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

Publication year range
1.
Mol Cell ; 60(2): 195-207, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26474064

ABSTRACT

Cancer cells adapt metabolically to proliferate under nutrient limitation. Here we used combined transcriptional-metabolomic network analysis to identify metabolic pathways that support glucose-independent tumor cell proliferation. We found that glucose deprivation stimulated re-wiring of the tricarboxylic acid (TCA) cycle and early steps of gluconeogenesis to promote glucose-independent cell proliferation. Glucose limitation promoted the production of phosphoenolpyruvate (PEP) from glutamine via the activity of mitochondrial PEP-carboxykinase (PCK2). Under these conditions, glutamine-derived PEP was used to fuel biosynthetic pathways normally sustained by glucose, including serine and purine biosynthesis. PCK2 expression was required to maintain tumor cell proliferation under limited-glucose conditions in vitro and tumor growth in vivo. Elevated PCK2 expression is observed in several human tumor types and enriched in tumor tissue from non-small-cell lung cancer (NSCLC) patients. Our results define a role for PCK2 in cancer cell metabolic reprogramming that promotes glucose-independent cell growth and metabolic stress resistance in human tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Gene Expression Regulation, Neoplastic , Gluconeogenesis/genetics , Lung Neoplasms/metabolism , Neoplasms/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Adaptation, Physiological/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Citric Acid Cycle/genetics , Glucose/deficiency , Glutamine/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Metabolomics , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/pathology , Phosphoenolpyruvate/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Purines/biosynthesis , Pyruvic Acid/metabolism , Serine/biosynthesis
2.
Biochem J ; 475(18): 2969-2983, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30135087

ABSTRACT

AMP-activated protein kinase (AMPK) is a key regulator of cellular and systemic energy homeostasis which achieves this through the phosphorylation of a myriad of downstream targets. One target is TBC1D1 a Rab-GTPase-activating protein that regulates glucose uptake in muscle cells by integrating insulin signalling with that promoted by muscle contraction. Ser237 in TBC1D1 is a target for phosphorylation by AMPK, an event which may be important in regulating glucose uptake. Here, we show AMPK heterotrimers containing the α1, but not the α2, isoform of the catalytic subunit form an unusual and stable association with TBC1D1, but not its paralogue AS160. The interaction between the two proteins is direct, involves a dual interaction mechanism employing both phosphotyrosine-binding (PTB) domains of TBC1D1 and is increased by two different pharmacological activators of AMPK (AICAR and A769962). The interaction enhances the efficiency by which AMPK phosphorylates TBC1D1 on its key regulatory site, Ser237 Furthermore, the interaction is reduced by a naturally occurring R125W mutation in the PTB1 domain of TBC1D1, previously found to be associated with severe familial obesity in females, with a concomitant reduction in Ser237 phosphorylation. Our observations provide evidence for a functional difference between AMPK α-subunits and extend the repertoire of protein kinases that interact with substrates via stabilisation mechanisms that modify the efficacy of substrate phosphorylation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , GTPase-Activating Proteins/metabolism , Mutation, Missense , Obesity/enzymology , AMP-Activated Protein Kinases/genetics , Amino Acid Substitution , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Female , GTPase-Activating Proteins/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Transgenic , Obesity/genetics , Phosphorylation , Ribonucleotides/genetics , Ribonucleotides/metabolism , Sex Characteristics
3.
J Cell Sci ; 126(Pt 9): 1931-41, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23444368

ABSTRACT

Insulin enhances the uptake of glucose into adipocytes and muscle cells by promoting the redistribution of the glucose transporter isoform 4 (GLUT4) from intracellular compartments to the cell surface. Rab GTPases regulate the trafficking itinerary of GLUT4 and several have been found on immunopurified GLUT4 vesicles. Specifically, Rab14 has previously been implicated in GLUT4 trafficking in muscle although its role, if any, in adipocytes is poorly understood. Analysis of 3T3-L1 adipocytes using confocal microscopy demonstrated that endogenous GLUT4 and endogenous Rab14 exhibited a partial colocalisation. However, when wild-type Rab14 or a constitutively-active Rab14Q70L mutant were overexpressed in these cells, the colocalisation with both GLUT4 and IRAP became extensive. Interestingly, this colocalisation was restricted to enlarged 'ring-like' vesicular structures (mean diameter 1.3 µm), which were observed in the presence of overexpressed wild-type Rab14 and Rab14Q70L, but not an inactive Rab14S25N mutant. These enlarged vesicles contained markers of early endosomes and were rapidly filled by GLUT4 and transferrin undergoing endocytosis from the plasma membrane. The Rab14Q70L mutant reduced basal and insulin-stimulated cell surface GLUT4 levels, probably by retaining GLUT4 in an insulin-insensitive early endosomal compartment. Furthermore, shRNA-mediated depletion of Rab14 inhibited the transit of GLUT4 through early endosomal compartments towards vesicles and tubules in the perinuclear region. Given the previously reported role of Rab14 in trafficking between endosomes and the Golgi complex, we propose that the primary role of Rab14 in GLUT4 trafficking is to control the transit of internalised GLUT4 from early endosomes into the Golgi complex, rather than direct GLUT4 translocation to the plasma membrane.


Subject(s)
Adipocytes/metabolism , Cell Membrane/metabolism , Endosomes/metabolism , Glucose Transporter Type 4/metabolism , Golgi Apparatus/metabolism , rab GTP-Binding Proteins/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Amino Acid Substitution , Animals , Cell Membrane/genetics , Endocytosis/drug effects , Endocytosis/physiology , Endosomes/genetics , Glucose Transporter Type 4/genetics , Golgi Apparatus/genetics , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Mice , Mutation, Missense , Protein Transport/physiology , rab GTP-Binding Proteins/genetics
4.
Biochem J ; 455(2): 195-206, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23905686

ABSTRACT

PIKfyve (FYVE domain-containing phosphatidylinositol 3-phosphate 5-kinase), the lipid kinase that phosphorylates PtdIns3P to PtdIns(3,5)P2, has been implicated in insulin-stimulated glucose uptake. We investigated whether PIKfyve could also be involved in contraction/AMPK (AMP-activated protein kinase)-stimulated glucose uptake in skeletal muscle. Incubation of rat epitrochlearis muscles with YM201636, a selective PIKfyve inhibitor, reduced contraction- and AICAriboside (5-amino-4-imidazolecarboxamide riboside)-stimulated glucose uptake. Consistently, PIKfyve knockdown in C2C12 myotubes reduced AICAriboside-stimulated glucose transport. Furthermore, muscle contraction increased PtdIns(3,5)P2 levels and PIKfyve phosphorylation. AMPK phosphorylated PIKfyve at Ser307 both in vitro and in intact cells. Following subcellular fractionation, PIKfyve recovery in a crude intracellular membrane fraction was increased in contracting versus resting muscles. Also in opossum kidney cells, wild-type, but not S307A mutant, PIKfyve was recruited to endosomal vesicles in response to AMPK activation. We propose that PIKfyve activity is required for the stimulation of skeletal muscle glucose uptake by contraction/AMPK activation. PIKfyve is a new AMPK substrate whose phosphorylation at Ser307 could promote PIKfyve translocation to endosomes for PtdIns(3,5)P2 synthesis to facilitate GLUT4 (glucose transporter 4) translocation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinase/metabolism , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Cell Line , Glucose Transporter Type 4/metabolism , Humans , Insulin/metabolism , Male , Opossums , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Rats , Rats, Wistar
5.
FASEB J ; 26(2): 513-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22002906

ABSTRACT

Inward rectifier potassium channels of the Kir2 subfamily are important determinants of the electrical activity of brain and muscle cells. Genetic mutations in Kir2.1 associate with Andersen-Tawil syndrome (ATS), a familial disorder leading to stress-triggered periodic paralysis and ventricular arrhythmia. To identify the molecular mechanisms of this stress trigger, we analyze Kir channel function and localization electrophysiologically and by time-resolved confocal microscopy. Furthermore, we employ a mathematical model of muscular membrane potential. We identify a novel corticoid signaling pathway that, when activated by glucocorticoids, leads to enrichment of Kir2 channels in the plasma membranes of mammalian cell lines and isolated cardiac and skeletal muscle cells. We further demonstrate that activation of this pathway can either partly restore (40% of cases) or further impair (20% of cases) the function of mutant ATS channels, depending on the particular Kir2.1 mutation. This means that glucocorticoid treatment might either alleviate or deteriorate symptoms of ATS depending on the patient's individual Kir2.1 genotype. Thus, our findings provide a possible explanation for the contradictory effects of glucocorticoid treatment on symptoms in patients with ATS and may open new pathways for the design of personalized medicines in ATS therapy.


Subject(s)
Andersen Syndrome/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Andersen Syndrome/drug therapy , Andersen Syndrome/genetics , Animals , Female , Glucocorticoids/therapeutic use , Guinea Pigs , HEK293 Cells , HeLa Cells , Humans , Immediate-Early Proteins/metabolism , In Vitro Techniques , Mutant Proteins/genetics , Mutant Proteins/metabolism , Myocytes, Cardiac/metabolism , Oocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Stress, Physiological , Xenopus laevis
6.
J Biol Chem ; 286(15): 13647-56, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21335550

ABSTRACT

The Forkhead box transcription factor FoxO1 regulates metabolic gene expression in mammals. FoxO1 activity is tightly controlled by phosphatidylinositol 3-kinase (PI3K) signaling, resulting in its phosphorylation and nuclear exclusion. We sought here to determine the mechanisms involved in glucose and insulin-stimulated nuclear shuttling of FoxO1 in pancreatic ß cells and its consequences for preproinsulin (Ins1, Ins2) gene expression. Nuclear-localized endogenous FoxO1 translocated to the cytosol in response to elevated glucose (3 versus 16.7 mM) in human islet ß cells. Real-time confocal imaging of nucleo-cytosolic shuttling of a FoxO1-EGFP chimera in primary mouse and clonal MIN6 ß cells revealed a time-dependent glucose-responsive nuclear export, also mimicked by exogenous insulin, and blocked by suppressing insulin secretion. Constitutively active PI3K or protein kinase B/Akt exerted similar effects, while inhibitors of PI3K, but not of glycogen synthase kinase-3 or p70 S6 kinase, blocked nuclear export. FoxO1 overexpression reversed the activation by glucose of pancreatic duodenum homeobox-1 (Pdx1) transcription. Silencing of FoxO1 significantly elevated the expression of mouse Ins2, but not Ins1, mRNA at 3 mM glucose. Putative FoxO1 binding sites were identified in the distal promoter of rodent Ins2 genes and direct binding of FoxO1 to the Ins2 promoter was demonstrated by chromatin immunoprecipitation. A 915-bp glucose-responsive Ins2 promoter was inhibited by constitutively active FoxO1, an effect unaltered by simultaneous overexpression of PDX1. We conclude that nuclear import of FoxO1 contributes to the suppression of Pdx1 and Ins2 gene expression at low glucose, the latter via a previously unsuspected and direct physical interaction with the Ins2 promoter.


Subject(s)
Cell Nucleus/metabolism , Cytosol/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/physiology , Insulin-Secreting Cells/metabolism , Insulin/biosynthesis , Response Elements/physiology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Cell Line , Cell Nucleus/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Glucose/metabolism , Glucose/pharmacology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sweetening Agents/metabolism , Sweetening Agents/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Cell Physiol Biochem ; 25(2-3): 187-94, 2010.
Article in English | MEDLINE | ID: mdl-20110679

ABSTRACT

The excitatory amino-acid transporter EAAT4 (SLC1A6), a Na(+),glutamate cotransporter expressed mainly in Purkinje cells, serves to clear glutamate from the synaptic cleft. EAAT4 activity is stimulated by the serum and glucocorticoid inducible kinase SGK1. SGK1-dependent regulation of the Na(+),glucose transporter SGLT1 (SLC5A1) and the creatine transporter CreaT (SLC6A8) has recently been shown to involve the mammalian phosphatidylinositol-3-phosphate-5-kinase PIKfyve (PIP5K3). The present experiments thus explored whether SGK1-dependent EAAT4-regulation similarly involves PIKfyve. In Xenopus oocytes expressing EAAT4, but not in water injected oocytes, glutamate induced a current which was significantly enhanced by coexpression of PIKfyve and SGK1. The glutamate induced current in Xenopus oocytes coexpressing EAAT4 and both, PIKfyve and SGK1, was significantly larger than the current in Xenopus oocytes expressing EAAT4 together with either kinase alone. Coexpression of the inactive SGK1 mutant (K127N)SGK1 did not significantly alter glutamate induced current in EAAT4-expressing Xenopus oocytes and abolished the stimulation of glutamate induced current by coexpression of PIKfyve. The stimulating effect of PIKfyve was abrogated by replacement of the serine with alanine in the SGK consensus sequence ((S318A)PIKfyve). Furthermore, coexpression of (S318A)PIKfyve significantly blunted the stimulating effect of SGK1 on EAAT4 activity. The observations disclose that PIKfyve indeed participates in the regulation of EAAT4.


Subject(s)
Excitatory Amino Acid Transporter 4/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Electrophysiology , Glutamic Acid/pharmacology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , Oocytes/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Xenopus laevis/metabolism
8.
Biochem Biophys Res Commun ; 397(4): 650-5, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20513353

ABSTRACT

PIKfyve is a protein and lipid kinase that plays an important role in membrane trafficking, including TGN to endosome retrograde sorting and in insulin-stimulated translocation of the GLUT4 glucose transporter from intracellular storage vesicles to the plasma membrane. We have previously demonstrated that PIKfyve is phosphorylated in response to insulin in a PI3-kinase and protein kinase B (PKB)-dependent manner. However, it has been implied that this was not due to direct phosphorylation of PIKfyve by PKB, but as a result of an insulin-induced PIKfyve autophosphorylation event. Here we demonstrate that purified PIKfyve is phosphorylated in vitro by a recombinant active PKB on two separate serine residues, S318 and S105, which flank the N-terminal FYVE domain of the protein. Only S318, however, becomes phosphorylated in intact cells stimulated with insulin. We further demonstrate that S318 is phosphorylated in response to hyperosmotic stress in a PI3-kinase- and PKB-independent manner. Importantly, the effects of insulin and sorbitol were not prevented by the presence of an ATP-competitive PIKfyve inhibitor (YM20163) or in a mutant PIKfyve lacking both lipid and protein kinase activity. Our results confirm, therefore, that PIKfyve is directly phosphorylated by PKB on a single serine residue in response to insulin and are not due to autophosphorylation of the enzyme. We further reveal that two stimuli known to promote glucose uptake in cells, both stimulate phosphorylation of PIKfyve on S318 but via distinct signal transduction pathways.


Subject(s)
Insulin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , CHO Cells , Cricetinae , Cricetulus , Insulin/pharmacology , Mice , Osmotic Pressure , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/genetics , Serine/metabolism , Sorbitol/pharmacology
9.
Circ Res ; 103(12): 1451-7, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-19008479

ABSTRACT

Physical and emotional stress is accompanied by release of stress hormones such as the glucocorticoid cortisol. This hormone upregulates the serum- and glucocorticoid-inducible kinase (SGK)1, which in turn stimulates I(Ks), a slow delayed rectifier potassium current that mediates cardiac action potential repolarization. Mutations in I(Ks) channel alpha (KCNQ1, KvLQT1, Kv7.1) or beta (KCNE1, IsK, minK) subunits cause long QT syndrome (LQTS), an inherited cardiac arrhythmia associated with increased risk of sudden death. Together with the GTPases RAB5 and RAB11, SGK1 facilitates membrane recycling of KCNQ1 channels. Here, we show altered SGK1-dependent regulation of LQTS-associated mutant I(Ks) channels. Whereas some mutant KCNQ1 channels had reduced basal activity but were still activated by SGK1, currents mediated by KCNQ1(Y111C) or KCNQ1(L114P) were paradoxically reduced by SGK1. Heteromeric channels coassembled of wild-type KCNQ1 and the LQTS-associated KCNE1(D76N) mutant were similarly downregulated by SGK1 because of a disrupted RAB11-dependent recycling. Mutagenesis experiments indicate that stimulation of I(Ks) channels by SGK1 depends on residues H73, N75, D76, and P77 in KCNE1. Identification of the I(Ks) recycling pathway and its modulation by stress-stimulated SGK1 provides novel mechanistic insight into potentially fatal cardiac arrhythmias triggered by physical or psychological stress.


Subject(s)
Endosomes/genetics , KCNQ1 Potassium Channel/genetics , Long QT Syndrome/genetics , Long QT Syndrome/metabolism , Mutation/genetics , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Voltage-Gated/genetics , Xenopus Proteins/genetics , Animals , COS Cells , Chlorocebus aethiops , Endosomes/metabolism , Female , KCNQ1 Potassium Channel/physiology , Oocytes/metabolism , Potassium Channels, Inwardly Rectifying/physiology , Potassium Channels, Voltage-Gated/physiology , Protein Subunits/genetics , Protein Subunits/physiology , Xenopus Proteins/physiology , Xenopus laevis
10.
Sci Rep ; 10(1): 17953, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087848

ABSTRACT

Proteins involved in the spaciotemporal regulation of GLUT4 trafficking represent potential therapeutic targets for the treatment of insulin resistance and type 2 diabetes. A key regulator of insulin- and exercise-stimulated glucose uptake and GLUT4 trafficking is TBC1D1. This study aimed to identify proteins that regulate GLUT4 trafficking and homeostasis via TBC1D1. Using an unbiased quantitative proteomics approach, we identified proteins that interact with TBC1D1 in C2C12 myotubes including VPS13A and VPS13C, the Rab binding proteins EHBP1L1 and MICAL1, and the calcium pump SERCA1. These proteins associate with TBC1D1 via its phosphotyrosine binding (PTB) domains and their interactions with TBC1D1 were unaffected by AMPK activation, distinguishing them from the AMPK regulated interaction between TBC1D1 and AMPKα1 complexes. Depletion of VPS13A or VPS13C caused a post-transcriptional increase in cellular GLUT4 protein and enhanced cell surface GLUT4 levels in response to AMPK activation. The phenomenon was specific to GLUT4 because other recycling proteins were unaffected. Our results provide further support for a role of the TBC1D1 PTB domains as a scaffold for a range of Rab regulators, and also the VPS13 family of proteins which have been previously linked to fasting glycaemic traits and insulin resistance in genome wide association studies.


Subject(s)
GTPase-Activating Proteins/pharmacology , Glucose Transporter Type 4/metabolism , Homeostasis/drug effects , Homeostasis/genetics , Muscle Fibers, Skeletal/metabolism , Proteins/pharmacology , Vesicular Transport Proteins/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Type 2 , GTPase-Activating Proteins/physiology , HEK293 Cells , Humans , Insulin Resistance , Male , Mice, Transgenic , Proteins/physiology , Vesicular Transport Proteins/physiology
11.
Cell Physiol Biochem ; 24(5-6): 361-8, 2009.
Article in English | MEDLINE | ID: mdl-19910676

ABSTRACT

The Na(+),glutamate cotransporter EAAT2 is expressed in astrocytes and clears glutamate from the synaptic cleft. EAAT2 dependent currrent is stimulated by the serum and glucocorticoid inducible kinase SGK1. Phosphorylation targets of SGK1 include the human phosphatidylinositol-3-phosphate-5-kinase PIKfyve (PIP5K3). Nothing is known, however, on the role of PIKfyve in the regulation of EAAT2. The present experiments thus explored, whether PIKfyve expression modifies EAAT2 dependent currrent and protein abundance in the cell membrane. In Xenopus oocytes expressing EAAT2 but not in water injected oocytes application of glutamate (2 mM) induced an inward current (I(glu)). Coexpression of either, SGK1 or PIKfyve, significantly enhanced I(glu) in EAAT2 expressing oocytes. I(glu) was significantly higher in Xenopus oocytes coexpressing EAAT2, SGK1 and PIKfyve than in Xenopus oocytes expressing EAAT2 and either, SGK1 or PIKfyve, alone. Additional coexpression of the inactive mutant of the serum and glucocorticoid inducible kinase (K127N)SGK1 did not significantly alter I(glu) in EAAT2 expressing oocytes and significantly decreased I(glu) in oocytes coexpressing EAAT2 together with PIKfyve. The stimulating effect of PIKfyve on I(glu) was abrogated by replacement of the serine in the SGK consensus sequence by alanine ((S318A)PIKfyve). Furthermore, additional coexpression of (S318A)PIKfyve virtually abolished I(glu) in Xenopus oocytes coexpressing SGK1 and EAAT2. Confocal microscopy reveals that PIKfyve enhances the EAAT2 protein abundance in the cell membrane. The observations disclose that PIKfyve indeed participates in the regulation of EAAT2.


Subject(s)
Excitatory Amino Acid Transporter 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Amino Acid Substitution , Animals , Astrocytes/metabolism , Electrophysiological Phenomena , Glutamic Acid/pharmacology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mutagenesis, Site-Directed , Oocytes/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Xenopus
12.
Biochem Biophys Res Commun ; 390(3): 952-7, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19852935

ABSTRACT

The cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP-activated Cl(-) channel critically important in Cl(-) secreting epithelia. Mutations in the CFTR gene, such as (DeltaF508)CFTR leads to cystic fibrosis, a severe disease with defective Cl(-) secretion. CFTR is stimulated by the serum and glucocorticoid-inducible kinase SGK1. The SGK1 dependent regulation of several carriers and channels involves the phosphatidylinositol-3-phosphate-5-kinase PIKfyve, which similarly mediates the regulation of glucose carriers by PKB/Akt. The present study was thus performed to elucidate whether PKB/Akt and PIKfyve are regulators of CFTR. To this end CFTR or (DeltaF508)CFTR were expressed in Xenopus oocytes alone or together with PKB, PIKfyve or the SGK1/PKB resistant mutant (S318A)PIKfyve, and the current generated by cAMP upregulation with 10muM forskolin+1mM IBMX determined utilizing dual electrode voltage clamp. As a result, forskolin/IBMX treatment triggered a current (I(cAMP)) in CFTR-expressing Xenopus oocytes, but not in oocytes expressing (DeltaF508)CFTR. Coexpression of PKB/Akt and PIKfyve, but not of (S318A)PIKfyve, stimulated I(cAMP) in CFTR-expressing ( approximately 2- to 3-fold) but not in (DeltaF508)CFTR-expressing or water injected Xenopus oocytes. Immunohistochemistry revealed that the coexpression of PIKfyve, but not of (S318A)PIKfyve, enhanced the CFTR protein abundance but not the (DeltaF508)CFTR protein abundance in CFTR or (DeltaF508)CFTR-expressing oocytes. The present observations reveal a novel powerful regulator of intact but not of defective CFTR.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Phosphatidylinositol 3-Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Humans , Oocytes , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Two-Hybrid System Techniques , Up-Regulation , Xenopus laevis
13.
Circ Res ; 100(5): 686-92, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17293474

ABSTRACT

Stress-dependent regulation of cardiac action potential duration is mediated by the sympathetic nervous system and the hypothalamic-pituitary-adrenal axis. It is accompanied by an increased magnitude of the slow outward potassium ion current, I(Ks). KCNQ1 and KCNE1 subunits coassemble to form the I(Ks) channel. Mutations in either subunit cause long QT syndrome, an inherited cardiac arrhythmia associated with an increased risk of sudden cardiac death. Here we demonstrate that exocytosis of KCNQ1 proteins to the plasma membrane requires the small GTPase RAB11, whereas endocytosis is dependent on RAB5. We further demonstrate that RAB-dependent KCNQ1/KCNE1 exocytosis is enhanced by the serum- and glucocorticoid-inducible kinase 1, and requires phosphorylation and activation of phosphoinositide 3-phosphate 5-kinase and the generation of PI(3,5)P(2). Identification of KCNQ1/KCNE1 recycling and its modulation by serum- and glucocorticoid-inducible kinase 1-phosphoinositide 3-phosphate 5-kinase -PI(3,5)P(2) provides a mechanistic insight into stress-induced acceleration of cardiac repolarization.


Subject(s)
Endocytosis/physiology , KCNQ1 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism , Transport Vesicles/metabolism , Animals , COS Cells , Chlorocebus aethiops , Exocytosis/physiology , Female , Ion Channel Gating/physiology , Protein Transport/physiology , Xenopus
14.
Nephrol Dial Transplant ; 24(11): 3288-96, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19556298

ABSTRACT

BACKGROUND: Cellular insulin resistance is the hallmark of type 2 diabetes and predominantly affects adipose and muscle cells. The saturated free fatty acid palmitate is elevated in insulin-resistant states and may directly contribute to cellular insulin resistance. A spectrum of renal disease is associated with increased markers of insulin resistance, although direct causal mechanisms are not known. In the kidney, glomerular podocytes are novel insulin-sensitive cells that have the ability to rapidly transport glucose. In this study, we tested the hypothesis that palmitate would induce insulin resistance in podocytes. METHODS: Conditionally immortalized human podocytes were cultured for up to 24 h with 375-750 muM palmitate. Functional effects on glucose uptake and ceramide production were measured. Gene expression was investigated using a focused gene array, and protein signalling and trafficking were studied with Western blotting and immunofluorescence. RESULTS: We found that palmitate blocked insulin-stimulated glucose uptake in human podocytes. This was associated with increased ceramide production, and use of the ceramide inhibitors myriocin and fumonisin B1 partially recovered the insulin sensitivity. At the level of transcription, palmitate downregulated genes associated with several pathways involved in insulin signalling. At the protein level, phosphorylation of the insulin receptor, IRS1 and PKB was reduced and there was impaired translocation of GLUT4 to the cell surface. CONCLUSION: This is the first study to demonstrate a direct effect of saturated fatty acids on podocyte function. These findings may represent a novel link between systemic insulin resistance and the development of nephropathy.


Subject(s)
Diabetic Nephropathies/etiology , Fatty Acids/pharmacology , Insulin Resistance , Podocytes/drug effects , Adenosine Triphosphate/analysis , Cell Line , Cell Survival/drug effects , Ceramides/biosynthesis , Gene Expression Profiling , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Humans , Palmitic Acid/pharmacology , Podocytes/physiology , Signal Transduction/drug effects
15.
PLoS One ; 14(4): e0214610, 2019.
Article in English | MEDLINE | ID: mdl-30969984

ABSTRACT

Glycogen synthase kinase-3 (GSK3) is over-expressed and hyperactivated in non-small cell lung carcinoma (NSCLC) and plays a role in ensuring the correct alignment of chromosomes on the metaphase plate during mitosis through regulation of microtubule stability. This makes the enzyme an attractive target for cancer therapy. We examined the effects of a selective cell-permeant GSK3 inhibitor (CHIR99021), used alone or in combination with paclitaxel, using an in vitro cell growth assay, a quantitative chromosome alignment assay, and a tumor xenograft model. CHIR99021 inhibits the growth of human H1975 and H1299 NSCLC cell lines in a synergistic manner with paclitaxel. CHIR99021 and paclitaxel promoted a synergistic defect in chromosomal alignment when compared to each compound administered as monotherapy. Furthermore, we corroborated our in vitro findings in a mouse tumor xenograft model. Our results demonstrate that a GSK3 inhibitor and paclitaxel act synergistically to inhibit the growth of NSCLC cells in vitro and in vivo via a mechanism that may involve converging modes of action on microtubule spindle stability and thus chromosomal alignment during metaphase. Our findings provide novel support for the use of the GSK3 inhibitor, CHIR99021, alongside taxol-based chemotherapy in the treatment of human lung cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Glycogen Synthase Kinase 3/metabolism , Lung Neoplasms/drug therapy , Paclitaxel/therapeutic use , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chromosome Aberrations/drug effects , Drug Synergism , Drug Therapy, Combination , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Humans , Male , Mice , Mice, Nude , Paclitaxel/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism
16.
BMC Cell Biol ; 9: 25, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18492238

ABSTRACT

BACKGROUND: Insulin stimulates glucose uptake by adipocytes through increasing translocation of the glucose transporter GLUT4 from an intracellular compartment to the plasma membrane. Fusion of GLUT4-containing vesicles at the cell surface is thought to involve phospholipase D activity, generating the signalling lipid phosphatidic acid, although the mechanism of action is not yet clear. RESULTS: Here we report the identification of a putative phosphatidic acid-binding motif in a GLUT4 intracellular loop. Mutation of this motif causes a decrease in the insulin-induced exposure of GLUT4 at the cell surface of 3T3-L1 adipocytes via an effect on vesicle fusion. CONCLUSION: The potential phosphatidic acid-binding motif identified in this study is unique to GLUT4 among the sugar transporters, therefore this motif may provide a unique mechanism for regulating insulin-induced translocation by phospholipase D signalling.


Subject(s)
Amino Acid Motifs/genetics , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , 3T3-L1 Cells , Adipocytes , Animals , Biological Transport, Active , Cytoplasmic Vesicles/genetics , Cytoplasmic Vesicles/metabolism , Glucose/metabolism , Glucose Transporter Type 4/chemistry , Insulin/metabolism , Membrane Fusion , Mice , Mutagenesis , Mutant Proteins , Mutation , Peptide Library , Transfection
17.
Diabetes ; 56(4): 1127-35, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17395751

ABSTRACT

The leading causes of albuminuria and end-stage renal failure are secondary to abnormalities in the production or cellular action of insulin, including diabetes and hyperinsulinemic metabolic syndrome. The human glomerular podocyte is a critical cell for maintaining the filtration barrier of the kidney and preventing albuminuria. We have recently shown this cell to be insulin sensitive with respect to glucose uptake, with kinetics similar to muscle cells. We now show that the podocyte protein nephrin is essential for this process. Conditionally immortalized podocytes from two different patients with nephrin mutations (natural human nephrin mutant models) were unresponsive to insulin. Knocking nephrin down with siRNA in wild-type podocytes abrogated the insulin response, and stable nephrin transfection of nephrin-deficient podocytes rescued their insulin response. Mechanistically, we show that nephrin allows the GLUT1- and GLUT4-rich vesicles to fuse with the membrane of this cell. Furthermore, we show that the COOH of nephrin interacts with the vesicular SNARE protein VAMP2 in vitro and ex vivo (using yeast-2 hybrid and coimmunoprecipitation studies). This work demonstrates a previously unsuspected role of nephrin in vesicular docking and insulin responsiveness of podocytes.


Subject(s)
Kidney Glomerulus/physiology , Membrane Proteins/physiology , Podocytes/physiology , Biological Transport , Child , Deoxyglucose/metabolism , Humans , Kidney Failure, Chronic/physiopathology , Kidney Glomerulus/physiopathology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Nephrotic Syndrome/physiopathology , RNA, Small Interfering/genetics , Vesicle-Associated Membrane Protein 2/physiology
18.
Diabetes ; 55(5): 1300-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16644686

ABSTRACT

Insulin stimulates glucose transport in skeletal muscle by glucose transporter GLUT4 translocation to sarcolemma and membrane invaginations, the t-tubules. Although muscle glucose uptake plays a key role in insulin resistance and type 2 diabetes, the dynamics of GLUT4 translocation and the signaling involved are not well described. We have now developed a confocal imaging technique to follow trafficking of green fluorescent protein-labeled proteins in living muscle fibers in situ in anesthetized mice. Using this technique, by imaging the dynamics of GLUT4 translocation and phosphatidylinositol 3,4,5 P(3) (PIP(3)) production in response to insulin, here, for the first time, we delineate the temporal and spatial distribution of these processes in a living animal. We find a 10-min delay of maximal GLUT4 recruitment and translocation to t-tubules compared with sarcolemma. Time-lapse imaging of a fluorescent dye after intravenous injection shows that this delay is similar to the time needed for insulin diffusion into the t-tubule system. Correspondingly, immunostaining of muscle fibers shows that insulin receptors are present throughout the t-tubule system. Finally, PIP(3) production, an early event in insulin signaling, progresses slowly along the t-tubules with a 10-min delay between maximal PIP(3) production at sarcolemma compared with deep t-tubules following the appearance of dye-labeled insulin. Our findings in living mice indicate a major role of the t-tubules in insulin signaling in skeletal muscle and show a diffusion-associated delay in insulin action between sarcolemma and inner t-tubules.


Subject(s)
Insulin/pharmacology , Muscle, Skeletal/physiology , Animals , Biological Transport , Fluorescent Dyes , Glucose Transporter Type 4/metabolism , Kinetics , Mice , Microscopy, Confocal , Microtubules/metabolism , Phosphatidylinositol Phosphates/metabolism , Receptor, Insulin/metabolism , Sarcolemma/metabolism
19.
Diabetes ; 54(11): 3095-102, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16249431

ABSTRACT

Microalbuminuria is significant both as the earliest stage of diabetic nephropathy and as an independent cardiovascular risk factor in nondiabetic subjects, in whom it is associated with insulin resistance. The link between disorders of cellular insulin metabolism and albuminuria has been elusive. Here, we report using novel conditionally immortalized human podocytes in vitro and human glomeruli ex vivo that the podocyte, the principal cell responsible for prevention of urinary protein loss, is insulin responsive and able to approximately double its glucose uptake within 15 min of insulin stimulation. Conditionally immortalized human glomerular endothelial cells do not respond to insulin, suggesting that insulin has a specific effect on the podocyte in the glomerular filtration barrier. The insulin response of the podocyte occurs via the facilitative glucose transporters GLUT1 and GLUT4, and this process is dependent on the filamentous actin cytoskeleton. Insulin responsiveness in this key structural component of the glomerular filtration barrier may have central relevance for understanding of diabetic nephropathy and for the association of albuminuria with states of insulin resistance.


Subject(s)
Insulin/pharmacology , Podocytes/drug effects , Podocytes/metabolism , Actins/metabolism , Animals , Biological Transport/drug effects , Cell Line , Cytoskeleton , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Gene Silencing , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Mice , Phosphorylation/drug effects , Podocytes/ultrastructure , Protein Transport , Signal Transduction/drug effects
20.
Biochem J ; 388(Pt 2): 713-20, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15713122

ABSTRACT

Serine and threonine phosphorylation of IRS-1 (insulin receptor substrate-1) has been reported to decrease its ability to be tyrosine-phosphorylated by the insulin receptor. Insulin itself may negatively regulate tyrosine phosphorylation of IRS-1 through a PI3K (phosphoinositide 3-kinase)-dependent feedback pathway. In the present study, we examined the regulation and role of IRS-1 serine phosphorylation in the modulation of IRS-1 tyrosine phosphorylation in physiologically relevant cells, namely freshly isolated primary adipocytes. We show that insulin-stimulated phosphorylation of Ser312 and Ser616 in IRS-1 was relatively slow, with maximal phosphorylation achieved after 20 and 5 min respectively. The effect of insulin on phosphorylation of both these sites required the activation of PI3K and the MAPKs (mitogen-activated protein kinases) ERK1/2 (extracellular-signal-regulated kinase 1 and 2), but not the activation of mTOR (mammalian target of rapamycin)/p70S6 kinase, JNK (c-Jun N-terminal kinase) or p38MAPK. Although inhibition of PI3K and ERK1/2 both substantially decreased insulin-stimulated phosphorylation of Ser312 and Ser616, only wortmannin enhanced insulin-stimulated tyrosine phosphorylation of IRS-1. Furthermore, inhibition of mTOR/p70S6 kinase, JNK or p38MAPK had no effect on insulin-stimulated IRS-1 tyrosine phosphorylation. The differential effect of inhibition of ERK1/2 on insulin-stimulated IRS-1 phosphorylation of Ser312/Ser616 and tyrosine indicates that these events are independent of each other and that phosphorylation of Ser312/Ser616 is not responsible for the negative regulation of IRS-1 tyrosine phosphorylation mediated by PI3K in primary adipocytes.


Subject(s)
Adipocytes/metabolism , Phosphoproteins/metabolism , Androstadienes/pharmacology , Animals , Butadienes/pharmacology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , Insulin/physiology , Insulin Receptor Substrate Proteins , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase 4 , Male , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Nitriles/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Time Factors , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL