ma et al. 2006). SGK1 phosphorylation increases after tetanization (Ma et al. 2006) and transfection of constitutively active SGK1 upregulates postsynaptic density-95 expression in the hippocampus and impairs the expression, but not the induction, of long-term potentiation (Ma et al. 2006). Transfection of wild-type SGK1 improves, and transfection of inactive SGK1 decreases, the learning abilities of rats (Lang et al. 2006). In those animals transfection of inactive SGK1 impairs spatial learning, fear-conditioning learning, and novel object recognition learning (Lang et al. 2006). Moreover, hippocampal SGK1 mRNA abundance is increased in fast-learning when compared to slow-learning rats (Lang et al. 2006). Transfection of inactive SGK1 to hippocampal neurons further impaired, whereas transfection of constitutively active SGK1 enhanced, fear retention (Lee et al. 2007). SGK1 downregulates Hairy and Enhancer of split 5 (Hes5) resulting in enhanced fear retention, whereas over-expression of Hes5 negatively regulates contextual fear memory formation (Lee et al. 2007). SGK1 stimulates dendrite growth, which may impact on learning abilities (Lang et al. 2006). The role of SGK1 in memory consolidation may further relate to its effect on glutamate receptors. SGK isoforms upregulate AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolic acid) and kainate receptors and are thus expected to enhance the excitatory effects of glutamate (Lang et al. 2006, 2009). The kainate receptor is particularly important for synaptic transmission and plasticity in the hippocampus (Dargan & Amici, 2009). SGK1 contributes to the glucocorticoid stimulation of cerebral GluR6 expression (Lang et al. 2006). Accordingly, SGK1 participates in the neuronal effects of glucocorticoids, which enhance the neuronal responsiveness but by the same token favour neurotoxicity (Lang et al. 2006). On the other hand, SGK1 stimulates glutamate transporters (Lang et al. 2006), which clear the synaptic clefts from glutamate and thus terminate excitation (Benarroch, 2010). SGK phosphorylates guanosine nucleotide dissociation inhibitor (GDI) thus increasing the formation of GDI-Rab4 complexes, which facilitate the Rab4-mediated recycling of AMPARs to the synaptic membrane (Liu et al. 2010). SGK1-mediated spatial memory facilitation and neuronal plasticity may further involve phosphorylation of IKKα with subsequent stimulation of NFκB and triggering of NFκB-dependent expression of genes such as NR2A and NR2B (Tai et al. 2009). In addition, SGK1 could modify excitation by regulating Kv channels (Lang et al. 2006, 2009), which participate in the maintenance of neuronal membrane potential (Pongs, 2008). SGK1 presumably participates in the signalling of brain-derived neurotrophic factor (BDNF), which stimulates the PI3-kinase and thus probably SGK1 (Lang et al. 2006). BDNF influences neuronal survival, plasticity, mood and long-lasting memory formation (Lang et al. 2006). PI3-kinase mediates the BDNF-dependent spatial memory formation in rats (Lang et al. 2006). Hypothalamic SGK1 presumably participates in the regulation of food intake (Nonogaki et al. 2006). Putative role of SGK1 in neuronal disease Lack of SGK may blunt the glutamate action but at the same time decrease glutamate clearance from the synaptic cleft. As glutamate may exert neurotoxic effects (Lau & Tymianski, 2010), altered function or regulation of glutamate transporters and glutamate receptors may influence neuroexcitotoxicity. Decreased expression of glutamate transporters favours extracellular glutamate accumulation, excitotoxicity and eventually neuronal cell death (Rothstein et al. 1996). Loss of EAAT2 function may contribute to amyotrophic lateral sclerosis (ALS); loss of EAAT4 function correlates with loss of Purkinje cells during ischaemia and defective glutamate transport may contribute to retinopathy following glaucoma or to diabetic retinopathy (Lang et al. 2006). Whether or not decreased stimulation of the carriers by SGK1 leads to pathophysiologically relevant decline of transporter function remains to be shown. AMPA receptor upregulation contributes to ischaemic-induced cell death (Lang et al. 2006). AMPA and kainate receptors further participate in the pathophysiology of ALS and schizophrenia (Lang et al. 2006). Deranged function of GluR3 is involved in the autoimmune disease Rasmussen's encephalitis, a disorder leading to inflammation of the brain, severe epilepsy, hemiplegia and dementia (Lang et al. 2006). Upregulation of kainate receptors supports epileptic activity (Lang et al. 2006). Deranged SGK1-dependent regulation of AMPA or kainate receptors could, at least in theory, participate in the pathophysiology of those diseases. Beyond its effects on glutamate receptors and carriers, SGK1 influences further carriers relevant for neuronal function, such as the creatine transporter CreaT (Lang et al. 2006). Individuals with defective CreaT suffer from mental retardation (Lang et al. 2006). SGK1 upregulates the inositol transporter SMIT (Klaus et al. 2008), which participates in neuronal cell volume regulation (Lang et al. 1998). SGK1 could contribute to the glucocorticoid effects during cerebral injury including stroke, seizure, or hypoglycaemia (Lang et al. 2006). SGK isoforms participate in the signalling of brain-derived neurotrophic factor and could thus contribute to BDNF signalling in several neuropsychiatric disorders, such as schizophrenia, depression and Alzheimer's disease (Lang et al. 2006; Lang et al. 2007). SGK1 could further contribute to the effects of antidepressants, antipsychotic drugs and electroconvulsive treatment (Lang et al. 2006). SGK1 phosphorylates tau and may thus contribute to the pathophysiology of Alzheimer's disease, which is paralleled by hyperphosphorylation of tau (Lang et al. 2006). Vascular and meningeal alterations similar to those in Alzheimer's disease are observed in transgenic mice over-expressing transforming growth factor beta (TGFβ), which is a powerful stimulator of SGK1 expression (Lang et al. 2006). On the other hand, TGFβ has been claimed to be neuroprotective by decreasing the accumulation of β-amyloid peptide (Lang et al. 2006) SGK1 phosphorylates huntingtin and may thus counteract huntingtin toxicity (Lang et al. 2006; Warby et al. 2009). SGK1 is further implicated in the pathophysiology of Parkinson's disease (Sakai et al. 2007). In a model of that disease, genomic upregulation of SGK1 coincides with the onset of dopaminergic cell death (Lang et al. 2006). Enhanced SGK1 expression has been observed in Rett syndrome (RTT), a disorder with severe mental retardation (Lang et al. 2006). SGK1 deficiency may participate in a major depressive disorder, which is frequently paralleled by insufficient glucocorticoid signalling (Sato et al. 2008). SGK1 may further be involved in anxiety disorders, as phosphatidylinositide-dependent kinase deficiency in mice increases anxiety and decreases GABA and serotonin abundance in the amygdala (Ackermann et al. 2008). SGK1 transcription is enhanced during cerebral ischaemia (Lang et al. 2006) and several effects of SGK1 could facilitate cellular survival during energy deprivation. Stimulation of creatine uptake by the Na+-coupled creatine transporter CreaT (Lang et al. 2006), for instance, enhances the capacity to bind phosphate to creatine and thus increases the cellular ability to store rapidly available energy. Stimulation of the glucose transporter GLUT1 (Palmada et al. 2006) provides neurons with fuel for anaerobic glycolysis. During ischaemia, the stimulation of the Na+/K+-ATPase by SGK1 is a double-edged sword. On the one hand the stimulation of the energy-consuming Na+/K+-ATPase leads to acceleration of energy depletion. On the other hand, it counteracts the decline of pump activity and the subsequent cellular K+ loss, depolarization, entry of Cl− and cell swelling – known consequences of energy depletion. Beyond its influence on transporters and stimulation of Na+/K+-ATPase, SGK1 inhibits apoptosis and thus prolongs cell survival by influencing several signalling pathways as described elsewhere (Lang et al. 2006, 2009). As apparent from the SGK1 knockout mice (see above), the lack of SGK1 does not lead to profound derangements of neuronal function. Obviously, the SGK1-sensitive functions could be sufficiently stimulated by related kinases, such as the other SGK or Akt/PKB isoforms. However, under conditions leading to marked upregulation of SGK1 this kinase may take a leading part in the regulation of cerebral function. In summary, compelling evidence suggests a role for SGKs in the pathophysiology of brain disease. However, it is not certain, to what extent SGK1 protects from or stimulates cell death or is just an innocent bystander in those disorders. Clearly, we are presently far from understanding the contribution of SGK1 to the deterioration or maintenance of neuronal function and additional experimental effort is needed to unravel the contribution of this multifunctional kinase to cerebral health and disease. References Ackermann TF, Hortnagl H, Wolfer DP, Colacicco G, Sohr R, Lang F, Hellweg R & Lang UE (2008). Phosphatidylinositide dependent kinase deficiency increases anxiety and decreases GABA and serotonin abundance in the amygdala. Cell Physiol Biochem 22, 735– 744. Befort K, Filliol D, Ghate A, Darcq E, Matifas A, Muller J, Lardenois A, Thibault C, Dembele D, Le Merrer J, Becker JA, Poch O & Kieffer BL (2008). Mu-opioid receptor activation induces transcriptional plasticity in the central extended amygdala. Eur J Neurosci 27, 2973– 2984. Benarroch EE (2010). Glutamate transporters: diversity, function, and involvement in neurologic disease. Neurology 74, 259– 264. Boehmer C, Laufer J, Jeyaraj S, Klaus F, Lindner R, Lang F & Palmada M (2008a). Modulation of the voltage-gated potassium channel Kv1.5 by the SGK1 protein kinase involves inhibition of channel ubiquitination. Cell Physiol Biochem 22, 591– 600. Boehmer C, Palmada M, Klaus F, Jeyaraj S, Lindner R, Laufer J, Daniel H & Lang F (2008b). The peptide transporter PEPT2 is targeted by the protein kinase SGK1 and the scaffold protein NHERF2. Cell Physiol Biochem 22, 705– 714. Bourassa EA & Speth RC (2010). Water deprivation increases angiotensin-converting enzyme but not AT1 receptor expression in brainstem and paraventricular nucleus of the hypothalamus of the rat. Brain Res 1319, 83– 91. Brocker C, Lassen N, Estey T, Pappa A, Cantore M, Orlova VV, Chavakis T, Kavanagh KL, Oppermann U & Vasiliou V (2010). Aldehyde dehydrogenase 7A1 (ALDH7A1) is a novel enzyme involved in cellular defense against hyperosmotic stress. J Biol Chem 285; 18452– 18463. Cserr HF, DePasquale M, Nicholson C, Patlak CS, Pettigrew KD & Rice ME (1991). Extracellular volume decreases while cell volume is maintained by ion uptake in rat brain during acute hypernatremia. J Physiol 442, 277– 295. Dargan SL & Amici M (2009). Role of kainate autoreceptors in short-term plasticity at hippocampal mossy fiber synapses. J Neurosci 29, 5713– 5715. De Luca LA Jr, Pereira-Derderian DT, Vendramini RC, David RB & Menani JV (2010). Water deprivation-induced sodium appetite. Physiol Behav 100, 535– 544. Firestone GL, Giampaolo JR & O’Keeffe BA (2003). Stimulus-dependent regulation of serum and glucocorticoid inducible protein kinase (SGK) transcription, subcellular localization and enzymatic activity. Cell Physiol Biochem 13, 1– 12. Gehring EM, Zurn A, Klaus F, Laufer J, Sopjani M, Lindner R, Strutz-Seebohm N, Tavare JM, Boehmer C, Palmada M, Lang UE, Seebohm G & Lang F (2009). Regulation of the glutamate transporter EAAT2 by PIKfyve. Cell Physiol Biochem 24, 361– 368. Hindmarch C, Fry M, Yao ST, Smith PM, Murphy D & Ferguson AV (2008). Microarray analysis of the transcriptome of the subfornical organ in the rat: regulation by fluid and food deprivation. Am J Physiol Regul Integr Comp Physiol 295, R1914– R1920. Hoffmann EK, Lambert IH & Pedersen SF (2009). Physiology of cell volume regulation in vertebrates. Physiol Rev 89, 193– 277. Huguet G, Aldavert-Vera L, Kádár E, Peña de Ortiz S, Morgado-Bernal I & Segura-Torres P (2009). Intracranial self-stimulation to the lateral hypothalamus, a memory improving treatment, results in hippocampal changes in gene expression. Neuroscience 162, 359– 374. James AB, Conway AM & Morris BJ (2006). Regulation of the neuronal proteasome by Zif268 (Egr1). J Neurosci 26, 1624– 1634. Kalinichev M, Robbins MJ, Hartfield EM, Maycox PR, Moore SH, Savage KM, Austin NE & Jones DN (2008). Comparison between intraperitoneal and subcutaneous phencyclidine administration in Sprague–Dawley rats: A locomotor activity and gene induction study. Prog Neuropsychopharmacol Biol Psychiatry 32, 414– 422. Klaus F, Palmada M, Lindner R, Laufer J, Jeyaraj S, Lang F & Boehmer C (2008). Up-regulation of hypertonicity-activated myo-inositol transporter SMIT1 by the cell volume-sensitive protein kinase SGK1. J Physiol 586, 1539– 1547. Krueger B, Haerteis S, Yang L, Hartner A, Rauh R, Korbmacher C & Diakov A (2009). Cholesterol depletion of the plasma membrane prevents activation of the epithelial sodium channel (ENaC) by SGK1. Cell Physiol Biochem 24, 605– 618. Lang F, Artunc F & Vallon V (2009). The physiological impact of the serum and glucocorticoid-inducible kinase SGK1. Curr Opin Nephrol Hypertens 18, 439– 448. Lang F, Bohmer C, Palmada M, Seebohm G, Strutz-Seebohm N & Vallon V (2006) (Patho)physiological significance of the serum- and glucocorticoid-inducible kinase isoforms. Physiol Rev 86, 1151– 1178. Lang F, Busch GL, Ritter M, Völkl H, Waldegger S, Gulbins E & Häussinger D (1998). Functional significance of cell volume regulatory mechanisms. Physiol Rev 78, 247– 306. Lang UE, Puls I, Muller DJ, Strutz-Seebohm N & Gallinat J (2007). Molecular mechanisms of schizophrenia. Cell Physiol Biochem 20, 687– 702. Lau A & Tymianski M (2010). Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch 460, 525– 542. Laufer J, Boehmer C, Jeyaraj S, Knuwer M, Klaus F, Lindner R, Palmada M & Lang F (2009). The C-terminal PDZ-binding motif in the Kv1.5 potassium channel governs its modulation by the Na+/H+ exchanger regulatory factor 2. Cell Physiol Biochem 23, 25– 36. Lee CT, Ma YL & Lee EH (2007). Serum- and glucocorticoid-inducible kinase1 enhances contextual fear memory formation through down-regulation of the expression of Hes5. J Neurochem 100, 1531– 1542. Lee HJ, Palkovits M & Young WS III (2006). miR-7b, a microRNA up-regulated in the hypothalamus after chronic hyperosmolar stimulation, inhibits Fos translation. Proc Natl Acad Sci U S A 103, 15669– 15674. Liu W, Yuen EY & Yan Z (2010). The stress hormone corticosterone increases synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors via serum- and glucocorticoid-inducible kinase (SGK) regulation of the GDI-Rab4 complex. J Biol Chem 285, 6101– 6108. Ma YL, Tsai MC, Hsu WL & Lee EH (2006). SGK protein kinase facilitates the expression of long-term potentiation in hippocampal neurons. Learn Mem 13, 114– 118. Mutsuga N, Shahar T, Verbalis JG, Xiang CC, Brownstein MJ & Gainer H (2005). Regulation of gene expression in magnocellular neurons in rat supraoptic nucleus during sustained hypoosmolality. Endocrinology 146, 1254– 1267. Nonogaki K, Ohashi-Nozue K & Oka Y (2006). Induction of hypothalamic serum- and glucocorticoid-induced protein kinase-1 gene expression and its relation to plasma des-acyl ghrelin in energy homeostasis in mice. Biochem Biophys Res Commun 344, 696– 699. Ochoa JG (2004). Electrolyte therapy for refractory seizures in familial dysautonomia. Epilepsia 45, 1461– 1462. Osehobo EP & Andrew RD (1993). Osmotic effects upon the theta rhythm, a natural brain oscillation in the hippocampal slice. Exp Neurol 124, 192– 199. Palmada M, Boehmer C, Akel A, Rajamanickam J, Jeyaraj S, Keller K & Lang F (2006). SGK1 kinase upregulates GLUT1 activity and plasma membrane expression. Diabetes 55, 421– 427. Pereira-Derderian DT, Vendramini RC, Menani JV & Luca Junior LA (2010). Water deprivation-induced sodium appetite and differential expression of encephalic c-Fos immunoreactivity in the spontaneously hypertensive rat. Am J Physiol Regul Integr Comp Physiol 298, R1298– R1309. Peterson TR, Laplante M, Thoreen CC, Sancak Y, Kang SA, Kuehl WM, Gray NS & Sabatini DM (2009). DEPTOR is an mTOR inhibitor frequently overexpressed in multiple myeloma cells and required for their survival. Cell 137, 873– 886. Pongs O (2008). Regulation of excitability by potassium channels. Results Probl Cell Differ 44, 145– 161. Ramos A, Ho WC, Forte S, Dickson K, Boutilier J, Favell K & Barker PA (2007). Hypo-osmolar stress induces p75NTR expression by activating Sp1-dependent transcription. J Neurosci 27, 1498– 1506. Robbins MJ, Critchlow HM, Lloyd A, Cilia J, Clarke JD, Bond B, Jones DN & Maycox PR (2008). Differential expression of IEG mRNA in rat brain following acute treatment with clozapine or haloperidol: a semi-quantitative RT-PCR study. J Psychopharmacol 22, 536– 542. Rothstein JD, Dykes-Hoberg M, Pardo CA, Bristol LA, Jin L, Kuncl RW, Kanai Y, Hediger MA, Wang Y, Schielke JP & Welty DF (1996). Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron 16, 675– 686. Sakai R, Irie Y, Murata T, Ishige A, Anjiki N & Watanabe K (2007). Toki-to protects dopaminergic neurons in the substantia nigra from neurotoxicity of MPTP in mice. Phytother Res 21, 868– 873. Sarabdjitsingh RA, Isenia S, Polman A, Mijalkovic J, Lachize S, Datson N, De Kloet ER & Meijer OC (2010). Disrupted corticosterone pulsatile patterns attenuate responsiveness to glucocorticoid signaling in rat brain. Endocrinology 151, 1177– 1186. Sato H, Horikawa Y, Iizuka K, Sakurai N, Tanaka T, Shihara N, Oshima A, Takeda J & Mikuni M (2008). Large-scale analysis of glucocorticoid target genes in rat hypothalamus. J Neurochem 106, 805– 814. Schuetz F, Kumar S, Poronnik P & Adams DJ (2008). Regulation of the voltage-gated K+ channels KCNQ2/3 and KCNQ3/5 by serum- and glucocorticoid-regulated kinase-1. Am J Physiol Cell Physiol 295, C73– C80. Scott V & Brown CH (2010). State-dependent plasticity in vasopressin neurones: dehydration-induced changes in activity patterning. J Neuroendocrinol 22, 343– 354. Sheng H, Sun T, Cong B, He P, Zhang Y, Yan J, Lu C & Ni X (2008). Corticotropin-releasing hormone stimulates SGK-1 kinase expression in cultured hippocampal neurons via CRH-R1. Am J Physiol Endocrinol Metab 295, E938– E946. Silver SM, Sterns RH & Halperin ML (1996). Brain swelling after dialysis: old urea or new osmoles Am J Kidney Dis 28, 1– 13. Tai DJC, Su CC, Ma YL & Lee EHY (2009). SGK1 phosphorylation of IκB kinase α and p300 up-regulates NF-κB activity and increases N-methyl-d-aspartate receptor NR2A and NR2B expression. J Biol Chem 284, 4073– 4089. Thornton SN (2010). Thirst and hydration: Physiology and consequences of dysfunction. Physiol Behav 100, 15– 21. Verkman AS (2005). Novel roles of aquaporins revealed by phenotype analysis of knockout mice. Rev Physiol Biochem Pharmacol 155, 31– 55. Wang D, Yan B, Rajapaksha WR & Fisher TE (2009). The expression of voltage-gated Ca2+ channels in pituicytes and the up-regulation of L-type Ca2+ channels during water deprivation. J Neuroendocrinol 21, 858– 866. Warby SC, Doty CN, Graham RK, Shively J, Singaraja RR & Hayden MR (2009). Phosphorylation of huntingtin reduces the accumulation of its nuclear fragments. Mol Cell Neurosci 40, 121– 127. Citing Literature Volume588, Issue18September 2010Pages 3349-3354 ReferencesRelatedInformation
最长约 10秒,即可获得该文献文件

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
濮阳傲易发布了新的文献求助10
2秒前
3秒前
愤怒的网络完成签到,获得积分10
5秒前
复杂函发布了新的文献求助10
9秒前
11秒前
11秒前
12秒前
14秒前
14秒前
秀丽香露发布了新的文献求助10
16秒前
Clara发布了新的文献求助10
16秒前
苏苏完成签到,获得积分10
17秒前
18秒前
wanci应助剥橘子高手采纳,获得30
18秒前
20秒前
韧迹发布了新的文献求助10
21秒前
禾禾发布了新的文献求助10
22秒前
复杂函完成签到,获得积分10
27秒前
2799完成签到,获得积分10
32秒前
王茹梦发布了新的文献求助10
33秒前
禾禾完成签到,获得积分20
33秒前
姬如雪儿完成签到 ,获得积分10
33秒前
可乐完成签到,获得积分10
34秒前
38秒前
42秒前
44秒前
46秒前
啦啦啦发布了新的文献求助10
47秒前
CodeCraft应助禾禾采纳,获得10
50秒前
Zzy完成签到 ,获得积分10
52秒前
56秒前
yumi关注了科研通微信公众号
58秒前
1分钟前
ding应助科研通管家采纳,获得10
1分钟前
1分钟前
SciGPT应助科研通管家采纳,获得10
1分钟前
1分钟前
yufanhui应助科研通管家采纳,获得30
1分钟前
小蘑菇应助科研通管家采纳,获得10
1分钟前
小准应助科研通管家采纳,获得10
1分钟前
高分求助中
请在求助之前详细阅读求助说明!!!! 20000
One Man Talking: Selected Essays of Shao Xunmei, 1929–1939 1000
The Three Stars Each: The Astrolabes and Related Texts 900
Yuwu Song, Biographical Dictionary of the People's Republic of China 700
[Lambert-Eaton syndrome without calcium channel autoantibodies] 520
Pressing the Fight: Print, Propaganda, and the Cold War 500
Bernd Ziesemer - Maos deutscher Topagent: Wie China die Bundesrepublik eroberte 500
热门求助领域 (近24小时)
化学 材料科学 医学 生物 有机化学 工程类 生物化学 纳米技术 物理 内科学 计算机科学 化学工程 复合材料 遗传学 基因 物理化学 催化作用 电极 光电子学 量子力学
热门帖子
关注 科研通微信公众号,转发送积分 2471257
求助须知:如何正确求助?哪些是违规求助? 2137961
关于积分的说明 5447789
捐赠科研通 1861848
什么是DOI,文献DOI怎么找? 925987
版权声明 562740
科研通“疑难数据库(出版商)”最低求助积分说明 495302