Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Proc Natl Acad Sci U S A ; 117(33): 20292-20297, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32747529

ABSTRACT

Nuclear Factor of Activated T cells 5 (NFAT5) is a transcription factor (TF) that mediates protection from adverse effects of hypertonicity by increasing transcription of genes, including those that lead to cellular accumulation of protective organic osmolytes. NFAT5 has three intrinsically ordered (ID) activation domains (ADs). Using the NFAT5 N-terminal domain (NTD), which contains AD1, as a model, we demonstrate by biophysical methods that the NTD senses osmolytes and hypertonicity, resulting in stabilization of its ID regions. In the presence of sufficient NaCl or osmolytes, trehalose and sorbitol, the NFAT5 NTD undergoes a disorder-to-order shift, adopting higher average secondary and tertiary structure. Thus, NFAT5 is activated by the stress that it protects against. In its salt and/or osmolyte-induced more ordered conformation, the NTD interacts with several proteins, including HMGI-C, which is known to protect against apoptosis. These findings raise the possibility that the increased intracellular ionic strength and elevated osmolytes caused by hypertonicity activate and stabilize NFAT5.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Transcription Factors/chemistry , Escherichia coli/metabolism , Osmotic Pressure , Protein Binding , Protein Folding , Sodium Chloride , Sorbitol , Transcription Factors/metabolism , Trehalose
2.
Physiol Genomics ; 48(11): 835-849, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27764768

ABSTRACT

NFAT5 is a transcription factor originally identified because it is activated by hypertonicity and that activation increases expression of genes that protect against the adverse effects of the hypertonicity. However, its targets also include genes not obviously related to tonicity. The transactivating domain of NFAT5 is contained in its COOH-terminal region, which is predicted to be unstructured. Unstructured regions are common in transcription factors particularly in transactivating domains where they can bind co-regulatory proteins essential to their function. To identify potential binding partners of NFAT5 from either cytoplasmic or nuclear HEK293 cell extracts, we used peptide affinity chromatography followed by mass spectrometry. Peptide aptamer-baits consisted of overlapping 20 amino acid peptides within the predicted COOH-terminal unstructured region of NFAT5. We identify a total of 351 unique protein preys that associate with at least one COOH-terminal peptide bait from NFAT5 in either cytoplasmic or nuclear extracts from cells incubated at various tonicities (NaCl varied). In addition to finding many proteins already known to associate with NFAT5, we found many new ones whose function suggest novel aspects of NFAT5 regulation, interaction, and function. Relatively few of the proteins pulled down by peptide baits from NFAT5 are generally involved in transcription, and most, therefore, are likely to be specifically related to the regulation of NFAT5 or its function. The novel associated proteins are involved with cancer, effects of hypertonicity on chromatin, development, splicing of mRNA, transcription, and vesicle trafficking.


Subject(s)
Chromatography, Affinity/methods , NFATC Transcription Factors/metabolism , Peptides/metabolism , Amino Acid Sequence , Cell Extracts , HEK293 Cells , Humans , NFATC Transcription Factors/chemistry , Osmosis , Protein Binding , Protein Domains , Protein Interaction Maps , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Sodium Chloride/pharmacology
3.
Physiol Genomics ; 48(4): 290-305, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26757802

ABSTRACT

NFAT5 is an osmoregulated transcription factor that particularly increases expression of genes involved in protection against hypertonicity. Transcription factors often contain unstructured regions that bind co-regulatory proteins that are crucial for their function. The NH2-terminal region of NFAT5 contains regions predicted to be intrinsically disordered. We used peptide aptamer-based affinity chromatography coupled with mass spectrometry to identify protein preys pulled down by one or more overlapping 20 amino acid peptide baits within a predicted NH2-terminal unstructured region of NFAT5. We identify a total of 467 unique protein preys that associate with at least one NH2-terminal peptide bait from NFAT5 in either cytoplasmic or nuclear extracts from HEK293 cells treated with elevated, normal, or reduced NaCl concentrations. Different sets of proteins are pulled down from nuclear vs. cytoplasmic extracts. We used GeneCards to ascertain known functions of the protein preys. The protein preys include many that were previously known, but also many novel ones. Consideration of the novel ones suggests many aspects of NFAT5 regulation, interaction and function that were not previously appreciated, for example, hypertonicity inhibits NFAT5 by sumoylating it and the NFAT5 protein preys include components of the CHTOP complex that desumoylate proteins, an action that should contribute to activation of NFAT5.


Subject(s)
Proteins/metabolism , Transcription Factors/metabolism , Cell Nucleus/metabolism , Chromatography, Affinity/methods , Cytoplasm/metabolism , HEK293 Cells , Humans , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptides/metabolism , Protein Interaction Mapping/methods , Tandem Mass Spectrometry/methods , Transcription Factors/chemistry
4.
Proc Natl Acad Sci U S A ; 110(18): 7482-7, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23589856

ABSTRACT

Glycerophosphocholine (GPC) is high in cells of the renal inner medulla where high interstitial NaCl and urea power concentration of the urine. GPC protects inner medullary cells against the perturbing effects of high NaCl and urea by stabilizing intracellular macromolecules. Degradation of GPC is catalyzed by the glycerophosphocholine phosphodiesterase activity of glycerophosphodiester phosphodiesterase domain containing 5 (GDPD5). We previously found that inhibitory posttranslational modification (PTM) of GDPD5 contributes to high NaCl- and urea-induced increase of GPC. The purpose of the present studies was to identify the PTM(s). We find at least three such PTMs in HEK293 cells: (i) Formation of a disulfide bond between C25 and C571. High NaCl and high urea increase reactive oxygen species (ROS). The ROS increase disulfide bonding between GDPD5-C25 and -C571, which inhibits GDPD5 activity, as supported by the findings that the antioxidant N-acetylcysteine prevents high NaCl- and urea-induced inhibition of GDPD5; GDPD5-C25S/C571S mutation or over expression of peroxiredoxin increases GDPD5 activity; H2O2 inhibits activity of wild type GDPD5, but not of GDPD5-C25S/C571S; and peroxiredoxin is relatively low in the renal inner medulla where GPC is high. (ii) Dephosphorylation of GDPD5-T587. GDPD5 threonine 587 is constitutively phosphorylated. High NaCl and high urea dephosphorylate GDPD5-T587. Mutation of GDPD5-T587 to alanine, which cannot be phosphorylated, decreases GPC-PDE activity of GDPD5. (iii) Alteration at an unknown site mediated by CDK1. Inhibition of CDK1 protein kinase reduces GDE-PDE activity of GDPD5 without altering phosphorylation at T587, and CDK1/5 inhibitor reduces activity of GDPD5- C25S/C571S-T587A.


Subject(s)
Glycerylphosphorylcholine/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Protein Processing, Post-Translational/drug effects , Sodium Chloride/pharmacology , Urea/pharmacology , Amino Acid Sequence , Animals , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Glycosylation/drug effects , HEK293 Cells , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Mass Spectrometry , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Mutant Proteins/metabolism , Mutation/genetics , Peptides/chemistry , Peptides/metabolism , Peroxiredoxins/metabolism , Phosphorylation/drug effects , Phosphothreonine/metabolism , Protein Kinase Inhibitors/pharmacology
5.
Physiol Genomics ; 47(10): 500-13, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26220925

ABSTRACT

High extracellular NaCl is known to change expression of numerous genes, many of which are regulated by the osmoprotective transcription factor nuclear factor of activated T cells-5 (NFAT5). In the present study we employed RNA-Seq to provide a comprehensive, unbiased account of genes regulated by high NaCl in mouse embryonic fibroblast cells (MEFs). To identify genes regulated by NFAT5 we compared wild-type MEFs (WT-MEFs) to MEFs in which mutation of the NFAT5 gene inhibits its transcriptional activity (Null-MEFs). In WT-MEFs adding NaCl to raise osmolality from 300 to 500 mosmol/kg for 24 h increases expression of 167 genes and reduces expression of 412. Raising osmolality through multiple passages (adapted cells) increases expression of 196 genes and reduces expression of 528. In Null-MEFs, after 24 h of high NaCl, expression of 217 genes increase and 428 decrease, while in adapted Null-MEFs 143 increase and 622 decrease. Fewer than 10% of genes are regulated in common between WT- and null-MEFs, indicating a profound difference in regulation of high-NaCl induced genes induced by NFAT5 compared with those induced in the absence of NFAT5. Based on our findings we suggest a mechanism for this phenomenon, which had previously been unexplained. The NFAT5 DNA-binding motif (osmotic response element) is overrepresented in the vicinity of genes that NFAT5 upregulates, but not genes that it downregulates. We used Gene Ontology and manual curation to determine the function of the genes targeted by NFAT5, revealing many novel consequences of NFAT5 transcriptional activity.


Subject(s)
Gene Expression Regulation/drug effects , Sequence Analysis, RNA/methods , Sodium Chloride/pharmacology , Adaptation, Physiological/drug effects , Animals , Binding Sites , Down-Regulation/drug effects , Embryo, Mammalian/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Ontology , Interferon Regulatory Factor-1/metabolism , Mice , Models, Biological , Mutation/genetics , NFATC Transcription Factors/genetics , Transcription, Genetic/drug effects , Up-Regulation/drug effects
6.
Am J Physiol Renal Physiol ; 308(2): F140-8, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25391900

ABSTRACT

High NaCl in the renal medullary interstitial fluid powers the concentration of urine but can damage cells. The transcription factor nuclear factor of activated T cells 5 (NFAT5) activates the expression of osmoprotective genes. We studied whether PKC-α contributes to the activation of NFAT5. PKC-α protein abundance was greater in the renal medulla than in the cortex. Knockout of PKC-α reduced NFAT5 protein abundance and expression of its target genes in the inner medulla. In human embryonic kidney (HEK)-293 cells, high NaCl increased PKC-α activity, and small interfering RNA-mediated knockdown of PKC-α attenuated high NaCl-induced NFAT5 transcriptional activity. Expression of ERK1/2 protein and phosphorylation of ERK1/2 were higher in the renal inner medulla than in the cortex. Knockout of PKC-α decreased ERK1/2 phosphorylation in the inner medulla, as did knockdown of PKC-α in HEK-293 cells. Also, knockdown of ERK2 reduced high NaCl-dependent NFAT5 transcriptional activity in HEK-293 cells. Combined knockdown of PKC-α and ERK2 had no greater effect than knockdown of either alone. Knockdown of either PKC-α or ERK2 reduced the high NaCl-induced increase of NFAT5 transactivating activity. We have previously found that the high NaCl-induced increase of phosphorylation of Ser(591) on Src homology 2 domain-containing phosphatase 1 (SHP-1-S591-P) contributes to the activation of NFAT5 in cell culture, and here we found high levels of SHP-1-S591-P in the inner medulla. PKC-α has been previously shown to increase SHP-1-S591-P, which raised the possibility that PKC-α might be acting through SHP-1. However, we did not find that knockout of PKC-α in the renal medulla or knockdown in HEK-293 cells affected SHP-1-S591-P. We conclude that PKC-α contributes to high NaCl-dependent activation of NFAT5 through ERK1/2 but not through SHP-1-S591.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Kidney/enzymology , MAP Kinase Signaling System , NFATC Transcription Factors/metabolism , Protein Kinase C-alpha/metabolism , Animals , HEK293 Cells , Humans , Mice, Inbred C57BL , Phosphorylation , Sodium Chloride/metabolism
7.
Protein Expr Purif ; 115: 141-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26256058

ABSTRACT

Hypertonicity stimulates Nuclear Factor of Activated T-cells 5 (NFAT5) nuclear localization and transactivating activity. Many transcription factors are known to contain intrinsically disordered regions (IDRs) which become more structured with local environmental changes such as osmolality, temperature and tonicity. The transactivating domain of NFAT5 is predicted to be intrinsically disordered under normal tonicity, and under high NaCl, the activity of this domain is increased. To study the binding of co-regulatory proteins at IDRs a cDNA construct expressing the NFAT5 TAD was created and transformed into Escherichia coli cells. Transformed E. coli cells were mass produced by fermentation and extracted by cell lysis to release the NFAT5 TAD. The NFAT5 TAD was subsequently purified using a His-tag column, cation exchange chromatography as well as hydrophobic interaction chromatography and then characterized by mass spectrometry (MS).


Subject(s)
Intrinsically Disordered Proteins/isolation & purification , Intrinsically Disordered Proteins/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transcription Factors/isolation & purification , Transcription Factors/metabolism , Amino Acid Sequence , Escherichia coli/genetics , Fermentation , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
8.
Am J Physiol Cell Physiol ; 307(5): C442-54, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24965592

ABSTRACT

High extracellular NaCl, such as in the renal medulla, can perturb and even kill cells, but cells mount protective responses that enable them to survive and function. Many high-NaCl-induced perturbations and protective responses are known, but the signaling pathways involved are less clear. Change in protein phosphorylation is a common mode of cell signaling, but there was no unbiased survey of protein phosphorylation in response to high NaCl. We used stable isotopic labeling of amino acids in cell culture coupled to mass spectrometry to identify changes in protein phosphorylation in human embryonic kidney (HEK 293) cells exposed to high NaCl. We reproducibly identify >8,000 unique phosphopeptides in 4 biological replicate samples with a 1% false discovery rate. High NaCl significantly changed phosphorylation of 253 proteins. Western analysis and targeted ion selection mass spectrometry confirm a representative sample of the phosphorylation events. We analyze the affected proteins by functional category to infer how altered protein phosphorylation might signal cellular responses to high NaCl, including alteration of cell cycle, cyto/nucleoskeletal organization, DNA double-strand breaks, transcription, proteostasis, metabolism of mRNA, and cell death.


Subject(s)
Extracellular Fluid/metabolism , Membrane Proteins/metabolism , Sodium Chloride/toxicity , Chromatography, Liquid , Extracellular Fluid/drug effects , HEK293 Cells , Humans , Phosphorylation/drug effects , Phosphorylation/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Sodium Chloride/chemistry , Tandem Mass Spectrometry
9.
Proc Natl Acad Sci U S A ; 108(29): 12155-60, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21712438

ABSTRACT

Separate reports that hypertonicity activates p38 via a Rac1-OSM-MEKK3-MKK3-p38 pathway and that p38α contributes to activation of TonEBP/OREBP led us to the hypothesis that Rac1 might activate TonEBP/OREBP via p38. The present studies examine that possibility. High NaCl is hypertonic. We find that siRNA knockdown of Rac1 reduces high NaCl-induced increase of TonEBP/OREBP transcriptional activity (by reducing its transactivating activity but not its nuclear localization). Similarly, siRNA knockdown of osmosensing scaffold for MEKK3 (OSM) also reduces high NaCl-dependent TonEBP/OREBP transcriptional and transactivating activities. Simultaneous siRNA knockdown of Rac1 and OSM is not additive in reduction of TonEBP/OREBP transcriptional activity, indicating a common pathway. However, siRNA knockdown of MKK3 does not reduce TonEBP/OREBP transcriptional activity, although siRNA knockdown of MKK6 does. Nevertheless, the effect of Rac1 on TonEBP/OREBP is also independent of MKK6 because it occurs in MKK6-null cells. Furthermore, we find that siRNA knockdown of Rac1 or OSM actually increases activity (phosphorylation) of p38, rather than decreasing it, as previously reported. Thus, the effect of Rac1 on TonEBP/OREBP is independent of p38. We find instead that phospholipase C-γ1 (PLC-γ1) is involved. When transfected into PLC-γ1-null mouse embryonic fibroblast cells, catalytically active Rac1 does not increase TonEBP/OREBP transcriptional activity unless PLC-γ1 is reconstituted. Similarly, dominant-negative Rac1 also does not inhibit TonEBP/OREBP in PLC-γ1-null cells unless PLC-γ1 is reconstituted. We conclude that Rac1/OSM supports TonEBP/OREBP activity and that this activity is mediated via PLC-γ1, not p38.


Subject(s)
Gene Expression Regulation/genetics , Microfilament Proteins/metabolism , Neuropeptides/metabolism , Phospholipase C gamma/metabolism , Transcription Factors/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Cell Line , Gene Knockdown Techniques , Humans , Luciferases , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/metabolism , Mice , Mitogen-Activated Protein Kinase 14/metabolism , Neuropeptides/genetics , RNA, Small Interfering/genetics , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
10.
Am J Physiol Renal Physiol ; 305(3): F362-9, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23720348

ABSTRACT

Activation of the transcription factor NFAT5 by high NaCl involves changes in phosphorylation. By siRNA screening, we previously found that protein targeting to glycogen (PTG), a regulatory subunit of protein phosphatase1 (PP1), contributes to regulation of high NaCl-induced NFAT5 transcriptional activity. The present study addresses the mechanism involved. We find that high NaCl-induced inhibition of PTG elevates NFAT5 activity by increasing NFAT5 transactivating activity, protein abundance, and nuclear localization. PTG acts via a catalytic subunit PP1γ. PTG associates physically with PP1γ, and NaCl reduces both this association and remaining PTG-associated PP1γ activity. High NaCl-induced phosphorylation of p38, ERK, and SHP-1 contributes to activation of NFAT5. Knockdown of PTG does not affect phosphorylation of p38 or ERK. However, PTG and PP1γ bind to SHP-1, and knockdown of either PTG or PP1γ increases high NaCl-induced phosphorylation of SHP-1-S591, which inhibits SHP-1. Mutation of SHP-1-S591 to alanine, which cannot be phosphorylated, increases inhibition of NFAT5 by SHP-1. Thus high NaCl reduces the stimulatory effect of PTG and PP1γ on SHP-1, which in turn reduces the inhibitory effect of SHP-1 on NFAT5. Our findings add to the known functions of PTG, which was previously recognized only for its glycogenic activity.


Subject(s)
Protein Phosphatase 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Sodium Chloride/pharmacology , Transcription Factors/metabolism , Blotting, Western , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , MAP Kinase Signaling System/physiology , Nuclear Localization Signals/drug effects , Plasmids , Polymerase Chain Reaction , Protein Phosphatase 1/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Am J Physiol Renal Physiol ; 304(7): F908-17, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23324178

ABSTRACT

High NaCl activates the transcription factor nuclear factor of activated T cells 5 (NFAT5), leading to increased transcription of osmoprotective target genes. Kinases PKA, PI3K, AKT1, and p38α were known to contribute to the high NaCl-induced increase of NFAT5 activity. We now identify another kinase, GSK-3ß. siRNA-mediated knock-down of GSK-3ß increases NFAT5 transcriptional and transactivating activities without affecting high NaCl-induced nuclear localization of NFAT5 or NFAT5 protein expression. High NaCl increases phosphorylation of GSK-3ß-S9, which inhibits GSK-3ß. In GSK-3ß-null mouse embryonic fibroblasts transfection of GSK-3ß, in which serine 9 is mutated to alanine, so that it cannot be inhibited by phosphorylation at that site, inhibits high NaCl-induced NFAT5 transcriptional activity more than transfection of wild-type GSK-3ß. High NaCl-induced phosphorylation of GSK-3ß-S9 depends on PKA, PI3K, and AKT, but not p38α. Overexpression of PKA catalytic subunit α or of catalytically active AKT1 reduces inhibition of NFAT5 by GSK-3ß, but overexpression of p38α together with its catalytically active upstream kinase, MKK6, does not. Thus, GSK-3ß normally inhibits NFAT5 by suppressing its transactivating activity. When activated by high NaCl, PKA, PI3K, and AKT1, but not p38α, increase phosphorylation of GSK-3ß-S9, which reduces the inhibitory effect of GSK-3ß on NFAT5, and thus contributes to activation of NFAT5.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Mice , Mitogen-Activated Protein Kinase 14/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Sodium Chloride/administration & dosage
12.
Proc Natl Acad Sci U S A ; 107(15): 7072-7, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20351292

ABSTRACT

Hypertonicity activates the transcription factor TonEBP/OREBP, resulting in increased expression of osmoprotective genes, including those responsible for accumulation of organic osmolytes and heat-shock proteins. Phosphorylation of TonEBP/OREBP contributes to its activation. Several of the kinases that are involved were previously identified, but the phosphatases were not. In the present studies we screened a genomewide human phosphatase siRNA library in human embryonic kidney (HEK)293 cells for effects on TonEBP/OREBP transcriptional activity. We found that siRNAs against 57 phosphatases significantly alter TonEBP/OREBP transcriptional activity during normotonicity (290 mosmol/kg) or hypertonicity (500 mosmol/kg, NaCl added) or both. Most siRNAs increase TonEBP/OREBP activity, implying that the targeted phosphatases normally reduce that activity. We further studied in detail SHP-1, whose knockdown by its specific siRNA increases TonEBP/OREBP transcriptional activity at 500 mosmol/kg. We confirmed that SHP-1 is inhibitory by overexpressing it, which reduces TonEBP/OREBP transcriptional activity at 500 mosmol/kg. SHP-1 dephosphorylates TonEBP/OREBP at a known regulatory site, Y143, both in vivo and in vitro. It inhibits TonEBP/OREBP by both reducing TonEBP/OREBP nuclear localization, which is Y143 dependent, and by lowering high NaCl-induced TonEBP/OREBP transactivating activity. SHP-1 coimmunoprecipitates with TonEBP/OREBP and vice versa, suggesting that they are physically associated in the cell. High NaCl inhibits the effect of SHP-1 on TonEBP/OREBP by increasing phosphorylation of SHP-1 on Ser591, which reduces its phosphatase activity and localization to the nucleus. Thus, TonEBP/OREBP is extensively regulated by phosphatases, including SHP-1, whose inhibition by high NaCl increases phosphorylation of TonEBP/OREBP at Y143, contributing to the nuclear localization and activation of TonEBP/OREBP.


Subject(s)
Gene Expression Regulation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Transcription Factors/metabolism , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Library , Heat-Shock Proteins/metabolism , Humans , Models, Biological , Osmosis , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , RNA, Small Interfering/metabolism , Sodium Chloride/chemistry
13.
Proc Natl Acad Sci U S A ; 107(2): 906-11, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-20080774

ABSTRACT

High NaCl elevates activity of the osmoprotective transcription factor TonEBP/OREBP by increasing its phosphorylation, transactivating activity, and localization to the nucleus. We investigated the possible role in this activation of phospholipase C-gamma1 (PLC-gamma1), which has a predicted binding site at TonEBP/OREBP-phospho-Y143. We find the following. (i) Activation of TonEBP/OREBP transcriptional activity by high NaCl is reduced in PLC-gamma1 null cells and in HEK293 cells in which PLC-gamma1 is knocked down by a specific siRNA. (ii) High NaCl increases phosphorylation of TonEBP/OREBP at Y143. (iii) Wild-type PLC-gamma1 coimmunoprecipitates with wild-type TonEBP/OREBP but not TonEBP/OREBP-Y143A, and the coimmunoprecipitation is increased by high NaCl. (iv) PLC-gamma1 is part of the protein complex that associates with TonEBP/OREBP at its DNA binding site. (v) Knockdown of PLC-gamma1 or overexpression of a PLC-gamma1-SH3 deletion mutant reduces high NaCl-dependent TonEBP/OREBP transactivating activity. (vi) Nuclear localization of PLC-gamma1 is increased by high NaCl. (vii) High NaCl-induced nuclear localization of TonEBP/OREBP is reduced if cells lack PLC-gamma1, if PLC-gamma1 mutated in its SH2C domain is overexpressed, or if Y143 in TonEBP/OREBP is mutated to alanine. (viii) Expression of recombinant PLC-gamma1 restores nuclear localization of wild-type TonEBP/OREBP in PLC-gamma1 null cells but not of TonEBP/OREBP-Y143A. (ix) The PLC-gamma1 phospholipase inhibitor U72133 inhibits nuclear localization of TonEBP/OREBP but not the increase of its transactivating activity. We conclude that, when NaCl is elevated, TonEBP/OREBP becomes phosphorylated at Y143, resulting in binding of PLC-gamma1 to that site, which contributes to TonEBP/OREBP transcriptional activity, transactivating activity, and nuclear localization.


Subject(s)
Phospholipase C gamma/physiology , Signal Transduction/physiology , Sodium Chloride/pharmacology , Transcription Factors/metabolism , Blotting, Western , Cell Line , Enzyme Activation , Humans , Kidney/enzymology , Kinetics , Nuclear Proteins/drug effects , Nuclear Proteins/metabolism , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Phosphorylation , Protein Binding , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription, Genetic
14.
Am J Physiol Cell Physiol ; 303(10): C1061-9, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22992674

ABSTRACT

The transcription factor nuclear factor of activated T cell 5 (NFAT5) is activated by the stress of hypertonicity (e.g., high NaCl). Increased expression of NFAT5 target genes causes accumulation of protective organic osmolytes and heat shock proteins. Under normotonic conditions (∼300 mosmol/kgH(2)O), NFAT5 is distributed between the nucleus and cytoplasm, hypertonicity causes it to translocate into the nucleus, and hypotonicity causes it to translocate into the cytoplasm. The mechanism of translocation is complex and not completely understood. NFAT5-T298 is a known contact site of NFAT5 with its specific DNA element [osmotic response element (ORE)]. In the present study, we find that mutation of NFAT5-T298 to alanine or aspartic acid not only reduces binding of NFAT5 to OREs (EMSA) but also proportionately reduces high NaCl-induced nuclear translocation of NFAT5. Combined mutation of other NFAT5 DNA contact sites (R293A/E299A/R302A) also greatly reduces both specific DNA binding and nuclear localization of NFAT5. NFAT5-T298 is a potential phosphorylation site, but, using protein mass spectrometry, we do not find phosphorylation at NFAT5-T298. Further, decreased high NaCl-induced nuclear localization of NFAT5 mutated at T298 does not involve previously known regulatory mechanisms, including hypotonicity-induced export of NFAT5, regulated by phosphorylation of NFAT5-S155, XPO1 (CRM1/exportin1)-mediated export of NFAT5 from the nucleus, or hypertonicity-induced elevation of NUP88, which enhances nuclear localization of NFAT5. We conclude that specific DNA binding of NFAT5 contributes to its nuclear localization, by mechanisms, as yet undetermined, but independent of ones previously described to regulate NFAT5 distribution.


Subject(s)
DNA/metabolism , Protein Transport/drug effects , Sodium Chloride/pharmacology , Transcription Factors/metabolism , Alternative Splicing , Animals , Cell Line , Gene Expression Regulation/physiology , Humans , Karyopherins/genetics , Karyopherins/metabolism , Mice , Mutation , Osmotic Pressure , Protein Binding , Protein Isoforms , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sodium Chloride/chemistry , Transcription Factors/genetics , Exportin 1 Protein
15.
Physiol Genomics ; 44(21): 1063-71, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22991206

ABSTRACT

Mammalian cells are normally stressed by high interstitial NaCl in the renal medulla and by lesser elevation of NaCl in several other tissues. High NaCl damages proteins and DNA and can kill cells. Known protective responses include nuclear translocation of the transcription factor NFAT5 and other proteins. In order better to understand the extent and significance of changes in nuclear protein abundance, we extracted nuclear and cytoplasmic proteins separately from HEK293 cells and measured by LC-MS/MS (iTRAQ) changes of abundance of proteins in the extracts in response to high NaCl at three time points: 1 h, 8 h, and adapted for two passages. We confidently identified a total of 3,190 proteins; 163 proteins changed significantly at least at one time point in the nucleus. We discerned the biological significance of the changes by Gene Ontology and protein network analysis. Proteins that change in the nucleus include ones involved in protein folding and localization, microtubule-based process, regulation of cell death, cytoskeleton organization, DNA metabolic process, RNA processing, and cell cycle. Among striking changes in the nucleus, we found a decrease of all six 14-3-3 isoforms; dynamic changes of "cytoskeletal" proteins, suggestive of nucleoskeletal reorganization; rapid decrease of tubulins; and dynamic changes of heat shock proteins. Identification of these changes of nuclear protein abundance enhances our understanding of high NaCl-induced cellular stress, and provides leads to previously unknown damages and protective responses.


Subject(s)
Nuclear Proteins/metabolism , Sodium Chloride/metabolism , Cell Nucleus/metabolism , HEK293 Cells , Humans , Mass Spectrometry , Proteomics
16.
Am J Physiol Renal Physiol ; 303(5): F674-80, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22718889

ABSTRACT

Oxidative stress damages cells. NaCl and urea are high in renal medullary interstitial fluid, which is necessary to concentrate urine, but which causes oxidative stress by elevating reactive oxygen species (ROS). Here, we measured the antioxidant enzyme superoxide dismutases (SODs, MnSOD, and Cu/ZnSOD) and catalase in mouse kidney that might mitigate the oxidative stress. MnSOD protein increases progressively from the cortex to the inner medulla, following the gradient of increasing NaCl and urea. MnSOD activity increases proportionately, but MnSOD mRNA does not. Water restriction, which elevates renal medullary NaCl and urea, increases MnSOD protein, accompanied by a proportionate increase in MnSOD enzymatic activity in the inner medulla, but not in the cortex or the outer medulla. In contrast, Cu/ZnSOD and TNF-α (an important regulator of MnSOD) do not vary between the regions of the kidney, and expression of catalase protein actually decreases from the cortex to the inner medulla. Water restriction increases activity of mitochondrial enzymes that catalyze production of ROS in the inner medulla, but reduces NADPH oxidase activity there. We also examined the effect of high NaCl and urea on MnSOD in Madin-Darby canine kidney (MDCK) cells. High NaCl and high urea both increase MnSOD in MDCK cells. This increase in MnSOD protein apparently depends on the elevation of ROS since it is eliminated by the antioxidant N-acetylcysteine, and it occurs without raising osmolality when ROS are elevated by antimycin A or xanthine oxidase plus xanthine. We conclude that ROS, induced by high NaCl and urea, increase MnSOD activity in the renal inner medulla, which moderates oxidative stress.


Subject(s)
Kidney/enzymology , Reactive Oxygen Species/metabolism , Water Deprivation/physiology , Acetylcysteine/pharmacology , Animals , Antimycin A/pharmacology , Catalase , Cells, Cultured , Dogs , Female , Male , Mice , NADPH Oxidases/biosynthesis , Osmolar Concentration , Oxidative Stress/drug effects , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Urea/pharmacology , Xanthine/pharmacology , Xanthine Oxidase/pharmacology
17.
FASEB J ; 24(11): 4325-35, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20585028

ABSTRACT

The transcription factor TonEBP/OREBP promotes cell survival during osmotic stress. High NaCl-induced phosphorylation of TonEBP/OREBP at tyrosine-143 was known to be an important factor in increasing its activity in cell culture. We now find that TonEBP/OREBP also is phosphorylated at tyrosine-143 in rat renal inner medulla, dependent on the interstitial osmolality. c-Abl seemed likely to be the kinase that phosphorylates TonEBP/OREBP because Y143 is in a consensus c-Abl phosphorylation site. We now confirm that, as follows. High NaCl increases c-Abl activity. Specific inhibition of c-Abl by imatinib, siRNA, or c-Abl kinase dead drastically reduces high NaCl-induced TonEBP/OREBP activity by reducing its nuclear location and transactivating activity. c-Abl associates with TonEBP/OREBP (coimmunoprecipitation) and phosphorylates TonEBP/OREBP-Y143 both in cell and in vitro. High NaCl-induced activation of ataxia telangiectasia mutated, previously known to contribute to activation of TonEBP/OREBP, depends on c-Abl activity. Thus, c-Abl is the kinase responsible for high NaCl-induced phosphorylation of TonEBP/OREBP-Y143, which contributes to its increased activity.


Subject(s)
NFATC Transcription Factors/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Amino Acid Sequence , Animals , Benzamides , Cell Line , Enzyme Activation/drug effects , HEK293 Cells , HeLa Cells , Humans , Imatinib Mesylate , Kidney Medulla/metabolism , Male , Molecular Sequence Data , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Transport/physiology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/genetics , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Sequence Alignment , Sodium Chloride/pharmacology , Transcriptional Activation/physiology , Tyrosine/metabolism
18.
Proc Natl Acad Sci U S A ; 105(31): 11026-31, 2008 Aug 05.
Article in English | MEDLINE | ID: mdl-18667693

ABSTRACT

Glycerophosphocholine (GPC) is an abundant osmoprotective renal medullary organic osmolyte. We previously found that its synthesis from phosphatidylcholine is catalyzed by tonicity-regulated activity of the phospholipase B, neuropathy target esterase. We also found that its degradation is catalyzed by glycerophosphocholine phosphodiesterase (GPC-PDE) activity and that elevating osmolality from 300 to 500 mosmol/kg by adding NaCl or urea, inhibits GPC-PDE activity, which contributes to the resultant increase of GPC. In the present studies we identify GDPD5 (glycerophosphodiester phosphodiesterase domain containing 5) as a GPC-PDE that is rapidly inhibited by high NaCl or urea. Recombinant GDPD5 colocalizes with neuropathy target esterase in the perinuclear region of HEK293 cells, and immuno-precipitated recombinant GDPD5 degrades GPC in vitro. The in vitro activity is reduced when the cells from which the GDPD5 is immuno-precipitated have been exposed to high NaCl or urea. In addition, high NaCl decreases mRNA abundance of GDPD5 via an increase of its degradation rate, although high urea does not. At 300 mosmol/kg siRNA knockdown of GDPD5 increases GPC in mouse inner medullary collecting duct-3 cells, and over expression of recombinant GDPD5 increases cellular GPC-PDE activity, accompanied by decreased GPC. We conclude that GDPD5 is a GPC-PDE that contributes to osmotic regulation of cellular GPC.


Subject(s)
Glycerylphosphorylcholine/metabolism , Kidney/metabolism , Phosphoric Diester Hydrolases/metabolism , Water-Electrolyte Balance/physiology , Analysis of Variance , Animals , Blotting, Western , Cell Line , DNA Primers , Humans , Immunoprecipitation , Mice , Microscopy, Fluorescence , Phosphoric Diester Hydrolases/genetics , RNA Interference , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sodium Chloride/pharmacology , Urea/pharmacology
19.
Proc Natl Acad Sci U S A ; 105(14): 5620-5, 2008 Apr 08.
Article in English | MEDLINE | ID: mdl-18367666

ABSTRACT

High NaCl rapidly activates p38 MAPK by phosphorylating it, the phosphorylation presumably being regulated by a balance of kinases and phosphatases. Kinases are known, but the phosphatases are uncertain. Our initial purpose was to identify the phosphatases. We find that in HEK293 cells transient overexpression of MAPK phosphatase-1 (MKP-1), a dual-specificity phosphatase, inhibits high NaCl-induced phosphorylation of p38, and that overexpression of a dominant negative mutant of MKP-1 does the opposite. High NaCl lowers MKP-1 activity by increasing reactive oxygen species, which directly inhibit MKP-1, and by reducing binding of MKP-1 to p38. Because inhibition of p38 is reported to reduce hypertonicity-induced activation of the osmoprotective transcription factor, TonEBP/OREBP, we anticipated that MKP-1 expression might also. However, overexpression of MKP-1 has no significant effect on Ton EBP/OREBP activity. This paradox is explained by opposing effects of p38alpha and p38delta, both of which are activated by high NaCl and inhibited by MKP-1. Thus, we find that overexpression of p38alpha increases high NaCl-induced TonEBP/OREBP activity, but overexpression of p38delta reduces it. Also, siRNA-mediated knockdown of p38delta enhances the activation of TonEBP/OREBP. We conclude that high NaCl inhibits MKP-1, which contributes to the activation of p38. However, opposing actions of p38alpha and p38delta negate any effect on TonEBP/OREBP activity. Thus, activation of p38 isoforms by hypertonicity does not contribute to activation of TonEBP/OREBP because of opposing effects of p38alpha and p38delta, and effects of inhibitors of p38 depend on which isoform is affected, which can be misleading.


Subject(s)
Dual Specificity Phosphatase 1/physiology , Mitogen-Activated Protein Kinase 13/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , NFATC Transcription Factors/metabolism , Sodium Chloride/pharmacology , Transcription Factors/metabolism , Cell Line , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/genetics , Enzyme Activation/drug effects , Humans , Mitogen-Activated Protein Kinase 13/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Phosphorylation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL