Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
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.
Proc Natl Acad Sci U S A ; 114(42): E8875-E8884, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28973931

ABSTRACT

G protein stimulatory α-subunit (Gαs)-coupled heptahelical receptors regulate cell processes largely through activation of protein kinase A (PKA). To identify signaling processes downstream of PKA, we deleted both PKA catalytic subunits using CRISPR-Cas9, followed by a "multiomic" analysis in mouse kidney epithelial cells expressing the Gαs-coupled V2 vasopressin receptor. RNA-seq (sequencing)-based transcriptomics and SILAC (stable isotope labeling of amino acids in cell culture)-based quantitative proteomics revealed a complete loss of expression of the water-channel gene Aqp2 in PKA knockout cells. SILAC-based quantitative phosphoproteomics identified 229 PKA phosphorylation sites. Most of these PKA targets are thus far unannotated in public databases. Surprisingly, 1,915 phosphorylation sites with the motif x-(S/T)-P showed increased phosphooccupancy, pointing to increased activity of one or more MAP kinases in PKA knockout cells. Indeed, phosphorylation changes associated with activation of ERK2 were seen in PKA knockout cells. The ERK2 site is downstream of a direct PKA site in the Rap1GAP, Sipa1l1, that indirectly inhibits Raf1. In addition, a direct PKA site that inhibits the MAP kinase kinase kinase Map3k5 (ASK1) is upstream of JNK1 activation. The datasets were integrated to identify a causal network describing PKA signaling that explains vasopressin-mediated regulation of membrane trafficking and gene transcription. The model predicts that, through PKA activation, vasopressin stimulates AQP2 exocytosis by inhibiting MAP kinase signaling. The model also predicts that, through PKA activation, vasopressin stimulates Aqp2 transcription through induction of nuclear translocation of the acetyltransferase EP300, which increases histone H3K27 acetylation of vasopressin-responsive genes (confirmed by ChIP-seq).


Subject(s)
Epithelial Cells/metabolism , Protein Kinase C/metabolism , Animals , Aquaporin 2/genetics , Aquaporin 2/metabolism , Chromatin Immunoprecipitation , Clustered Regularly Interspaced Short Palindromic Repeats , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , E1A-Associated p300 Protein/metabolism , Exocytosis/physiology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Knockout Techniques , Kidney/cytology , MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Phosphorylation , Protein Kinase C/genetics , Signal Transduction , Vasopressins/metabolism
3.
Proc Natl Acad Sci U S A ; 114(46): E9989-E9998, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29089413

ABSTRACT

Prior RNA sequencing (RNA-seq) studies have identified complete transcriptomes for most renal epithelial cell types. The exceptions are the cell types that make up the renal collecting duct, namely intercalated cells (ICs) and principal cells (PCs), which account for only a small fraction of the kidney mass, but play critical physiological roles in the regulation of blood pressure, extracellular fluid volume, and extracellular fluid composition. To enrich these cell types, we used FACS that employed well-established lectin cell surface markers for PCs and type B ICs, as well as a newly identified cell surface marker for type A ICs, c-Kit. Single-cell RNA-seq using the IC- and PC-enriched populations as input enabled identification of complete transcriptomes of A-ICs, B-ICs, and PCs. The data were used to create a freely accessible online gene-expression database for collecting duct cells. This database allowed identification of genes that are selectively expressed in each cell type, including cell-surface receptors, transcription factors, transporters, and secreted proteins. The analysis also identified a small fraction of hybrid cells expressing aquaporin-2 and anion exchanger 1 or pendrin transcripts. In many cases, mRNAs for receptors and their ligands were identified in different cells (e.g., Notch2 chiefly in PCs vs. Jag1 chiefly in ICs), suggesting signaling cross-talk among the three cell types. The identified patterns of gene expression among the three types of collecting duct cells provide a foundation for understanding physiological regulation and pathophysiology in the renal collecting duct.


Subject(s)
Aquaporin 2/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Collecting/metabolism , Kidney/metabolism , Sequence Analysis, RNA/methods , Transcriptome , Animals , Anion Exchange Protein 1, Erythrocyte/metabolism , Anion Transport Proteins/metabolism , Base Sequence , Biomarkers/metabolism , Gene Expression , Gene Expression Profiling , Jagged-1 Protein/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , RNA/metabolism , Receptor, Notch2/metabolism , Signal Transduction , Sulfate Transporters , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics
4.
Arterioscler Thromb Vasc Biol ; 37(3): 598-606, 2017 03.
Article in English | MEDLINE | ID: mdl-28062505

ABSTRACT

OBJECTIVE: Serum sodium concentration is maintained by osmoregulation within normal range of 135 to 145 mmol/L. Previous analysis of data from the ARIC study (Atherosclerosis Risk in Communities) showed association of serum sodium with the 10-year risk scores of coronary heart disease and stroke. Current study evaluated the association of within-normal-range serum sodium with cardiovascular risk factors. APPROACH AND RESULTS: Only participants who did not take cholesterol or blood pressure medications and had sodium within normal 135 to 145 mmol/L range were included (n=8615), and the cohort was stratified based on race, sex, and smoking status. Multiple linear regression analysis of data from ARIC study was performed, with adjustment for age, blood glucose, insulin, glomerular filtration rate, body mass index, waist to hip ratio, and calorie intake. The analysis showed positive associations with sodium of total cholesterol, low-density lipoprotein cholesterol, and total cholesterol to high-density lipoprotein cholesterol ratio; apolipoprotein B; and systolic and diastolic blood pressure. Increases in lipids and blood pressure associated with 10 mmol/L increase in sodium are similar to the increases associated with 7 to 10 years of aging. Analysis of sodium measurements made 3 years apart demonstrated that it is stable within 2 to 3 mmol/L, explaining its association with long-term health outcomes. Furthermore, elevated sodium promoted lipid accumulation in cultured adipocytes, suggesting direct causative effects on lipid metabolism. CONCLUSIONS: Serum sodium concentration is a cardiovascular risk factor even within the normal reference range. Thus, decreasing sodium to the lower end of the normal range by modification of water and salt intake is a personalizable strategy for decreasing cardiovascular risks.


Subject(s)
Blood Pressure , Cardiovascular Diseases/epidemiology , Lipids/blood , Sodium/blood , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Biomarkers/blood , Body Mass Index , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/physiopathology , Cross-Sectional Studies , Female , Humans , Male , Mice , Middle Aged , Reference Values , Retrospective Studies , Risk Factors , Time Factors , United States/epidemiology , Waist-Hip Ratio
5.
Proc Natl Acad Sci U S A ; 111(17): 6485-90, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24733925

ABSTRACT

Hypercoagulability increases risk of thrombi that cause cardiovascular events. Here we identify plasma sodium concentration as a factor that modulates blood coagulability by affecting the production of von Willebrand factor (vWF), a key initiator of the clotting cascade. We find that elevation of salt over a range from the lower end of what is normal in blood to the level of severe hypernatremia reversibly increases vWF mRNA in endothelial cells in culture and the rate of vWF secretion from them. The high NaCl increases expression of tonicity-regulated transcription factor NFAT5 and its binding to promoter of vWF gene, suggesting involvement of hypertonic signaling in vWF up-regulation. To elevate NaCl in vivo, we modeled mild dehydration, subjecting mice to water restriction (WR) by feeding them with gel food containing 30% water. Such WR elevates blood sodium from 145.1 ± 0.5 to 150.2 ± 1.3 mmol/L and activates hypertonic signaling, evidenced from increased expression of NFAT5 in tissues. WR increases vWF mRNA in liver and lung and raises vWF protein in blood. Immunostaining of liver revealed increased production of vWF protein by endothelium and increased number of microthrombi inside capillaries. WR also increases blood level of D-dimer, indicative of ongoing coagulation and thrombolysis. Multivariate regression analysis of clinical data from the Atherosclerosis Risk in Communities Study demonstrated that serum sodium significantly contributes to prediction of plasma vWF and risk of stroke. The results indicate that elevation of extracellular sodium within the physiological range raises vWF sufficiently to increase coagulability and risk of thrombosis.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Sodium/metabolism , Thrombophilia/complications , Thrombophilia/metabolism , Thrombosis/complications , Thrombosis/metabolism , von Willebrand Factor/metabolism , Animals , Dehydration/blood , Dehydration/complications , Dehydration/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Middle Aged , Osmosis/drug effects , Risk Factors , Signal Transduction/drug effects , Sodium/blood , Sodium Chloride/pharmacology , Stroke/blood , Stroke/complications , Stroke/metabolism , Thrombophilia/blood , Thrombosis/blood , Transcription Factors/metabolism
6.
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
7.
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
8.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Proc Natl Acad Sci U S A ; 108(51): 20796-801, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22106305

ABSTRACT

High concentration of NaCl increases DNA breaks both in cell culture and in vivo. The breaks remain elevated as long as NaCl concentration remains high and are rapidly repaired when the concentration is lowered. The exact nature of the breaks, and their location, has not been entirely clear, and it has not been evident how cells survive, replicate, and maintain genome integrity in environments like the renal inner medulla in which cells are constantly exposed to high NaCl concentration. Repair of the breaks after NaCl is reduced is accompanied by formation of foci containing phosphorylated H2AX (γH2AX), which occurs around DNA double-strand breaks and contributes to their repair. Here, we confirm by specific comet assay and pulsed-field electrophoresis that cells adapted to high NaCl have increased levels of double-strand breaks. Importantly, γH2AX foci that occur during repair of the breaks are nonrandomly distributed in the mouse genome. By chromatin immunoprecipitation using anti-γH2AX antibody, followed by massive parallel sequencing (ChIP-Seq), we find that during repair of double-strand breaks induced by high NaCl, γH2AX is predominantly localized to regions of the genome devoid of genes ("gene deserts"), indicating that the high NaCl-induced double-strand breaks are located there. Localization to gene deserts helps explain why the DNA breaks are less harmful than are the random breaks induced by genotoxic agents such as UV radiation, ionizing radiation, and oxidants. We propose that the universal presence of NaCl around animal cells has directly influenced the evolution of the structure of their genomes.


Subject(s)
DNA Breaks, Double-Stranded , DNA/drug effects , Sodium Chloride/chemistry , Animals , Bleomycin/pharmacology , Comet Assay/methods , DNA/genetics , DNA Damage , DNA Fragmentation , DNA Repair Enzymes/chemistry , Histones/chemistry , Kidney/metabolism , Mice , Models, Genetic , Oxidants/chemistry , Phosphorylation , Radiation, Ionizing , Ultraviolet Rays
16.
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
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL