1
|
Chou CL, Jayatissa NU, Kichula ET, Ou SM, Limbutara K, Knepper MA. Phosphoproteomic response to epidermal growth factor in native rat inner medullary collecting duct. Am J Physiol Renal Physiol 2025; 328:F29-F47. [PMID: 39508840 PMCID: PMC11918369 DOI: 10.1152/ajprenal.00182.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/25/2024] [Accepted: 11/01/2024] [Indexed: 12/18/2024] Open
Abstract
Epidermal growth factor (EGF) has important effects in the renal collecting duct to regulate salt and water transport. To identify elements of EGF-mediated signaling in the rat renal inner medullary collecting duct (IMCD), we carried out phosphoproteomic analysis. Biochemically isolated rat IMCD suspensions were treated with 1 µM of EGF or vehicle for 30 min. We performed comprehensive quantitative phosphoproteomics using tandem mass tag (TMT)-labeling of tryptic peptides followed by protein mass spectrometry. We present a data resource reporting all detected phosphorylation sites and their changes in response to EGF. For a total of 29,881 unique phosphorylation sites, 135 sites were increased and 119 sites were decreased based on stringent statistical analysis. The data are provided to users at https://esbl.nhlbi.nih.gov/Databases/EGF-phospho/. The analysis demonstrated that EGF signals through canonical EGF pathways in the renal IMCD. Analysis of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways in which EGF-regulated phosphoproteins are over-represented in native rat IMCD cells confirmed mapping to RAF-MEK-extracellular signal-regulated kinase (ERK) signaling but also pointed to a role for EGF in the regulation of protein translation. A large number of phosphoproteins regulated by EGF contained PDZ domains that are key elements of epithelial polarity determination. We also provide a collecting duct EGF-network map as a user-accessible web resource at https://esbl.nhlbi.nih.gov/Databases/EGF-network/. Overall, the phosphoproteomic data presented provide a useful resource for experimental design and modeling of signaling in the renal collecting duct.NEW & NOTEWORTHY EGF negatively regulates transepithelial water and salt transport across the kidney collecting duct. This study identified phosphoproteins affected by EGF stimulation in normal rat collecting ducts, providing insights into global cell signaling mechanisms. Bioinformatic analyses highlighted enhanced canonical ERK signaling alongside a diminished activity in the PI3K-Akt pathway, which is crucial for cell proliferation and survival. This EGF response differs somewhat from prior studies where both pathways were prominently activated.
Collapse
Affiliation(s)
- Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Nipun U Jayatissa
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Elena T Kichula
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Shuo-Ming Ou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Kavee Limbutara
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
2
|
Ando F, Hara Y, Uchida S. Identification of protein kinase A signalling molecules in renal collecting ducts. J Physiol 2024; 602:3057-3067. [PMID: 37013848 DOI: 10.1113/jp284178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Body water homeostasis is maintained by the correct balance between water intake and water loss through urine, faeces, sweat and breath. It is known that elevated circulating levels of the antidiuretic hormone vasopressin decrease urine volume to prevent excessive water loss from the body. Vasopressin/cAMP/protein kinase A (PKA) signalling is the canonical pathway in renal collecting ducts for phosphorylating aquaporin-2 (AQP2) water channels, which leads to the reabsorption of water from urine via AQP2. Although recent omics data have verified various downstream targets of PKA, crucial regulators that mediate PKA-induced AQP2 phosphorylation remain unknown, mainly because vasopressin is usually used to activate PKA as a positive control. Vasopressin is extremely potent and phosphorylates various PKA substrates non-specifically, making it difficult to narrow down the candidate mediators responsible for AQP2 phosphorylation. The intracellular localization of PKA is tightly regulated by its scaffold proteins, also known as A-kinase anchoring proteins (AKAPs). Furthermore, each AKAP has a target domain that determines its intracellular localization, enabling the creation of a local PKA signalling network. Although vasopressin activates most PKAs independently of their intracellular localization, some chemical compounds preferentially act on PKAs localized on AQP2-containing vesicles while simultaneously phosphorylating AQP2 and its surrounding PKA substrates. Immunoprecipitation with antibodies against phosphorylated PKA substrates followed by mass spectrometry analysis revealed that the PKA substrate in proximity to AQP2 was lipopolysaccharide-responsive and beige-like anchor (LRBA). Furthermore, Lrba knockout studies revealed that LRBA was required for vasopressin-induced AQP2 phosphorylation.
Collapse
Affiliation(s)
- Fumiaki Ando
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yu Hara
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shinichi Uchida
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
3
|
Park E, Yang CR, Raghuram V, Chen L, Chou CL, Knepper MA. Using CRISPR-Cas9/phosphoproteomics to identify substrates of calcium/calmodulin-dependent kinase 2δ. J Biol Chem 2023; 299:105371. [PMID: 37865316 PMCID: PMC10783575 DOI: 10.1016/j.jbc.2023.105371] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/30/2023] [Accepted: 10/10/2023] [Indexed: 10/23/2023] Open
Abstract
Ca2+/Calmodulin-dependent protein kinase 2 (CAMK2) family proteins are involved in the regulation of cellular processes in a variety of tissues including brain, heart, liver, and kidney. One member, CAMK2δ (CAMK2D), has been proposed to be involved in vasopressin signaling in the renal collecting duct, which controls water excretion through regulation of the water channel aquaporin-2 (AQP2). To identify CAMK2D target proteins in renal collecting duct cells (mpkCCD), we deleted Camk2d and carried out LC-MS/MS-based quantitative phosphoproteomics. Specifically, we used CRISPR/Cas9 with two different guide RNAs targeting the CAMK2D catalytic domain to create multiple CAMK2D KO cell lines. AQP2 protein abundance was lower in the CAMK2D KO cells than in CAMK2D-intact controls. AQP2 phosphorylation at Ser256 and Ser269 (normalized for total AQP2) was decreased. However, trafficking of AQP2 to and from the apical plasma membrane was sustained. Large-scale quantitative phosphoproteomic analysis (TMT-labeling) in the presence of the vasopressin analog dDAVP (0.1 nM, 30 min) allowed quantification of 11,570 phosphosites of which 169 were significantly decreased, while 206 were increased in abundance in CAMK2D KO clones. These data are available for browsing or download at https://esbl.nhlbi.nih.gov/Databases/CAMK2D-proteome/. Motif analysis of the decreased phosphorylation sites revealed a target preference of -(R/K)-X-X-p(S/T)-X-(D/E), matching the motif identified in previous in vitro phosphorylation studies using recombinant CAMK2D. Thirty five of the significantly downregulated phosphorylation sites in CAMK2D KO cells had exactly this motif and are judged to be likely direct CAMK2D targets. This adds to the list of known CAMK2D target proteins found in prior reductionist studies.
Collapse
Affiliation(s)
- Euijung Park
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Chin-Rang Yang
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA.
| |
Collapse
|
4
|
Huynh NV, Rehage C, Hyndman KA. Mild dehydration effects on the murine kidney single-nucleus transcriptome and chromatin accessibility. Am J Physiol Renal Physiol 2023; 325:F717-F732. [PMID: 37767569 PMCID: PMC11550884 DOI: 10.1152/ajprenal.00161.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Daily, we may experience mild dehydration with a rise in plasma osmolality that triggers the release of vasopressin. Although the effect of dehydration is well characterized in collecting duct principal cells (CDPCs), we hypothesized that mild dehydration (<12 h) results in many kidney cell-specific changes in transcriptomes and chromatin accessibility. Single-nucleus (sn) multiome (RNA-assay for transposase-accessible chromatin) sequencing and bulk RNA sequencing of kidneys from male and female mice that were mildly water deprived or not were compared. Water-deprived mice had a significant increase in plasma osmolality. sn-multiome-seq resulted in 19,837 nuclei that were annotated into 33 clusters. In CDPCs, aquaporin 2 (Aqp2) and aquaporin 3 (Apq3) were greater in dehydrated mice, but there were novel genes like gremlin 2 (Grem2; a cytokine) that were increased compared with ad libitum mice. The transcription factor cAMP-responsive element modulator (Crem) was greater in CDPCs of dehydrated mice, and the Crem DNA motif was more accessible. There were hundreds of sex- and dehydration-specific differentially expressed genes (DEGs) throughout the kidney, especially in the proximal tubules and thin limbs. In male mice, DEGs were enriched in pathways related to lipid metabolism, whereas female DEGs were enriched in organic acid metabolism. Many highly expressed genes had a positive correlation with increased chromatin accessibility, and mild dehydration exerted many transcriptional changes that we detected at the chromatin level. Even with a rise in plasma osmolality, male and female kidneys have distinct transcriptomes suggesting that there may be diverse mechanisms used to remain in fluid balance.NEW & NOTEWORTHY The kidney consists of >30 cell types that work collectively to maintain fluid-electrolyte balance. Kidney single-nucleus transcriptomes and chromatin accessibility profiles from male and female control (ad libitum water and food) or mildly dehydrated mice (ad libitum food, water deprivation) were determined. Mild dehydration caused hundreds of cell- and sex-specific transcriptomic changes, even though the kidney function to conserve water was the same.
Collapse
Affiliation(s)
- Nha Van Huynh
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Cassidy Rehage
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kelly A Hyndman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
5
|
Yanagawa H, Hara Y, Ando F, Suzuki S, Fujiki T, Oikawa D, Yui N, Mandai S, Mori Y, Susa K, Mori T, Sohara E, Tokunaga F, Uchida S. LRBA signalosomes activate vasopressin-induced AQP2 trafficking at recycling endosomes. J Physiol 2023; 601:5437-5451. [PMID: 37860942 DOI: 10.1113/jp285188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/04/2023] [Indexed: 10/21/2023] Open
Abstract
Aquaporin-2 (AQP2) water channels are proteins that are recycled between intracellular vesicles and the apical plasma membrane in renal collecting ducts. Lipopolysaccharide-responsive beige-like anchor protein (LRBA) is a protein kinase A (PKA) anchoring protein that creates compartmentalized PKA signalling responsible for AQP2 phosphorylation. In response to increased plasma osmolality, vasopressin/cyclic adenosine monophosphate (cAMP)/PKA signalling phosphorylates AQP2, promoting AQP2 trafficking into the apical plasma membrane and increasing water reabsorption from urine. However, the molecular mechanisms by which LRBA mediates vasopressin-induced AQP2 phosphorylation remain unknown. To investigate AQP2 intracellular localization and phosphorylation status in vivo, a density gradient ultracentrifugation technique was combined with an in situ proximity ligation assay, super-resolution structured illumination microscopy and immunoelectron microscopy. Most of the AQP2 was localized on the recycling endosome in the presence of tolvaptan, a vasopressin type 2 receptor (V2R) antagonist. Desmopressin, a V2R agonist, phosphorylated AQP2, translocating it from the recycling endosome to the apical plasma membrane. In contrast, LRBA was constitutively localized at the recycling endosome. Therefore, LRBA and AQP2 were well colocalized in the absence of vasopressin stimulation. The loss of LRBA/PKA signalling by Lrba knockout impaired vasopressin-induced AQP2 phosphorylation, resulting in AQP2 retention at the recycling endosome. Defective AQP2 trafficking caused low urinary concentrating ability in Lrba-/- mice. The LRBA-PKA complex created compartmentalized PKA signalling at the recycling endosome, which facilitated AQP2 phosphorylation in response to vasopressin. KEY POINTS: Membrane proteins are continuously internalized into the endosomal system via endocytosis, after which they are either recycled back to the plasma membrane or degraded at the lysosome. In T cells, lipopolysaccharide-responsive beige-like anchor protein (LRBA) binds directly to the cytotoxic T lymphocyte antigen 4 (CTLA-4), a checkpoint immune molecule, to prevent CTLA-4 lysosomal degradation and promote its vesicle recycling. LRBA has different physiological functions in renal collecting ducts. LRBA and aquaporin-2 (AQP2) water channels were colocalized on the recycling endosome in vivo in the absence of the anti-diuretic hormone vasopressin. LRBA promoted vasopressin-induced AQP2 trafficking, increasing water reabsorption from urine via AQP2. LRBA determined renal responsiveness to vasopressin at recycling endosomes. LRBA is a ubiquitously expressed anchor protein. LRBA signalosomes might regulate membrane trafficking of several constitutively recycled proteins at recycling endosomes.
Collapse
Affiliation(s)
- Hideki Yanagawa
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Yu Hara
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Fumiaki Ando
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Soichiro Suzuki
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Tamami Fujiki
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Daisuke Oikawa
- Department of Medical Biochemistry, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Naofumi Yui
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Shintaro Mandai
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Yutaro Mori
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Koichiro Susa
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Takayasu Mori
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Eisei Sohara
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| | - Fuminori Tokunaga
- Department of Medical Biochemistry, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Shinichi Uchida
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Bunkyo, Tokyo, Japan
| |
Collapse
|
6
|
Zhou X, Torres VE. Emerging therapies for autosomal dominant polycystic kidney disease with a focus on cAMP signaling. Front Mol Biosci 2022; 9:981963. [PMID: 36120538 PMCID: PMC9478168 DOI: 10.3389/fmolb.2022.981963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), with an estimated genetic prevalence between 1:400 and 1:1,000 individuals, is the third most common cause of end stage kidney disease after diabetes mellitus and hypertension. Over the last 3 decades there has been great progress in understanding its pathogenesis. This allows the stratification of therapeutic targets into four levels, gene mutation and polycystin disruption, proximal mechanisms directly caused by disruption of polycystin function, downstream regulatory and signaling pathways, and non-specific pathophysiologic processes shared by many other diseases. Dysfunction of the polycystins, encoded by the PKD genes, is closely associated with disruption of calcium and upregulation of cyclic AMP and protein kinase A (PKA) signaling, affecting most downstream regulatory, signaling, and pathophysiologic pathways altered in this disease. Interventions acting on G protein coupled receptors to inhibit of 3',5'-cyclic adenosine monophosphate (cAMP) production have been effective in preclinical trials and have led to the first approved treatment for ADPKD. However, completely blocking cAMP mediated PKA activation is not feasible and PKA activation independently from cAMP can also occur in ADPKD. Therefore, targeting the cAMP/PKA/CREB pathway beyond cAMP production makes sense. Redundancy of mechanisms, numerous positive and negative feedback loops, and possibly counteracting effects may limit the effectiveness of targeting downstream pathways. Nevertheless, interventions targeting important regulatory, signaling and pathophysiologic pathways downstream from cAMP/PKA activation may provide additive or synergistic value and build on a strategy that has already had success. The purpose of this manuscript is to review the role of cAMP and PKA signaling and their multiple downstream pathways as potential targets for emergent therapies for ADPKD.
Collapse
Affiliation(s)
- Xia Zhou
- Mayo Clinic, Department of Nephrology, Rochester, MN, United States
| | | |
Collapse
|
7
|
Leo KT, Chou CL, Yang CR, Park E, Raghuram V, Knepper MA. Bayesian analysis of dynamic phosphoproteomic data identifies protein kinases mediating GPCR responses. Cell Commun Signal 2022; 20:80. [PMID: 35659261 PMCID: PMC9164474 DOI: 10.1186/s12964-022-00892-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/28/2022] [Indexed: 11/18/2022] Open
Abstract
Background A major goal in the discovery of cellular signaling networks is to identify regulated phosphorylation sites (“phosphosites”) and map them to the responsible protein kinases. The V2 vasopressin receptor is a G-protein coupled receptor (GPCR) that is responsible for regulation of renal water excretion through control of aquaporin-2-mediated osmotic water transport in kidney collecting duct cells. Genome editing experiments have demonstrated that virtually all vasopressin-triggered phosphorylation changes are dependent on protein kinase A (PKA), but events downstream from PKA are still obscure. Methods Here, we used: 1) Tandem mass tag-based quantitative phosphoproteomics to experimentally track phosphorylation changes over time in native collecting ducts isolated from rat kidneys; 2) a clustering algorithm to classify time course data based on abundance changes and the amino acid sequences surrounding the phosphosites; and 3) Bayes’ Theorem to integrate the dynamic phosphorylation data with multiple prior “omic” data sets covering expression, subcellular location, known kinase activity, and characteristic surrounding sequences to identify a set of protein kinases that are regulated secondary to PKA activation. Results Phosphoproteomic studies revealed 185 phosphosites regulated by vasopressin over 15 min. The resulting groups from the cluster algorithm were integrated with Bayes’ Theorem to produce corresponding ranked lists of kinases likely responsible for each group. The top kinases establish three PKA-dependent protein kinase modules whose regulation mediate the physiological effects of vasopressin at a cellular level. The three modules are 1) a pathway involving several Rho/Rac/Cdc42-dependent protein kinases that control actin cytoskeleton dynamics; 2) mitogen-activated protein kinase and cyclin-dependent kinase pathways that control cell proliferation; and 3) calcium/calmodulin-dependent signaling. Conclusions Our findings identify a novel set of downstream small GTPase effectors and calcium/calmodulin-dependent kinases with potential roles in the regulation of water permeability through actin cytoskeleton rearrangement and aquaporin-2 trafficking. The proposed signaling network provides a stronger hypothesis for the kinases mediating V2 vasopressin receptor responses, encouraging future targeted examination via reductionist approaches. Furthermore, the Bayesian analysis described here provides a template for investigating signaling via other biological systems and GPCRs. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00892-6.
Collapse
|
8
|
Ostrom KF, LaVigne JE, Brust TF, Seifert R, Dessauer CW, Watts VJ, Ostrom RS. Physiological roles of mammalian transmembrane adenylyl cyclase isoforms. Physiol Rev 2022; 102:815-857. [PMID: 34698552 PMCID: PMC8759965 DOI: 10.1152/physrev.00013.2021] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/20/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Adenylyl cyclases (ACs) catalyze the conversion of ATP to the ubiquitous second messenger cAMP. Mammals possess nine isoforms of transmembrane ACs, dubbed AC1-9, that serve as major effector enzymes of G protein-coupled receptors (GPCRs). The transmembrane ACs display varying expression patterns across tissues, giving the potential for them to have a wide array of physiological roles. Cells express multiple AC isoforms, implying that ACs have redundant functions. Furthermore, all transmembrane ACs are activated by Gαs, so it was long assumed that all ACs are activated by Gαs-coupled GPCRs. AC isoforms partition to different microdomains of the plasma membrane and form prearranged signaling complexes with specific GPCRs that contribute to cAMP signaling compartments. This compartmentation allows for a diversity of cellular and physiological responses by enabling unique signaling events to be triggered by different pools of cAMP. Isoform-specific pharmacological activators or inhibitors are lacking for most ACs, making knockdown and overexpression the primary tools for examining the physiological roles of a given isoform. Much progress has been made in understanding the physiological effects mediated through individual transmembrane ACs. GPCR-AC-cAMP signaling pathways play significant roles in regulating functions of every cell and tissue, so understanding each AC isoform's role holds potential for uncovering new approaches for treating a vast array of pathophysiological conditions.
Collapse
Affiliation(s)
| | - Justin E LaVigne
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Tarsis F Brust
- Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida
| | - Roland Seifert
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California
| |
Collapse
|
9
|
Mehta YR, Lewis SA, Leo KT, Chen L, Park E, Raghuram V, Chou CL, Yang CR, Kikuchi H, Khundmiri S, Poll BG, Knepper MA. "ADPKD-omics": determinants of cyclic AMP levels in renal epithelial cells. Kidney Int 2022; 101:47-62. [PMID: 34757121 PMCID: PMC10671900 DOI: 10.1016/j.kint.2021.10.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/30/2021] [Accepted: 10/12/2021] [Indexed: 12/30/2022]
Abstract
The regulation of cyclic adenosine monophosphate (cAMP) levels in kidney epithelial cells is important in at least 2 groups of disorders, namely water balance disorders and autosomal dominant polycystic kidney disease. Focusing on the latter, we review genes that code for proteins that are determinants of cAMP levels in cells. We identify which of these determinants are expressed in the 14 kidney tubule segments using recently published RNA-sequencing and protein mass spectrometry data ("autosomal dominant polycystic kidney disease-omics"). This includes G protein-coupled receptors, adenylyl cyclases, cyclic nucleotide phosphodiesterases, cAMP transporters, cAMP-binding proteins, regulator of G protein-signaling proteins, G protein-coupled receptor kinases, arrestins, calcium transporters, and calcium-binding proteins. In addition, compartmentalized cAMP signaling in the primary cilium is discussed, and a specialized database of the proteome of the primary cilium of cultured "IMCD3" cells is provided as an online resource (https://esbl.nhlbi.nih.gov/Databases/CiliumProteome/). Overall, this article provides a general resource in the form of a curated list of proteins likely to play roles in determination of cAMP levels in kidney epithelial cells and, therefore, likely to be determinants of progression of autosomal dominant polycystic kidney disease.
Collapse
Affiliation(s)
- Yash R Mehta
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Spencer A Lewis
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kirby T Leo
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Euijung Park
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chin-Rang Yang
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hiroaki Kikuchi
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Syed Khundmiri
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian G Poll
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
10
|
Cid-Jofré V, Moreno M, Reyes-Parada M, Renard GM. Role of Oxytocin and Vasopressin in Neuropsychiatric Disorders: Therapeutic Potential of Agonists and Antagonists. Int J Mol Sci 2021; 22:ijms222112077. [PMID: 34769501 PMCID: PMC8584779 DOI: 10.3390/ijms222112077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/20/2021] [Accepted: 11/02/2021] [Indexed: 12/27/2022] Open
Abstract
Oxytocin (OT) and vasopressin (AVP) are hypothalamic neuropeptides classically associated with their regulatory role in reproduction, water homeostasis, and social behaviors. Interestingly, this role has expanded in recent years and has positioned these neuropeptides as therapeutic targets for various neuropsychiatric diseases such as autism, addiction, schizophrenia, depression, and anxiety disorders. Due to the chemical-physical characteristics of these neuropeptides including short half-life, poor blood-brain barrier penetration, promiscuity for AVP and OT receptors (AVP-R, OT-R), novel ligands have been developed in recent decades. This review summarizes the role of OT and AVP in neuropsychiatric conditions, as well as the findings of different OT-R and AVP-R agonists and antagonists, used both at the preclinical and clinical level. Furthermore, we discuss their possible therapeutic potential for central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Valeska Cid-Jofré
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
| | - Macarena Moreno
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
- Facultad de Ciencias Sociales, Escuela de Psicología, Universidad Bernardo OHiggins, Santiago 8370993, Chile
| | - Miguel Reyes-Parada
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Providencia 7500912, Chile
- Correspondence: (M.R.-P.); (G.M.R.)
| | - Georgina M. Renard
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
- Correspondence: (M.R.-P.); (G.M.R.)
| |
Collapse
|
11
|
Soares AG, Contreras J, Archer CR, Mironova E, Berdeaux R, Stockand JD, Abd El-Aziz TM. Stimulation of the Epithelial Na + Channel in Renal Principal Cells by Gs-Coupled Designer Receptors Exclusively Activated by Designer Drugs. Front Physiol 2021; 12:725782. [PMID: 34512393 PMCID: PMC8425396 DOI: 10.3389/fphys.2021.725782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/28/2021] [Indexed: 11/29/2022] Open
Abstract
The activity of the Epithelial Na+ Channel (ENaC) in renal principal cells (PC) fine-tunes sodium excretion and consequently, affects blood pressure. The Gs-adenylyl cyclase-cAMP signal transduction pathway is believed to play a central role in the normal control of ENaC activity in PCs. The current study quantifies the importance of this signaling pathway to the regulation of ENaC activity in vivo using a knock-in mouse that has conditional expression of Gs-DREADD (designer receptors exclusively activated by designer drugs; GsD) in renal PCs. The GsD mouse also contains a cAMP response element-luciferase reporter transgene for non-invasive bioluminescence monitoring of cAMP signaling. Clozapine N-oxide (CNO) was used to selectively and temporally stimulate GsD. Treatment with CNO significantly increased luciferase bioluminescence in the kidneys of PC-specific GsD but not control mice. CNO also significantly increased the activity of ENaC in principal cells in PC-specific GsD mice compared to untreated knock-in mice and CNO treated littermate controls. The cell permeable cAMP analog, 8-(4-chlorophenylthio)adenosine 3′,5′-cyclic monophosphate, significantly increased the activity and expression in the plasma membrane of recombinant ENaC expressed in CHO and COS-7 cells, respectively. Treatment of PC-specific GsD mice with CNO rapidly and significantly decreased urinary Na+ excretion compared to untreated PC-specific GsD mice and treated littermate controls. This decrease in Na+ excretion in response to CNO in PC-specific GsD mice was similar in magnitude and timing as that induced by the selective vasopressin receptor 2 agonist, desmopressin, in wild type mice. These findings demonstrate for the first time that targeted activation of Gs signaling exclusively in PCs is sufficient to increase ENaC activity and decrease dependent urinary Na+ excretion in live animals.
Collapse
Affiliation(s)
- Antonio G Soares
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Jorge Contreras
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Crystal R Archer
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Elena Mironova
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - James D Stockand
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Tarek Mohamed Abd El-Aziz
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Zoology Department, Faculty of Science, Minia University, Minya, Egypt
| |
Collapse
|
12
|
Salhadar K, Matthews A, Raghuram V, Limbutara K, Yang CR, Datta A, Chou CL, Knepper MA. Phosphoproteomic Identification of Vasopressin/cAMP/Protein Kinase A-Dependent Signaling in Kidney. Mol Pharmacol 2021; 99:358-369. [PMID: 32245905 PMCID: PMC8058505 DOI: 10.1124/mol.120.119602] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/24/2020] [Indexed: 12/15/2022] Open
Abstract
Water excretion by the kidney is regulated by the neurohypophyseal peptide hormone vasopressin through actions in renal collecting duct cells to regulate the water channel protein aquaporin-2. Vasopressin signaling is initiated by binding to a G-protein-coupled receptor called V2R, which signals through heterotrimeric G-protein subunit Gs α, adenylyl cyclase 6, and activation of the cAMP-regulated protein kinase (PKA). Signaling events coupling PKA activation and aquaporin-2 regulation were largely unknown until the advent of modern protein mass spectrometry techniques that allow proteome-wide quantification of protein phosphorylation changes (phosphoproteomics). This short review documents phosphoproteomic findings in collecting duct cells describing the response to V2R-selective vasopressin agonists and antagonists, the response to CRISPR-mediated deletion of PKA, results from in vitro phosphorylation studies using recombinant PKA, the response to the broad-spectrum kinase inhibitor H89 (N-[2-p-bromocinnamylamino-ethyl]-5-isoquinolinesulphonamide), and the responses underlying lithium-induced nephrogenic diabetes insipidus. These phosphoproteomic data sets have been made available online for modeling vasopressin signaling and signaling downstream from other G-protein-coupled receptors. SIGNIFICANCE STATEMENT: New developments in protein mass spectrometry are facilitating progress in identification of signaling networks. Using mass spectrometry, it is now possible to identify and quantify thousands of phosphorylation sites in a given cell type (phosphoproteomics). The authors describe the use of phosphoproteomics technology to identify signaling mechanisms downstream from a G-protein-coupled receptor, the vasopressin V2 subtype receptor, and its role of the regulation and dysregulation of water excretion in the kidney. Data from multiple phosphoproteomic data sets are provided as web-based resources.
Collapse
Affiliation(s)
- Karim Salhadar
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Allanah Matthews
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Kavee Limbutara
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Chin-Rang Yang
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Arnab Datta
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland
| |
Collapse
|
13
|
Raghuram V, Salhadar K, Limbutara K, Park E, Yang CR, Knepper MA. Protein kinase A catalytic-α and catalytic-β proteins have nonredundant regulatory functions. Am J Physiol Renal Physiol 2020; 319:F848-F862. [PMID: 33017189 PMCID: PMC7789987 DOI: 10.1152/ajprenal.00383.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/28/2020] [Accepted: 09/25/2020] [Indexed: 01/08/2023] Open
Abstract
Vasopressin regulates osmotic water transport in the renal collecting duct by protein kinase A (PKA)-mediated control of the water channel aquaporin-2 (AQP2). Collecting duct principal cells express two seemingly redundant PKA catalytic subunits, PKA catalytic α (PKA-Cα) and PKA catalytic β (PKA-Cβ). To identify the roles of these two protein kinases, we carried out deep phosphoproteomic analysis in cultured mpkCCD cells in which either PKA-Cα or PKA-Cβ was deleted using CRISPR-Cas9-based genome editing. Controls were cells carried through the genome editing procedure but without deletion of PKA. TMT mass tagging was used for protein mass spectrometric quantification. Of the 4,635 phosphopeptides that were quantified, 67 phosphopeptides were significantly altered in abundance with PKA-Cα deletion, whereas 21 phosphopeptides were significantly altered in abundance with PKA-Cβ deletion. However, only four sites were changed in both. The target proteins identified in PKA-Cα-null cells were largely associated with cell membranes and membrane vesicles, whereas target proteins in PKA-Cβ-null cells were largely associated with the actin cytoskeleton and cell junctions. In contrast, in vitro incubation of mpkCCD proteins with recombinant PKA-Cα and PKA-Cβ resulted in virtually identical phosphorylation changes. In addition, analysis of total protein abundances in in vivo samples showed that PKA-Cα deletion resulted in a near disappearance of AQP2 protein, whereas PKA-Cβ deletion did not decrease AQP2 abundance. We conclude that PKA-Cα and PKA-Cβ serve substantially different regulatory functions in renal collecting duct cells and that differences in phosphorylation targets may be due to differences in protein interactions, e.g., mediated by A-kinase anchor proteins, C-kinase anchoring proteins, or PDZ binding.
Collapse
Affiliation(s)
- Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Karim Salhadar
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Kavee Limbutara
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Euijung Park
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Chin-Rang Yang
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
14
|
Riffault M, Johnson GP, Owen MM, Javaheri B, Pitsillides AA, Hoey DA. Loss of Adenylyl Cyclase 6 in Leptin Receptor-Expressing Stromal Cells Attenuates Loading-Induced Endosteal Bone Formation. JBMR Plus 2020; 4:e10408. [PMID: 33210061 PMCID: PMC7657397 DOI: 10.1002/jbm4.10408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023] Open
Abstract
Bone marrow stromal/stem cells represent a quiescent cell population that replenish the osteoblast bone‐forming cell pool with age and in response to injury, maintaining bone mass and repair. A potent mediator of stromal/stem cell differentiation in vitro and bone formation in vivo is physical loading, yet it still remains unclear whether loading‐induced bone formation requires the osteogenic differentiation of these resident stromal/stem cells. Therefore, in this study, we utilized the leptin receptor (LepR) to identify and trace the contribution of bone marrow stromal cells to mechanoadaptation of bone in vivo. Twelve‐week‐old Lepr‐cre;tdTomato mice were subjected to compressive tibia loading with an 11 N peak load for 40 cycles, every other day for 2 weeks. Histological analysis revealed that Lepr‐cre;tdTomato+ cells arise perinatally around blood vessels and populate bone surfaces as lining cells or osteoblasts before a percentage undergo osteocytogenesis. Lepr‐cre;tdTomato+ stromal cells within the marrow increase in abundance with age, but not following the application of tibial compressive loading. Mechanical loading induces an increase in bone mass and bone formation parameters, yet does not evoke an increase in Lepr‐cre;tdTomato+ osteoblasts or osteocytes. To investigate whether adenylyl cyclase‐6 (AC6) in LepR cells contributes to this mechanoadaptive response, Lepr‐cre;tdTomato mice were further crossed with AC6fl/fl mice to generate a LepR+ cell‐specific knockout of AC6. These Lepr‐cre;tdTomato;AC6fl/fl animals have an attenuated response to compressive tibia loading, characterized by a deficient load‐induced osteogenic response on the endosteal bone surface. This, therefore, shows that Lepr‐cre;tdTomato+ cells contribute to short‐term bone mechanoadaptation. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mathieu Riffault
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin Ireland.,Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering, Trinity College Dublin Dublin Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER) Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Gillian P Johnson
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin Ireland.,Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering, Trinity College Dublin Dublin Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER) Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland.,Department of Mechanical, Aeronautical and Biomedical Engineering University of Limerick Limerick Ireland
| | - Madeline M Owen
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin Ireland.,Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering, Trinity College Dublin Dublin Ireland
| | - Behzad Javaheri
- Skeletal Biology Group, Comparative Biomedical Sciences The Royal Veterinary College London United Kingdom
| | - Andrew A Pitsillides
- Skeletal Biology Group, Comparative Biomedical Sciences The Royal Veterinary College London United Kingdom
| | - David A Hoey
- Trinity Centre for Biomedical Engineering Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin Ireland.,Department of Mechanical, Manufacturing, and Biomedical Engineering School of Engineering, Trinity College Dublin Dublin Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER) Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland.,Department of Mechanical, Aeronautical and Biomedical Engineering University of Limerick Limerick Ireland
| |
Collapse
|
15
|
Fenton RA, Murali SK, Kaji I, Akiba Y, Kaunitz JD, Kristensen TB, Poulsen SB, Dominguez Rieg JA, Rieg T. Adenylyl Cyclase 6 Expression Is Essential for Cholera Toxin-Induced Diarrhea. J Infect Dis 2020; 220:1719-1728. [PMID: 30624615 DOI: 10.1093/infdis/jiz013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/07/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cholera toxin (CT)-induced diarrhea is mediated by cyclic adenosine monophosphate (cAMP)-mediated active Cl- secretion via the cystic fibrosis transmembrane conductance regulator (CFTR). Although the constitutive activation of adenylyl cyclase (AC) in response to CT is due to adenosine diphosphate ribosylation of the small G protein α-subunit activating CFTR with consequent secretory diarrhea, the AC isoform(s) involved remain unknown. METHODS We generated intestinal epithelial cell-specific adenylyl cyclase 6 (AC6) knockout mice to study its role in CT-induced diarrhea. RESULTS AC6 messenger RNA levels were the highest of all 9 membrane-bound AC isoforms in mouse intestinal epithelial cells. Intestinal epithelial-specific AC6 knockout mice (AC6loxloxVillinCre) had undetectable AC6 levels in small intestinal and colonic epithelial cells. No significant differences in fluid and food intake, plasma electrolytes, intestinal/colon anatomy and morphology, or fecal water content were observed between genotypes. Nevertheless, CT-induced fluid accumulation in vivo was completely absent in AC6loxloxVillinCre mice, associated with a lack of forskolin- and CT-induced changes in the short-circuit current (ISC) of the intestinal mucosa, impaired cAMP generation in acutely isolated small intestinal epithelial cells, and significantly impaired apical CFTR levels in response to forskolin. CONCLUSIONS AC6 is a novel target for the treatment of CT-induced diarrhea.
Collapse
Affiliation(s)
| | - Sathish K Murali
- Department of Biomedicine, Aarhus University, Denmark.,University of South Florida, Tampa
| | - Izumi Kaji
- Greater Los Angeles VA Healthcare System, California.,Department of Medicine, University of California, Los Angeles
| | - Yasutada Akiba
- Greater Los Angeles VA Healthcare System, California.,Department of Medicine, University of California, Los Angeles
| | - Jonathan D Kaunitz
- Greater Los Angeles VA Healthcare System, California.,Department of Medicine, University of California, Los Angeles
| | | | | | | | | |
Collapse
|
16
|
Sussman CR, Wang X, Chebib FT, Torres VE. Modulation of polycystic kidney disease by G-protein coupled receptors and cyclic AMP signaling. Cell Signal 2020; 72:109649. [PMID: 32335259 DOI: 10.1016/j.cellsig.2020.109649] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a systemic disorder associated with polycystic liver disease (PLD) and other extrarenal manifestations, the most common monogenic cause of end-stage kidney disease, and a major burden for public health. Many studies have shown that alterations in G-protein and cAMP signaling play a central role in its pathogenesis. As for many other diseases (35% of all approved drugs target G-protein coupled receptors (GPCRs) or proteins functioning upstream or downstream from GPCRs), treatments targeting GPCR have shown effectiveness in slowing the rate of progression of ADPKD. Tolvaptan, a vasopressin V2 receptor antagonist is the first drug approved by regulatory agencies to treat rapidly progressive ADPKD. Long-acting somatostatin analogs have also been effective in slowing the rates of growth of polycystic kidneys and liver. Although no treatment has so far been able to prevent the development or stop the progression of the disease, these encouraging advances point to G-protein and cAMP signaling as a promising avenue of investigation that may lead to more effective and safe treatments. This will require a better understanding of the relevant GPCRs, G-proteins, cAMP effectors, and of the enzymes and A-kinase anchoring proteins controlling the compartmentalization of cAMP signaling. The purpose of this review is to provide an overview of general GPCR signaling; the function of polycystin-1 (PC1) as a putative atypical adhesion GPCR (aGPCR); the roles of PC1, polycystin-2 (PC2) and the PC1-PC2 complex in the regulation of calcium and cAMP signaling; the cross-talk of calcium and cAMP signaling in PKD; and GPCRs, adenylyl cyclases, cyclic nucleotide phosphodiesterases, and protein kinase A as therapeutic targets in ADPKD.
Collapse
Affiliation(s)
- Caroline R Sussman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Xiaofang Wang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
17
|
Albertoni Borghese MF, Ortiz MDC, Marinoni RC, Oronel LH, Palamidessi M, Yarza CA, Di Siervi N, Davio C, Majowicz MP. Inhibition of Endothelin system during the postnatal nephrogenic period in the rat. Its relationship with hypertension and renal disease in adulthood. PLoS One 2020; 15:e0229756. [PMID: 32126132 PMCID: PMC7053749 DOI: 10.1371/journal.pone.0229756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/12/2020] [Indexed: 12/26/2022] Open
Abstract
The aim of this work was to study the effect of a high sodium (HS) diet on blood pressure and renal function in male adult rats that have been treated with a dual Endothelin receptor antagonist (ERA) during their early postnatal period (day 1 to 20 of life). Male Sprague-Dawley rats were divided in four groups: CNS: control rats with normosodic diet; ERANS: ERA-treated rats with normosodic diet; CHS: control rats with high sodium diet; ERAHS: ERA-treated rats with HS diet. Systolic blood pressure (SBP) was recorded before and after the diet and 24-hour metabolic cage studies were performed. AQP2 and α-ENac expressions were measured by western blot and real time PCR in the renal medulla. Vasopressin (AVP) pathway was evaluated by measuring V2 receptor and adenylyl cyclase 6 (AC6) expression and cAMP production in the renal medulla. Pre-pro ET-1mRNA was also evaluated in the renal medulla. Only rats that had been treated with an ERA during their postnatal period increased their SBP after consumption of a HS diet, showing an impaired capacity to excrete sodium and water, i.e. developing salt sensitivity. This salt sensitivity would be mediated by an increase in renomedullary expression and activity of AQP2 and α-ENaC as a consequence of increased AC6 expression and cAMP production and/or a decreased ET-1 production in the renal medulla. The knowledge of the molecular mechanisms underlying the perinatal programming of salt sensitive hypertension will allow the development of reprogramming strategies in order to avoid this pathology.
Collapse
Affiliation(s)
- María Florencia Albertoni Borghese
- Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María del Carmen Ortiz
- Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rocío C. Marinoni
- Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Lucas H. Oronel
- Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Milena Palamidessi
- Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carolina A. Yarza
- Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nicolás Di Siervi
- CONICET, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos Davio
- CONICET, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mónica P. Majowicz
- Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
18
|
|
19
|
Gonzalez AA, Salinas-Parra N, Cifuentes-Araneda F, Reyes-Martinez C. Vasopressin actions in the kidney renin angiotensin system and its role in hypertension and renal disease. VITAMINS AND HORMONES 2019; 113:217-238. [PMID: 32138949 DOI: 10.1016/bs.vh.2019.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vasopressin, also named antidiuretic hormone (ADH), arginine vasopressin (AVP) is the main hormone responsible for water maintenance in the body through the antidiuretic actions in the kidney. The posterior pituitary into the blood releases vasopressin formed in the hypothalamus. Hypothalamic osmotic neurons are responsible to initiate the cascade for AVP actions. The effects of AVP peptide includes activation of V2 receptors which stimulate the formation of cyclic AMP (cAMP) and phosphorylation of water channels aquaporin 2 (AQP2) in the collecting duct. AVP also has vasoconstrictor effects through V1a receptors in the vasculature, while V1b is found in the nervous system. V1a and b receptors increases intracellular Ca2+ while activation of V2 receptors of signaling pathways are related to cAMP-dependent phosphorylation in kidney collecting ducts acting in coordination to stimulate water and electrolyte homeostasis. AVP potentiate formation of intratubular angiotensin II (Ang II) through V2 receptors-dependent distal tubular renin formation, contributing to Na+ reabsorption. On the same way, Ang II receptors are able to potentiate the effects of V2-dependent stimulation of AQP2 abundance in the plasma membrane. The role of AVP in hypertension and renal disease has been demonstrated in pathological states with the involvement of V2 receptors in the progression of kidney damage in diabetes and also on the stimulation of intracellular pathways linked to the development of polycystic kidney.
Collapse
Affiliation(s)
- Alexis A Gonzalez
- Instituto de Química Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.
| | - Nicolas Salinas-Parra
- Instituto de Química Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | | | | |
Collapse
|
20
|
Deshpande V, Kao A, Raghuram V, Datta A, Chou CL, Knepper MA. Phosphoproteomic identification of vasopressin V2 receptor-dependent signaling in the renal collecting duct. Am J Physiol Renal Physiol 2019; 317:F789-F804. [PMID: 31313956 PMCID: PMC6843035 DOI: 10.1152/ajprenal.00281.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 06/28/2019] [Accepted: 07/04/2019] [Indexed: 12/15/2022] Open
Abstract
Vasopressin controls water balance largely through PKA-dependent effects to regulate the collecting duct water channel aquaporin-2 (AQP2). Although considerable information has accrued regarding the regulation of water and solute transport in collecting duct cells, information is sparse regarding the signaling connections between PKA and transport responses. Here, we exploited recent advancements in protein mass spectrometry to perform a comprehensive, multiple-replicate analysis of changes in the phosphoproteome of native rat inner medullary collecting duct cells in response to the vasopressin V2 receptor-selective agonist 1-desamino-8D-arginine vasopressin. Of the 10,738 phosphopeptides quantified, only 156 phosphopeptides were significantly increased in abundance, and only 63 phosphopeptides were decreased, indicative of a highly selective response to vasopressin. The list of upregulated phosphosites showed several general characteristics: 1) a preponderance of sites with basic (positively charged) amino acids arginine (R) and lysine (K) in position -2 and -3 relative to the phosphorylated amino acid, consistent with phosphorylation by PKA and/or other basophilic kinases; 2) a greater-than-random likelihood of sites previously demonstrated to be phosphorylated by PKA; 3) a preponderance of sites in membrane proteins, consistent with regulation by membrane association; and 4) a greater-than-random likelihood of sites in proteins with class I COOH-terminal PDZ ligand motifs. The list of downregulated phosphosites showed a preponderance of those with proline in position +1 relative to the phosphorylated amino acid, consistent with either downregulation of proline-directed kinases (e.g., MAPKs or cyclin-dependent kinases) or upregulation of one or more protein phosphatases that selectively dephosphorylate such sites (e.g., protein phosphatase 2A). The phosphoproteomic data were used to create a web resource for the investigation of G protein-coupled receptor signaling and regulation of AQP2-mediated water transport.
Collapse
Affiliation(s)
- Venkatesh Deshpande
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Anika Kao
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Arnab Datta
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
21
|
Sung CC, Chen L, Limbutara K, Jung HJ, Gilmer GG, Yang CR, Lin SH, Khositseth S, Chou CL, Knepper MA. RNA-Seq and protein mass spectrometry in microdissected kidney tubules reveal signaling processes initiating lithium-induced nephrogenic diabetes insipidus. Kidney Int 2019; 96:363-377. [PMID: 31146973 PMCID: PMC6650374 DOI: 10.1016/j.kint.2019.02.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 01/17/2019] [Accepted: 02/07/2019] [Indexed: 12/14/2022]
Abstract
Lithium salts, used for treating bipolar disorder, frequently induce nephrogenic diabetes insipidus (NDI) thereby limiting therapeutic success. NDI is associated with loss of expression of the gene coding for the molecular water channel, aquaporin-2, in the renal collecting duct (CD). Here, we use systems biology methods in a well-established rat model of lithium-induced NDI to identify signaling pathways activated at the onset of polyuria. Using single-tubule RNA-Seq, full transcriptomes were determined in microdissected cortical collecting ducts (CCDs) of rats after 72 hours without or with initiation of lithium chloride administration. Transcriptome-wide changes in mRNA abundances were mapped to gene sets associated with curated canonical signaling pathways, showing evidence for activation of NF-κB signaling with induction of genes coding for multiple chemokines and most components of the Major Histocompatibility Complex Class I antigen-presenting complex. Administration of anti-inflammatory doses of dexamethasone to lithium chloride-treated rats countered the loss of aquaporin-2. RNA-Seq also confirmed prior evidence of a shift from quiescence into the cell cycle with arrest. Time course studies demonstrated an early (12 hour) increase in multiple immediate early response genes including several transcription factors. Protein mass spectrometry in microdissected CCDs provided corroborative evidence and identified decreased abundance of several anti-oxidant proteins. Thus, in the context of prior observations, our study can be best explained by a model in which lithium increases ERK activation leading to induction of NF-κB signaling and an inflammatory-like response that represses Aqp2 transcription.
Collapse
Affiliation(s)
- Chih-Chien Sung
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA; Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kavee Limbutara
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Hyun Jun Jung
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Gabrielle G Gilmer
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chin-Rang Yang
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Shih-Hua Lin
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sookkasem Khositseth
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA; Department of Pediatrics, Faculty of Medicine, Thammasat University (Rangsit Campus), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
22
|
Rogers RT, Sun MA, Yue Q, Bao HF, Sands JM, Blount MA, Eaton DC. Lack of urea transporters, UT-A1 and UT-A3, increases nitric oxide accumulation to dampen medullary sodium reabsorption through ENaC. Am J Physiol Renal Physiol 2019; 316:F539-F549. [PMID: 30539654 PMCID: PMC6459308 DOI: 10.1152/ajprenal.00166.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 11/19/2018] [Accepted: 11/30/2018] [Indexed: 12/31/2022] Open
Abstract
Although the role of urea in urine concentration is known, the effect of urea handling by the urea transporters (UTs), UT-A1 and UT-A3, on sodium balance remains elusive. Serum and urinary sodium concentration is similar between wild-type mice (WT) and UT-A3 null (UT-A3 KO) mice; however, mice lacking both UT-A1 and UT-A3 (UT-A1/A3 KO) have significantly lower serum sodium and higher urinary sodium. Protein expression of renal sodium transporters is unchanged among all three genotypes. WT, UT-A3 KO, and UT-A1/A3 KO acutely respond to hydrochlorothiazide and furosemide; however, UT-A1/A3 KO fail to show a diuretic or natriuretic response following amiloride administration, indicating that baseline epithelial Na+ channel (ENaC) activity is impaired. UT-A1/A3 KO have more ENaC at the apical membrane than WT mice, and single-channel analysis of ENaC in split-open inner medullary collecting duct (IMCD) isolated in saline shows that ENaC channel density and open probability is higher in UT-A1/A3 KO than WT. UT-A1/A3 KO excrete more urinary nitric oxide (NO), a paracrine inhibitor of ENaC, and inner medullary nitric oxide synthase 1 mRNA expression is ~40-fold higher than WT. Because endogenous NO is unstable, ENaC activity was reassessed in split-open IMCD with the NO donor PAPA NONOate [1-propanamine-3-(2-hydroxy-2-nitroso-1-propylhydrazine)], and ENaC activity was almost abolished in UT-A1/A3 KO. In summary, loss of both UT-A1 and UT-A3 (but not UT-A3 alone) causes elevated medullary NO production and salt wasting. NO inhibition of ENaC, despite elevated apical accumulation of ENaC in UT-A1/A3 KO IMCD, appears to be the main contributor to natriuresis in UT-A1/A3 KO mice.
Collapse
Affiliation(s)
- Richard T Rogers
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
| | - Michael A Sun
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
| | - Qiang Yue
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| | - Hui-Fang Bao
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| | - Jeff M Sands
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| | - Mitsi A Blount
- Renal Division, Department of Medicine, Emory University School of Medicine , Atlanta, Georgia
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| | - Douglas C Eaton
- Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| |
Collapse
|
23
|
Ramkumar N, Stuart D, Mironova E, Abraham N, Gao Y, Wang S, Lakshmipathi J, Stockand JD, Kohan DE. Collecting duct principal, but not intercalated, cell prorenin receptor regulates renal sodium and water excretion. Am J Physiol Renal Physiol 2018; 315:F607-F617. [PMID: 29790390 PMCID: PMC6172572 DOI: 10.1152/ajprenal.00122.2018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/09/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022] Open
Abstract
The collecting duct is the predominant nephron site of prorenin and prorenin receptor (PRR) expression. We previously demonstrated that the collecting duct PRR regulates epithelial Na+ channel (ENaC) activity and water transport; however, which cell type is involved remains unclear. Herein, we examined the effects of principal cell (PC) or intercalated cell (IC) PRR deletion on renal Na+ and water handling. PC or IC PRR knockout (KO) mice were obtained by crossing floxed PRR mice with mice harboring Cre recombinase under the control of the AQP2 or B1 subunit of the H+ ATPase promoters, respectively. PC KO mice had reduced renal medullary ENaC-α abundance and increased urinary Na+ losses on a low-Na+ diet compared with controls. Conversely, IC KO mice had no apparent differences in Na+ balance or ENaC abundance compared with controls. Acute treatment with prorenin increased ENaC channel number and open probability in acutely isolated cortical collecting ducts from control and IC PRR KO, but not PC PRR KO, mice. Furthermore, compared with controls, PC KO, but not IC KO mice, had increased urine volume, reduced urine osmolality, and reduced abundance of renal medullary AQP2. Taken together, these findings indicate that PC, but not IC, PRR modulates ENaC activity, urinary Na+ excretion, and water transport.
Collapse
Affiliation(s)
- Nirupama Ramkumar
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center , Salt Lake City, Utah
| | - Deborah Stuart
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center , Salt Lake City, Utah
| | - Elena Mironova
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center , San Antonio, Texas
| | - Nikita Abraham
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center , Salt Lake City, Utah
| | - Yang Gao
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center , Salt Lake City, Utah
| | - Shuping Wang
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center , Salt Lake City, Utah
| | - Jayalakshmi Lakshmipathi
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center , Salt Lake City, Utah
| | - James D Stockand
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center , San Antonio, Texas
| | - Donald E Kohan
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center , Salt Lake City, Utah
- Department of Veterans Affairs Medical Center , Salt Lake City, Utah
| |
Collapse
|
24
|
Adenylyl cyclase 6 is required for maintaining acid-base homeostasis. Clin Sci (Lond) 2018; 132:1779-1796. [PMID: 29941522 DOI: 10.1042/cs20180060] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 06/15/2018] [Accepted: 06/25/2018] [Indexed: 12/14/2022]
Abstract
Adenylyl cyclase (AC) isoform 6 (AC6) is highly expressed throughout the renal tubule and collecting duct (CD), catalyzes the synthesis of cAMP and contributes to various aspects of renal transport. Several proteins involved in acid-base homeostasis are regulated by cAMP. In the present study, we assess the relative contribution of AC6 to overall acid-base regulation using mice with global deletion of AC6 (AC6-/-) or newly generated mice lacking AC6 in the renal tubule and CD (AC6loxloxPax8Cre). Higher energy expenditure in AC6-/- relative to wild-type (WT) mice, was associated with lower urinary pH, mild alkalosis in conjunction with elevated blood HCO3- concentrations, and significantly higher renal abundance of the H+-ATPase B1 subunit. In contrast with WT mice, AC6-/- mice have a less pronounced increase in urinary pH after 8 days of HCO3- challenge, which is associated with increased blood pH and HCO3- concentrations. Immunohistochemistry demonstrated that AC6 was expressed in intercalated cells (IC), but subcellular distribution of the H+-ATPase B1 subunit, pendrin, and the anion exchangers 1 and 2 in AC6-/- mice was normal. In the AC6-/- mice, H+-ATPase B1 subunit levels after HCO3- challenge were greater, which correlated with a higher number of type A IC. In contrast with the AC6-/- mice, AC6loxloxPax8Cre mice had normal urinary pH under baseline conditions but higher blood HCO3- than controls after HCO3- challenge. In conclusion, AC6 is required for maintaining normal acid-base homeostasis and energy expenditure. Under baseline conditions, renal AC6 is redundant for acid-base balance but becomes important under alkaline conditions.
Collapse
|
25
|
Gonzalez AA, Salinas-Parra N, Leach D, Navar LG, Prieto MC. PGE 2 upregulates renin through E-prostanoid receptor 1 via PKC/cAMP/CREB pathway in M-1 cells. Am J Physiol Renal Physiol 2017; 313:F1038-F1049. [PMID: 28701311 DOI: 10.1152/ajprenal.00194.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/23/2017] [Accepted: 07/10/2017] [Indexed: 12/12/2022] Open
Abstract
During the early phase of ANG II-dependent hypertension, tubular PGE2 is increased. Renin synthesis and secretion in the collecting duct (CD) are upregulated by ANG II, contributing to further intratubular ANG II formation. However, what happens first and whether the triggering mechanism is independent of tubular ANG II remain unknown. PGE2 stimulates renin synthesis in juxtaglomerular cells via E-prostanoid (EP) receptors through the cAMP/cAMP-responsive element-binding (CREB) pathway. EP receptors are also expressed in the CD. Here, we tested the hypothesis that renin is upregulated by PGE2 in CD cells. The M-1 CD cell line expressed EP1, EP3, and EP4 but not EP2. Dose-response experiments, in the presence of ANG II type 1 receptor blockade with candesartan, demonstrated that 10-6 M PGE2 maximally increases renin mRNA (approximately 4-fold) and prorenin/renin protein levels (approximately 2-fold). This response was prevented by micromolar doses of SC-19220 (EP1 antagonist), attenuated by the EP4 antagonist, L-161982, and exacerbated by the highly selective EP3 antagonist, L-798106 (~10-fold increase). To evaluate further the signaling pathway involved, we used the PKC inhibitor calphostin C and transfections with PKCα dominant negative. Both strategies blunted the PGE2-induced increases in cAMP levels, CREB phosphorylation, and augmentation of renin. Knockdown of the EP1 receptor and CREB also prevented renin upregulation. These results indicate that PGE2 increases CD renin expression through the EP1 receptor via the PKC/cAMP/CREB pathway. Therefore, we conclude that during the early stages of ANG II-dependent hypertension, there is augmentation of PGE2 that stimulates renin in the CD, resulting in increased tubular ANG II formation and further stimulation of renin.
Collapse
Affiliation(s)
- Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile;
| | - Nicolas Salinas-Parra
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Dan Leach
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana; and
| | - L Gabriel Navar
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana; and.,Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Minolfa C Prieto
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana; and.,Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
26
|
Poulsen SB, Kristensen TB, Brooks HL, Kohan DE, Rieg T, Fenton RA. Role of adenylyl cyclase 6 in the development of lithium-induced nephrogenic diabetes insipidus. JCI Insight 2017; 2:e91042. [PMID: 28405619 DOI: 10.1172/jci.insight.91042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Psychiatric patients treated with lithium (Li+) may develop nephrogenic diabetes insipidus (NDI). Although the etiology of Li+-induced NDI (Li-NDI) is poorly understood, it occurs partially due to reduced aquaporin-2 (AQP2) expression in the kidney collecting ducts. A mechanism postulated for this is that Li+ inhibits adenylyl cyclase (AC) activity, leading to decreased cAMP, reduced AQP2 abundance, and less membrane targeting. We hypothesized that Li-NDI would not develop in mice lacking AC6. Whole-body AC6 knockout (AC6-/-) mice and potentially novel connecting tubule/principal cell-specific AC6 knockout (AC6loxloxCre) mice had approximately 50% lower urine osmolality and doubled water intake under baseline conditions compared with controls. Dietary Li+ administration increased water intake and reduced urine osmolality in control, AC6-/-, and AC6loxloxCre mice. Consistent with AC6-/- mice, medullary AQP2 and pS256-AQP2 abundances were lower in AC6loxloxCre mice compared with controls under standard conditions, and levels were further reduced after Li+ administration. AC6loxloxCre and control mice had a similar increase in the numbers of proliferating cell nuclear antigen-positive cells in response to Li+. However, AC6loxloxCre mice had a higher number of H+-ATPase B1 subunit-positive cells under standard conditions and after Li+ administration. Collectively, AC6 has a minor role in Li-NDI development but may be important for determining the intercalated cell-to-principal cell ratio.
Collapse
Affiliation(s)
- Søren Brandt Poulsen
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,VA San Diego Healthcare System, San Diego, California, USA
| | | | - Heddwen L Brooks
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - Timo Rieg
- VA San Diego Healthcare System, San Diego, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Robert A Fenton
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
27
|
Dessauer CW, Watts VJ, Ostrom RS, Conti M, Dove S, Seifert R. International Union of Basic and Clinical Pharmacology. CI. Structures and Small Molecule Modulators of Mammalian Adenylyl Cyclases. Pharmacol Rev 2017; 69:93-139. [PMID: 28255005 PMCID: PMC5394921 DOI: 10.1124/pr.116.013078] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adenylyl cyclases (ACs) generate the second messenger cAMP from ATP. Mammalian cells express nine transmembrane AC (mAC) isoforms (AC1-9) and a soluble AC (sAC, also referred to as AC10). This review will largely focus on mACs. mACs are activated by the G-protein Gαs and regulated by multiple mechanisms. mACs are differentially expressed in tissues and regulate numerous and diverse cell functions. mACs localize in distinct membrane compartments and form signaling complexes. sAC is activated by bicarbonate with physiologic roles first described in testis. Crystal structures of the catalytic core of a hybrid mAC and sAC are available. These structures provide detailed insights into the catalytic mechanism and constitute the basis for the development of isoform-selective activators and inhibitors. Although potent competitive and noncompetitive mAC inhibitors are available, it is challenging to obtain compounds with high isoform selectivity due to the conservation of the catalytic core. Accordingly, caution must be exerted with the interpretation of intact-cell studies. The development of isoform-selective activators, the plant diterpene forskolin being the starting compound, has been equally challenging. There is no known endogenous ligand for the forskolin binding site. Recently, development of selective sAC inhibitors was reported. An emerging field is the association of AC gene polymorphisms with human diseases. For example, mutations in the AC5 gene (ADCY5) cause hyperkinetic extrapyramidal motor disorders. Overall, in contrast to the guanylyl cyclase field, our understanding of the (patho)physiology of AC isoforms and the development of clinically useful drugs targeting ACs is still in its infancy.
Collapse
Affiliation(s)
- Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Val J Watts
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Rennolds S Ostrom
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Marco Conti
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Stefan Dove
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Roland Seifert
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| |
Collapse
|
28
|
Tamma G, Di Mise A, Ranieri M, Geller A, Tamma R, Zallone A, Valenti G. The V2 receptor antagonist tolvaptan raises cytosolic calcium and prevents AQP2 trafficking and function: an in vitro and in vivo assessment. J Cell Mol Med 2017; 21:1767-1780. [PMID: 28326667 PMCID: PMC5571526 DOI: 10.1111/jcmm.13098] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/21/2016] [Indexed: 01/07/2023] Open
Abstract
Tolvaptan, a selective vasopressin V2 receptor antagonist, is a new generation diuretic. Its clinical efficacy is in principle due to impaired vasopressin‐regulated water reabsorption via aquaporin‐2 (AQP2). Nevertheless, no direct in vitro evidence that tolvaptan prevents AQP2‐mediated water transport, nor that this pathway is targeted in vivo in patients with syndrome of inappropriate antidiuresis (SIAD) has been provided. The effects of tolvaptan on the vasopressin–cAMP/PKA signalling cascade were investigated in MDCK cells expressing endogenous V2R and in mouse kidney. In MDCK, tolvaptan prevented dDAVP‐induced increase in ser256‐AQP2 and osmotic water permeability. A similar effect on ser256‐AQP2 was found in V1aR −/− mice, thus confirming the V2R selectively. Of note, calcium calibration in MDCK showed that tolvaptan per se caused calcium mobilization from the endoplasmic reticulum resulting in a significant increase in basal intracellular calcium. This effect was only observed in cells expressing the V2R, indicating that it requires the tolvaptan–V2R interaction. Consistent with this finding, tolvaptan partially reduced the increase in ser256‐AQP2 and the water permeability in response to forskolin, a direct activator of adenylyl cyclase (AC), suggesting that the increase in intracellular calcium is associated with an inhibition of the calcium‐inhibitable AC type VI. Furthermore, tolvaptan treatment reduced AQP2 excretion in two SIAD patients and normalized plasma sodium concentration. These data represent the first detailed demonstration of the central role of AQP2 blockade in the aquaretic effect of tolvaptan and underscore a novel effect in raising intracellular calcium that can be of significant clinical relevance.
Collapse
Affiliation(s)
- Grazia Tamma
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Annarita Di Mise
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Marianna Ranieri
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | | | - Roberto Tamma
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Alberta Zallone
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Giovanna Valenti
- Department of Biosciences Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
29
|
Olesen ETB, Fenton RA. Aquaporin-2 membrane targeting: still a conundrum. Am J Physiol Renal Physiol 2017; 312:F744-F747. [PMID: 28179252 DOI: 10.1152/ajprenal.00010.2017] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/01/2017] [Accepted: 02/01/2017] [Indexed: 11/22/2022] Open
Abstract
The targeting of the water channel aquaporin-2 (AQP2) to the apical plasma membrane of kidney collecting duct principal cells is regulated mainly by the antidiuretic peptide hormone arginine vasopressin (AVP). This process is of crucial importance for the maintenance of body water homeostasis. In this brief review we assess the role of cyclic adenosine monophosphate (cAMP) and discuss the emerging concept that type 2 AVP receptor (V2R)-mediated AQP2 trafficking is cAMP-independent.
Collapse
Affiliation(s)
- Emma T B Olesen
- Department of Biomedical Sciences, Faculty of Health Sciences, Copenhagen University, Copenhagen, Denmark; .,Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; and
| | - Robert A Fenton
- Department of Biomedicine, InterPrET Center, Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
30
|
Jung HJ, Kwon TH. Molecular mechanisms regulating aquaporin-2 in kidney collecting duct. Am J Physiol Renal Physiol 2016; 311:F1318-F1328. [PMID: 27760771 DOI: 10.1152/ajprenal.00485.2016] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/11/2016] [Accepted: 10/11/2016] [Indexed: 01/04/2023] Open
Abstract
The kidney collecting duct is an important renal tubular segment for regulation of body water homeostasis and urine concentration. Water reabsorption in the collecting duct principal cells is controlled by vasopressin, a peptide hormone that induces the osmotic water transport across the collecting duct epithelia through regulation of water channel proteins aquaporin-2 (AQP2) and aquaporin-3 (AQP3). In particular, vasopressin induces both intracellular translocation of AQP2-bearing vesicles to the apical plasma membrane and transcription of the Aqp2 gene to increase AQP2 protein abundance. The signaling pathways, including AQP2 phosphorylation, RhoA phosphorylation, intracellular calcium mobilization, and actin depolymerization, play a key role in the translocation of AQP2. This review summarizes recent data demonstrating the regulation of AQP2 as the underlying molecular mechanism for the homeostasis of water balance in the body.
Collapse
Affiliation(s)
- Hyun Jun Jung
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|
31
|
Soleimani M, Barone S, Xu J, Alshahrani S, Brooks M, McCormack FX, Smith RD, Zahedi K. Prostaglandin-E2 Mediated Increase in Calcium and Phosphate Excretion in a Mouse Model of Distal Nephron Salt Wasting. PLoS One 2016; 11:e0159804. [PMID: 27442254 PMCID: PMC4956050 DOI: 10.1371/journal.pone.0159804] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/10/2016] [Indexed: 01/29/2023] Open
Abstract
Contribution of salt wasting and volume depletion to the pathogenesis of hypercalciuria and hyperphosphaturia is poorly understood. Pendrin/NCC double KO (pendrin/NCC-dKO) mice display severe salt wasting under basal conditions and develop profound volume depletion, prerenal renal failure, and metabolic alkalosis and are growth retarded. Microscopic examination of the kidneys of pendrin/NCC-dKO mice revealed the presence of calcium phosphate deposits in the medullary collecting ducts, along with increased urinary calcium and phosphate excretion. Confirmatory studies revealed decreases in the expression levels of sodium phosphate transporter-2 isoforms a and c, increases in the expression of cytochrome p450 family 4a isotypes 12 a and b, as well as prostaglandin E synthase 1, and cyclooxygenases 1 and 2. Pendrin/NCC-dKO animals also had a significant increase in urinary prostaglandin E2 (PGE-2) and renal content of 20-hydroxyeicosatetraenoic acid (20-HETE) levels. Pendrin/NCC-dKO animals exhibit reduced expression levels of the sodium/potassium/2chloride co-transporter 2 (NKCC2) in their medullary thick ascending limb. Further assessment of the renal expression of NKCC2 isoforms by quantitative real time PCR (qRT-PCR) reveled that compared to WT mice, the expression of NKCC2 isotype F was significantly reduced in pendrin/NCC-dKO mice. Provision of a high salt diet to rectify volume depletion or inhibition of PGE-2 synthesis by indomethacin, but not inhibition of 20-HETE generation by HET0016, significantly improved hypercalciuria and salt wasting in pendrin/NCC dKO mice. Both high salt diet and indomethacin treatment also corrected the alterations in NKCC2 isotype expression in pendrin/NCC-dKO mice. We propose that severe salt wasting and volume depletion, irrespective of the primary originating nephron segment, can secondarily impair the reabsorption of salt and calcium in the thick ascending limb of Henle and/or proximal tubule, and reabsorption of sodium and phosphate in the proximal tubule via processes that are mediated by PGE-2.
Collapse
Affiliation(s)
- Manoocher Soleimani
- Center on Genetics of Transport, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, United States of America
- * E-mail:
| | - Sharon Barone
- Center on Genetics of Transport, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, United States of America
| | - Jie Xu
- Center on Genetics of Transport, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Saeed Alshahrani
- Department of Pharmacology and Cell Biophysics and, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Marybeth Brooks
- Center on Genetics of Transport, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Francis X. McCormack
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Roger D. Smith
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Kamyar Zahedi
- Center on Genetics of Transport, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Departments of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
- Research Services, Veterans Affairs Medical Center, Cincinnati, OH, United States of America
| |
Collapse
|
32
|
Mamenko M, Dhande I, Tomilin V, Zaika O, Boukelmoune N, Zhu Y, Gonzalez-Garay ML, Pochynyuk O, Doris PA. Defective Store-Operated Calcium Entry Causes Partial Nephrogenic Diabetes Insipidus. J Am Soc Nephrol 2016; 27:2035-2048. [PMID: 26574044 PMCID: PMC4926963 DOI: 10.1681/asn.2014121200] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 09/23/2015] [Indexed: 12/12/2022] Open
Abstract
Store-operated calcium entry (SOCE) is the mechanism by which extracellular signals elicit prolonged intracellular calcium elevation to drive changes in fundamental cellular processes. Here, we investigated the role of SOCE in the regulation of renal water reabsorption, using the inbred rat strain SHR-A3 as an animal model with disrupted SOCE. We found that SHR-A3, but not SHR-B2, have a novel truncating mutation in the gene encoding stromal interaction molecule 1 (STIM1), the endoplasmic reticulum calcium (Ca(2+)) sensor that triggers SOCE. Balance studies revealed increased urine volume, hypertonic plasma, polydipsia, and impaired urinary concentrating ability accompanied by elevated circulating arginine vasopressin (AVP) levels in SHR-A3 compared with SHR-B2. Isolated, split-open collecting ducts (CD) from SHR-A3 displayed decreased basal intracellular Ca(2+) levels and a major defect in SOCE. Consequently, AVP failed to induce the sustained intracellular Ca(2+) mobilization that requires SOCE in CD cells from SHR-A3. This effect decreased the abundance of aquaporin 2 and enhanced its intracellular retention, suggesting impaired sensitivity of the CD to AVP in SHR-A3. Stim1 knockdown in cultured mpkCCDc14 cells reduced SOCE and basal intracellular Ca(2+) levels and prevented AVP-induced translocation of aquaporin 2, further suggesting the effects in SHR-A3 result from the expression of truncated STIM1. Overall, these results identify a novel mechanism of nephrogenic diabetes insipidus and uncover a role of SOCE in renal water handling.
Collapse
Affiliation(s)
| | - Isha Dhande
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Viktor Tomilin
- Department of Integrative Biology and Pharmacology, and Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, and
| | | | - Yaming Zhu
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas; and
| | - Manuel L Gonzalez-Garay
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas; and
| | | | - Peter A Doris
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas; and
| |
Collapse
|
33
|
Cebotaru L, Cebotaru V, Wang H, Arend LJ, Guggino WB. STIM1fl/fl Ksp-Cre Mouse has Impaired Renal Water Balance. Cell Physiol Biochem 2016; 39:172-82. [PMID: 27336410 DOI: 10.1159/000445614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND/AIM STIM1 is as an essential component in store operated Ca2+ entry. However give the paucity of information on the role of STIM1 in kidney, the aim was to study the function of STIM1 in the medulla of the kidney. METHODS we crossed a Ksp-cre mouse with another mouse containing two loxP sites flanking Exon 6 of STIM1. The Ksp-cre mouse is based upon the Ksp-cadherin gene promoter which expresses cre recombinase in developing nephrons, collecting ducts (SD) and thick ascending limbs (TAL) of the loop of Henle. RESULTS The offspring of these mice are viable without gross morphological changes, however, we noticed that the STIM1 Ksp-cre knockout mice produced more urine compared to control. To examine this more carefully, we fed mice low (LP) and high protein (HP) diets respectively. When mice were fed HP diet STIM1 ko mice had significantly increased urinary volume and lower specific gravity compared to wt mice. In STIM1 ko mice fed HP diet urine creatinine and urea were significantly lower compared to wt mice fed HP diet, however the fractional excretion was the same. CONCLUSION These data support the idea that STIM1 ko mice have impaired urinary concentrating ability when challenged with HP diet is most likely caused by impaired Ca2+-dependent signal transduction through the vasopressin receptor cascade.
Collapse
Affiliation(s)
- Liudmila Cebotaru
- Division of Gastroenterology, Department of Medicine, Division of Nephrology, University of Maryland, Baltimore, USA
| | | | | | | | | |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Renal collecting ducts maintain NaCl homeostasis by fine-tuning urinary excretion to balance dietary salt intake. This review focuses on recent studies on transcellular Cl secretion by collecting ducts, its regulation and its role in cyst growth in autosomal dominant polycystic kidney disease (ADPKD). RECENT FINDINGS Lumens of nonperfused rat medullary collecting ducts collapse in control media but expand with fluid following treatment with cAMP, demonstrating the capacity for both salt absorption and secretion. Recently, inhibition of apical epithelial Na channels (ENaC) unmasked Cl secretion in perfused mouse cortical collecting ducts (CCDs), involving Cl uptake by basolateral NKCC1 and efflux through apical Cl channels. AVP, the key hormone for osmoregulation, promotes cystic fibrosis transmembrane conductance regulator (CFTR)-mediated Cl secretion. In addition, prostaglandin E2 stimulates Cl secretion through both CFTR and Ca-activated Cl channels. SUMMARY Renal Cl secretion has been commonly overlooked because of the overwhelming capacity for the nephron to reabsorb NaCl from the glomerular filtrate. In ADPKD, Cl secretion plays a central role in the accumulation of cyst fluid and the remarkable size of the cystic kidneys. Investigation of renal Cl secretion may provide a better understanding of NaCl homeostasis and identify new approaches to reduce cyst growth in PKD.
Collapse
|
35
|
|
36
|
Wu M, Yu S. New Insights into the Molecular Mechanisms Targeting Tubular Channels/Transporters in PKD Development. KIDNEY DISEASES 2016; 2:128-135. [PMID: 27921040 DOI: 10.1159/000444839] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/18/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (PKD) or autosomal recessive PKD is caused by a mutation in the PKD1, PKD2 or PKHD1 gene, which encodes polycystin-1, polycystin-2 or fibrocystin, respectively. Embryonic and postnatal mutation studies show that transport or channel function is dysregulated before the initiation of cystogenesis, suggesting that the abnormality of transport or channel function plays a critical role in the pathology of PKD. SUMMARY Polycystin-2 by itself is a calcium-permeable cation channel, and its channel function can be regulated by polycystin-1 or fibrocystin. In this paper, we reviewed the current knowledge about calcium transports and cyclic adenosine monophosphate (cAMP)-driven chloride transports in PKD. In addition, the function and the underlining mechanism of glucose transporters, phosphate transporters and water channels in PKD are also discussed. KEY MESSAGES Abnormalities in calcium handling and exuberant cAMP-dependent cystic fibrosis transmembrane conductance regulator-mediated fluid secretion in the collecting duct are the most important issues in the pathogenesis of PKD.
Collapse
Affiliation(s)
- Ming Wu
- Kidney Institute of PLA, Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, PR China
| | - Shengqiang Yu
- Kidney Institute of PLA, Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, PR China
| |
Collapse
|
37
|
Zhou L, Panasiuk A, Downton M, Zhao D, Yang B, Jia Z, Yang T. Systemic PPARγ deletion causes severe disturbance in fluid homeostasis in mice. Physiol Genomics 2015; 47:541-7. [PMID: 26330489 DOI: 10.1152/physiolgenomics.00066.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/27/2015] [Indexed: 01/29/2023] Open
Abstract
The pharmacological action of peroxisome proliferator-activated receptor (PPAR)γ in promoting sodium and water retention is well documented as highlighted by the major side-effect of body weight gain and edema associated with thiazolidinedione use. However, a possible physiological role of PPARγ in regulation of fluid metabolism has not been reported by previous studies. Here we analyzed fluid metabolism in inducible whole-body PPARγ knockout mice. The null mice developed severe polydipsia and polyuria, reduced urine osmolality, and modest hyperphagia. The phenomenon persisted during 3 days of pair feeding and pair drinking, accompanied by progressive weight loss. After 24 h water deprivation, the null mice had a lower urine osmolality, a higher urine volume, a greater weight loss, and a greater rise in hematocrit than the floxed control. Urinary vasopressin (AVP) excretion was not different between the genotypes under basal condition or after WD. The response of urine osmolality to acute and chronic 1-desamino-8-D-arginine vasopressin treatment was attenuated in the null mice, but the total abundance or phosphorylation of aquaporin 2 (AQP2) in the kidney or AVP-induced cAMP production in inner medullary collecting duct suspensions was unaffected. Overall, PPARγ participates in physiological control of fluid homeostasis through an unknown mechanism involving cAMP/AQP2-independent enhancement of AVP response.
Collapse
Affiliation(s)
- Li Zhou
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China; Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Alexandra Panasiuk
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Maicy Downton
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Daqiang Zhao
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhanjun Jia
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Tianxin Yang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China; Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| |
Collapse
|
38
|
Kortenoeven MLA, Pedersen NB, Rosenbaek LL, Fenton RA. Vasopressin regulation of sodium transport in the distal nephron and collecting duct. Am J Physiol Renal Physiol 2015; 309:F280-99. [DOI: 10.1152/ajprenal.00093.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/27/2015] [Indexed: 12/22/2022] Open
Abstract
Arginine vasopressin (AVP) is released from the posterior pituitary gland during states of hyperosmolality or hypovolemia. AVP is a peptide hormone, with antidiuretic and antinatriuretic properties. It allows the kidneys to increase body water retention predominantly by increasing the cell surface expression of aquaporin water channels in the collecting duct alongside increasing the osmotic driving forces for water reabsorption. The antinatriuretic effects of AVP are mediated by the regulation of sodium transport throughout the distal nephron, from the thick ascending limb through to the collecting duct, which in turn partially facilitates osmotic movement of water. In this review, we will discuss the regulatory role of AVP in sodium transport and summarize the effects of AVP on various molecular targets, including the sodium-potassium-chloride cotransporter NKCC2, the thiazide-sensitive sodium-chloride cotransporter NCC, and the epithelial sodium channel ENaC.
Collapse
Affiliation(s)
- M. L. A. Kortenoeven
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus, Denmark
| | - N. B. Pedersen
- Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; and
| | - L. L. Rosenbaek
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - R. A. Fenton
- Department of Biomedicine and Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus, Denmark
| |
Collapse
|
39
|
Gonzalez AA, Liu L, Lara LS, Bourgeois CRT, Ibaceta-Gonzalez C, Salinas-Parra N, Gogulamudi VR, Seth DM, Prieto MC. PKC-α-dependent augmentation of cAMP and CREB phosphorylation mediates the angiotensin II stimulation of renin in the collecting duct. Am J Physiol Renal Physiol 2015; 309:F880-8. [PMID: 26268270 DOI: 10.1152/ajprenal.00155.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/06/2015] [Indexed: 11/22/2022] Open
Abstract
In contrast to the negative feedback of angiotensin II (ANG II) on juxtaglomerular renin, ANG II stimulates renin in the principal cells of the collecting duct (CD) in rats and mice via ANG II type 1 (AT1R) receptor, independently of blood pressure. In vitro data indicate that CD renin is augmented by AT1R activation through protein kinase C (PKC), but the exact mechanisms are unknown. We hypothesize that ANG II stimulates CD renin synthesis through AT1R via PKC and the subsequent activation of cAMP/PKA/CREB pathway. In M-1 cells, ANG II increased cAMP, renin mRNA (3.5-fold), prorenin, and renin proteins, as well as renin activity in culture media (2-fold). These effects were prevented by PKC inhibition with calphostin C, PKC-α dominant negative, and by PKA inhibition. Forskolin-induced increases in cAMP and renin expression were prevented by calphostin C. PKC inhibition and Ca2+ depletion impaired ANG II-mediated CREB phosphorylation and upregulation of renin. Adenylate cyclase 6 (AC) siRNA remarkably attenuated the ANG II-dependent upregulation of renin mRNA. Physiological activation of AC with vasopressin increased renin expression in M-1 cells. The results suggest that the ANG II-dependent upregulation of renin in the CD depends on PKC-α, which allows the augmentation of cAMP production and activation of PKA/CREB pathway via AC6. This study defines the intracellular signaling pathway involved in the ANG II-mediated stimulation of renin in the CD. This is a novel mechanism responsible for the regulation of local renin-angiotensin system in the distal nephron.
Collapse
Affiliation(s)
- Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile; Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana; and
| | - Liu Liu
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana; and
| | - Lucienne S Lara
- Instituto de Ciencias Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana; and
| | - Camille R T Bourgeois
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana; and
| | | | - Nicolas Salinas-Parra
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | | | - Dale M Seth
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana; and
| | - Minolfa C Prieto
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana; and
| |
Collapse
|
40
|
Ranieri M, Tamma G, Di Mise A, Russo A, Centrone M, Svelto M, Calamita G, Valenti G. Negative feedback from CaSR signaling to aquaporin-2 sensitizes vasopressin to extracellular Ca2. J Cell Sci 2015; 128:2350-60. [PMID: 25977473 DOI: 10.1242/jcs.168096] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 05/05/2015] [Indexed: 01/10/2023] Open
Abstract
We previously described that high luminal Ca(2+) in the renal collecting duct attenuates short-term vasopressin-induced aquaporin-2 (AQP2) trafficking through activation of the Ca(2+)-sensing receptor (CaSR). Here, we evaluated AQP2 phosphorylation and permeability, in both renal HEK-293 cells and in the dissected inner medullary collecting duct, in response to specific activation of CaSR with NPS-R568. In CaSR-transfected cells, CaSR activation drastically reduced the basal levels of AQP2 phosphorylation at S256 (AQP2-pS256), thus having an opposite effect to vasopressin action. When forskolin stimulation was performed in the presence of NPS-R568, the increase in AQP2-pS256 and in the osmotic water permeability were prevented. In the freshly isolated inner mouse medullar collecting duct, stimulation with forskolin in the presence of NPS-R568 prevented the increase in AQP2-pS256 and osmotic water permeability. Our data demonstrate that the activation of CaSR in the collecting duct prevents the cAMP-dependent increase in AQP2-pS256 and water permeability, counteracting the short-term vasopressin response. By extension, our results suggest the attractive concept that CaSR expressed in distinct nephron segments exerts a negative feedback on hormones acting through cAMP, conferring high sensitivity of hormone to extracellular Ca(2+).
Collapse
Affiliation(s)
- Marianna Ranieri
- Dept of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Aldo Moro, 70125, Bari, Italy
| | - Grazia Tamma
- Dept of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Aldo Moro, 70125, Bari, Italy Istituto Nazionale di Biostrutture e Biosistemi (I.N.B.B.) 00136, Rome, Italy
| | - Annarita Di Mise
- Dept of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Aldo Moro, 70125, Bari, Italy
| | - Annamaria Russo
- Dept of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Aldo Moro, 70125, Bari, Italy
| | - Mariangela Centrone
- Dept of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Aldo Moro, 70125, Bari, Italy
| | - Maria Svelto
- Dept of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Aldo Moro, 70125, Bari, Italy Istituto Nazionale di Biostrutture e Biosistemi (I.N.B.B.) 00136, Rome, Italy Centro di Eccellenza di Genomica in campo Biomedico ed Agrario (CEGBA) 70125, Bari, Italy
| | - Giuseppe Calamita
- Dept of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Aldo Moro, 70125, Bari, Italy Istituto Nazionale di Biostrutture e Biosistemi (I.N.B.B.) 00136, Rome, Italy
| | - Giovanna Valenti
- Dept of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Aldo Moro, 70125, Bari, Italy Istituto Nazionale di Biostrutture e Biosistemi (I.N.B.B.) 00136, Rome, Italy Centro di Eccellenza di Genomica in campo Biomedico ed Agrario (CEGBA) 70125, Bari, Italy
| |
Collapse
|
41
|
Kittikulsuth W, Stuart D, Van Hoek AN, Kohan DE. Lack of an effect of nephron-specific deletion of adenylyl cyclase 3 on renal sodium and water excretion or arterial pressure. Physiol Rep 2015; 3:3/3/e12316. [PMID: 25747587 PMCID: PMC4393152 DOI: 10.14814/phy2.12316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Adenylyl cyclase (AC)-stimulated cAMP plays a key role in modulating transport and channel activity along the nephron. However, the role of individual adenylyl cyclase isoforms in such regulation is largely unknown. Since adenylyl cyclase 3 (AC3) is expressed throughout nephron, we investigated its role in the physiologic regulation of renal Na+ and water transport. Mice were generated with inducible nephron knockout of AC3 (AC3 KO) by breeding mice with loxP-flanked critical exons in the Adcy3 gene with mice expressing Pax8-rtTA and LC-1 transgenes. After doxycycline treatment at 1 month of age, nephron AC3 KO mice had 100% Adcy3 gene recombination in all renal tubule segments, but not in glomeruli. Sodium intake, urinary Na+ excretion, glomerular filtration rate, and blood pressure were similar between nephron KO mice and the controls during normal, high, and low Na+ diets. Plasma renin concentration was not different between the two groups during varied Na+ intake. Moreover, there were no differences in urine volume and urine osmolality between the two genotypes during normal or restricted water intake. In conclusion, these data suggested that AC3 is not involved in the physiological regulation of nephron Na+ and water handling.
Collapse
Affiliation(s)
- Wararat Kittikulsuth
- Division of Nephrology, University of Utah Health Sciences Center Salt Lake City Veterans Affairs Medical Center, Salt Lake City, UT
| | - Deborah Stuart
- Division of Nephrology, University of Utah Health Sciences Center Salt Lake City Veterans Affairs Medical Center, Salt Lake City, UT
| | - Alfred N Van Hoek
- Division of Nephrology, University of Utah Health Sciences Center Salt Lake City Veterans Affairs Medical Center, Salt Lake City, UT
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Sciences Center Salt Lake City Veterans Affairs Medical Center, Salt Lake City, UT
| |
Collapse
|
42
|
Fenton RA, Murray F, Dominguez Rieg JA, Tang T, Levi M, Rieg T. Renal phosphate wasting in the absence of adenylyl cyclase 6. J Am Soc Nephrol 2014; 25:2822-34. [PMID: 24854272 PMCID: PMC4243352 DOI: 10.1681/asn.2013101102] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 03/31/2014] [Indexed: 11/03/2022] Open
Abstract
Parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF-23) enhance phosphate excretion by the proximal tubule of the kidney by retrieval of the sodium-dependent phosphate transporters (Npt2a and Npt2c) from the apical plasma membrane. PTH activates adenylyl cyclase (AC) through PTH 1 receptors and stimulates the cAMP/PKA signaling pathway. However, the precise role and isoform(s) of AC in phosphate homeostasis are not known. We report here that mice lacking AC6 (AC6(-/-)) have increased plasma PTH and FGF-23 levels compared with wild-type (WT) mice but comparable plasma phosphate concentrations. Acute activation of the calcium-sensing receptor or feeding a zero phosphate diet almost completely suppressed plasma PTH levels in both AC6(-/-) and WT mice, indicating a secondary cause for hyperparathyroidism. Pharmacologic blockade of FGF receptors resulted in a comparable increase in plasma phosphate between genotypes, whereas urinary phosphate remained significantly higher in AC6(-/-) mice. Compared with WT mice, AC6(-/-) mice had reduced renal Npt2a and Npt2c protein abundance, with approximately 80% of Npt2a residing in lysosomes. WT mice responded to exogenous PTH with redistribution of Npt2a from proximal tubule microvilli to intracellular compartments and lysosomes alongside a PTH-induced dose-response relationship for fractional phosphate excretion and urinary cAMP excretion. These responses were absent in AC6(-/-) mice. In conclusion, AC6 in the proximal tubule modulates cAMP formation, Npt2a trafficking, and urinary phosphate excretion, which are highlighted by renal phosphate wasting in AC6(-/-) mice.
Collapse
Affiliation(s)
- Robert A Fenton
- Interactions of Proteins in Epithelial Transport Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Fiona Murray
- Department of Medicine, University of California San Diego, La Jolla, California
| | | | - Tong Tang
- Veterans Affairs San Diego Healthcare System, San Diego, California; and
| | - Moshe Levi
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Timo Rieg
- Department of Medicine, University of California San Diego, La Jolla, California; Veterans Affairs San Diego Healthcare System, San Diego, California; and
| |
Collapse
|
43
|
Ramkumar N, Stuart D, Rees S, Hoek AV, Sigmund CD, Kohan DE. Collecting duct-specific knockout of renin attenuates angiotensin II-induced hypertension. Am J Physiol Renal Physiol 2014; 307:F931-8. [PMID: 25122048 DOI: 10.1152/ajprenal.00367.2014] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The physiological and pathophysiological significance of collecting duct (CD)-derived renin, particularly as it relates to blood pressure (BP) regulation, is unknown. To address this question, we generated CD-specific renin knockout (KO) mice and examined BP and renal salt and water excretion. Mice containing loxP-flanked exon 1 of the renin gene were crossed with mice transgenic for aquaporin-2-Cre recombinase to achieve CD-specific renin KO. Compared with controls, CD renin KO mice had 70% lower medullary renin mRNA and 90% lower renin mRNA in microdissected cortical CD. Urinary renin levels were significantly lower in KO mice (45% of control levels) while plasma renin concentration was significantly higher in KO mice (63% higher than controls) during normal-Na intake. While no observable differences were noted in BP between the two groups with varying Na intake, infusion of angiotensin II at 400 ng·kg(-1)·min(-1) resulted in an attenuated hypertensive response in the KO mice (mean arterial pressure 111 ± 4 mmHg in KO vs. 128 ± 3 mmHg in controls). Urinary renin excretion and epithelial Na(+) channel (ENaC) remained significantly lower in the KO mice following ANG II infusion compared with controls. Furthermore, membrane-associated ENaC protein levels were significantly lower in KO mice following ANG II infusion. These findings suggest that CD renin modulates BP in ANG II-infused hypertension and these effects are associated with changes in ENaC expression.
Collapse
Affiliation(s)
- Nirupama Ramkumar
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| | - Deborah Stuart
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| | - Sara Rees
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| | - Alfred Van Hoek
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| | - Curt D Sigmund
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Donald E Kohan
- Division of Nephrology and Hypertension, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| |
Collapse
|
44
|
Cano-Peñalver JL, Griera M, Serrano I, Rodríguez-Puyol D, Dedhar S, de Frutos S, Rodríguez-Puyol M. Integrin-linked kinase regulates tubular aquaporin-2 content and intracellular location: a link between the extracellular matrix and water reabsorption. FASEB J 2014; 28:3645-59. [PMID: 24784577 DOI: 10.1096/fj.13-249250] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
One of the clinical alterations observed in chronic renal disease (CRD) is the impaired urine concentration, known as diabetes insipidus (DI). Tubulointerstitial fibrosis of the kidney is also a pathological finding observed in CRD and involves composition of extracellular matrix (ECM). However, an association between these two events has not been elucidated. In this study, we showed that the extracellular-to-intracellular scaffold protein integrin-linked kinase (ILK) regulates expression of tubular water channel aquaporin-2 (AQP2) and its apical membrane presence in the renal tubule. Basally, polyuria and decreased urine osmolality were present in ILK conditional-knockdown (cKD-ILK) adult mice compared with nondepleted ILK littermates. No changes were observed in arginine-vasopressin (AVP) blood levels, renal receptor (V2R), or AQP3 expression. However, tubular AQP2 was decreased in expression and apical membrane presence in cKD-ILK mice, where the canonical V2R/cAMP axis activation is still functional, but independent of the absence of ILK. Thus, cKD-ILK constitutes a nephrogenic diabetes insipidus (NDI) model. AQP2 and ILK colocalize in cultured inner medullary collecting duct (mIMCD3) cells. Specific ILK siRNAs and collagen I (Col) decrease ILK and AQP2 levels and AQP2 presence on the membrane of tubular mIMCD3 cells, which impairs the capacity of the cells to transport water under hypotonic stress. The present work points to ILK as a therapeutic target in NDI.
Collapse
Affiliation(s)
- Jose Luis Cano-Peñalver
- Department of Systems Biology, Physiology Unit, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercedes Griera
- Department of Systems Biology, Physiology Unit, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Serrano
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, British Columbia, Canada; and
| | - Diego Rodríguez-Puyol
- Instituto Reina Sofia de Investigación Renal and Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, Madrid, Spain; Biomedical Research Foundation and Department of Nephrology, Hospital Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Shoukat Dedhar
- Department of Integrative Oncology, British Columbia Cancer Research Center, Vancouver, British Columbia, Canada; and
| | - Sergio de Frutos
- Department of Systems Biology, Physiology Unit, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, Madrid, Spain;
| | - Manuel Rodríguez-Puyol
- Department of Systems Biology, Physiology Unit, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain; Instituto Reina Sofia de Investigación Renal and Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
45
|
Kittikulsuth W, Stuart D, Kohan DE. Adenylyl cyclase 4 does not regulate collecting duct water and sodium handling. Physiol Rep 2014; 2:e00277. [PMID: 24760529 PMCID: PMC4002255 DOI: 10.1002/phy2.277] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Adenylyl cyclase (AC)‐stimulated cAMP is a key mediator of collecting duct (CD) Na and water transport. AC isoforms 3, 4, and 6 are expressed in the CD. Our group demonstrated that AC6, but not AC3, is involved in regulating CD Na and water transport. However, the role of AC4 in such regulation remains unknown. Therefore, we generated mice with loxP‐flanked critical exons in the Adcy4 gene and bred with mice expressing the aquaporin‐2/Cre recombinase transgene to yield CD principal cell‐specific knockout of AC4 (CD AC4 KO). Isolated inner medullary CD showed 100% genomic target gene recombination in CD AC4 KO mice, while microdissected cortical CD and renal papillary AC4 mRNA was significantly reduced in CD AC4 KO mice. CD AC4 KO had no effect on vasopressin (AVP)‐stimulated cAMP generation in the inner medulla. Water intake, urine volume, and urine osmolality were similar between CD AC4 KO and control mice during normal or restricted water intake. Sodium intake, urinary Na excretion, and blood pressure on a normal‐, high‐, or low‐Na diet were not affected by CD AC4 KO. Moreover, there were no differences in plasma AVP or plasma renin concentration between CD AC4 KO and control mice. In summary, these data suggest that CD AC4 does not play a role in the physiologic regulation of CD Na and water handling. Principal cells in the collecting duct express adenylyl cyclase 4 (AC4), however, the role of AC4 in the regulation of collecting duct function is unknown. We made mice with collecting duct principal cell‐specific deletion of AC4 and found that these mice have no alterations in arterial pressure or urinary sodium, potassium, or water excretion under varying physiological conditions.
Collapse
Affiliation(s)
- Wararat Kittikulsuth
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City Veterans Affairs Medical Center, Salt Lake City, Utah
| | | | | |
Collapse
|
46
|
Rees S, Kittikulsuth W, Roos K, Strait KA, Van Hoek A, Kohan DE. Adenylyl cyclase 6 deficiency ameliorates polycystic kidney disease. J Am Soc Nephrol 2014; 25:232-7. [PMID: 24158982 PMCID: PMC3904559 DOI: 10.1681/asn.2013010077] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 08/01/2013] [Indexed: 12/13/2022] Open
Abstract
cAMP is an important mediator of cystogenesis in polycystic kidney disease (PKD). Several adenylyl cyclase (AC) isoforms could mediate cAMP accumulation in PKD, and identification of a specific pathogenic AC isoform is of therapeutic interest. We investigated the role of AC6 in a mouse model of PKD that is homozygous for the loxP-flanked PKD1 gene and heterozygous for an aquaporin-2-Cre recombinase transgene to achieve collecting duct-specific gene targeting. Collecting duct-specific knockout of polycystin-1 caused massive renal cyst formation, kidney enlargement, and severe kidney failure, with a mean survival time of 2 months. In contrast, coincident collecting duct-specific knockout of polycystin-1 and AC6 (also homozygous for the floxed ADCY6 gene) markedly decreased kidney size and cystogenesis, improved renal function, reduced activation of the B-Raf/ERK/MEK pathway, and greatly increased survival. Absence of collecting duct AC6 did not alter urinary cAMP excretion or kidney cAMP concentration. In conclusion, AC6 is a key mediator of cyst formation and renal injury in a model of PKD.
Collapse
MESH Headings
- Adenylyl Cyclases/deficiency
- Adenylyl Cyclases/genetics
- Adenylyl Cyclases/physiology
- Animals
- Aquaporin 2/genetics
- Cyclic AMP/physiology
- Disease Models, Animal
- Female
- Genotype
- Integrases/genetics
- Kidney/pathology
- Kidney Failure, Chronic/etiology
- Kidney Failure, Chronic/prevention & control
- Kidney Tubules, Collecting/physiopathology
- MAP Kinase Signaling System
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Polycystic Kidney, Autosomal Dominant/enzymology
- Polycystic Kidney, Autosomal Dominant/genetics
- Polycystic Kidney, Autosomal Dominant/metabolism
- Polycystic Kidney, Autosomal Dominant/pathology
- TRPP Cation Channels/deficiency
- TRPP Cation Channels/genetics
- Transgenes
Collapse
Affiliation(s)
- Sara Rees
- Division of Nephrology, University of Utah Health Sciences Center and Salt Lake City Veterans Affairs Medical Center, Salt Lake City, Utah
| | | | | | | | | | | |
Collapse
|
47
|
Rieg T, Kohan DE. Regulation of nephron water and electrolyte transport by adenylyl cyclases. Am J Physiol Renal Physiol 2014; 306:F701-9. [PMID: 24477683 DOI: 10.1152/ajprenal.00656.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adenylyl cyclases (AC) catalyze formation of cAMP, a critical component of G protein-coupled receptor signaling. So far, nine distinct membrane-bound AC isoforms (AC1-9) and one soluble AC (sAC) have been identified and, except for AC8, all of them are expressed in the kidney. While the role of ACs in renal cAMP formation is well established, we are just beginning to understand the function of individual AC isoforms, particularly with regard to hormonal regulation of transporter and channel phosphorylation, membrane abundance, and trafficking. This review focuses on the role of different AC isoforms in regulating renal water and electrolyte transport in health as well as potential pathological implications of disordered AC isoform function. In particular, we focus on modulation of transporter and channel abundance, activity, and phosphorylation, with an emphasis on studies employing genetically modified animals. As will be described, it is now evident that specific AC isoforms can exert unique effects in the kidney that may have important implications in our understanding of normal physiology as well as disease pathogenesis.
Collapse
Affiliation(s)
- Timo Rieg
- Dept. of Medicine, Div. of Nephrology/Hypertension, Univ. of California San Diego and VA San Diego Healthcare System; 3350 La Jolla Village Dr. (9151 San Diego, CA 92161.
| | | |
Collapse
|
48
|
Kittikulsuth W, Stuart D, Van Hoek AN, Stockand JD, Bugaj V, Mironova E, Blount MA, Kohan DE. Lack of an effect of collecting duct-specific deletion of adenylyl cyclase 3 on renal Na+ and water excretion or arterial pressure. Am J Physiol Renal Physiol 2014; 306:F597-607. [PMID: 24431204 DOI: 10.1152/ajprenal.00505.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
cAMP is a key mediator of connecting tubule and collecting duct (CD) Na(+) and water reabsorption. Studies performed in vitro have suggested that CD adenylyl cyclase (AC)3 partly mediates the actions of vasopressin; however, the physiological role of CD AC3 has not been determined. To assess this, mice were developed with CD-specific disruption of AC3 [CD AC3 knockout (KO)]. Inner medullary CDs from these mice exhibited 100% target gene recombination and had reduced ANG II- but not vasopressin-induced cAMP accumulation. However, there were no differences in urine volume, urinary urea excretion, or urine osmolality between KO and control mice during normal water intake or varying degrees of water restriction in the presence or absence of chronic vasopressin administration. There were no differences between CD AC3 KO and control mice in arterial pressure or urinary Na(+) or K(+) excretion during a normal or high-salt diet, whereas plasma renin and vasopressin concentrations were similar between the two genotypes. Patch-clamp analysis of split-open cortical CDs revealed no difference in epithelial Na(+) channel activity in the presence or absence of vasopressin. Compensatory changes in AC6 were not responsible for the lack of a renal phenotype in CD AC3 KO mice since combined CD AC3/AC6 KO mice had similar arterial pressure and renal Na(+) and water handling compared with CD AC6 KO mice. In summary, these data do not support a significant role for CD AC3 in the regulation of renal Na(+) and water excretion in general or vasopressin regulation of CD function in particular.
Collapse
Affiliation(s)
- Wararat Kittikulsuth
- Div. of Nephrology, Univ. of Utah Health Sciences Center, 1900 East 30 North, Salt Lake City, UT 84132.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kortenoeven MLA, Sinke AP, Hadrup N, Trimpert C, Wetzels JFM, Fenton RA, Deen PMT. Demeclocycline attenuates hyponatremia by reducing aquaporin-2 expression in the renal inner medulla. Am J Physiol Renal Physiol 2013; 305:F1705-18. [PMID: 24154696 DOI: 10.1152/ajprenal.00723.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Binding of vasopressin to its type 2 receptor in renal collecting ducts induces cAMP signaling, transcription and translocation of aquaporin (AQP)2 water channels to the plasma membrane, and water reabsorption from the prourine. Demeclocycline is currently used to treat hyponatremia in patients with the syndrome of inappropriate antidiuretic hormone secretion (SIADH). Demeclocycline's mechanism of action, which is poorly understood, is studied here. In mouse cortical collecting duct (mpkCCD) cells, which exhibit deamino-8-D-arginine vasopressin (dDAVP)-dependent expression of endogenous AQP2, demeclocycline decreased AQP2 abundance and gene transcription but not its protein stability. Demeclocycline did not affect vasopressin type 2 receptor localization but decreased dDAVP-induced cAMP generation and the abundance of adenylate cyclase 3 and 5/6. The addition of exogenous cAMP partially corrected the demeclocycline effect. As in patients, demeclocycline increased urine volume, decreased urine osmolality, and reverted hyponatremia in an SIADH rat model. AQP2 and adenylate cyclase 5/6 abundances were reduced in the inner medulla but increased in the cortex and outer medulla, in the absence of any sign of toxicity. In conclusion, our in vitro and in vivo data indicate that demeclocycline mainly attenuates hyponatremia in SIADH by reducing adenylate cyclase 5/6 expression and, consequently, cAMP generation, AQP2 gene transcription, and AQP2 abundance in the renal inner medulla, coinciding with a reduced vasopressin escape response in other collecting duct segments.
Collapse
Affiliation(s)
- Marleen L A Kortenoeven
- no. 286, Dept. of Physiology, Radboud Univ. Medical Centre, PO Box 9101, Nijmegen 6500 HB, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
50
|
Roos KP, Bugaj V, Mironova E, Stockand JD, Ramkumar N, Rees S, Kohan DE. Adenylyl cyclase VI mediates vasopressin-stimulated ENaC activity. J Am Soc Nephrol 2013; 24:218-27. [PMID: 23264685 PMCID: PMC3559481 DOI: 10.1681/asn.2012050449] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Accepted: 10/22/2012] [Indexed: 12/11/2022] Open
Abstract
Vasopressin modulates sodium reabsorption in the collecting duct through adenylyl cyclase-stimulated cyclic AMP, which exists as multiple isoforms; the specific isoform involved in vasopressin-stimulated sodium transport is unknown. To assess this, we studied mice deficient in adenylyl cyclase type VI specifically in the principal cells of the collecting duct. Knockout mice had increased urine volume and reduced urine sodium concentration, but regardless of the level of sodium intake, they did not exhibit significant alterations in urinary sodium excretion, arterial pressure, or pulse rate. Plasma renin concentration was elevated in knockout mice, however, suggesting a compensatory response. Valsartan significantly reduced arterial pressure in knockout mice but not in controls. Knockout mice had decreased renal cortical mRNA content of all three epithelial sodium channel (ENaC) isoforms, and total cell sodium channel isoforms α and γ were reduced in these animals. Patch-clamp analysis of split-open cortical collecting ducts revealed no difference in baseline activity of sodium channels, but knockout mice had abolished vasopressin-stimulated ENaC open probability and apical membrane channel number. In summary, these data suggest that adenylyl cyclase VI mediates vasopressin-stimulated ENaC activity in the kidney.
Collapse
Affiliation(s)
- Karl P. Roos
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| | - Vladislav Bugaj
- Department of Physiology, University of Texas Health Sciences Center, San Antonio, Texas
| | - Elena Mironova
- Department of Physiology, University of Texas Health Sciences Center, San Antonio, Texas
| | - James D. Stockand
- Department of Physiology, University of Texas Health Sciences Center, San Antonio, Texas
| | - Nirupama Ramkumar
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| | - Sara Rees
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| | - Donald E. Kohan
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah; and
| |
Collapse
|