Skip to main content
Physiological Genomics logoLink to Physiological Genomics
. 2009 Dec 8;40(3):167–183. doi: 10.1152/physiolgenomics.00148.2009

Proteomic profiling of nuclei from native renal inner medullary collecting duct cells using LC-MS/MS

Dmitry Tchapyjnikov 1, Yuedan Li 1, Trairak Pisitkun 1, Jason D Hoffert 1, Ming-Jiun Yu 1, Mark A Knepper 1,
PMCID: PMC2825761  PMID: 19996160

Abstract

Vasopressin is a peptide hormone that regulates renal water excretion in part through its actions on the collecting duct. The regulation occurs in part via control of transcription of genes coding for the water channels aquaporin-2 (Aqp2) and aquaporin-3 (Aqp3). To identify transcription factors expressed in collecting duct cells, we have carried out LC-MS/MS-based proteomic profiling of nuclei isolated from native rat inner medullary collecting ducts (IMCDs). To maximize the number of proteins identified, we matched spectra to rat amino acid sequences using three different search algorithms (SEQUEST, InsPecT, and OMSSA). All searches were coupled to target-decoy methodology to limit false-discovery identifications to 2% of the total for single-peptide identifications. In addition, we developed a computational tool (ProMatch) to identify and eliminate ambiguous identifications. With this approach, we identified >3,500 proteins, including 154 proteins classified as “transcription factor” proteins (Panther Classification System). Among these, are members of CREB, ETS, RXR, NFAT, HOX, GATA, EBOX, EGR, MYT1, KLF, and CP2 families, which were found to have evolutionarily conserved putative binding sites in the 5′-flanking region or first intron of the Aqp2 gene, as well as members of EBOX, NR2, GRE, MAZ, KLF, and SP1 families corresponding to conserved sites in the 5′-flanking region of the Aqp3 gene. In addition, several novel phosphorylation sites in nuclear proteins were identified using the neutral loss-scanning LC-MS3 technique. The newly identified proteins have been incorporated into the IMCD Proteome Database (http://dir.nhlbi.nih.gov/papers/lkem/imcd/).

Keywords: vasopressin, transcription, aquaporin


the inner medullary collecting duct (IMCD) is the final portion of the renal collecting duct system. It is responsible for the controlled reabsorption of water from the tubule lumen into the interstitial space of the kidney for eventual return to the bloodstream. The main controlling factor is the peptide hormone vasopressin. Vasopressin mediates rapid regulation of IMCD water permeability by triggering redistribution of the water channel protein aquaporin-2 (Aqp2) from a largely intracellular location to the apical plasma membrane through vesicular trafficking (30). Addition of water channels to the apical plasma membrane increases its permeability to water, allowing accelerated osmotic water transport. In addition to this “classic” mode of regulation, vasopressin has long term effects on the renal collecting duct to increase the total abundance of the Aqp2 protein (10) as well as that of its basolateral counterpart aquaporin-3 (Aqp3) (13).

Vasopressin has been demonstrated to increase transcription of the Aqp2 gene (22, 28, 52), resulting in increased levels of Aqp2 mRNA (12, 16, 51) and protein (31) in kidney tissue. Water restriction increases, while water loading decreases Aqp2 mRNA levels in rat kidney (9, 29, 40). Administration of an orally acting vasopressin V2 antagonist decreased Aqp2 mRNA (16), a finding subsequently confirmed by Christensen et al. (9) and Murillo-Carretero et al. (29). Christensen et al. (9) showed in addition that treatment of rats with a vasopressin V2 receptor antagonist caused renal Aqp2 mRNA levels to fall within 30 min.

In addition to Aqp2, Aqp3 gene expression is regulated by vasopressin as well, with marked increases in levels of Aqp3 mRNA (12, 29) and Aqp3 protein (13, 45). However, the role of transcriptional mechanisms is largely unexplored for Aqp3.

The transcriptional network that governs long-term responses to vasopressin is largely unknown. Recently, we used mRNA profiling (Affymetrix) of native rat IMCD and mouse mpkCCD collecting duct cells, coupled with computational analysis (Genomatix) to identify conserved transcriptional regulator binding site motifs in the 5′-flanking region of the Aqp2 gene to identify transcriptional regulators (TRs) that potentially regulate Aqp2 gene expression (54). The findings demonstrated SF1, NFAT, FKHD, ETS, RXR, AP2, CREB, GATA, SRF, HOX, and EBOX family TR binding sites as likely components of the transcriptional network responsible for regulation of Aqp2 gene transcription. Although these conserved binding site motifs can predict what TR families may be involved in transcriptional regulation of Aqp2 and other genes, identification of the actual TR proteins expressed in the IMCD is a necessary intermediate step to the design of studies needed to fully resolve transcriptional regulatory networks involved in regulation of Aqp2 gene expression. We have previously carried out extensive transcriptomic profiling of IMCD cells using Affymetrix arrays to learn what transcripts are expressed in the IMCD (46) (see “IMCD Transcriptome Database”: http://dir.nhlbi.nih.gov/papers/lkem/imcdtr/). However, relative transcript levels are not necessarily predictive of the level of the corresponding proteins in cells and direct detection by mass spectrometry is desirable. The low abundance of many TR proteins relative to other categories of proteins has made them difficult to detect by protein mass spectrometry and the current IMCD Proteome Database (33) contains only a few TR proteins such as Stat2 and Pax8, detected in whole cell analysis. Notably, a transcription factor whose role is already established, namely Creb1 (22, 28, 52), was not detected using proteomic methods. This problem can be addressed biochemically by isolating nuclei from IMCD cells to enrich nuclear proteins prior to mass spectrometric identification, making it more likely that TRs and other nuclear proteins will be detected. In this study, we have isolated nuclei from native rat IMCDs and carried out proteomic and phosphoproteomic profiling using 1-D SDS-PAGE followed by LC-MS/MS. To increase the number of proteins identified we utilized a computational protocol that includes a combination of three search algorithms (SEQUEST, InsPecT, and OMSSA) for matching mass spectra to rat protein sequences. To limit false-positive identifications we used target-decoy analysis to set the false discovery rate (FDR) to <2% of the total for single-peptide identifications. In addition we developed a novel computational tool (ProMatch) to identify ambiguous identifications, i.e., tryptic or chymotryptic peptides that are the same in two or more proteins. Finally, since regulation of transcription is in part mediated by regulated phosphorylation of various nuclear proteins, we also carried out MS-based experiments to identify phosphoproteins in nuclei from native IMCD cells.

METHODS

IMCD Sample Preparation and Isolation of Nuclei

Twenty male Sprague-Dawley rats were euthanized (National Heart, Lung, and Blood Institute Animal Care and Use Committee-approved protocol H-0110) and their renal inner medullas (IMs) were resected and pooled. The IMs were minced and digested in a solution containing 250 mM sucrose, 2,000 units/ml hyaluronidase, and 3 mg/ml collagenase B for 75 min at 37°C with continuous stirring. IMCDs were sedimented at 70 g for 20 s, and the supernatant containing non-IMCD elements was discarded. IMCD pellet was resuspended in a homogenization solution [250 ml sucrose solution containing phosphatase inhibitor (Halt phosphatase inhibitor, Thermo Scientific, Waltham, MA) and protease inhibitor (Complete mini, Roche Diagnostics, Indianapolis, IN)]. Previous studies have shown that IMCDs isolated by this technique are at least 85% pure (46), viable, and vasopressin-responsive (35).

Isolated IMCDs were suspended in the homogenization solution and homogenized using a Potter-Elvehjem motor-driven homogenizer on ice for a total of 2 min in 15 s intervals. The homogenized solutions were centrifuged at 1,000 g for 30 min at 4°C. The supernatant from the previous step, which contained the cytoplasmic fraction of lysed IMCD cells, was kept. The cell lysis solutions Cytoplasmic Extraction Reagent I and II (CER I and CERII) from the NE-PER Nuclear and Cytoplasmic Extraction Reagents kit (Thermo Scientific) were then added, according to instructions, to the pellet now containing nuclei and unbroken IMCD cells followed by centrifugation for 5 min at 16,000 g. CERI and CERII were again added to the pellet at half the original volumes and the sample was centrifuged as above. All supernatants containing the cytoplasm of lysed cells were pooled (designated “Cyto”). Nuclear extraction solution was then added to the pellet, and the sample was centrifuged according to the kit instructions. The supernatant from the final centrifugation contained soluble nuclear proteins (designated “NE”) and the pellet contained nuclear membrane/ER proteins (designated “NP”).

One-dimensional SDS-PAGE and In-gel Digestion

One-dimensional (1-D) SDS-PAGE was performed on samples from NE, NP, and Cyto fractions (400 μg protein each) using three separate 10% polyacrylamide Ready Gels (Bio-Rad, Hercules, CA). The gels were stained with Imperial Protein Stain (Thermo Scientific). Each gel was sliced into 40 fractions and cut into 1 mm3 blocks. In-gel digestion was performed as described previously (34). Briefly, 25 mM NH4HCO3/50% acetonitrile (ACN) was added to each sample three times for 10 min each to dehydrate the gels and samples were dried via a Speed Vac. 10 mM DTT in 25 mM NH4HCO3 was added to each sample at 56°C for 1 h followed by the addition of 55 mM iodoacetamide in 25 mM NH4HCO3 for 45 min in the dark. The gel blocks were then dried via a Speed Vac. The gel pieces were digested overnight at 37°C with 12.5 ng/μl of Sequencing Grade Modified Trypsin (Promega, Madison, WI). Following the in-gel digestion, peptides were extracted with 50% ACN/0.1% formic acid (FA) then desalted using a ZipTip C18 pipette tip (Millipore, Billerica, MA). In addition, a separate set of 40 NE fractions was in-gel digested overnight at 25°C with 12.5 ng/μl of Sequencing Grade Chymotrypsin (Roche Diagnostics, Indianapolis, IN). In total, 120 samples were analyzed by LC-MS/MS.

Nanospray LC-MS/MS

Each peptide sample was analyzed once via 1-D nanospray LC-MS/MS with a modified ProteomeX 2D LC/MS workstation utilizing a linear ion trap mass spectrometer, LTQ-FT (Thermo, San Jose, CA) (20). In an alternating fashion, two Zorbax 300SB-C18 peptide traps (Agilent Technologies, Wilmington, DE) chromatographically separated the peptides. A nanospray ionization source and a reverse-phase PicoFrit column [BioBasic C18, 75 μm × 10 cm, tip = 15 μm (New Objective, Woburn, MA)] were used. The peptides were eluted via an increasing 0–60% gradient of solvent B in solvent A (A = 0.1% FA, B = 100% ACN/0.1% FA) over a 30 min period at a flow rate of ∼200 nl/min.

Post-LC-MS/MS Analysis

Search algorithms.

To maximize the number of peptide identifications, we analyzed the data using three different search algorithms. In addition to utilizing two peak matching algorithms viz. SEQUEST (15) and the Open Mass Spectrometry Search Algorithm (OMSSA) (17), we also employed InsPecT (44), a so-called “hybrid” algorithm that performs partial de novo sequencing generating 3-amino acid tags that are used to narrow down the possible peptide candidates before performing the final peak matching. A fixed +57 Da modification on cysteine and variable +14 Da cysteine and +16 Da methionine modifications were included as part of the search.

The raw data files were searched, using the three search algorithms, against a concatenated forward and reverse database that included the most recent RefSeq rat protein database from the National Center for Biotechnology Information appended with a list of common contaminants (pig and bovine trypsin as well as human isoforms of keratin). Peptide identifications were sorted from best to worst using the following parameters: XCorr (SEQUEST), P value (InsPecT), and E-value (OMSSA). Target-decoy analysis was performed to limit global FDR for each peptide identified using the following formula: FDR = 2R/(F + R), where R is the number of accumulated peptide hits from reversed “decoy” sequences and F is the number of accumulated peptide hits from forward “target” sequences (14). All peptides passing the 2% FDR filter from the three different search algorithms were merged at the spectral level. For a given spectrum, a valid identification was defined if at least 67% of the passed search algorithms agree on the identification (an agreement of 3 out of 3, 2 out of 3, 2 out of 2, or 1 out of 1). If no agreement among the three search algorithms was found, the single best identification (based on FDR) across all algorithms for a given spectrum was selected. Unresolved spectra were discarded.

Peptide to protein matching.

The same peptide may map to more than one protein. However, peak matching programs like SEQUEST often report a single match even when the peptide could match to multiple protein candidates. To detect ambiguous identifications, we have used an in-house program written in Java called ProMatch (executable file available upon request). In this program, each peptide identified is matched against all proteins in the rat RefSeq database, every protein whose amino acid subsequence identically matches to that peptide is extracted.

The following rules were used in reporting the peptide-based protein identifications. A peptide that matched only to a single protein was defined as a “unique peptide” and reported as such. A peptide that matched to multiple proteins was defined as a “nonunique peptide.” If that peptide matched to multiple proteins that are splicing variants or products of alternative transcription start sites in the same gene, it was considered as a unique peptide since it maps to only a single gene. For this determination, we extracted the nucleotide sequences of all the proteins that matched to a particular peptide and then applied the ClustalW2 sequence alignment program (European Bioinformatics Institute) to compare them. Proteins with a similarity score ≥90 were considered to be derived from the same gene and a single RefSeq accession number is reported for all. A protein identification was labeled as “unambiguous” if it was derived from at least one unique peptide. A protein identification was labeled as “ambiguous” if it was derived exclusively from one or more nonunique peptides derived from different genes. Ambiguous protein identifications were not reported.

Phosphopeptide Analysis

Sample preparation.

IMCDs were isolated and pooled from 10 male Sprague-Dawley rats. Nuclear isolation was performed as described above. The NE and NP fractions (250 and 500 μg, respectively) were resuspended with 6 M guanidine solution. Protein samples were reduced with 10 mM DTT solution at 56°C for 1 h then alkylated with 40 mM iodoacetamide solution in the dark at room temperature for 1 h. DTT solution (40 mM) was added after alkylation to quench excess iodoacetamide. Samples were diluted in 25 mM NH4HCO3 solution and digested with trypsin (1:30 wt/wt) overnight at 37°C. Peptide samples were desalted with a 1 ml HLB cartridge (Oasis, Milford, MA) and then dried via a Speed Vac.

Enrichment of phosphopeptides.

Dried peptide samples were resuspended in 5% acetic acid and Ga3+-IMAC (Phosphopeptide Isolation Kit, Thermo Scientific) was performed as described previously (20). The IMAC flow-through was subjected to an additional phosphopeptide isolation protocol using TopTips with TiO2 Material (PolyLC, Columbia, MD). Phosphopeptide samples were dried with a Speed Vac, resuspended in 0.5% FA, and desalted with ZipTip C18 pipette tips (Millipore).

LC-MS3 analysis.

The neutral loss scanning LC-MS3 analysis was performed as described (20). Briefly, isolated phosphopeptide samples were analyzed on an Agilent 1100 nanoflow system (Agilent Technologies, Palo Alto, CA) connected to a Finnigan LTQ-FT mass spectrometer (Thermo Electron, San Jose, CA) equipped with a nanoelectrospray ion source. The LTQ was used for the full MS scan and subsequent spectra (MS2 and MS3). MS3 scans were triggered when the presence of a neutral loss peak (−98, −49, or −32.7 m/z from precursor ion) was detected in MS2 scans.

Post LC-MS3 analysis.

SEQUEST, InsPecT, and OMSSA searches were performed against the concatenated forward and reverse database as described above. A fixed +57 Da cysteine modification and variable +16 Da methionine and +80 Da phosphorylation on serine, threonine, and tyrosine modifications were included. In addition, a variable loss of water −18 Da modification was included for serine and threonine when searching MS3 spectra. Identified peptides (both phosphopeptides and nonphosphopeptides) were filtered for a 2% FDR using the target-decoy approach described above. The PhosphoPIC program was used to extract and compile MS2 and MS3 spectra of phosphopeptides (21). Phosphorylation site assignment was performed using Ascore (2), Phosphate Localization Score (PLscore) (1), and PhosphoScore (39).

Immunoblotting

Samples were solubilized in 1.5% SDS/Tris, pH 6.8. A BCA assay (Thermo Scientific) using BSA as the standard was used to determine total protein concentrations. Samples were then diluted in Laemmli buffer (10 mM Tris, pH 6.8, 1.5% SDS, 6% glycerol, 0.05% bromphenol blue, and 40 mM DTT). Proteins (5 μg of each sample) were separated by 1-D SDS-PAGE and transferred to nitrocellulose membranes. The membranes were blocked with blocking buffer (LI-COR Biotechnology, Lincoln, NE) then exposed overnight to primary antibodies diluted in the blocking buffer. After a series of washes, membranes were incubated with secondary antibodies as previously described (35) prior to image acquisition on an Odyssey Infrared Imaging System (LI-COR Biotechnology).

Antibodies

Antibodies used are listed as follows: aldose reductase (Alr2) (sc-17735, Santa Cruz Biotechnology, Santa Cruz, CA), BRM/SWI2-related gene 1 (Brg1) (sc-17796, Santa Cruz Biotechnology), and cyclic AMP response element binding protein 1 (Creb1) (9197, Cell Signaling, Danvers, MA). Species-specific secondary antibodies were obtained from Rockland Immunochemicals (Gilbertsville, PA).

Transcription Factor Binding Site Analysis

The 5′-flanking region (1,000 nucleotides) and first intron of Aqp2 and Aqp3 gene of human, rat, and mouse were extracted using UCSC Genome Browser (http://genome.ucsc.edu/). To identify conserved transcription factor binding sites (TFBS), the 5′-flanking region, and first intron sequences were analyzed using the online Genomatix software suite (http://www.genomatix.de/) as previously described (54). Sequence matches to a particular TFBS matrix were scored and filtered based on MatInd algorithm (36).

RESULTS AND DISCUSSION

Confirmation of Nuclear Isolation and Purification from Native Rat IMCD

Common markers for the cytoplasm and nucleus were used to determine the efficacy of the nuclear isolation and enrichment procedure. Figure 1 demonstrates that the nuclear proteins BRM/SWI2-related gene 1 (Brg1) and cyclic-AMP response element binding protein 1 (Creb1) were almost exclusively present in the nuclear fractions. In contrast, the cytosolic marker aldose reductase (Alr2) was far more abundant in the cytoplasmic fractions.

Fig. 1.

Fig. 1.

Quality control for nuclear isolation. Nuclear marker Brg1 is found only in the nuclear extract (NE) fraction. Another nuclear marker Creb1 is chiefly present in the NE fraction but is also found in the nuclear pellet (NP) fraction. Cytoplasmic marker (Alr2) is predominantly present in the cytoplasmic fractions. (Cytoplasm I = Potter-Elvehjem supernatant, Cytoplasm II = CERI/CERII supernatant.) Note that 5 μg of each sample was loaded. In this study we carried out MS analysis of the pooled Cytoplasm I and II (Cyto) as well as NE and NP fractions.

Proteomic Profiling of Rat IMCD Fractions

LC-MS/MS analysis was performed on three fractions derived from the nuclear isolation protocol: the soluble nuclear fraction (NE), the nuclear membrane pellet (NP), and the pooled cytoplasmic fractions (Cyto). Figure 2 summarizes the proteomic and bioinformatic workflow used for these samples. While the sample preparation protocol is relatively standard, we customized the post-LC-MS/MS data analysis for this study. Specifically, three different search algorithms (SEQUEST, InsPecT, and OMSSA) were used to increase the number of peptides identified from the spectra that were generated. The ProMatch algorithm was then used to check whether identified peptide sequences were unique to a single identified protein to allow ambiguous identifications to be eliminated.

Fig. 2.

Fig. 2.

Proteomic work flow. Graphical representation of the complete workflow used for this proteomic study. The ovals represent the experimental samples, spectra, or proteomic data. Shaded boxes represent the biochemical and computational methods that were applied to the biological samples and data.

A total of 2,172 proteins were identified in the NE fraction, 1,306 proteins were identified in the NP fraction, and 1,555 proteins were identified in the cytoplasmic fraction. For all three fractions we imposed a single-peptide FDR of 2% or less using a target-decoy algorithm (Fig. 3A). A total of 3,531 proteins were identified in one or more of the three fractions (all protein identifications are listed in Supplementary Table S1).1 Overall, 45% of protein identifications in this study are based on spectra for 2 or more peptides (Supplementary Table S1). Figure 3B summarizes the number of proteins discovered by each of the three search algorithms (SEQUEST, InsPecT, and OMSSA) in the NE fraction. Also in the NE fraction, 690 proteins were identified from chymotrypsin-digested samples. This yielded 184 proteins that were not found using trypsin and confirmed the presence of 506 proteins found in the NE trypsin dataset (Fig. 3C). It is evident from Fig. 3, B and C, that the use of multiple search algorithms and different proteases are beneficial in increasing the number as well as the confidence of unique peptides and proteins identified. Among the 3,531 protein identifications made in this study, 2,163 (61%) were not identified in the IMCD in previous studies and now have been added to the IMCD Proteome Database (http://dir.nhlbi.nih.gov/papers/lkem/imcd/).

Fig. 3.

Fig. 3.

Venn diagram of proteomic results. A: the number of proteins identified in the NE, NP, and cytoplasmic fractions (Cyto) with an estimated single-peptide false discovery rate specified as 2% or less. A total of 3,531 proteins were identified in one or more of the 3 fractions. B: the number of proteins identified in the NE fraction by each searching algorithm (SEQUEST, InsPecT, and OMSSA). Intersections between each circle show the number of proteins identified by a combination of search algorithms. C: the number of proteins identified in the NE fraction using 2 different proteases (trypsin and chymotrypsin).

Transcription Factors Identified by LC-MS/MS

To enumerate the transcription factors found in the rat IMCD in this study, proteins identified in all the samples were combined into one dataset (Supplementary Table S1). One hundred fifty-four proteins (4.4% of total) were classified as “transcription factor” proteins (Table 1) using the Panther Classification System (http://www.pantherdb.org/). Figure 4 shows a Venn diagram of the distribution of transcription factors identified in the NE, NP, and Cyto fractions, with the majority of these proteins found in the NE fraction. One hundred twenty-eight of these proteins (83%) were new identifications for the IMCD Proteome Database.

Table 1.

Transcription factors

Accession No. Gene Name Sample
CREB transcription factors
NP_001002809 Atf6b (Crebl1) cAMP responsive element binding protein-like 1 NP
NP_604392 Creb1 cAMP responsive element binding protein 1 isoform A NE
NP_001099749 RGD1565584 similar to tyrosine kinase-associated leucine zipper protein LAZipII Cyto
Basic helix-loop-helix transcription factors
NP_036913 Arnt2 aryl hydrocarbon receptor nuclear translocator 2 Cyto
XP_001072661 LOC684826 similar to Basic helix-loop-helix transcription factor scleraxis NP
NP_001020878 Tfeb transcription factor EB NP
NP_113965 Usf1 upstream stimulatory factor 1 NE
NP_112401 Usf2 upstream transcription factor 2 NE
Homeobox transcription factors
NP_476450 Barhl1 BarH-like homeobox 1 NP
XP_001058888 Chx10 similar to Homeobox protein CHX10 (Ceh-10 homeodomain-containing homolog) isoform 2 NE
XP_347164 Cutl1 similar to Homeobox protein cut-like 1 (CCAAT displacement protein) (CDP) (Homeobox protein Cux) NE
NP_036713 Hoxa2 homeo box A2 NE
NP_001017480 Hoxb7 homeo box B7 NE
XP_001069122 LOC688999 similar to Homeobox protein Hox-D8 (Hox-4.3) (Hox-5.4) NE
NP_113925 Nkx6-1 NK6 homeobox 1 Cyto
NP_001099831 Pax2 paired box 2 NE
NP_037133 Pax6 paired box 6 NE
NP_112403 Pax8 paired box 8 NE
NP_001094151 Pbx1 preB-cell leukemia homeobox 1 isoform b NE
XP_001081290 RGD1562142 similar to Homeobox protein Hox-B6 (Hox-2.2) (MH-22A) NE
XP_001081293 RGD1562292 similar to Homeobox protein Hox-B5 (Hox-2.1) (MU-1) (H24.1) Cyto
XP_226533 RGD1562686 similar to genetic suppressor element 1 NE
XP_001073119 RGD1563929 similar to OG2 homeobox Cyto
XP_234082 RGD1564874 similar to NK1 transcription factor related 2-like,b Cyto
XP_001058915 Zfhx4 similar to zinc finger homeodomain 4 NE
NP_001040562 Zhx3 zinc fingers and homeoboxes 3 NE, NP
Nuclear hormone receptors
NP_599227 Dnttip1 deoxynucleotidyltransferase, terminal, interacting protein 1 NE
NP_001008511 Esrra estrogen related receptor, alpha NE
NP_976081 Esrrg estrogen-related receptor gamma NE
NP_113814 Nr1 h2 nuclear receptor subfamily 1, group H, member 2 NP
NP_542956 Nr2f2 nuclear receptor subfamily 2, group F, member 2 NE
NP_036708 Nr3c1 nuclear receptor subfamily 3, group C, member 1 NP
NP_478117 Sf1 splicing factor 1 isoform 2 NP
Zinc finger transcription factors
NP_001099559 Ash2l ash2 (absent, small, or homeotic)-like NE
NP_446008 Bud31 BUD31 homolog NE
NP_114012 Ctcf CCCTC-binding factor (zinc finger protein) NE, NP
NP_001101986 Dpf2 D4, zinc and double PHD fingers family 2 NE, NP
NP_001099893 Evi1 ecotropic viral integration site 1 NE
NP_001029098 Fhl1 four and a half LIM domains 1 isoform 1 Cyto
NP_001101293 Gfi1b growth factor independent 1B transcription repressor NE
XP_218939 Hbxap similar to remodeling and spacing factor 1 Cyto
NP_001100647 Jarid1b jumonji, AT rich interactive domain 1B NP
NP_112397 Klf10 Kruppel-like factor 10 NE
NP_445988 Klf15 Kruppel-like factor 15 Cyto
XP_001072513 LOC684924 similar to zinc finger protein 449 NP
XP_001062820 LOC685209 similar to Zinc finger BED domain containing protein 4 Cyto
XP_001067699 LOC688634 similar to Histone acetyltransferase MYST4 (MYST protein 4) (MOZ, YBF2/SAS3, SAS2 and TIP60 protein 4) (Querkopf protein) NE
NP_001094368 Mtf2 metal response element binding transcription factor 2 NE
NP_851595 Myst2 MYST histone acetyltransferase 2 NE
NP_001099501 Np nucleoside phosphorylase Cyto
NP_001103962 Phf14 PHD finger protein 14 Cyto
NP_001100812 Phf2 PHD finger protein 2 NE
NP_001071116 Prdm2 retinoblastoma protein-binding zinc finger protein Cyto
NP_001005893 Repin1 replication initiator 1 NE
XP_219346 RGD1305903 similar to mesenchymal stem cell protein DSC43 NE
XP_001061052 RGD1310645 similar to Zinc finger protein 407 NE,NP
XP_001057325 RGD1359201 similar to CG15011-PA Cyto
XP_001067122 RGD1560834 similar to zinc finger and BTB domain containing 41 Cyto
XP_233885 RGD1561526 similar to zinc finger protein 512 NP
XP_234528 RGD1563945 similar to PHD finger protein 16 NE
XP_001076564 RGD1564807 similar to zinc finger protein, subfamily 1A, 5 NP
NP_001128016 RGD1565545 hypothetical protein LOC308337 Cyto
NP_997714 Ring1 ring finger protein 1 NE
NP_001094197 Rufy1 RUN and FYVE domain containing 1 NE
NP_695222 St18 suppression of tumorigenicity 18 NE
XP_223196 Wdfy3 similar to WD repeat and FYVE domain containing 3 NP
NP_775412 Yy1 YY1 transcription factor NE, NP
NP_001100567 Zbtb11 zinc finger and BTB domain containing 11 NE
NP_001099350 Zbtb20 zinc finger and BTB domain containing 20 NE, NP
NP_001124009 Zbtb39 zinc finger and BTB domain containing 39 Cyto
NP_446454 Zbtb7a zinc finger and BTB domain containing 7a NE
NP_001010963 Zc3 h15 erythropoietin 4 immediate early response NE
NP_001028873 Zeb2 zinc finger E-box binding homeobox 2 NE, Cyto
NP_001101283 Zfp217 zinc finger protein 217 NE
XP_001059668 Zfp236 similar to Zinc finger protein 236 NE, Cyto
NP_001100701 Zfp278 zinc finger protein 278 Cyto
NP_001100591 Zfp316 zinc finger protein 316 NE
NP_001128057 Zfp426l2 zinc finger protein 426 NP
NP_877975 Zfp472 zinc finger protein 472 NE
NP_714953 Zfp709 zinc finger protein 709 NP
NP_001101279 Znf512b zinc finger protein 512B NE
NP_001019429 Znf574 zinc finger protein 574 Cyto
NP_001132956 Znf644 zinc finger protein 644 NP, Cyto
NP_001073676 Znf652 zinc finger protein 652 NP
XP_001080236 Zzef1 similar to zinc finger, ZZ type with EF hand domain 1 Cyto
Other transcription factors
XP_219832 Aff2 similar to AF4/FMR2 family member 2 (Fragile X mental retardation protein 2 homolog) NP
XP_341162 Ahctf1 similar to transcription factor ELYS NE, Cyto
NP_446456 Cand1 cullin-associated and neddylation-dissociated 1 NE, NP, Cyto
NP_001101391 Casp8ap2 caspase 8 associated protein 2 NE
NP_001013209 Cbfb core-binding factor, beta subunit NE
NP_445979 Cdc5l CDC5 cell division cycle 5-like NE
NP_001028242 Cip29 cytokine induced protein 29 kDa NE
NP_446118 Dll3 delta-like 3 Cyto
NP_001011914 Dr1 down-regulator of transcription 1 NE
NP_942046 Eif3 h eukaryotic translation initiation factor 3, subunit H Cyto
NP_001102912 Erh enhancer of rudimentary homolog NE
NP_001100552 Etv5 ets variant 5 Cyto
NP_001014192 Fam48a transcription factor (p38 interacting protein) NE
NP_001012137 Fus fusion, derived from t(12;16) malignant liposarcoma NE, Cyto
NP_001041351 Ilf2 (Nf45) interleukin enhancer binding factor 2 NE,NP
XP_001076183 LOC500591 similar to calmodulin-binding transcription activator 1 Cyto
XP_001069951 LOC503338 similar to HBxAg transactivated protein 2 Cyto
XP_001057305 LOC681004 similar to GC-rich sequence DNA-binding factor homolog isoform 2 NE
XP_001081073 LOC688079 similar to TAF15 RNA polymerase II, TATA box binding protein (TBP)-associated factor NE
NP_001103001 LOC689296 hypothetical protein LOC689296 Cyto
NP_001101145 Maml3 mastermind like 3 NE
NP_001011999 Morf4l1 mortality factor 4 like 1 NE
NP_001007715 Morf4l2 mortality factor 4 like 2 Cyto
NP_113754 Nfib nuclear factor I/B NE
NP_113755 Nfic nuclear factor I/C NE
XP_342347 Nfkb1 similar to Nuclear factor NF-kappa-B p105 subunit (DNA-binding factor KBF1) (EBP-1) NE, NP
NP_036997 Nfya nuclear transcription factor Y, alpha NE
NP_113741 Nfyb nuclear transcription factor-Y beta NE
NP_001004206 Pa2 g4 ErbB3-binding protein 1 NE, NP
NP_001099311 Ptrf polymerase I and transcript release factor NE,NP, Cyto
NP_071786 Pttg1 pituitary tumor-transforming 1 NP
XP_001063244 Pura similar to Transcriptional activator protein Pur-alpha (Purine-rich single-stranded DNA-binding protein alpha) NE, NP, Cyto
NP_001017503 Purb purine rich element binding protein B NE, Cyto
XP_001071121 Rb1 similar to Retinoblastoma-associated protein (PP105) (RB) NE, NP
NP_001099414 Rfx1 regulatory factor X, 1 (influences HLA class II expression) NE
XP_001056829 RGD1308009 similar to CCR4-NOT transcription complex, subunit 1 isoform a NE, Cyto
XP_001080950 RGD1311017 similar to RIKEN cDNA 4732497O03 NP
XP_001061499 RGD1560020 similar to Myb proto-oncogene protein (C-myb) NE
NP_001102749 RGD1561926 hypothetical protein LOC500695 NP
XP_220843 RGD1562272 similar to TAF11 RNA polymerase II, TATA box binding protein (TBP)-associated factor Cyto
XP_228426 RGD1564748 similar to Mortality factor 4-like protein 2 (MORF-related gene X protein) NP
NP_001012004 Rnf25 ring finger protein 25 NE
NP_001008290 Sbds Shwachman-Bodian-Diamond syndrome homolog NE, NP
NP_001007713 Sdpr serum deprivation response NP
NP_001100834 Sfmbt2 Scm-like with four mbt domains 2 NE
NP_001102231 Sin3a SIN3 homolog A, transcription regulator NE,NP
NP_001108510 Sin3b SIN3 homolog B, transcription regulator NE, NP, Cyto
NP_062064 Smad2 SMAD family member 2 NP
NP_037227 Smad3 SMAD family member 3 NE
NP_062148 Smad4 SMAD family member 4 NE
NP_110485 Smad7 SMAD family member 7 NE
NP_112383 Ssrp1 Structure specific recognition protein 1 NE, NP
NP_036879 Stat3 signal transducer and activator of transcription 3 NE, NP
NP_001011979 Tardbp TAR DNA binding protein NE, NP, Cyto
NP_001099921 Tbx15 T-box 15 NE
NP_001100504 Tbx4 T-box 4 NE
NP_001124046 Tcf20 transcription factor 20 NP
NP_001128186 Tcfcp2 transcription factor CP2 NE, NP
NP_001100640 Tcfcp2l1 transcription factor CP2-like 1 NP
NP_001032431 Tcfcp2l2 transcription factor CP2-like 2 Cyto
XP_001069278 Tead1 similar to Transcriptional enhancer factor TEF-1 (TEA domain family member 1) isoform 2 NE, NP
NP_001100317 Tfdp2 transcription factor Dp-2 (E2F dimerization partner 2) Cyto
NP_001009693 Thrap3 thyroid hormone receptor associated protein 3 NE, NP
NP_001020136 Trappc2 trafficking protein particle complex 2 NE
NP_001128309 Trps1 trichorhinophalangeal syndrome I homolog NP, Cyto
XP_001076644 Ubp1 similar to upstream binding protein 1 (LBP-1a) NP
NP_001099193 Ubtf upstream binding transcription factor, RNA polymerase I isoform 1 NE, NP
NP_620809 Xab2 XPA-binding protein 2 NE
NP_113751 Ybx1 Y box binding protein 1 NE, NP, Cyto
XP_001079683 Ybx2 similar to Y-box-binding protein 2 (Germ cell-specific Y-box-binding protein) (FRGY2 homolog) NE

NE, soluble nuclear fraction; NP, nuclear membrane pellet; Cyto, pooled cytoplasmic fractions.

Fig. 4.

Fig. 4.

Venn diagram of the distribution of transcription factors identified in the NE, NP, and Cyto fractions.

Computational Analysis of Conserved Transcription Factor Binding Elements in Aqp2 and Aqp3 Genes

To relate the transcription factors found (Table 1) to their possible roles in regulation of Aqp2 and Aqp3 gene transcription, we have mapped conserved binding element motifs present in the 5′-flanking region as well as the first intron of rat Aqp2 and Aqp3 genes (Fig. 5), based on analysis using the Genomatix software suite (methods). In the following, we summarize these findings in the context of existing literature.

Fig. 5.

Fig. 5.

Transcription factor binding site analysis. The 1,000 bp 5′-flanking region and 1st intron of the Aqp2 and Aqp3 genes of human, rat, and mouse were analyzed for conserved transcription factor binding sites (TFBS) using Genomatix database and software suite. This figure represents only Rat Aqp2 and Aqp3 genes. Conserved TFBS were found in the 1,000 bp 5′-flanking region of the Aqp2 and Aqp3 genes and the 1st intron of the Aqp2, however, no conserved TFBS were found in the 1st intron of the Aqp3 gene. Transcription factors identified in this study that potentially bind to these TFBS are shown above each TFBS. Abbreviations for Genomatix TFBS family name: V$SF1F = Vertebrate steroidogenic factor; V$NFAT = Nuclear factor of activated T-cells; V$FKHD = Fork head domain factors; V$ETSF = Human and murine ETS1 factors; V$RXRF = RXR heterodimer binding sites; V$AP2F = Activator protein 2; V$CREB = cAMP-responsive element binding proteins; V$GATA = GATA binding factors; V$SRFF = Serum response element binding factor; V$HOXF = Paralog hox genes 1–8 from the four hox clusters A, B, C, D; V$EBOX = E-box binding factors; V$EGRF = EGR/nerve growth factor induced protein C & related factors; V$MYT1 = MYT1 C2HC zinc finger protein; V$KLFS = Krueppel like transcription factors; V$CP2F = CP2-erythrocyte Factor related to drosophila Elf1; V$CEBP = Ccaat/Enhancer Binding Protein; V$PARF = PAR/bZIP family; V$NR2F = Nuclear receptor subfamily 2 factors; V$HAND = Twist subfamily of class B bHLH transcription factors; V$GREF = Glucocorticoid responsive and related elements; V$MAZF = Myc associated zinc fingers; V$ZBPF = Zinc binding protein factors; and V$SP1F = GC-Box factors SP1/GC.

Transcription Factors Potentially Involved in Aqp2 Gene Transcription

Figure 5A shows the transcription factors found in this study (listed in Table 1) that potentially bind conserved transcription factor binding elements in the first 1,000 bp of the 5′-flanking region or first intron of the Aqp2 gene, based on computational analysis using the Genomatix suite. As discussed in the following some of these have been examined experimentally in prior studies.

CREB.

A conserved CREB binding motif (aka CRE) is present at −222 bp from the transcription start site. Originally identified by Uchida and colleagues (48), the importance of this site has been thoroughly documented by the subsequent studies of Hozawa et al. (22), Matsumura et al. (28), Yasui et al. (52), and Cai et al. (5). There are several genes in mammalian genomes that code for proteins that could potentially bind to CRE, including the index protein Creb1, which is activated by phosphorylation at Ser-133 by Ca-calmodulin-dependent kinases, ribosomal S6 kinase, or protein kinase A (41). In this study we identify two of these, Creb1 and Crebl1 (also known as “cAMP responsive element binding protein-like 1”). These contain the so-called “bZIP domain,” comprising a basic region and a leucine zipper region. Crebl1 acts in the unfolded protein response (UPR) pathway by activating UPR target genes induced during endoplasmic reticulum (ER) stress (24). It is a single-pass integral membrane protein that undergoes regulated intramembrane proteolysis in the Golgi during the UPR to yield an ∼400 amino acid cytoplasmic product that translocates into the nucleus.

GATA.

Centered at −214 bp from the transcription start site is a conserved putative GATA sequence, a binding motif for zinc-finger transcription factors of the GATA family. Uchida et al. (47) identified Gata3 mRNA in microdissected collecting ducts and demonstrated that overexpression of Gata3 enhanced Aqp2 promoter-reporter activity. Both Gata2 and Gata3 are expressed in IMCD at many fold above the median signal (46). In the present study, we identified another GATA family member, Trps1 (Tricho-rhino-phalangeal syndrome type I protein), in nuclei from native IMCD cells. This protein is known to bind relatively specifically to GATA sequences and represses GATA-regulated genes (27). Interestingly Rai et al. (37) identified, in the 5′-flanking region of the Aqp2 gene, a region overlapping this GATA site that contained an unidentified cis-element with an apparent negative regulatory role on transcriptional activity. However, in a later study overexpression of the Gata3 transcription factor increased Aqp2 transcription pointing to an enhancer role for this binding element (47), in seeming contradiction to the findings of Rai et al. (37). A possible explanation for this conundrum is that Trps1 normally maintains repression at the GATA site, blocking the enhancer activity of Gata2 and Gata3 that would otherwise occur.

HOX.

In the present study we also identified by mass spectrometry two transcription factors that potentially bind to a conserved HOX binding element centered at −201 bp from the transcription start site, viz., Hoxa2 and Hoxb7. Homeobox or HOX transcription factors are recognized to be involved in renal tubule segmentation as well as collecting duct development (32). The 5′-flanking region of Hoxb7 has been used to target transgene expression specifically to collecting duct cells in mice (38, 42).

NFAT.

Centered at −469 bp upstream from the transcription start site is a potential NFAT binding element. Several NFAT proteins including TonEBP (Nfat5) (19) and the calcineurin-dependent Nfat proteins (Nfatc1–4) (26) have been implicated in Aqp2 transcriptional regulation. Although we did not identify these transcription factors in the present study, we found a protein (Nf45) with weak homology to them that has been previously implicated in regulation of interleukin-2 transcription in lymphocytes (23). Although mRNA levels do not necessarily correlate with protein expression, Affymetrix microarray studies of native IMCD cells demonstrated that Nfat5 (also called TonEBP) is expressed with a signal 12.5-fold above the median signal for all transcripts (46). Interestingly, the same study showed that another Nfat protein is strongly expressed in rat renal IMCD, viz. Nfatc3, which is regulated by intracellular calcium. A rise in calcium activates the Ca-dependent protein phosphatase, calcineurin, which dephosphorylates the Nfat protein allowing it to translocate into the nucleus. A member of this family has recently been implicated in the transcriptional control of the Aqp2 gene (26). The involvement of Ca2+-calcineurin-sensitive regulation of Aqp2 gene transcription identifies another mechanism by which vasopressin can regulate Aqp2 expression, since several laboratories have demonstrated that activation of the V2 vasopressin receptor is associated with an elevation of intracellular calcium (6, 8, 11, 35, 43, 53).

ETS.

Centered at −455 bp from the transcriptional start site of the Aqp2 gene is a conserved Ets binding motif that we identified in a previous study (54) as playing an enhancer role in cell-specific expression of Aqp2, and possibly playing a role in the vasopressin response. In the present study, MS analysis identified Etv5 (ets variant gene 5 or PEA3), a member of the ets family. This transcription factor is known to be selectively expressed in ureteric bud (the developmental precursor of the collecting duct system) (7).

RXR.

Centered at −342 bp from the transcription start site is a conserved putative RXR binding motif. RXR binding elements bind dimers of ligand-activated transcription factors, usually RAR/RXR heterodimers. These transcription factors are known to be involved in development of the ureteric bud (4). In addition, RXR can heterodimerize with vitamin D receptors, peroxisome proliferator activator receptors, thyroid receptors, and other ligand-activated nuclear receptors. MS analysis in the present paper found one such ligand-activated transcription factor, viz. Nr1h2 (nuclear receptor subfamily 1 group H member 2, also known as “liver X receptor”). Interestingly, this is one of two transcription factor genes (with Elf1) whose mRNA levels correlated negatively with Aqp2 mRNA levels among subcloned mpkCCD collecting duct cell lines (54). Its endogenous ligand is at present unknown.

EBOX.

A conserved EBOX motif is located at −67 bp relative to the transcription start site of Aqp2. Two transcription factors were identified that potentially bind this site, namely Usf1 and Usf2. These are classical basic helix-loop-helix transcription factors known to be involved in transcriptional regulation in a variety of tissues.

Potential binding elements in the first intron.

We identified four potential transcription factor binding elements in a conserved region of the first intron of the Aqp2 gene (Fig. 5A), viz. binding sites for EGR, MYT1, KLF, and CP2 family transcription factors. The transcription factors identified in IMCD nuclei that correspond to these sites (Zbtb7a, St18, Klf15, Tcfcp2, and Tcfcp2l1) are all of the zinc-finger or the CP2 classes of transcription factors and typically play repressor roles. Klf15 has been previously implicated in regulation of the ClC-K1 chloride channel of the thin ascending limb of Henle (49). Tcfp2l1 is associated with normal duct development in both the salivary gland and kidney (50).

Transcription Factors Potentially Involved in Aqp3 Gene Transcription

Figure 5B shows the transcription factors found in this study (Table 1) that potentially bind conserved transcription factor binding elements in the first 1,000 bp of the 5′-flanking region or first intron of the Aqp3 gene, based on computational analysis using the Genomatix suite. No conserved binding motifs were found in the first intron.

The 5′-flanking region of the Aqp3 gene contains several conserved transcription factor binding element motifs seen in the Aqp2 5′-flanking region or first intron including two EBOX motifs (potentially binding Usf1 and Usf2), an NR2 nuclear receptor site (potentially binding the orphan nuclear receptor Nrf2f), and two KLF sites (potentially binding Klf15). These sites are potentially responsible for the coordinate regulation of Aqp2 and Aqp3 seen in response to vasopressin (45). In addition, the 5′-flanking region of the Aqp3 gene contains some interesting unique sites that may provide some of the explanation for the difference in the tissue distribution of Aqp3 expression vs. Aqp2 expression, as well as differences in regulation. Foremost among these unique sites is a putative GRE (glucocorticoid response element), which can bind either the glucocorticoid receptor (Nr3c1) or the mineralocorticoid receptor (Nr3c2). Although only the former was found by MS analysis of the nuclear fraction in the current study, the latter is also known to be strongly expressed in the rat renal IMCD (46), providing a potential explanation for the large increase in Aqp3 protein abundance seen in the renal collecting duct in response to treatment of rats with the mineralocorticoid, aldosterone (25).

In addition, the 5′-flanking region of the Aqp3 gene contains two highly conserved “MAZ” or “MYC-associated zinc finger” sites at −62 and −79 bp relative to the transcriptional start site. Genomatix analysis predicts that one transcription factor from Table 1 would bind to these sites, viz. Zbtb19 (aka Zfp278 or “protein kinase A RI subunit α-associated protein”). Like other zinc-finger transcription factors, this transcription factor is associated with repressor activity. Finally, the 5′-flanking region of the Aqp3 gene contains a highly conserved SP1 binding element motif at −62 bp, which potentially binds another zinc-finger transcription factor found in this study by mass spectrometry, viz. Klf10 (also known as “TGF-β-inducible early growth response protein 1”), that likely manifests repressor function.

Transcriptional Co-regulators and Nucleic Acid Binding Proteins Identified by Proteomic Analysis of IMCD

Transcriptional co-regulators are listed in Table 2. Proteins classified as “nucleic acid binding” proteins by Panther analysis are listed in Supplementary Table S2. This category includes basal transcription factors, helicases, RNA polymerases, and other ubiquitous proteins. Both lists are made up largely of proteins that would be found in any cell type.

Table 2.

Transcriptional co-regulators

Accession No. Gene Name Sample
NP_062093 Aes amino-terminal enhancer of split NP
NP_001101094 Arid5b AT rich interactive domain 5B (Mrf1 like) NP
NP_001101923 Cbfa2t3 core-binding factor, runt domain, alpha subunit 2, translocated to, 3 NE
NP_445981 Ciita class II transactivator NE
NP_596872 Crebbp CREB binding protein NE
NP_445787 Ctbp2 C-terminal binding protein 2 NE
XP_341396 Dcp1a similar to mRNA decapping enzyme 1A (Smad4-interacting transcriptional co-activator) NE
NP_001102523 Dtx3l deltex 3-like NE
NP_001100027 Edf1 endothelial differentiation-related factor 1 NE
NP_001100774 Ell elongation factor RNA polymerase II Cyto
XP_576312 Ep300 similar to E1A binding protein p300 NE
NP_598232 Hdgfrp2 hepatoma-derived growth factor-related protein 2 NE
NP_001009625 Ifi35 interferon-induced protein 35 NE
XP_001061895 LOC360888 similar to lin-9 homolog NE
XP_001055295 LOC679225 similar to Ladybird homeobox corepressor 1 NP
XP_001054608 LOC679693 similar to Mediator of RNA polymerase II transcription subunit 12 isoform 1 NE
XP_001080941 LOC688035 similar to transformation related protein 53 binding protein 1 NE
XP_001066953 LOC689062 similar to nuclear DNA-binding protein NE
NP_001019932 Lrrfip2 leucine rich repeat (in FLII) interacting protein 2 NE, NP
NP_001100271 Med17 mediator complex subunit 17 NE
NP_001100035 Med27 mediator complex subunit 27 NP
NP_114010 Ncoa2 nuclear receptor coactivator 2 NP
XP_221724 Nrip1 similar to nuclear receptor interacting protein 1 NE
NP_001101968 Nrip3 nuclear receptor interacting protein 3 NE
NP_001094308 Prkcbp1 protein kinase C binding protein 1 NE
NP_786941 Psip1 PC4 and SFRS1 interacting protein 1 NE, NP
NP_001100644 Rbbp5 retinoblastoma binding protein 5 NP
NP_001101164 Rfx5 regulatory factor X, 5 (influences HLA class II expression) NE
XP_221241 RGD1304870 similar to transcription elongation factor B (SIII), polypeptide 2 NP
XP_223519 RGD1561605 similar to Ada2b CG9638-PA, isoform A Cyto
NP_671706 Ruvbl1 RuvB-like 1 NE, NP
NP_001020576 Ruvbl2 RuvB-like 2 NE, NP
NP_071789 Safb scaffold attachment factor B NE, Cyto
NP_073185 Snd1 staphylococcal nuclease domain containing 1 NE, Cyto
NP_001009618 Sub1 RNA polymerase II transcriptional coactivator NE, NP
NP_001100731 Supt16 h suppressor of Ty 16 homolog NE, NP
NP_001101374 Tgs1 trimethylguanosine synthase homolog NE, NP
XP_233081 Thoc2 similar to THO complex subunit 2 (Tho2) NE
NP_446368 Trim28 tripartite motif-containing 28 NE, NP, Cyto
XP_345267 Trim33 similar to tripartite motif protein 33 NE
NP_001101666 Wtip WT1-interacting protein Cyto
NP_976244 Zmynd11 zinc finger, MYND domain containing 11 delta isoform NE

Phosphoproteomic Analysis

Figure 6 summarizes the workflow utilized for the discovery of phosphoproteins in NE and NP fractions from rat IMCD. Three phosphorylation site verification tools were used [Ascore (2), Phosphate Localization Score (PLscore) (1), and PhosphoScore (39)] to increase our confidence in the identified phosphorylation sites. Table 3 summarizes the 122 phosphorylation sites found. Of those, 63 were previously unidentified phosphorylation sites (not present in Phosphosite, http://www.phosphosite.org). Eight phosphorylation sites in six proteins were identified as a “transcription factor” or “transcription co-regulator” by Panther, namely Bclaf1 (S512), Hbxap (S608 and S1352), Lrrfip2 (S133), Ptrf (S169), Safb (S309), and Trim28 (S52 and S474). The presence of phosphorylated COOH-terminal tails of two aquaporins in the nuclear fractions raises the possibility of a role for regulated intramembrane proteolysis (3) of aquaporin proteins in transcriptional regulation in the IMCD, a possibility that needs further investigation. However, we cannot rule out the presence of these aquaporin phosphopeptides in nuclear fractions owing to contamination with small amounts of ER membranes, which are essentially extensions of the nuclear envelope.

Fig. 6.

Fig. 6.

Phosphoproteomic work flow. Graphical representation of the sample preparation, LC-MS3, and bioinformatic steps taken in the nuclear phosphoproteomic study. Ovals represent the sample or proteomic data. Shaded boxes represent the biochemical and computational methods that were applied to the sample or proteomic data. The work flow utilized 2 phosphopeptide isolation kits (IMAC and TiO2 tips) to maximize phosphopeptide recovery and 3 phosphorylation site verification programs (Ascore, PLscore, and PhosphoScore) to ensure phosphosite validity.

Table 3.

Phosphorylation sites identified in nuclear fractions

Accession No. Gene Name Sequence PhosphoSite
NP_001037859 Ablim1 actin-binding LIM protein 1 S*GLHRPVSTDFAQYNSYGDVSGGVR S311^
TLS*PTPSAEGFQDGR S115^
XP_574618 Ahnak similar to AHNAK nucleoprotein isoform 1 FKAEAALPS*PK S2257
XP_216539 Aim1l similar to absent in melanoma 1 AVRDYCT*PR T1271^
NP_001029156 Ank3 ankyrin 3, epithelial isoform 2 RQS*FTSLALR S1458^
NP_036910 Aqp1 aquaporin 1 VWTSGQVEEYDLDADDINS*R S262^
NP_037041 Aqp2 aquaporin 2 (collecting duct) QS*VELHS*PQSLPR S256, S261
RQS*VELHSPQSLPR S256
RQSVELHS*PQSLPR S261
NP_001029312 Baiap2l1 BAI1-associated protein 2-like 1 SIS*TVDLTEK S423
NP_001041317 Bclaf1 BCL2-associated transcription factor 1 LKELFDYS*PPLHK S512^
NP_001102045 Brd3 bromodomain containing 3 SES*PPPLSEPK S263^
NP_114175 Calm1 calmodulin 1 HVMT*NLGEKLT*DEEVDEMIR T111^, T118
NP_446072 Cdc42bpb Cdc42-binding protein kinase beta HSTPSNSSNPSGPPS*PNSPHR S1692
NP_001101634 Cgnl1 cingulin-like 1 EGVGEETLS*PR S251^
NCFPKPCGS*QPNS*PTPEDLAK S198, S202
NCFPKPCGSQPNS*PT*PEDLAK S202, T204^
NP_001007146 Ctnna1 catenin (cadherin-associated protein), alpha 1, 102 kDa TPEELDDS*DFETEDFDVRSRTS*VQTEDDQLIAGQSAR S643S657
NP_001101210 Ctnnd1 catenin (cadherin associated protein), delta 1 GS*LASLDSLRK S346
GSLAS*LDSLRK S349
VGGS*SVDLHR S268
XP_225259 Dsp similar to desmoplakin isoform I isoform 2 NLTIRS*SSLS*DPLEESSPIAAIFDTENLEK S2613, S2617
NP_114008 Dvl1 dishevelled, dsh homolog 1 HHFLGIS*IVGQS*NDR S265^, S270^
NP_001013122 Eef1d eukaryotic translation elongation factor 1 delta GATPAEDDEDNDIDLFGS*DEEEEDKEAAR S531
KGATPAEDDEDNDIDLFGS*DEEEEDKEAAR S531
NP_067713 Epb4.1l1 erythrocyte protein band 4.1-like 1 isoform L HQAS*INELKR S510
XP_217039 eplin similar to Epithelial protein lost in neoplasm (mEPLIN) GENEETLGRPAQPPSAGETPHS*PGVEDAPIAKRLS*ENSCSLDDLEIGAGHLSSSAFNSEK S488S225
SEAQQPIYTKPLS*PDAR S360
SPKPLS*PSLR S608
TSSVKS*PKPLS*PSLRK S603^, S608
TSSVKS*PKPLSPS*LRK S603^, S610^
NP_001124037 Fam83 h family with sequence similarity 83, member H KGS*PTPAYPER S860
NP_001128071 Flna filamin, alpha RAPS*VANVGSHCDLSLK S2144
XP_218939 Hbxap similar to remodeling and spacing factor 1 IES*DEEEDFENVGK S1352^
TSVDKDIQRLS*PIPEEVVR S608^
NP_001020580 Hdac1 histone deacetylase 1 MLPHAPGVQMQAIPEDAIPEES*GDEDEEDPDKR S393
NP_446159 Hdgf hepatoma-derived growth factor AGDMLEDS*PKRPK S165
GNAEGS*S*DEEGKLVIDEPAKEK S132, S133
RAGDMLEDS*PKRPK S165
NP_579819 Hist1 h1d histone cluster 1, H1d KAS*GPPVSELITK S36
RKAS*GPPVSELITK S36
NP_073177 Hist1 h4b histone cluster 1, H4b RIS*GLIYEETR S48
NP_001101168 Hist2 h3c2 histone cluster 2, H3c2 FQSSAVMALQEASEAY*LVGLFEDTNLCAIHAK Y100^
NP_058944 Hnrnpa1 heterogeneous nuclear ribonucleoprotein A1 SES*PKEPEQLR S6
SES*PKEPEQLRK S6
NP_476482 Hnrnpk heterogeneous nuclear ribonucleoprotein K IIPTLEEGLQLPS*PTATSQLPLESDAVECLNYQHYK S116^
NP_114176 Hspb1 heat shock protein 1 QLS*S*GVSEIR S85, S86^
QLS*SGVSEIR S85
SPS*WEPFR S15
SPS*WEPFRDWYPAHSR S15
NP_001101412 Lad1 ladinin LPS*VEEAEVSKPSPPASKDEGEEFQAILR S62
NP_001002016 Lmna lamin A isoform C2 LRLS*PS*PTSQR S389, S391
LRLS*PSPTSQR S389
LRLSPS*PTSQR S391
NP_001001515 Lmo7 LIM domain 7 SRS*TTELNDPLIEK S1476^
XP_001080824 LOC360570 similar to myosin XVIIIa RFS*FSQR S140
SLAPDLS*DDEHDPVDSISRPR S2020^
XP_341387 LOC361100 similar to topoisomerase (DNA) II beta KTS*FDQDSDVDIFPSDFTSEPPALPR S367^
KTSFDQDS*DVDIFPSDFTSEPPALPR S372
KVVEPANS*DS*DSELGNIPK S312^, S314^
VKAS*PITNDGEDEFVPS*DGIDKDEYAFSPGK S189, S202^
VKAS*PITNDGEDEFVPSDGIDKDEYAFSPGK S189
XP_573444 LOC498222 similar to specifically androgen-regulated protein APAQPAS*PALVR S396^
XP_001056725 LOC681423 similar to delangin isoform A AITSLLGGGS*PK S2431^
XP_001061576 LOC682450 similar to CG31613-PA FQS*S*AVIALQEASEAYLVGLFEDTNLCAIHAK S87^, S88^
XP_001068152 LOC684233 similar to Putative RNA-binding protein 15 (RNA-binding motif protein 15) (One-twenty two protein) DRT*PPLLYRLLLLERPS*PVRDR T567S701^
XP_001071565 LOC684681 similar to Histone H1.2 (H1 VAR.1) (H1c) S*ET*APAAPAAAPPAEKAPAK S108^, T110^
XP_001055673 LOC685179 similar to SWI/SNF-related matrix-associated actin-dependent regulator of chromatin c2 isoform b isoform 2 DMDEPS*PVPNVEEVTLPK S347^
XP_001078152 LOC686579 similar to AHNAK nucleoprotein isoform 2 isoform 1 KGDRS*PEPGQTWAHEVFSSR S94
NP_001019932 Lrrfip2 leucine rich repeat (in FLII) interacting protein 2 RGS*GDTSSLIDPDTSLSELR S133^
NP_037198 Map2 microtubule-associated protein 2 ARVDHGAEIITQS*PSR S1780
NP_113708 Myh10 myosin, heavy chain 10, nonmuscle QLHIEGAS*LELS*DDDTESKQLHIEGASLELS*DDDTESK S1952^, S1956S1956
RQLHIEGASLELS*DDDTESK S1956
NP_037326 Myh9 myosin, heavy chain 9, nonmuscle GTGDCS*DEEVDGKADGADAK S1944
NP_631977 Nexn nexilin (F actin binding protein) isoform b AEIKDMLAS*DEEEEPSKVEK S16
NP_037124 Npm1 nucleophosmin (nucleolar phosphoprotein B23, numatrin) CGSGPVHISGQHLVAVEEDAES*EDEDEEDVK S125
NP_073636 Nucks1 nuclear casein kinase and cyclin-dependent kinase substrate 1 TSAS*PPLEK S223^
XP_218972 Numa1 similar to nuclear mitotic apparatus protein 1 AAQLQGS*PAPEKGEVLGDALQLDTLKQEAAK S416^
NP_001106799 Pde4d phosphodiesterase 4D isoform 1 KSRMS*WPSSFQGLR S137^
NP_071796 Plec1 plectin 1 isoform 1 KQIT*VEELVR T4033^
SSS*VGSSSSYPISSAVPR S4389
NP_001101576 Plxnb2 plexin B2 ADPAY*RCVWCRGQNR Y779^
XP_343923 Polr2a similar to DNA-directed RNA polymerase II largest subunit (RPB1) GHGGCGRY*QPRIR Y187^
NP_001099311 Ptrf polymerase I and transcript release factor LPAKLS*VSK S169^
NP_001013225 Rbm39 RNA binding motif protein 39 DKS*PVREPIDNLTPEER S136
XP_578547 RGD1562028 similar to Mitogen-activated protein kinase kinase kinase kinase 5 (MAPK/ERK kinase kinase kinase 5) (MEK kinase kinase 5) (MEKKK 5) TAS*EINFDKLQFEPPLRK S316^
XP_001075668 RGD1564067 hypothetical protein T*KVEEEPPPPPPPPPPPPPPPPPR T104^
NP_071789 Safb scaffold attachment factor B APTAAPS*PEPR S309
XP_217021 Sbf1 similar to SET binding factor 1 isoform a LGLGTLSS*SLS*R S1142^, S1145^
NP_476489 Sept2 septin 2 IYHLPDAES*DEDEDFKEQTR S218
IYHLPDAESDEDEDFKEQT*R T228^
NP_789826 Sept9 septin 9 isoform 2 LVDTLSQRS*PKPSLR S67^
NP_001009255 Sfrs9 splicing factor, arginine/serine rich 9 GS*PHYFSPFRPY S211
NP_690921 Slc4a9 solute carrier family 4, sodium bicarbonate cotransporter, member 9 WS*APHVPTLALPSLQK S89^
NP_037165 Slc5a1 solute carrier family 5 (sodium/glucose cotransporter), member 1 MT*KEEEEAMKLK T622^
NP_062251 Snip SNAP25-interacting protein RCRVVT*DTLAQIRR T853^
NP_001011981 Snx17 sorting nexin 17 S*QEYKIVLRK S194^
NP_741984 Spna2 alpha-spectrin 2 WRS*LQQLAEER S1217
NP_001101456 Srrm1 serine/arginine repetitive matrix 1 EKS*PELPEPSVR S220^
RYS*PPIQR S601
NP_001099736 Tjp1 tight junction protein 1 VQIPVSHPDPDPVS*DNEDDSYDEDVHDPR S113^
VQIPVSHPDPDPVSDNEDDS*YDEDVHDPR S119^
NP_446225 Tjp2 tight junction protein 2 KVQVAPLQGS*PPLSHDDR S107
NP_037019 Tmpo thymopoietin GPPDFSSDEEREPT*PVLGSGASVGR T74
NP_446368 Trim28 tripartite motif-containing 28 RPAASSAAAASASASS*PAGGGGEAQELLEHCGVCR S52^
SRS*GEGEVSGLMR S474^
NP_001026829 Trip12 thyroid hormone receptor interactor 12 S*ESPPAELPSLR S310^
NP_037202 Utrn utrophin AAQAS*LSALNDPSAVEQALQEK S933^
NP_973718 Xirp2 xin actin-binding repeat containing 2 CFET*QPLY*VIR T507^, Y511^
NP_001101423 Zcchc11 zinc finger, CCHC domain containing 11 FILTS*GKPPTIVCS*ICK S768^, S777^
^

Previously unidentified.

Kinases in Nuclear Fractions

Table 4 categorizes the kinases found in this study, which include 58 kinases present in the nuclear extract fraction. While all are candidates for regulatory roles in transcription in IMCD, some already have well documented roles in other tissues. For example, all three of the kinases known to phosphorylate Creb1 at Ser133 are present in either the NE or NP fraction, viz. protein kinase A catalytic subunit α (Prkaca), calcium/calmodulin-dependent protein kinase II δ and γ (Camk2d and Camk2g), and ribosomal protein S6 kinase (Rps6ka5 and Rps6kc1) (41). In addition several MAP kinases were detected, including Erk1 (Mapk3), Erk2 (Mapk1), Erk3 (Mapk6), and p38α (Mapk14), which are known to phosphorylate transcription factors of the ETS, AP1, and GATA families (Phosphosite: http://www.phosphosite.org/). Finally, the Hipk2 protein phosphorylates a number of transcription factors including p53, Pax6, and Zbtb4 (Phosphosite).

Table 4.

Protein kinases in nuclear fractions

Accession No. Gene Name Sample
AGC Ser/Thr protein kinase family
NP_001101987 Ccdc88b coiled-coil domain containing 88B NP
NP_446072 Cdc42bpb Cdc42-binding protein kinase beta NE, NP, Cyto
NP_001025082 Cit citron (rho-interacting, serine/threonine kinase 21) NE
NP_112358 Grk1 G protein-coupled receptor kinase 1 NE, NP, Cyto
NP_001100737 Lats2 large tumor suppressor 2 NE
NP_851603 Mast1 microtubule associated serine/threonine kinase 1 NE
NP_001099225 Pkn2 protein kinase N2 NE
NP_001094392 Prkaca cAMP-dependent protein kinase catalytic subunit alpha NP, Cyto
NP_112360 Rock1 Rho-associated coiled-coil containing protein kinase 1 NE, Cyto
NP_037154 Rock2 Rho-associated coiled-coil containing protein kinase 2 NE, Cyto
NP_001101518 Rps6ka5 ribosomal protein S6 kinase, polypeptide 5 NP
NP_001099454 Rps6kc1 ribosomal protein S6 kinase, 52 kDa, polypeptide 1 NP
NP_001076805 Stk38l serine/threonine kinase 38 like NE
Atypical: PDK/BCKDK protein kinase family
NP_110499 Pdk2 pyruvate dehydrogenase kinase, isozyme 2 NE
Atypical: PI3/PI4-kinase family
NP_001100291 Atm ataxia telangiectasia mutated homolog NE
NP_063971 Mtor FK506 binding protein 12-rapamycin associated protein 1 NE, Cyto
NP_001101797 Prkdc protein kinase, DNA activated, catalytic polypeptide NE
CAMK Ser/Thr protein kinase family
NP_036651 Camk2d calcium/calmodulin-dependent protein kinase II delta NE, NP, Cyto
NP_598289 Camk2 g calcium/calmodulin-dependent protein kinase II gamma NE
NP_071991 Dapk3 Death-associated protein kinase 3 NE
NP_067731 Mark2 MAP/microtubule affinity-regulating kinase 2 NE
NP_570105 Mark3 MAP/microtubule affinity-regulating kinase 3 NE
NP_001007618 Nuak2 NUAK family, SNF1-like kinase, 2 NP, Cyto
NP_001009362 Pask PAS domain containing serine/threonine kinase NP
NP_062015 Prkaa1 protein kinase, AMP-activated, alpha 1 catalytic subunit NP
XP_230713 RGD1565143 Sperm motility kinase X NE
NP_001011900 Tssk1 testis-specific serine kinase 1 NE
CK1 Ser/Thr protein kinase family
NP_446067 Csnk1a1 casein kinase 1, alpha 1 NE, NP
NP_001012194 Vrk1 vaccinia related kinase 1 NE, NP
CMGC Ser/Thr protein kinase family
NP_543161 Cdk5 cyclin-dependent kinase 5 NP
NP_059040 Gsk3a glycogen synthase kinase 3 alpha NE
NP_114469 Gsk3b glycogen synthase kinase 3 beta NE
NP_001102092 Hipk2 homeodomain interacting protein kinase 2 NE
NP_446294 Mapk1 mitogen activated protein kinase 1 NE, NP
NP_112282 Mapk14 mitogen-activated protein kinase 14 NP
NP_059043 Mapk3 mitogen activated protein kinase 3 NE
NP_113810 Mapk6 mitogen-activated protein kinase 6 NE
NP_001093976 Pctk3 PCTAIRE protein kinase 3 NE, NP, Cyto
NP_001011923 Prpf4b PRP4 premRNA processing factor 4 homolog B NE, NP
NP_001020897 Srpk1 SFRS protein kinase 1 NE
NEK Ser/Thr protein kinase family
XP_224971 Nek3 similar to Serine/threonine-protein kinase Nek3 (NimA-related protein kinase 3) NP
NP_001099274 Nek8 NIMA-related kinase 8 NE
STE Ser/Thr protein kinase family
NP_596900 Ilk integrin linked kinase NE, NP
XP_001079680 LOC687705 similar to misshapen-like kinase 1 isoform 1 isoform 1 NE
NP_113831 Map2k1 mitogen-activated protein kinase kinase 1 NP
NP_579817 Map2k2 mitogen activated protein kinase kinase 2 NE
NP_001094144 Map2k3 mitogen activated protein kinase kinase 3 NP
NP_001100374 Map4k4 mitogen-activated protein kinase kinase kinase kinase 4 NE
NP_001101664 Oxsr1 oxidative-stress responsive 1 NP
NP_445758 Pak2 p21-activated kinase 2 NP
NP_001099708 Pak4 p21 (CDKN1A)-activated kinase 4 NE
XP_001070192 RGD1563568 similar to serine/threonine protein kinase MASK NE, NP
NP_062222 Slk serine/threonine kinase 2 NE, NP, Cyto
XP_577078 Stk10 similar to Serine/threonine-protein kinase 10 (Lymphocyte-oriented kinase) NE
NP_001120966 Stk24 serine/threonine kinase 24 NE
NP_113923 Stk3 serine/threonine kinase 3 NP
NP_062235 Stk39 Ste-20 related kinase (SPAK) NP
NP_775449 Taok1 TAO kinase 1 NE
Tyr protein kinase family
XP_001079831 Abl1 similar to Proto-oncogene tyrosine-protein kinase ABL1 (p150) NE
NP_001025210 Csk c-src tyrosine kinase NE,NP
NP_001102447 Epha2 ephrin receptor EphA2 NE, NP, Cyto
NP_599158 Epha7 ephrin receptor EphA7 NE
NP_001120791 Ephb2 Eph receptor B2 NE, Cyto
NP_077059 Fgr proto-oncogene tyrosine-protein kinase Fgr NE
NP_077344 Frk fyn-related kinase NP, Cyto
NP_036887 Fyn FYN oncogene related to SRC, FGR, YES NE
NP_058767 Insr insulin receptor NP
NP_445918 Jak1 Janus kinase 1 NP
NP_110484 Lyn Yamaguchi sarcoma viral (v-yes-1) oncogene homolog isoform A NE,NP
NP_001100785 Pragmin pragmin NE, Cyto
NP_114183 Src v-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog NP, Cyto
NP_001099207 Tek TEK tyrosine kinase, endothelial NE
NP_001100482 Tnk1 tyrosine kinase, nonreceptor, 1 NP
NP_150640 Yes1 Yamaguchi sarcoma viral oncogene homolog 1 NE, NP, Cyto
Other protein kinases
NP_112628 Camkk2 calcium/calmodulin-dependent protein kinase 2 beta NP
NP_446276 Csnk2a1 casein kinase 2, alpha 1 polypeptide NE, Cyto
NP_001100879 Csnk2a2 casein kinase 2, alpha prime polypeptide NE, Cyto
NP_062208 Eif2ak2 eukaryotic translation initiation factor 2-alpha kinase 2 NP
XP_226572 RGD1306091 similar to cDNA sequence BC021891 NP
XP_577704 RGD1560972 similar to serine/threonine kinase NE
XP_001076790 RGD1562028 similar to Mitogen-activated protein kinase kinase kinase kinase 5 (MEK kinase kinase 5) (MEKKK 5) NE
XP_001080090 RGD1562674 similar to kinase suppressor of ras 2 NE

In general, of the 82 kinases listed in Table 4, 36 of them are classified by Gene Ontology (component) as located in “nucleus.” Among these are kinases that have housekeeping roles in the nucleus, such as DNA repair (Atm and Prkdc), mRNA splicing (Stk3), and cell cycle regulation (Cit, Cdk5, Gsk3β, and Mtor).

Phosphatases

Table 5 categorizes the 17 phosphatase proteins found in nuclear fractions in this study. These proteins include receptor and nonreceptor tyrosine phosphatases, serine/threonine phosphatases, and dual-specificity phosphatases. All could potentially play roles in transcriptional regulation in the collecting duct.

Table 5.

Protein phosphatases in nuclear fractions

Accession No Gene Name Sample
Dual-specificity phosphatases
NP_001012352 Dusp26 Dual-specificity phosphatase 26 NP
XP_001071732 Dusp27 PREDICTED: similar to Dual specificity protein phosphatase 13 (Testis- and skeletal-muscle-specific DSP) NE
NP_001013065 Mtm1 X-linked myotubular myopathy gene 1 NP
NP_001101593 Mtmr2 myotubularin-related protein 2 NE
Receptor tyrosine phosphatases
NP_062122 Ptprf protein tyrosine phosphatase, receptor type, F NE, NP
NP_599183 Ptprg protein tyrosine phosphatase, receptor type, G NE
NP_058965 Ptprj protein tyrosine phosphatase, receptor type, J NP
XP_001063025 Ptpru PREDICTED: similar to protein tyrosine phosphatase, receptor type, L NE
Nonreceptor tyrosine phosphatases
NP_446360 Ptpn6 protein tyrosine phosphatase, nonreceptor type 6 NE, NP
NP_037220 Ptpn11 protein tyrosine phosphatase, nonreceptor type 11 NE, NP
XP_213997 Ptpn13 PREDICTED: similar to Tyrosine-protein phosphatase nonreceptor type 13 NE
NP_001100670 Ptpn14 protein tyrosine phosphatase, nonreceptor type 14 NP
Serine/threonine phosphatases
NP_037197 Ppp1cb protein phosphatase 1, catalytic subunit, beta NE, Cyto
NP_071943 Ppp1 ml protein phosphatase 1, catalytic subunit, gamma isoform NE
NP_058735 Ppp2ca protein phosphatase 2a, catalytic subunit, alpha isoform NE
NP_476481 Ppp2r1a alpha isoform of regulatory subunit A, protein phosphatase 2 NE, NP, Cyto
NP_852044 Ppp2r5b protein phosphatase 2, regulatory subunit B', beta isoform NE

General Observations

In addition to investigating the regulation of the water channels Aqp2 and Aqp3, a general goal of our studies is the development of tools, including proteomics databases, that can be useful to the kidney research community. In so doing, we have developed several proteomics and transcriptomic databases profiling genes expressed in the IMCD, thick ascending limb and proximal tubule (https:http://dirweb.nhlbi.nih.gov/Centers/CBPC/LKEM_G/LKEM/Pages/--TranscriptomicandProteomicDatabases.aspx). Our nuclear proteomics data have been added to an upgraded IMCD Proteome Database website (http://dir.nhlbi.nih.gov/papers/lkem/imcd/).

An additional goal has been to develop improved computational approaches to increase the yield of peptides that can be identified from high-quality spectra without a high rate of false positive identifications. In this study, the use of multiple search algorithms [SEQUEST (15), InsPecT (44), and OMSSA (17)] gave substantial numbers of identifications unique to each search algorithm. The largest increment in identifications was found with the InsPecT algorithm, a hybrid approach that uses de novo sequencing of a portion of each peptide to narrow the search space for pattern matching. InsPecT allowed the identification of a subset of peptides often missed by strictly peak-matching software such as SEQUEST or OMSSA. Further steps, such as target-decoy analysis as well as performing ambiguity checks on identified peptides using an in-house algorithm ProMatch, limited the number of false positive identifications and eliminated ambiguous identifications. We emphasize that target-decoy analysis has been demonstrated to be superior in limiting false positive identifications compared with use of the “two-peptide rule,” requiring protein identifications to those established by two or more peptides (18).

Overall, with the new data presented in this study, we have laid the groundwork for future studies of the role of vasopressin and other hormones in transcriptional regulation in renal collecting duct cells. The data provide a guide to proteins present in the nucleus of the “cells of interest” for such studies, which become candidates for genetic manipulation in cell culture and mouse models. The ultimate goal is to identify the relevant transcriptional regulatory networks involved in the control of Aqp2 and Aqp3 gene transcription. By sharing our data in a publicly accessible database, we encourage other investigators to partake in these investigations.

DISCLOSURES

No conflicts of interest are declared by the authors.

Supplementary Material

Table S1
tableS1.pdf (43KB, pdf)
Table S2
tableS2.pdf (40KB, pdf)

Footnotes

1

The online version of this article contains supplemental material.

REFERENCES

  • 1. Albuquerque CP, Smolka MB, Payne SH, Bafna V, Eng J, Zhou H. A multidimensional chromatography technology for in-depth phosphoproteome analysis. Mol Cell Proteomics 7: 1389–1396, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Beausoleil SA, Villen J, Gerber SA, Rush J, Gygi SP. A probability-based approach for high-throughput protein phosphorylation analysis and site localization. Nat Biotechnol 24: 1285–1292, 2006. [DOI] [PubMed] [Google Scholar]
  • 3. Biemesderfer D. Regulated intramembrane proteolysis of megalin: linking urinary protein and gene regulation in proximal tubule? Kidney Int 69: 1717–1721, 2006. [DOI] [PubMed] [Google Scholar]
  • 4. Burrow CR. Retinoids and renal development. Exp Nephrol 8: 219–225, 2000. [DOI] [PubMed] [Google Scholar]
  • 5. Cai Q, McReynolds MR, Keck M, Greer KA, Hoying JB, Brooks HL. Vasopressin receptor subtype 2 activation increases cell proliferation in the renal medulla of AQP1 null mice. Am J Physiol Renal Physiol 293: F1858–F1864, 2007. [DOI] [PubMed] [Google Scholar]
  • 6. Champigneulle A, Siga E, Vassent G, Imbert-Teboul M. V-2-like vasopressin receptor mobilizes intracellular Ca2+ in medullary collecting tubules. Am J Physiol Renal Fluid Electrolyte Physiol 265: F35–F45, 1993. [DOI] [PubMed] [Google Scholar]
  • 7. Chen Q, Xu S, Huang S, Zhang A, Feng Q, Guo X, Guo M, Chen R, Yang T. Suppression subtractive hybridization analysis of gene expression during late kidney development identifies the developmentally regulated gene rPEA3. Nephron Exp Nephrol 111: e103–e115, 2009. [DOI] [PubMed] [Google Scholar]
  • 8. Chou CL, Yip KP, Michea L, Kador K, Ferraris J, Wade JB, Knepper MA. Regulation of aquaporin-2 trafficking by vasopressin in renal collecting duct: Roles of ryanodine-sensitive Ca2+ stores and calmodulin. J Biol Chem 275: 36839–36846, 2000. [DOI] [PubMed] [Google Scholar]
  • 9. Christensen BM, Marples D, Jensen UB, Frokiaer J, Sheikh-Hamad D, Knepper M, Nielsen S. Acute effects of vasopressin V2-receptor antagonist on kidney AQP2 expression and subcellular distribution. Am J Physiol Renal Physiol 275: F285–F297, 1998. [DOI] [PubMed] [Google Scholar]
  • 10. DiGiovanni SR, Nielsen S, Christensen EI, Knepper MA. Regulation of collecting duct water channel expression by vasopressin in Brattleboro rat. Proc Natl Acad Sci USA 91: 8984–8988, 1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Ecelbarger CA, Chou CL, Lolait SJ, Knepper MA, DiGiovanni SR. Evidence for dual signaling pathways for V2 vasopressin receptor in rat inner medullary collecting duct. Am J Physiol Renal Fluid Electrolyte Physiol 270: F623–F633, 1996. [DOI] [PubMed] [Google Scholar]
  • 12. Ecelbarger CA, Nielsen S, Olson BR, Murase T, Baker EA, Knepper MA, Verbalis JG. Role of renal aquaporins in escape from vasopressin-induced antidiuresis in rat. J Clin Invest 99: 1852–1863, 1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Ecelbarger CA, Terris J, Frindt G, Echevarria M, Marples D, Nielsen S, Knepper MA. Aquaporin-3 water channel localization and regulation in rat kidney. Am J Physiol Renal Fluid Electrolyte Physiol 269: F663–F672, 1995. [DOI] [PubMed] [Google Scholar]
  • 14. Elias JE, Gygi SP. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat Methods 4: 207–214, 2007. [DOI] [PubMed] [Google Scholar]
  • 15. Eng JK, McCormack AL, Yates JR., III An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J Am Soc Mass Spectrom 5: 976–989, 1994. [DOI] [PubMed] [Google Scholar]
  • 16. Fujita N, Ishikawa SE, Sasaki S, Fujisawa G, Fushimi K, Marumo F, Saito T. Role of water channel AQP-CD in water retention in SIADH and cirrhotic rats. Am J Physiol Renal Fluid Electrolyte Physiol 269: F926–F931, 1995. [DOI] [PubMed] [Google Scholar]
  • 17. Geer LY, Markey SP, Kowalak JA, Wagner L, Xu M, Maynard DM, Yang X, Shi W, Bryant SH. Open mass spectrometry search algorithm. J Proteome Res 3: 958–964, 2004. [DOI] [PubMed] [Google Scholar]
  • 18. Gupta N, Pevzner PA. False discovery rates of protein identifications: a strike against the two-peptide rule. J Proteome Res 8: 4173–4181, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Hasler U, Jeon US, Kim JA, Mordasini D, Kwon HM, Feraille E, Martin PY. Tonicity-responsive enhancer binding protein is an essential regulator of aquaporin-2 expression in renal collecting duct principal cells. J Am Soc Nephrol 17: 1521–1531, 2006. [DOI] [PubMed] [Google Scholar]
  • 20. Hoffert JD, Pisitkun T, Wang G, Shen RF, Knepper MA. Quantitative phosphoproteomics of vasopressin-sensitive renal cells: regulation of aquaporin-2 phosphorylation at two sites. Proc Natl Acad Sci USA 103: 7159–7164, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Hoffert JD, Wang G, Pisitkun T, Shen RF, Knepper MA. An automated platform for analysis of phosphoproteomic datasets: application to kidney collecting duct phosphoproteins. J Proteome Res 6: 3501–3508, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Hozawa S, Holtzman EJ, Ausiello DA. cAMP motifs regulating transcription in the aquaporin-2 gene. Am J Physiol Cell Physiol 270: C1695–C1702, 1996. [DOI] [PubMed] [Google Scholar]
  • 23. Kao PN, Chen L, Brock G, Ng J, Kenny J, Smith AJ, Corthesy B. Cloning and expression of cyclosporin A- and FK506-sensitive nuclear factor of activated T-cells: NF45 and NF90. J Biol Chem 269: 20691–20699, 1994. [PubMed] [Google Scholar]
  • 24. Kitamura M. Endoplasmic reticulum stress and unfolded protein response in renal pathophysiology: Janus faces. Am J Physiol Renal Physiol 295: F323–F334, 2008. [DOI] [PubMed] [Google Scholar]
  • 25. Kwon TH, Nielsen J, Masilamani S, Hager H, Knepper MA, Frokiaer J, Nielsen S. Regulation of collecting duct AQP3 expression: response to mineralocorticoid. Am J Physiol Renal Physiol 283: F1403–F1421, 2002. [DOI] [PubMed] [Google Scholar]
  • 26. Li SZ, McDill BW, Kovach PA, Ding L, Go WY, Ho SN, Chen F. Calcineurin-NFATc signaling pathway regulates AQP2 expression in response to calcium signals and osmotic stress. Am J Physiol Cell Physiol 292: C1606–C1616, 2007. [DOI] [PubMed] [Google Scholar]
  • 27. Malik TH, Shoichet SA, Latham P, Kroll TG, Peters LL, Shivdasani RA. Transcriptional repression and developmental functions of the atypical vertebrate GATA protein TRPS1. EMBO J 20: 1715–1725, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Matsumura Y, Uchida S, Rai T, Sasaki S, Marumo F. Transcriptional regulation of aquaporin-2 water channel gene by cAMP. J Am Soc Nephrol 8: 861–867, 1997. [DOI] [PubMed] [Google Scholar]
  • 29. Murillo-Carretero MI, Ilundain AA, Echevarria M. Regulation of aquaporin mRNA expression in rat kidney by water intake. J Am Soc Nephrol 10: 696–703, 1999. [DOI] [PubMed] [Google Scholar]
  • 30. Nielsen S, Chou CL, Marples D, Christensen EI, Kishore BK, Knepper MA. Vasopressin increases water permeability of kidney collecting duct by inducing translocation of aquaporin-CD water channels to plasma membrane. Proc Natl Acad Sci USA 92: 1013–1017, 1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Nielsen S, DiGiovanni SR, Christensen EI, Knepper MA, Harris HW. Cellular and subcellular immunolocalization of vasopressin-regulated water channel in rat kidney. Proc Natl Acad Sci USA 90: 11663–11667, 1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Patterson LT, Potter SS. Atlas of Hox gene expression in the developing kidney. Dev Dyn 229: 771–779, 2004. [DOI] [PubMed] [Google Scholar]
  • 33. Pisitkun T, Bieniek J, Tchapyjnikov D, Wang G, Wu WW, Shen RF, Knepper MA. High-throughput identification of IMCD proteins using LC-MS/MS. Physiol Genomics 25: 263–276, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Pisitkun T, Shen RF, Knepper MA. Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci USA 101: 13368–13373, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Pisitkun T, Jacob V, Schleicher SM, Chou CL, Yu MJ, Knepper MA. Akt and ERK1/2 pathways are components of the vasopressin signaling network in rat native IMCD. Am J Physiol Renal Physiol 295: F1030–F1043, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Quandt K, Frech K, Karas H, Wingender E, Werner T. MatInd and MatInspector: new fast and versatile tools for detection of consensus matches in nucleotide sequence data. Nucleic Acids Res 23: 4878–4884, 1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Rai T, Uchida S, Marumo F, Sasaki S. Cloning of rat and mouse aquaporin-2 gene promoters and identification of a negative cis-regulatory element. Am J Physiol Renal Physiol 273: F264–F273, 1997. [DOI] [PubMed] [Google Scholar]
  • 38. Rubera I, Loffing J, Palmer LG, Frindt G, Fowler-Jaeger N, Sauter D, Carroll T, McMahon A, Hummler E, Rossier BC. Collecting duct-specific gene inactivation of alphaENaC in the mouse kidney does not impair sodium and potassium balance. J Clin Invest 112: 554–565, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Ruttenberg BE, Pisitkun T, Knepper MA, Hoffert JD. PhosphoScore: an open-source phosphorylation site assignment tool for MSn data. J Proteome Res 7: 3054–3059, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Saito T, Ishikawa SE, Sasaki S, Fujita N, Fushimi K, Okada K, Takeuchi K, Sakamoto A, Ookawara S, Kaneko T, Marumo F, Saito T. Alteration in water channel AQP-2 by removal of AVP stimulation in collecting duct cells of dehydrated rats. Am J Physiol Renal Physiol 272: F183–F191, 1997. [DOI] [PubMed] [Google Scholar]
  • 41. Shaywitz AJ, Greenberg ME. CREB: a stimulus-induced transcription factor activated by a diverse array of extracellular signals. Annu Rev Biochem 68: 821–861, 1999. [DOI] [PubMed] [Google Scholar]
  • 42. Srinivas S, Goldberg MR, Watanabe T, D'Agati V, al-Awqati Q, Costantini F. Expression of green fluorescent protein in the ureteric bud of transgenic mice: a new tool for the analysis of ureteric bud morphogenesis. Dev Genet 24: 241–251, 1999. [DOI] [PubMed] [Google Scholar]
  • 43. Star RA, Nonoguchi H, Balaban R, Knepper MA. Calcium and cyclic adenosine monophosphate as second messengers for vasopressin in the rat inner medullary collecting duct. J Clin Invest 81: 1879–1888, 1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Tanner S, Shu H, Frank A, Wang LC, Zandi E, Mumby M, Pevzner PA, Bafna V. InsPecT: identification of posttranslationally modified peptides from tandem mass spectra. Anal Chem 77: 4626–4639, 2005. [DOI] [PubMed] [Google Scholar]
  • 45. Terris J, Ecelbarger CA, Nielsen S, Knepper MA. Long-term regulation of four renal aquaporins in rat. Am J Physiol Renal Fluid Electrolyte Physiol 271: F414–F422, 1996. [DOI] [PubMed] [Google Scholar]
  • 46. Uawithya P, Pisitkun T, Ruttenberg BE, Knepper MA. Transcriptional profiling of native inner medullary collecting duct cells from rat kidney. Physiol Genomics 32: 229–253, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Uchida S, Matsumura Y, Rai T, Sasaki S, Marumo F. Regulation of aquaporin-2 gene transcription by GATA-3. Biochem Biophys Res Commun 232: 65–68, 1997. [DOI] [PubMed] [Google Scholar]
  • 48. Uchida S, Sasaki S, Fushimi K, Marumo F. Isolation of human Aquaporin-CD gene. J Biol Chem 269: 23451–23455, 1994. [PubMed] [Google Scholar]
  • 49. Uchida S, Tanaka Y, Ito H, Saitoh-Ohara F, Inazawa J, Yokoyama KK, Sasaki S, Marumo F. Transcriptional regulation of the CLC-K1 promoter by myc-associated zinc finger protein and kidney-enriched Kruppel-like factor, a novel zinc finger repressor. Mol Cell Biol 20: 7319–7331, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Yamaguchi Y, Yonemura S, Takada S. Grainyhead-related transcription factor is required for duct maturation in the salivary gland and the kidney of the mouse. Development 133: 4737–4748, 2006. [DOI] [PubMed] [Google Scholar]
  • 51. Yasui M, Marples D, Belusa R, Eklof AC, Celsi G, Nielsen S, Aperia A. Development of urinary concentrating capacity: role of aquaporin-2. Am J Physiol Renal Fluid Electrolyte Physiol 271: F461–F468, 1996. [DOI] [PubMed] [Google Scholar]
  • 52. Yasui M, Zelenin SM, Celsi G, Aperia A. Adenylate cyclase-coupled vasopressin receptor activates AQP2 promoter via a dual effect on CRE and AP1 elements. Am J Physiol Renal Physiol 272: F443–F450, 1997. [DOI] [PubMed] [Google Scholar]
  • 53. Yip KP. Coupling of vasopressin-induced intracellular Ca2+ mobilization and apical exocytosis in perfused rat kidney collecting duct. J Physiol 538: 891–899, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Yu MJ, Miller RL, Uawithya P, Rinschen MM, Khositseth S, Braucht DW, Chou CL, Pisitkun T, Nelson RD, Knepper MA. Systems-level analysis of cell-specific AQP2 gene expression in renal collecting duct. Proc Natl Acad Sci USA 106: 2441–2446, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Table S1
tableS1.pdf (43KB, pdf)
Table S2
tableS2.pdf (40KB, pdf)

Articles from Physiological Genomics are provided here courtesy of American Physiological Society

RESOURCES