1
|
Kraus A, Skoczynski K, Brötsch M, Burzlaff N, Leipziger J, Schiffer M, Büttner-Herold M, Buchholz B. P2Y2R and Cyst Growth in Polycystic Kidney Disease. J Am Soc Nephrol 2024; 35:1351-1365. [PMID: 38848134 PMCID: PMC11452133 DOI: 10.1681/asn.0000000000000416] [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: 11/24/2023] [Accepted: 05/30/2024] [Indexed: 06/09/2024] Open
Abstract
Key Points Polycystic kidney disease (PKD) is characterized by continuous cyst growth, which results in a decline in kidney function. Deletion of P2Y2R and pharmacological antagonism of purinergic signaling significantly reduced cyst growth in an orthologous PKD mouse model. P2Y2R was expressed in cysts of human PKD nephrectomies, which makes P2Y2R a reasonable target for treatment of PKD. Background Autosomal dominant polycystic kidney disease (ADPKD) is characterized by multiple bilateral kidney cysts that gradually enlarge, resulting in a decline in kidney function. Cyst growth is significantly driven by ATP-dependent chloride secretion mediated by the ion channel TMEM16A. This pathway is further augmented in advanced stages of the disease by hypoxia and activation of hypoxia-inducible factor (HIF)-1α . The mechanisms by which ATP leads to activation of TMEM16A and how HIF-1α contributes to cyst growth in vivo have remained elusive. Methods Mice with an inducible tubule-specific deletion of Pkd1 were compared with mice with an additional codeletion of the purinergic receptor P2y2r . Furthermore, animals were challenged by pharmacological activation of HIF-1α and Pkd1 -deficient mice were treated with suramin, an antagonist of purinergic signaling. In addition, expression of P2Y2R, TMEM16A, and HIF-1α was analyzed in nephrectomy samples from 27 patients with ADPKD. Results Genetic deletion of P2y2r significantly inhibited cyst growth in vivo . In addition, aggravation of the polycystic phenotype mediated by pharmacological activation of HIF-1α was reduced by deletion of P2y2r . Application of suramin to pharmacologically inhibit purinergic signaling also suppressed cyst enlargement in vivo . Analysis of kidney samples from 27 patients with ADPKD revealed significant expression of P2Y2R at the luminal site of the cyst-lining epithelium. Conclusions P2Y2R was significantly expressed in human and mouse polycystic kidneys. Deletion and antagonism of P2Y2R reduced cyst enlargement in an ADPKD mouse model.
Collapse
Affiliation(s)
- Andre Kraus
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and University Hospital, Erlangen, Germany
| | - Kathrin Skoczynski
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and University Hospital, Erlangen, Germany
| | - Martin Brötsch
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Nicolai Burzlaff
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jens Leipziger
- Department of Biomedicine, Physiology, Aarhus University, Aarhus, Denmark
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and University Hospital, Erlangen, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and University Hospital, Erlangen, Germany
| | - Bjoern Buchholz
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and University Hospital, Erlangen, Germany
| |
Collapse
|
2
|
van Megen WH, Canki E, Wagenaar VHA, van Waes CRMM, Peters DJM, Van Asbeck-Van der Wijst J, Hoenderop JGJ. Fluid shear stress stimulates ATP release without regulating purinergic gene expression in the renal inner medullary collecting duct. FASEB J 2023; 37:e23232. [PMID: 37819258 DOI: 10.1096/fj.202301434r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
In the kidney, the flow rate of the pro-urine through the renal tubules is highly variable. The tubular epithelial cells sense these variations in pro-urinary flow rate in order to regulate various physiological processes, including electrolyte reabsorption. One of the mechanosensitive pathways activated by flow is the release of ATP, which can then act as a autocrine or paracrine factor. Increased ATP release is observed in various kidney diseases, among others autosomal dominant polycystic kidney disease (ADPKD). However, the mechanisms underlying flow-induced ATP release in the collecting duct, especially in the inner medullary collecting duct, remain understudied. Using inner medullary collecting duct 3 (IMCD3) cells in a microfluidic setup, we show here that administration of a high flow rate for 1 min results in an increased ATP release compared to a lower flow rate. Although the ATP release channel pannexin-1 contributed to flow-induced ATP release in Pkd1-/- IMCD3 cells, it did not in wildtype IMCD3 cells. In addition, flow application increased the expression of the putative ATP release channel connexin-30.3 (CX30.3) in wildtype and Pkd1-/- IMCD3 cells. However, CX30.3 knockout IMCD3 cells exhibited a similar flow-induced ATP release as wildtype IMCD3 cells, suggesting that CX30.3 does not drive flow-induced ATP release in wildtype IMDC3 cells. Collectively, our results show differential mechanisms underlying flow-induced ATP release in wildtype and Pkd1-/- IMCD3 cells and further strengthen the link between ADPKD and pannexin-1-dependent ATP release.
Collapse
Affiliation(s)
- Wouter H van Megen
- Department of Medical Biosciences, Radboudumc, Nijmegen, The Netherlands
| | - Esra Canki
- Department of Medical Biosciences, Radboudumc, Nijmegen, The Netherlands
| | - Vera H A Wagenaar
- Department of Medical Biosciences, Radboudumc, Nijmegen, The Netherlands
| | | | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
3
|
Kulthinee S, Tasanarong A, Franco M, Navar LG. Interaction of Angiotensin II AT1 Receptors with Purinergic P2X Receptors in Regulating Renal Afferent Arterioles in Angiotensin II-Dependent Hypertension. Int J Mol Sci 2023; 24:11413. [PMID: 37511174 PMCID: PMC10380633 DOI: 10.3390/ijms241411413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
In angiotensin II (Ang II)-dependent hypertension, Ang II activates angiotensin II type 1 receptors (AT1R) on renal vascular smooth muscle cells, leading to renal vasoconstriction with eventual glomerular and tubular injury and interstitial inflammation. While afferent arteriolar vasoconstriction is initiated by the increased intrarenal levels of Ang II activating AT1R, the progressive increases in arterial pressure stimulate the paracrine secretion of adenosine triphosphate (ATP), leading to the purinergic P2X receptor (P2XR)-mediated constriction of afferent arterioles. Thus, the afferent arteriolar tone is maintained by two powerful systems eliciting the co-existing activation of P2XR and AT1R. This raises the conundrum of how the AT1R and P2XR can both be responsible for most of the increased renal afferent vascular resistance existing in angiotensin-dependent hypertension. Its resolution implies that AT1R and P2XR share common receptor or post receptor signaling mechanisms which converge to maintain renal vasoconstriction in Ang II-dependent hypertension. In this review, we briefly discuss (1) the regulation of renal afferent arterioles in Ang II-dependent hypertension, (2) the interaction of AT1R and P2XR activation in regulating renal afferent arterioles in a setting of hypertension, (3) mechanisms regulating ATP release and effect of angiotensin II on ATP release, and (4) the possible intracellular pathways involved in AT1R and P2XR interactions. Emerging evidence supports the hypothesis that P2X1R, P2X7R, and AT1R actions converge at receptor or post-receptor signaling pathways but that P2XR exerts a dominant influence abrogating the actions of AT1R on renal afferent arterioles in Ang II-dependent hypertension. This finding raises clinical implications for the design of therapeutic interventions that will prevent the impairment of kidney function and subsequent tissue injury.
Collapse
Affiliation(s)
- Supaporn Kulthinee
- Department of Physiology, Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Adis Tasanarong
- Chulabhorn International College of Medicine, Thammasat University, Klong Luang 12120, Thailand
| | - Martha Franco
- Department of Cardio-Renal Physiopathology, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| | - Luis Gabriel Navar
- Department of Physiology, Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
4
|
Zhou JX, Torres VE. Autosomal Dominant Polycystic Kidney Disease Therapies on the Horizon. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:245-260. [PMID: 37088527 DOI: 10.1053/j.akdh.2023.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/21/2022] [Accepted: 01/06/2023] [Indexed: 04/25/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the formation of numerous kidney cysts which leads to kidney failure. ADPKD is responsible for approximately 10% of patients with kidney failure. Overwhelming evidence supports that vasopressin and its downstream cyclic adenosine monophosphate signaling promote cystogenesis, and targeting vasopressin 2 receptor with tolvaptan and other antagonists ameliorates cyst growth in preclinical studies. Tolvaptan is the only drug approved by Food and Drug Administration to treat ADPKD patients at the risk of rapid disease progression. A major limitation of the widespread use of tolvaptan is aquaretic events. This review discusses the potential strategies to improve the tolerability of tolvaptan, the progress on the use of an alternative vasopressin 2 receptor antagonist lixivaptan, and somatostatin analogs. Recent advances in understanding the pathophysiology of PKD have led to new approaches of treatment via targeting different signaling pathways. We review the new pharmacotherapies and dietary interventions of ADPKD that are promising in the preclinical studies and investigated in clinical trials.
Collapse
|
5
|
Shihan M, Novoyatleva T, Lehmeyer T, Sydykov A, Schermuly RT. Role of the Purinergic P2Y2 Receptor in Pulmonary Hypertension. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182111009. [PMID: 34769531 PMCID: PMC8582672 DOI: 10.3390/ijerph182111009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 11/29/2022]
Abstract
Pulmonary arterial hypertension (PAH), group 1 pulmonary hypertension (PH), is a fatal disease that is characterized by vasoconstriction, increased pressure in the pulmonary arteries, and right heart failure. PAH can be described by abnormal vascular remodeling, hyperproliferation in the vasculature, endothelial cell dysfunction, and vascular tone dysregulation. The disease pathomechanisms, however, are as yet not fully understood at the molecular level. Purinergic receptors P2Y within the G-protein-coupled receptor family play a major role in fluid shear stress transduction, proliferation, migration, and vascular tone regulation in systemic circulation, but less is known about their contribution in PAH. Hence, studies that focus on purinergic signaling are of great importance for the identification of new therapeutic targets in PAH. Interestingly, the role of P2Y2 receptors has not yet been sufficiently studied in PAH, whereas the relevance of other P2Ys as drug targets for PAH was shown using specific agonists or antagonists. In this review, we will shed light on P2Y receptors and focus more on the P2Y2 receptor as a potential novel player in PAH and as a new therapeutic target for disease management.
Collapse
|
6
|
Praetorius H. The bacteria and the host: a story of purinergic signaling in urinary tract infections. Am J Physiol Cell Physiol 2021; 321:C134-C146. [PMID: 33979212 DOI: 10.1152/ajpcell.00054.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The local environment forces a selection of bacteria that might invade the urinary tract, allowing only the most virulent to access the kidney. Quite similar to the diet in setting the stage for the gut microbiome, renal function determines the conditions for bacteria-host interaction in the urinary tract. In the kidney, the term local environment or microenvironment is completely justified because the environment literally changes within a few micrometers. The precise composition of the urine is a function of the epithelium lining the microdomain, and the microenvironment in the kidney shows more variation in the content of nutrients, ion composition, osmolality, and pH than any other site of bacteria-host interaction. This review will cover some of the aspects of bacterial-host interaction in this unique setting and how uropathogenic bacteria can alter the condition for bacteria-host interaction. There will be a particular focus on the recent findings regarding how bacteria specifically trigger host paracrine signaling, via release of extracellular ATP and activation of P2 purinergic receptors. These finding will be discussed from the perspective of severe urinary tract infections, including pyelonephritis and urosepsis.
Collapse
|
7
|
Himmel NJ, Rogers RT, Redd SK, Wang Y, Blount MA. Purinergic signaling is enhanced in the absence of UT-A1 and UT-A3. Physiol Rep 2021; 9:e14636. [PMID: 33369887 PMCID: PMC7769175 DOI: 10.14814/phy2.14636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 11/24/2022] Open
Abstract
ATP is an important paracrine regulator of renal tubular water and urea transport. The activity of P2Y2 , the predominant P2Y receptor of the medullary collecting duct, is mediated by ATP, and modulates urinary concentration. To investigate the role of purinergic signaling in the absence of urea transport in the collecting duct, we studied wild-type (WT) and UT-A1/A3 null (UT-A1/A3 KO) mice in metabolic cages to monitor urine output, and collected tissue samples for analysis. We confirmed that UT-A1/A3 KO mice are polyuric, and concurrently observed lower levels of urinary cAMP as compared to WT, despite elevated serum vasopressin (AVP) levels. Because P2Y2 inhibits AVP-stimulated transport by dampening cAMP synthesis, we suspected that, similar to other models of AVP-resistant polyuria, purinergic signaling is increased in UT-A1/A3 KO mice. In fact, we observed that both urinary ATP and purinergic-mediated prostanoid (PGE2 ) levels were elevated. Collectively, our data suggest that the reduction of medullary osmolality due to the lack of UT-A1 and UT-A3 induces an AVP-resistant polyuria that is possibly exacerbated by, or at least correlated with, enhanced purinergic signaling.
Collapse
Affiliation(s)
- Nathaniel J. Himmel
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Richard T. Rogers
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Sara K. Redd
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Yirong Wang
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Mitsi A. Blount
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
- Department of PhysiologyEmory University School of MedicineAtlantaGAUSA
| |
Collapse
|
8
|
Dalghi MG, Montalbetti N, Carattino MD, Apodaca G. The Urothelium: Life in a Liquid Environment. Physiol Rev 2020; 100:1621-1705. [PMID: 32191559 PMCID: PMC7717127 DOI: 10.1152/physrev.00041.2019] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/02/2020] [Accepted: 03/14/2020] [Indexed: 02/08/2023] Open
Abstract
The urothelium, which lines the renal pelvis, ureters, urinary bladder, and proximal urethra, forms a high-resistance but adaptable barrier that surveils its mechanochemical environment and communicates changes to underlying tissues including afferent nerve fibers and the smooth muscle. The goal of this review is to summarize new insights into urothelial biology and function that have occurred in the past decade. After familiarizing the reader with key aspects of urothelial histology, we describe new insights into urothelial development and regeneration. This is followed by an extended discussion of urothelial barrier function, including information about the roles of the glycocalyx, ion and water transport, tight junctions, and the cellular and tissue shape changes and other adaptations that accompany expansion and contraction of the lower urinary tract. We also explore evidence that the urothelium can alter the water and solute composition of urine during normal physiology and in response to overdistension. We complete the review by providing an overview of our current knowledge about the urothelial environment, discussing the sensor and transducer functions of the urothelium, exploring the role of circadian rhythms in urothelial gene expression, and describing novel research tools that are likely to further advance our understanding of urothelial biology.
Collapse
Affiliation(s)
- Marianela G Dalghi
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nicolas Montalbetti
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
9
|
Conversion of extracellular ATP into adenosine: a master switch in renal health and disease. Nat Rev Nephrol 2020; 16:509-524. [PMID: 32641760 DOI: 10.1038/s41581-020-0304-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2020] [Indexed: 12/22/2022]
Abstract
ATP and its ultimate degradation product adenosine are potent extracellular signalling molecules that elicit a variety of pathophysiological functions in the kidney through the activation of P2 and P1 purinergic receptors, respectively. Extracellular purines can modulate immune responses, balancing inflammatory processes and immunosuppression; indeed, alterations in extracellular nucleotide and adenosine signalling determine outcomes of inflammation and healing processes. The functional activities of ectonucleotidases such as CD39 and CD73, which hydrolyse pro-inflammatory ATP to generate immunosuppressive adenosine, are therefore pivotal in acute inflammation. Protracted inflammation may result in aberrant adenosinergic signalling, which serves to sustain inflammasome activation and worsen fibrotic reactions. Alterations in the expression of ectonucleotidases on various immune cells, such as regulatory T cells and macrophages, as well as components of the renal vasculature, control purinergic receptor-mediated effects on target tissues within the kidney. The role of CD39 as a rheostat that can have an impact on purinergic signalling in both acute and chronic inflammation is increasingly supported by the literature, as detailed in this Review. Better understanding of these purinergic processes and development of novel drugs targeting these pathways could lead to effective therapies for the management of acute and chronic kidney disease.
Collapse
|
10
|
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
|
11
|
Zaika O, Tomilin VN, Pochynyuk O. Adenosine inhibits the basolateral Cl - ClC-K2/b channel in collecting duct intercalated cells. Am J Physiol Renal Physiol 2020; 318:F870-F877. [PMID: 31984792 DOI: 10.1152/ajprenal.00572.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adenosine plays an important role in various aspects of kidney physiology, but the specific targets and mechanisms of actions are not completely understood. The collecting duct has the highest expression of adenosine receptors, particularly adenosine A1 receptors (A1Rs). Interstitial adenosine levels are greatly increased up to a micromolar range in response to dietary salt loading. We have previously shown that the basolateral membrane of principal cells has primarily K+ conductance mediated by Kir4.1/5.1 channels to mediate K+ recycling and to set up a favorable driving force for Na+/K+ exchange (47). Intercalated cells express the Cl- ClC-K2/b channel mediating transcellular Cl- reabsorption. Using patch-clamp electrophysiology in freshly isolated mouse collecting ducts, we found that acute application of adenosine reversely inhibits ClC-K2/b open probability from 0.31 ± 0.04 to 0.17 ± 0.06 and to 0.10 ± 0.05 for 1 and 10 µM, respectively. In contrast, adenosine (10 µM) had no measureable effect on Kir4.1/5.1 channel activity in principal cells. The inhibitory effect of adenosine on ClC-K2/b was abolished in the presence of the A1R blocker 8-cyclopentyl-1,3-dipropylxanthine (10 µM). Consistently, application of the A1R agonist N6-cyclohexyladenosine (1 µM) recapitulated the inhibitory action of adenosine on ClC-K2/b open probability. The effects of adenosine signaling in the collecting duct were independent from its purinergic counterpartner, ATP, having no measurable actions on ClC-K2/b and Kir4.1/5.1. Overall, we demonstrated that adenosine selectively inhibits ClC-K2/b activity in intercalated cells by targeting A1Rs. We propose that inhibition of transcellular Cl- reabsorption in the collecting duct by adenosine would aid in augmenting NaCl excretion during high salt intake.
Collapse
Affiliation(s)
- Oleg Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Viktor N Tomilin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
12
|
Leipziger J, Praetorius H. Renal Autocrine and Paracrine Signaling: A Story of Self-protection. Physiol Rev 2020; 100:1229-1289. [PMID: 31999508 DOI: 10.1152/physrev.00014.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Autocrine and paracrine signaling in the kidney adds an extra level of diversity and complexity to renal physiology. The extensive scientific production on the topic precludes easy understanding of the fundamental purpose of the vast number of molecules and systems that influence the renal function. This systematic review provides the broader pen strokes for a collected image of renal paracrine signaling. First, we recapitulate the essence of each paracrine system one by one. Thereafter the single components are merged into an overarching physiological concept. The presented survey shows that despite the diversity in the web of paracrine factors, the collected effect on renal function may not be complicated after all. In essence, paracrine activation provides an intelligent system that perceives minor perturbations and reacts with a coordinated and integrated tissue response that relieves the work load from the renal epithelia and favors diuresis and natriuresis. We suggest that the overall function of paracrine signaling is reno-protection and argue that renal paracrine signaling and self-regulation are two sides of the same coin. Thus local paracrine signaling is an intrinsic function of the kidney, and the overall renal effect of changes in blood pressure, volume load, and systemic hormones will always be tinted by its paracrine status.
Collapse
Affiliation(s)
- Jens Leipziger
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; and Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| | - Helle Praetorius
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; and Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| |
Collapse
|
13
|
Zhang Y, Ecelbarger CM, Lesniewski LA, Müller CE, Kishore BK. P2Y 2 Receptor Promotes High-Fat Diet-Induced Obesity. Front Endocrinol (Lausanne) 2020; 11:341. [PMID: 32582029 PMCID: PMC7283874 DOI: 10.3389/fendo.2020.00341] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022] Open
Abstract
P2Y2, a G protein-coupled receptor (R), is expressed in all organs involved in the development of obesity and insulin resistance. To explore the role of it in diet-induced obesity, we fed male P2Y2-R whole body knockout (KO) and wild type (WT) mice (B6D2 genetic background) with regular diet (CNT; 10% calories as fat) or high-fat diet (HFD; 60% calories as fat) with free access to food and water for 16 weeks, and euthanized them. Adjusted for body weights (BW), KO mice consumed modestly, but significantly more HFD vs. WT mice, and excreted well-formed feces with no taint of fat or oil. Starting from the 2nd week, HFD-WT mice displayed significantly higher BW with terminal mean difference of 22% vs. HFD-KO mice. Terminal weights of white adipose tissue (WAT) were significantly lower in the HFD-KO vs. HFD-WT mice. The expression of P2Y2-R mRNA in WAT was increased by 2-fold in HFD-fed WT mice. Serum insulin, leptin and adiponectin levels were significantly elevated in the HFD-WT mice, but not in the HFD-KO mice. When induced in vitro, preadipocytes derived from KO mice fed regular diet did not differentiate and mature as robustly as those from the WT mice, as assessed by cellular expansion and accumulation of lipid droplets. Blockade of P2Y2-R by AR-C118925 in preadipocytes derived from WT mice prevented differentiation and maturation. Under basal conditions, KO mice had significantly higher serum triglycerides and showed slightly impaired lipid tolerance as compared to the WT mice. HFD-fed KO mice had significantly better glucose tolerance (GTT) as compared to HFD-fed WT mice. Whole body insulin sensitivity and mRNA expression of insulin receptor, IRS-1 and GLUT4 in WAT was significantly higher in HFD-fed KO mice vs. HFD-fed WT mice. On the contrary, the expression of pro-inflammatory molecules MCP-1, CCR2, CD68, and F4/80 were significantly higher in the WAT of HFD-fed WT vs. HFD-fed KO mice. These data suggest that P2Y2-R plays a significant role in the development of diet-induced obesity by promoting adipogenesis and inflammation, and altering the production of adipokines and lipids and their metabolism in adipose tissue, and thereby facilitates HFD-induced insulin resistance.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Veterans Affairs Salt Lake City Health Care System, Nephrology Research, Salt Lake City, UT, United States
- Departments of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Carolyn M. Ecelbarger
- Division of Endocrinology and Metabolism, Department of Medicine, Center for the Study of Sex Differences in Health, Aging, and Disease, Georgetown University, Washington, DC, United States
| | - Lisa A. Lesniewski
- Departments of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
- Department of Veterans Affairs Salt Lake City Health Care System, Geriatric Research, Education and Clinical Center, Salt Lake City, UT, United States
- Department of Nutrition and Integrative Physiology, University of Utah Health, Salt Lake City, UT, United States
- Center on Aging, University of Utah Health, Salt Lake City, UT, United States
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Bellamkonda K. Kishore
- Department of Veterans Affairs Salt Lake City Health Care System, Nephrology Research, Salt Lake City, UT, United States
- Departments of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
- Department of Nutrition and Integrative Physiology, University of Utah Health, Salt Lake City, UT, United States
- Center on Aging, University of Utah Health, Salt Lake City, UT, United States
- *Correspondence: Bellamkonda K. Kishore
| |
Collapse
|
14
|
Vallon V, Unwin R, Inscho EW, Leipziger J, Kishore BK. Extracellular Nucleotides and P2 Receptors in Renal Function. Physiol Rev 2019; 100:211-269. [PMID: 31437091 DOI: 10.1152/physrev.00038.2018] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The understanding of the nucleotide/P2 receptor system in the regulation of renal hemodynamics and transport function has grown exponentially over the last 20 yr. This review attempts to integrate the available data while also identifying areas of missing information. First, the determinants of nucleotide concentrations in the interstitial and tubular fluids of the kidney are described, including mechanisms of cellular release of nucleotides and their extracellular breakdown. Then the renal cell membrane expression of P2X and P2Y receptors is discussed in the context of their effects on renal vascular and tubular functions. Attention is paid to effects on the cortical vasculature and intraglomerular structures, autoregulation of renal blood flow, tubuloglomerular feedback, and the control of medullary blood flow. The role of the nucleotide/P2 receptor system in the autocrine/paracrine regulation of sodium and fluid transport in the tubular and collecting duct system is outlined together with its role in integrative sodium and fluid homeostasis and blood pressure control. The final section summarizes the rapidly growing evidence indicating a prominent role of the extracellular nucleotide/P2 receptor system in the pathophysiology of the kidney and aims to identify potential therapeutic opportunities, including hypertension, lithium-induced nephropathy, polycystic kidney disease, and kidney inflammation. We are only beginning to unravel the distinct physiological and pathophysiological influences of the extracellular nucleotide/P2 receptor system and the associated therapeutic perspectives.
Collapse
Affiliation(s)
- Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Robert Unwin
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Edward W Inscho
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Jens Leipziger
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Bellamkonda K Kishore
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| |
Collapse
|
15
|
Zhang Y, Hansson KM, Liu T, Magnell K, Huang Y, Carlson NG, Kishore BK. Genetic deletion of ADP-activated P2Y 12 receptor ameliorates lithium-induced nephrogenic diabetes insipidus in mice. Acta Physiol (Oxf) 2019; 225:e13191. [PMID: 30257062 DOI: 10.1111/apha.13191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/19/2018] [Accepted: 09/19/2018] [Indexed: 12/31/2022]
Abstract
AIM Therapeutic use of lithium in bipolar disorder is limited by the development of nephrogenic diabetes insipidus (NDI). We reported that pharmacological blockade of P2Y12 receptor (R) with clopidogrel or prasugrel significantly ameliorated lithium-induced NDI in rodents. Using mice genetically lacking P2Y12 -R we evaluated whether the observed amelioration is mediated through P2Y12 -R METHODS: P2ry12-/- mouse line (C57/BL6) was rederived from cryopreserved embryos of the knockout (KO) mice generated by Deltagen Inc. Syngeneic wild type (WT) mice obtained by heterozygous crossing were inbred. Groups of adult WT and KO mice were fed lithium-added (40 mmol LiCl/kg food) or regular diet, and euthanized after 2 or 4 weeks. Twenty-four hour urine samples and terminal blood and kidney samples were analyzed. RESULTS At both time points, lithium-induced polyuria and decrease in aquaporin-2 (AQP2) protein abundance in the kidney medulla were less marked in KO vs WT mice. Immunofluorescence microscopy revealed that lithium-induced alterations in the cellular disposition of AQP2 protein in the medullary collecting ducts of WT mice were blunted in KO mice. Serum lithium, sodium and osmolality were similar in both genotypes after lithium treatment. After 2 weeks, lithium induced marked increases in urinary excretion of Na, K, and arginine vasopressin in WT mice but not in KO mice. CONCLUSION Taken together, our data show that similar to pharmacological blockade, deletion of P2Y12 -R significantly ameliorates lithium-induced NDI, without reducing serum lithium levels. Hence, targeting P2Y12 -R with currently available drugs in the market offers a novel and safer method for treating NDI.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Internal Medicine; University of Utah Health Sciences Center; Salt Lake City Utah
- Nephrology Research, Department of Veterans Affairs Salt; Lake City Health Care System; Salt Lake City Utah
| | - Kenny M. Hansson
- Cardiovascular, Renal and Metabolism Innovative Medicines and Early Development Biotech Unit; AstraZeneca; Gothenburg Sweden
| | - Tao Liu
- Department of Internal Medicine; University of Utah Health Sciences Center; Salt Lake City Utah
- Nephrology Research, Department of Veterans Affairs Salt; Lake City Health Care System; Salt Lake City Utah
| | - Kerstin Magnell
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit; AstraZeneca; Gothenburg Sweden
| | - Yufeng Huang
- Department of Internal Medicine; University of Utah Health Sciences Center; Salt Lake City Utah
| | - Noel G. Carlson
- Center on Aging; University of Utah Health Sciences Center; Salt Lake City Utah
- Department of Neurobiology and Anatomy; University of Utah Health Sciences Center; Salt Lake City Utah
- Geriatric Research, Education and Clinical Center, Department of Veterans Affairs Salt; Lake City Health Care System; Salt Lake City Utah
| | - Bellamkonda K. Kishore
- Department of Internal Medicine; University of Utah Health Sciences Center; Salt Lake City Utah
- Nephrology Research, Department of Veterans Affairs Salt; Lake City Health Care System; Salt Lake City Utah
- Center on Aging; University of Utah Health Sciences Center; Salt Lake City Utah
- Department of Nutrition and Integrative Physiology; University of Utah College of Health; Salt Lake City Utah
| |
Collapse
|
16
|
Zhang Y, Riquier-Brison A, Liu T, Huang Y, Carlson NG, Peti-Peterdi J, Kishore BK. Genetic Deletion of P2Y 2 Receptor Offers Long-Term (5 Months) Protection Against Lithium-Induced Polyuria, Natriuresis, Kaliuresis, and Collecting Duct Remodeling and Cell Proliferation. Front Physiol 2018; 9:1765. [PMID: 30618788 PMCID: PMC6304354 DOI: 10.3389/fphys.2018.01765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/22/2018] [Indexed: 11/15/2022] Open
Abstract
Chronic lithium administration for the treatment of bipolar disorder leads to nephrogenic diabetes insipidus (NDI), characterized by polyuria, natriuresis, kaliuresis, and collecting duct remodeling and cell proliferation among other features. Previously, using a 2-week lithium-induced NDI model, we reported that P2Y2 receptor (R) knockout mice are significantly resistant to polyuria, natriuresis, kaliuresis, and decrease in AQP2 protein abundance in the kidney relative to wild type mice. Here we show this protection is long-lasting, and is also associated with significant amelioration of lithium-induced collecting duct remodeling and cell proliferation. Age-matched wild type and knockout mice were fed regular (n = 5/genotype) or lithium-added (40 mmol/kg chow; n = 10/genotype) diet for 5 months and euthanized. Water intake, urine output and osmolality were monitored once in every month. Salt blocks were provided to mice on lithium-diet to prevent sodium loss. At the end of 5 months mice were euthanized and serum and kidney samples were analyzed. There was a steady increase in lithium-induced polyuria, natriuresis and kaliuresis in wild type mice over the 5-month period. Increases in these urinary parameters were very low in lithium-fed knockout mice, resulting in significantly widening differences between the wild type and knockout mice. Terminal AQP2 and NKCC2 protein abundances in the kidney were significantly higher in lithium-fed knockout vs. wild type mice. There were no significant differences in terminal serum lithium or sodium levels between the wild type and knockout mice. Confocal immunofluorescence microscopy revealed that lithium-induced marked remodeling of collecting duct with significantly increased proportion of [H+]-ATPase-positive intercalated cells and decreased proportion of AQP2-positive principal cells in the wild type, but not in knockout mice. Lithium-induced collecting duct cell proliferation (indicated by Ki67 labeling), was significantly lower in knockout vs. wild type mice. This is the first piece of evidence that purinergic signaling is potentially involved in lithium-induced collecting duct remodeling and cell proliferation. Our results demonstrate that genetic deletion of P2Y2-R protects against the key structural and functional alterations in Li-induced NDI, and underscore the potential utility of targeting this receptor for the treatment of NDI in bipolar patients on chronic lithium therapy.
Collapse
Affiliation(s)
- Yue Zhang
- Nephrology Research, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, United States
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Anne Riquier-Brison
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, United States
| | - Tao Liu
- Nephrology Research, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, United States
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
| | - Yufeng Huang
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
| | - Noel G. Carlson
- Geriatric Research, Education and Clinical Center, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, United States
- Department of Neurobiology and Anatomy, University of Utah Health, Salt Lake City, UT, United States
- Center on Aging, University of Utah Health, Salt Lake City, UT, United States
| | - János Peti-Peterdi
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, United States
| | - Bellamkonda K. Kishore
- Nephrology Research, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, United States
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
- Center on Aging, University of Utah Health, Salt Lake City, UT, United States
- Department of Nutrition and Integrative Physiology, University of Utah Health, Salt Lake City, UT, United States
| |
Collapse
|
17
|
Zhang Y, Peti-Peterdi J, Brandes AU, Riquier-Brison A, Carlson NG, Müller CE, Ecelbarger CM, Kishore BK. Prasugrel suppresses development of lithium-induced nephrogenic diabetes insipidus in mice. Purinergic Signal 2017; 13:239-248. [PMID: 28233082 PMCID: PMC5432483 DOI: 10.1007/s11302-017-9555-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/23/2017] [Indexed: 12/17/2022] Open
Abstract
Previously, we localized ADP-activated P2Y12 receptor (R) in rodent kidney and showed that its blockade by clopidogrel bisulfate (CLPD) attenuates lithium (Li)-induced nephrogenic diabetes insipidus (NDI). Here, we evaluated the effect of prasugrel (PRSG) administration on Li-induced NDI in mice. Both CLPD and PRSG belong to the thienopyridine class of ADP receptor antagonists. Groups of age-matched adult male B6D2 mice (N = 5/group) were fed either regular rodent chow (CNT), or with added LiCl (40 mmol/kg chow) or PRSG in drinking water (10 mg/kg bw/day) or a combination of LiCl and PRSG for 14 days and then euthanized. Water intake and urine output were determined and blood and kidney tissues were collected and analyzed. PRSG administration completely suppressed Li-induced polydipsia and polyuria and significantly prevented Li-induced decreases in AQP2 protein abundance in renal cortex and medulla. However, PRSG either alone or in combination with Li did not have a significant effect on the protein abundances of NKCC2 or NCC in the cortex and/or medulla. Immunofluorescence microscopy revealed that PRSG administration prevented Li-induced alterations in cellular disposition of AQP2 protein in medullary collecting ducts. Serum Li, Na, and osmolality were not affected by the administration of PRSG. Similar to CLPD, PRSG administration had no effect on Li-induced increase in urinary Na excretion. However, unlike CLPD, PRSG did not augment Li-induced increase in urinary arginine vasopressin (AVP) excretion. Taken together, these data suggest that the pharmacological inhibition of P2Y12-R by the thienopyridine group of drugs may potentially offer therapeutic benefits in Li-induced NDI.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Internal Medicine and Center on Aging, University of Utah Health Sciences Center, Veterans Affairs Salt Lake City, Health Care System, 500 Foothill Drive (151M), Salt Lake City, UT, 84148, USA
| | - János Peti-Peterdi
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, University of Southern California, 1501 San Pablo Street, ZNI 313, Los Angeles, CA, 90033, USA
| | - Anna U Brandes
- Department of Internal Medicine and Center on Aging, University of Utah Health Sciences Center, Veterans Affairs Salt Lake City, Health Care System, 500 Foothill Drive (151M), Salt Lake City, UT, 84148, USA
| | - Anne Riquier-Brison
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, University of Southern California, 1501 San Pablo Street, ZNI 313, Los Angeles, CA, 90033, USA
| | - Noel G Carlson
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121, Bonn, Germany
| | - Christa E Müller
- Depatment of Neurobiology and Anatomy and Center on Aging, University of Utah Health Sciences Center, Geriatric Research, Education, and Clinical Center (GRECC) Veterans Affairs Salt Lake City Health Care System, 500 Foothill Drive (151B), Salt Lake City, UT, 84148, USA
| | - Carolyn M Ecelbarger
- Department of Medicine, Center for the Study of Sex Differences in Health, Aging, and Disease, Georgetown University, 4000 Reservoir Road NW Bldg D, Rm 392, Washington, DC, 20057, USA
| | - Bellamkonda K Kishore
- Department of Internal Medicine and Center on Aging, University of Utah Health Sciences Center, Veterans Affairs Salt Lake City, Health Care System, 500 Foothill Drive (151M), Salt Lake City, UT, 84148, USA.
| |
Collapse
|
18
|
Zhang Y, Peti-Peterdi J, Heiney KM, Riquier-Brison A, Carlson NG, Müller CE, Ecelbarger CM, Kishore BK. Clopidogrel attenuates lithium-induced alterations in renal water and sodium channels/transporters in mice. Purinergic Signal 2015; 11:507-18. [PMID: 26386699 PMCID: PMC4648798 DOI: 10.1007/s11302-015-9469-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/09/2015] [Indexed: 10/23/2022] Open
Abstract
Lithium (Li) administration causes deranged expression and function of renal aquaporins and sodium channels/transporters resulting in nephrogenic diabetes insipidus (NDI). Extracellular nucleotides (ATP/ADP/UTP), via P2 receptors, regulate these transport functions. We tested whether clopidogrel bisulfate (CLPD), an antagonist of ADP-activated P2Y(12) receptor, would affect Li-induced alterations in renal aquaporins and sodium channels/transporters. Adult mice were treated for 14 days with CLPD and/or Li and euthanized. Urine and kidneys were collected for analysis. When administered with Li, CLPD ameliorated polyuria, attenuated the rise in urine prostaglandin E2 (PGE2), and resulted in significantly higher urinary arginine vasopressin (AVP) and aldosterone levels as compared to Li treatment alone. However, urine sodium excretion remained elevated. Semi-quantitative immunoblotting revealed that CLPD alone increased renal aquaporin 2 (AQP2), Na-K-2Cl cotransporter (NKCC2), Na-Cl cotransporter (NCC), and the subunits of the epithelial Na channel (ENaC) in medulla by 25-130 %. When combined with Li, CLPD prevented downregulation of AQP2, Na-K-ATPase, and NKCC2 but was less effective against downregulation of cortical α- or γ-ENaC (70 kDa band). Thus, CLPD primarily attenuated Li-induced downregulation of proteins involved in water conservation (AVP-sensitive), with modest effects on aldosterone-sensitive proteins potentially explaining sustained natriuresis. Confocal immunofluorescence microscopy revealed strong labeling for P2Y(12)-R in proximal tubule brush border and blood vessels in the cortex and less intense labeling in medullary thick ascending limb and the collecting ducts. Therefore, there is the potential for CLPD to be directly acting at the tubule sites to mediate these effects. In conclusion, P2Y(12)-R may represent a novel therapeutic target for Li-induced NDI.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Internal Medicine and Center on Aging, University of Utah Health Sciences Center & Veterans Affairs Salt Lake City Health Care System, 500 Foothill Drive (151M), Salt Lake City, UT, 84148, USA
| | - János Peti-Peterdi
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, University of Southern California, 1501 San Pablo Street, ZNI 313, Los Angeles, CA, 90033, USA
| | - Kristina M Heiney
- Department of Internal Medicine and Center on Aging, University of Utah Health Sciences Center & Veterans Affairs Salt Lake City Health Care System, 500 Foothill Drive (151M), Salt Lake City, UT, 84148, USA
| | - Anne Riquier-Brison
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, University of Southern California, 1501 San Pablo Street, ZNI 313, Los Angeles, CA, 90033, USA
| | - Noel G Carlson
- Department of Neurobiology and Anatomy and Center on Aging Geriatric Research, Education, and Clinical Center (GRECC), University of Utah Health Sciences Center & Veterans Affairs Salt Lake City Health Care System, 500 Foothill Drive (151B), Salt Lake City, UT, 84148, USA
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121, Bonn, Germany
| | - Carolyn M Ecelbarger
- Department of Medicine, Center for the Study of Sex Differences in Health, Aging, and Disease, Georgetown University, 4000 Reservoir Road NW Bldg D, Rm 392, Washington, DC, 20057, USA
| | - Bellamkonda K Kishore
- Department of Internal Medicine and Center on Aging, University of Utah Health Sciences Center & Veterans Affairs Salt Lake City Health Care System, 500 Foothill Drive (151M), Salt Lake City, UT, 84148, USA.
| |
Collapse
|
19
|
Chebib FT, Sussman CR, Wang X, Harris PC, Torres VE. Vasopressin and disruption of calcium signalling in polycystic kidney disease. Nat Rev Nephrol 2015; 11:451-64. [PMID: 25870007 PMCID: PMC4539141 DOI: 10.1038/nrneph.2015.39] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic kidney disease and is responsible for 5-10% of cases of end-stage renal disease worldwide. ADPKD is characterized by the relentless development and growth of cysts, which cause progressive kidney enlargement associated with hypertension, pain, reduced quality of life and eventual kidney failure. Mutations in the PKD1 or PKD2 genes, which encode polycystin-1 (PC1) and polycystin-2 (PC2), respectively, cause ADPKD. However, neither the functions of these proteins nor the molecular mechanisms of ADPKD pathogenesis are well understood. Here, we review the literature that examines how reduced levels of functional PC1 or PC2 at the primary cilia and/or the endoplasmic reticulum directly disrupts intracellular calcium signalling and indirectly disrupts calcium-regulated cAMP and purinergic signalling. We propose a hypothetical model in which dysregulated metabolism of cAMP and purinergic signalling increases the sensitivity of principal cells in collecting ducts and of tubular epithelial cells in the distal nephron to the constant tonic action of vasopressin. The resulting magnified response to vasopressin further enhances the disruption of calcium signalling that is initiated by mutations in PC1 or PC2, and activates downstream signalling pathways that cause impaired tubulogenesis, increased cell proliferation, increased fluid secretion and interstitial inflammation.
Collapse
Affiliation(s)
- Fouad T Chebib
- Division of Nephrology and Hypertension, 200 First Street S. W., Mayo Clinic College of Medicine, Rochester, MN 55901, USA
| | - Caroline R Sussman
- Division of Nephrology and Hypertension, 200 First Street S. W., Mayo Clinic College of Medicine, Rochester, MN 55901, USA
| | - Xiaofang Wang
- Division of Nephrology and Hypertension, 200 First Street S. W., Mayo Clinic College of Medicine, Rochester, MN 55901, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension, 200 First Street S. W., Mayo Clinic College of Medicine, Rochester, MN 55901, USA
| | - Vicente E Torres
- Division of Nephrology and Hypertension, 200 First Street S. W., Mayo Clinic College of Medicine, Rochester, MN 55901, USA
| |
Collapse
|
20
|
Abed AB, Kavvadas P, Chadjichristos CE. Functional roles of connexins and pannexins in the kidney. Cell Mol Life Sci 2015; 72:2869-77. [PMID: 26082183 PMCID: PMC11113829 DOI: 10.1007/s00018-015-1964-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 12/22/2022]
Abstract
Kidneys are highly complex organs, playing a crucial role in human physiopathology, as they are implicated in vital processes, such as fluid filtration and vasomotor tone regulation. There is growing evidence that gap junctions are major determinants of renal physiopathology. It has been demonstrated that their expression or channel activity may vary depending on physiological and pathological situations within distinct renal compartments. While some studies have focused on the role of connexins in renal physiology, our knowledge regarding the functional relevance of pannexins is still very limited. In this paper, we provide an overview of the involvement of connexins, pannexins and their channels in various physiological processes related to different renal compartments.
Collapse
Affiliation(s)
- Ahmed B. Abed
- INSERM UMR-S1155, Batiment Recherche, Tenon Hospital, 4 rue de la Chine, 75020 Paris, France
- Sorbonne Universite´s, UPMC Univ Paris 6, Paris, France
| | - Panagiotis Kavvadas
- INSERM UMR-S1155, Batiment Recherche, Tenon Hospital, 4 rue de la Chine, 75020 Paris, France
| | | |
Collapse
|
21
|
Kishore BK, Carlson NG, Ecelbarger CM, Kohan DE, Müller CE, Nelson RD, Peti-Peterdi J, Zhang Y. Targeting renal purinergic signalling for the treatment of lithium-induced nephrogenic diabetes insipidus. Acta Physiol (Oxf) 2015; 214:176-88. [PMID: 25877068 PMCID: PMC4430398 DOI: 10.1111/apha.12507] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/08/2015] [Indexed: 12/26/2022]
Abstract
Lithium still retains its critical position in the treatment of bipolar disorder by virtue of its ability to prevent suicidal tendencies. However, chronic use of lithium is often limited by the development of nephrogenic diabetes insipidus (NDI), a debilitating condition. Lithium-induced NDI is due to resistance of the kidney to arginine vasopressin (AVP), leading to polyuria, natriuresis and kaliuresis. Purinergic signalling mediated by extracellular nucleotides (ATP/UTP), acting via P2Y receptors, opposes the action of AVP on renal collecting duct (CD) by decreasing the cellular cAMP and thus AQP2 protein levels. Taking a cue from this phenomenon, we discovered the potential involvement of ATP/UTP-activated P2Y2 receptor in lithium-induced NDI in rats and showed that P2Y2 receptor knockout mice are significantly resistant to Li-induced polyuria, natriuresis and kaliuresis. Extension of these studies revealed that ADP-activated P2Y12 receptor is expressed in the kidney, and its irreversible blockade by the administration of clopidogrel bisulphate (Plavix(®)) ameliorates Li-induced NDI in rodents. Parallel in vitro studies showed that P2Y12 receptor blockade by the reversible antagonist PSB-0739 sensitizes CD to the action of AVP. Thus, our studies unravelled the potential beneficial effects of targeting P2Y2 or P2Y12 receptors to counter AVP resistance in lithium-induced NDI. If established in further studies, our findings may pave the way for the development of better and safer methods for the treatment of NDI by bringing a paradigm shift in the approach from the current therapies that predominantly counter the anti-AVP effects to those that enhance the sensitivity of the kidney to AVP action.
Collapse
Affiliation(s)
- B. K. Kishore
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Nephrology Research, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA
- Center on Aging, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - N. G. Carlson
- Center on Aging, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Department of Neurobiology and Anatomy, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Geriatric Research, Education and Clinical Center, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA
| | - C. M. Ecelbarger
- Department of Medicine, Georgetown University, Washington, District of Columbia, USA
- Center for the Study of Sex Differences in Health, Aging, and Disease, Georgetown University, Washington, District of Columbia, USA
| | - D. E. Kohan
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Nephrology Research, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA
| | - C. E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - R. D. Nelson
- Department of Paediatrics, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - J. Peti-Peterdi
- Department of Physiology and Biophysics, and Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Y. Zhang
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Nephrology Research, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA
| |
Collapse
|
22
|
Zhang Y, Robson SC, Morris KL, Heiney KM, Dwyer KM, Kishore BK, Ecelbarger CM. Impaired natriuretic response to high-NaCl diet plus aldosterone infusion in mice overexpressing human CD39, an ectonucleotidase (NTPDase1). Am J Physiol Renal Physiol 2015; 308:F1398-408. [PMID: 25877509 DOI: 10.1152/ajprenal.00125.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 04/09/2015] [Indexed: 01/14/2023] Open
Abstract
Extracellular nucleotides acting through P2 receptors facilitate natriuresis. To define how purinergic mechanisms are involved in sodium homeostasis, we used transgenic (TG) mice that globally overexpress human CD39 (hCD39, NTPDase1), an ectonucleotidase that hydrolyzes extracellular ATP/ADP to AMP, resulting in an altered extracellular purine profile. On a high-sodium diet (HSD, 3.5% Na(+)), urine volume and serum sodium were significantly higher in TG mice but sodium excretion was unaltered. Furthermore, TG mice showed an attenuated fall in urine aldosterone with HSD. Western blot analysis revealed significantly lower densities (∼40%) of the β-subunit of the epithelial sodium channel (ENaC) in medulla, and the major band (85-kDa) of γ-ENaC in TG mice cortex. To evaluate aldosterone-independent differences, in a second experiment, aldosterone was clamped by osmotic minipump at 20 μg/day, and mice were fed either an HSD or a low-sodium diet (LSD, 0.03% Na(+)). Here, no differences in urine volume or osmolality, or serum aldosterone were found, but TG mice showed a modest, yet significant impairment in late natriuresis (days 3 and 4). Several major sodium transporters or channel subunits were differentially expressed between the genotypes. HSD caused a downregulation of Na-Cl cotransporter (NCC) in both genotypes; and had higher cortical levels of NCC, Na-K-ATPase (α-1 subunit), and α- and γ-ENaC. The Na-K-2Cl cotransporter (NKCC2) was downregulated by HSD in wild-type mice, but it increased in TG mice. In summary, our data support the concept that extracellular nucleotides facilitate natriuresis; they also reveal an aldosterone-independent downregulation of major renal sodium transporters and channel subunits by purinergic signaling.
Collapse
Affiliation(s)
- Yue Zhang
- Nephrology Research, Department of Veterans Administration Salt Lake City Health Care System, Salt Lake City, Utah; Department of Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Simon C Robson
- Transplant Institute and Gastroenterology Division, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Kaiya L Morris
- Nephrology Research, Department of Veterans Administration Salt Lake City Health Care System, Salt Lake City, Utah; Department of Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Kristina M Heiney
- Nephrology Research, Department of Veterans Administration Salt Lake City Health Care System, Salt Lake City, Utah; Department of Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Karen M Dwyer
- Immunology Research Center, Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, Australia
| | - Bellamkonda K Kishore
- Nephrology Research, Department of Veterans Administration Salt Lake City Health Care System, Salt Lake City, Utah; Department of Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah; Center on Aging, University of Utah Health Sciences Center, Salt Lake City, Utah;
| | - Carolyn M Ecelbarger
- Department of Medicine, Georgetown University, Washington, DC; and Center for the Study of Sex Differences in Health, Aging, and Disease, Georgetown University, Washington, District of Columbia
| |
Collapse
|
23
|
Zhang Y, Peti-Peterdi J, Müller CE, Carlson NG, Baqi Y, Strasburg DL, Heiney KM, Villanueva K, Kohan DE, Kishore BK. P2Y12 Receptor Localizes in the Renal Collecting Duct and Its Blockade Augments Arginine Vasopressin Action and Alleviates Nephrogenic Diabetes Insipidus. J Am Soc Nephrol 2015; 26:2978-87. [PMID: 25855780 DOI: 10.1681/asn.2014010118] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 01/27/2015] [Indexed: 11/03/2022] Open
Abstract
P2Y12 receptor (P2Y12-R) signaling is mediated through Gi, ultimately reducing cellular cAMP levels. Because cAMP is a central modulator of arginine vasopressin (AVP)-induced water transport in the renal collecting duct (CD), we hypothesized that if expressed in the CD, P2Y12-R may play a role in renal handling of water in health and in nephrogenic diabetes insipidus. We found P2Y12-R mRNA expression in rat kidney, and immunolocalized its protein and aquaporin-2 (AQP2) in CD principal cells. Administration of clopidogrel bisulfate, an irreversible inhibitor of P2Y12-R, significantly increased urine concentration and AQP2 protein in the kidneys of Sprague-Dawley rats. Notably, clopidogrel did not alter urine concentration in Brattleboro rats that lack AVP. Clopidogrel administration also significantly ameliorated lithium-induced polyuria, improved urine concentrating ability and AQP2 protein abundance, and reversed the lithium-induced increase in free-water excretion, without decreasing blood or kidney tissue lithium levels. Clopidogrel administration also augmented the lithium-induced increase in urinary AVP excretion and suppressed the lithium-induced increase in urinary nitrates/nitrites (nitric oxide production) and 8-isoprostane (oxidative stress). Furthermore, selective blockade of P2Y12-R by the reversible antagonist PSB-0739 in primary cultures of rat inner medullary CD cells potentiated the expression of AQP2 and AQP3 mRNA, and cAMP production induced by dDAVP (desmopressin). In conclusion, pharmacologic blockade of renal P2Y12-R increases urinary concentrating ability by augmenting the effect of AVP on the kidney and ameliorates lithium-induced NDI by potentiating the action of AVP on the CD. This strategy may offer a novel and effective therapy for lithium-induced NDI.
Collapse
Affiliation(s)
- Yue Zhang
- Nephrology Research and Department of Internal Medicine
| | - Janos Peti-Peterdi
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, University of Southern California, Los Angeles, California
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany; and
| | - Noel G Carlson
- Geriatric Research, Education and Clinical Center, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah; Department of Neurobiology and Anatomy, and Center on Aging, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Younis Baqi
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany; and Department of Chemistry, Faculty of Science, Sultan Qaboos University, Muscat, Oman
| | | | | | - Karie Villanueva
- Zilkha Neurogenetic Institute and Department of Physiology and Biophysics, University of Southern California, Los Angeles, California
| | | | - Bellamkonda K Kishore
- Nephrology Research and Department of Internal Medicine, Center on Aging, University of Utah Health Sciences Center, Salt Lake City, Utah;
| |
Collapse
|
24
|
Mamenko M, Zaika O, Boukelmoune N, O'Neil RG, Pochynyuk O. Deciphering physiological role of the mechanosensitive TRPV4 channel in the distal nephron. Am J Physiol Renal Physiol 2015; 308:F275-86. [PMID: 25503733 PMCID: PMC4329491 DOI: 10.1152/ajprenal.00485.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 12/08/2014] [Indexed: 12/14/2022] Open
Abstract
Long-standing experimental evidence suggests that epithelial cells in the renal tubule are able to sense osmotic and pressure gradients caused by alterations in ultrafiltrate flow by elevating intracellular Ca(2+) concentration. These responses are viewed as critical regulators of a variety of processes ranging from transport of water and solutes to cellular growth and differentiation. A loss in the ability to sense mechanical stimuli has been implicated in numerous pathologies associated with systemic imbalance of electrolytes and to the development of polycystic kidney disease. The molecular mechanisms conferring mechanosensitive properties to epithelial tubular cells involve activation of transient receptor potential (TRP) channels, such as TRPV4, allowing direct Ca(2+) influx to increase intracellular Ca(2+) concentration. In this review, we critically analyze the current evidence about signaling determinants of TRPV4 activation by luminal flow in the distal nephron and discuss how dysfunction of this mechanism contributes to the progression of polycystic kidney disease. We also review the physiological relevance of TRPV4-based mechanosensitivity in controlling flow-dependent K(+) secretion in the distal renal tubule.
Collapse
Affiliation(s)
- M Mamenko
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| | - O Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| | - N Boukelmoune
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| | - R G O'Neil
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| | - O Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| |
Collapse
|
25
|
Sim JH, Himmel NJ, Redd SK, Pulous FE, Rogers RT, Black LN, Hong SM, von Bergen TN, Blount MA. Absence of PKC-alpha attenuates lithium-induced nephrogenic diabetes insipidus. PLoS One 2014; 9:e101753. [PMID: 25006961 PMCID: PMC4090211 DOI: 10.1371/journal.pone.0101753] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 06/11/2014] [Indexed: 01/01/2023] Open
Abstract
Lithium, an effective antipsychotic, induces nephrogenic diabetes insipidus (NDI) in ∼40% of patients. The decreased capacity to concentrate urine is likely due to lithium acutely disrupting the cAMP pathway and chronically reducing urea transporter (UT-A1) and water channel (AQP2) expression in the inner medulla. Targeting an alternative signaling pathway, such as PKC-mediated signaling, may be an effective method of treating lithium-induced polyuria. PKC-alpha null mice (PKCα KO) and strain-matched wild type (WT) controls were treated with lithium for 0, 3 or 5 days. WT mice had increased urine output and lowered urine osmolality after 3 and 5 days of treatment whereas PKCα KO mice had no change in urine output or concentration. Western blot analysis revealed that AQP2 expression in medullary tissues was lowered after 3 and 5 days in WT mice; however, AQP2 was unchanged in PKCα KO. Similar results were observed with UT-A1 expression. Animals were also treated with lithium for 6 weeks. Lithium-treated WT mice had 19-fold increased urine output whereas treated PKCα KO animals had a 4-fold increase in output. AQP2 and UT-A1 expression was lowered in 6 week lithium-treated WT animals whereas in treated PKCα KO mice, AQP2 was only reduced by 2-fold and UT-A1 expression was unaffected. Urinary sodium, potassium and calcium were elevated in lithium-fed WT but not in lithium-fed PKCα KO mice. Our data show that ablation of PKCα preserves AQP2 and UT-A1 protein expression and localization in lithium-induced NDI, and prevents the development of the severe polyuria associated with lithium therapy.
Collapse
Affiliation(s)
- Jae H. Sim
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Nathaniel J. Himmel
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Sara K. Redd
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Fadi E. Pulous
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Richard T. Rogers
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Lauren N. Black
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Seongun M. Hong
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Tobias N. von Bergen
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mitsi A. Blount
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|
26
|
Burnstock G, Evans LC, Bailey MA. Purinergic signalling in the kidney in health and disease. Purinergic Signal 2014; 10:71-101. [PMID: 24265071 PMCID: PMC3944043 DOI: 10.1007/s11302-013-9400-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/24/2013] [Indexed: 12/21/2022] Open
Abstract
The involvement of purinergic signalling in kidney physiology and pathophysiology is rapidly gaining recognition and this is a comprehensive review of early and recent publications in the field. Purinergic signalling involvement is described in several important intrarenal regulatory mechanisms, including tuboglomerular feedback, the autoregulatory response of the glomerular and extraglomerular microcirculation and the control of renin release. Furthermore, purinergic signalling influences water and electrolyte transport in all segments of the renal tubule. Reports about purine- and pyrimidine-mediated actions in diseases of the kidney, including polycystic kidney disease, nephritis, diabetes, hypertension and nephrotoxicant injury are covered and possible purinergic therapeutic strategies discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK,
| | | | | |
Collapse
|
27
|
Abstract
UT-A and UT-B families of urea transporters consist of multiple isoforms that are subject to regulation of both acutely and by long-term measures. This chapter provides a brief overview of the expression of the urea transporter forms and their locations in the kidney. Rapid regulation of UT-A1 results from the combination of phosphorylation and membrane accumulation. Phosphorylation of UT-A1 has been linked to vasopressin and hyperosmolality, although through different kinases. Other acute influences on urea transporter activity are ubiquitination and glycosylation, both of which influence the membrane association of the urea transporter, again through different mechanisms. Long-term regulation of urea transport is most closely associated with the environment that the kidney experiences. Low-protein diets may influence the amount of urea transporter available. Conditions of osmotic diuresis, where urea concentrations are low, will prompt an increase in urea transporter abundance. Although adrenal steroids affect urea transporter abundance, conflicting reports make conclusions tenuous. Urea transporters are upregulated when P2Y2 purinergic receptors are decreased, suggesting a role for these receptors in UT regulation. Hypercalcemia and hypokalemia both cause urine concentration deficiencies. Urea transporter abundances are reduced in aging animals and animals with angiotensin-converting enzyme deficiencies. This chapter will provide information about both rapid and long-term regulation of urea transporters and provide an introduction into the literature.
Collapse
Affiliation(s)
- Janet D Klein
- Renal Division, Department of Medicine and Department of Physiology, Emory University School of Medicine, WMB Room 3319B, 1639 Pierce Drive, NE, Atlanta, GA, 30322, USA,
| |
Collapse
|
28
|
Zhang Y, Li L, Kohan DE, Ecelbarger CM, Kishore BK. Attenuation of lithium-induced natriuresis and kaliuresis in P2Y₂ receptor knockout mice. Am J Physiol Renal Physiol 2013; 305:F407-16. [PMID: 23739592 DOI: 10.1152/ajprenal.00464.2012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Whole body knockout (KO) of the P2Y₂ receptor (P2Y₂R) results in enhanced vasopressin V2 receptor activity and increased renal Na⁺ conservation. We hypothesized that P2Y₂R KO mice would be less sensitive to lithium-induced natriuresis and kaliuresis due to attenuated downregulation of one or more of the major renal Na⁺ or K⁺ transporter/channel proteins. KO and wild-type (WT) mice were fed a control or lithium-added diet (40 mmol/kg food) for 14 days. Lithium-induced natriuresis and kaliuresis were significantly (~25%) attenuated in KO mice. The subunits of the epithelial Na⁺ channel (ENaC) were variably affected by lithium and genotype, but, overall, medullary levels were decreased substantially by lithium (15-60%) in both genotypes. In contrast, cortical, β-, and γ-ENaC were increased by lithium (~50%), but only in WT mice. Moreover, an assessment of ENaC activity by benzamil sensitivity suggested that lithium increased ENaC activity in WT mice but in not KO mice. In contrast, medullary levels of Na⁺-K⁺-2Cl⁻ cotransporter 2 and cortical levels of the renal outer medullary K⁺ channel were not downregulated by lithium and were significantly (15-76%) higher in KO mice under both dietary conditions. In addition, under control conditions, tissue osmolality of the inner medulla as well as furosemide sensitivity were significantly higher in KO mice versus WT mice. Therefore, we suggest that increased expression of these proteins, particularly in the control state, reduces Na⁺ delivery to the distal nephron and provides a buffer to attenuate collecting duct-mediated natriuresis and kaliuresis. Additional studies are warranted to explore the potential therapeutic benefits of purinergic antagonism.
Collapse
Affiliation(s)
- Yue Zhang
- Nephrology Research, Department of Veterans Administration Salt Lake City Health Care System, Salt Lake City, UT 84148, USA
| | | | | | | | | |
Collapse
|
29
|
Pochynyuk O, Zaika O, O’Neil RG, Mamenko M. Novel insights into TRPV4 function in the kidney. Pflugers Arch 2013; 465:177-86. [PMID: 23207579 PMCID: PMC3562383 DOI: 10.1007/s00424-012-1190-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 11/16/2012] [Accepted: 11/19/2012] [Indexed: 10/27/2022]
Abstract
Kidneys are complex highly organized paired organs of nearly one million nephrons each. They rigorously process about 180 l of plasma daily to keep whole body homeostasis. To effectively perform such a titanic work, kidneys rely on mechanisms able to sense dynamic changes in composition and flow rates of protourine along the renal tubule. It is envisioned that Ca(2+)-permeable transient receptor potential (TRP) channels, and specifically mechanosensitive TRPV4, can serve to interpret these external mechanical cues in the form of elevated intracellular Ca(2+) concentration. This, in turn, initiates multiple cellular responses and adaptation mechanisms. The current review summarizes up-to-date knowledge about the sites of TRPV4 expression in renal tissue as well as discusses the functional role of the channel in cellular responses to hypotonicity and tubular flow. We will also provide insights as to how TRPV4 fits into classical polycystin mechanosensory complex in cilia and will speculate about previously underappreciated clinical implication of pharmacological TRPV4 targeting in treatment of polycystic kidney disease.
Collapse
Affiliation(s)
- Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin st., Houston TX 77030, USA; Phone: (713) 500-7466; Fax: (713) 500-7455
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin st., Houston, TX 77030 USA; Phone: (713) 500-6342; Fax: (713) 500-7455
| | - Roger G. O’Neil
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin st., Houston TX 77030, USA; Phone: (713) 500-6316; Fax: (713) 500-7455
| | - Mykola Mamenko
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin st., Houston TX 77030, USA; Phone: (713) 500-6342; Fax: (713) 500-7455
| |
Collapse
|
30
|
Abstract
Cellular release of nucleotides is of physiological importance to regulate and maintain cell function and integrity. Also in the tubular and collecting duct system of the kidney, nucleotides are released in response to changes in cell volume or luminal flow rate and act in a paracrine and autocrine way on basolateral and luminal P2Y receptors. Recent studies using gene knockout mice assigned a prominent role to G protein-coupled P2Y(2) receptors, which are activated by both ATP and UTP. The antidiuretic hormone, arginine-vasopressin (AVP), and possibly an increase in collecting duct cell volume induce ATP release. The subsequent activation of P2Y(2) receptors inhibits AVP-induced cAMP formation and water reabsorption, which stabilizes cell volume and facilitates water excretion. An increase in NaCl intake enhances luminal release of ATP and UTP in the aldosterone-sensitive distal nephron which by activating apical P2Y(2) receptors and phospholipase C lowers the open probability of the epithelial sodium channel ENaC, thereby facilitating sodium excretion. Thus, the renal ATP/UTP/P2Y(2) receptor system not only serves to preserve cell volume and integrity but is also regulated by stimuli that derive from body NaCl homeostasis. The system also inhibits ENaC activity during aldosterone escape, i.e. when sodium reabsorption via ENaC is inappropriately high. The P2Y(2) receptor tone inhibits the expression and activity of the Na-K-2Cl cotransporter NKCC2 in the thick ascending limb and mediates vasodilation. While the role of other P2Y receptors in the kidney is less clear, the ATP/UTP/P2Y(2) receptor system regulates NaCl and water homeostasis and blood pressure.
Collapse
Affiliation(s)
- Volker Vallon
- Department of Medicine, University of California San Diego, San Diego, CA 92161, USA; VA San Diego Healthcare System, San Diego California, San Diego, CA 92161, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
31
|
Zhang Y, Morris KL, Sparrow SK, Dwyer KM, Enjyoji K, Robson SC, Kishore BK. Defective renal water handling in transgenic mice over-expressing human CD39/NTPDase1. Am J Physiol Renal Physiol 2012; 303:F420-30. [PMID: 22622462 DOI: 10.1152/ajprenal.00060.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Ectonucleoside triphosphate diphosphohydrolase-1 hydrolyzes extracellular ATP and ADP to AMP. Previously, we showed that CD39 is expressed at several sites within the kidney and thus may impact the availability of type 2 purinergic receptor (P2-R) ligands. Because P2-Rs appear to regulate urinary concentrating ability, we have evaluated renal water handling in transgenic mice (TG) globally overexpressing hCD39. Under basal conditions, TG mice exhibited significantly impaired urinary concentration and decreased protein abundance of AQP2 in the kidney compared with wild-type (WT) mice. Urinary excretion of total nitrates/nitrites was significantly higher in TG mice, but the excretion of AVP or PGE(2) was equivalent to control WT mice. There were no significant differences in electrolyte-free water clearance or fractional excretion of sodium. Under stable hydrated conditions (gelled diet feeding), the differences between the WT and TG mice were negated, but the decrease in urine osmolality persisted. When water deprived, TG mice failed to adequately concentrate urine and exhibited impaired AVP responses. However, the increases in urinary osmolalities in response to subacute dDAVP or chronic AVP treatment were similar in TG and WT mice. These observations suggest that TG mice have impaired urinary concentrating ability despite normal AVP levels. We also note impaired AVP release in response to water deprivation but that TG kidneys are responsive to exogenous dDAVP or AVP. We infer that heightened nucleotide scavenging by increased levels of CD39 altered the release of endogenous AVP in response to dehydration. We propose that ectonucleotidases and modulated purinergic signaling impact urinary concentration and indicate potential utility of targeted therapy for the treatment of water balance disorders.
Collapse
Affiliation(s)
- Yue Zhang
- Nephrology Research, Department of Veterans Administration Salt Lake City Health Care System, Salt Lake City, UT 84148, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Zhang Y, Pop IL, Carlson NG, Kishore BK. Genetic deletion of the P2Y2 receptor offers significant resistance to development of lithium-induced polyuria accompanied by alterations in PGE2 signaling. Am J Physiol Renal Physiol 2011; 302:F70-7. [PMID: 21975874 DOI: 10.1152/ajprenal.00444.2011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lithium (Li)-induced polyuria is due to resistance of the medullary collecting duct (mCD) to the action of arginine vasopressin (AVP), apparently mediated by increased production of PGE(2). We previously reported that the P2Y(2) receptor (P2Y(2)-R) antagonizes the action of AVP on the mCD and may play a role in Li-induced polyuria by enhancing the production of PGE(2) in mCD. Hence, we hypothesized that genetic deletion of P2Y(2)-R should ameliorate Li-induced polyuria. Wild-type (WT) or P2Y(2)-R knockout (KO) mice were fed normal or Li-added diets for 14 days and euthanized. Li-induced polyuria, and decreases in urine osmolality and AQP2 protein abundance in the renal medulla, were significantly less compared with WT mice despite the lack of differences in Li intake or terminal serum or inner medullary tissue Li levels. Li-induced increased urinary excretion of PGE(2) was not affected in KO mice. However, prostanoid EP(3) receptor (EP3-R) protein abundance in the renal medulla of KO mice was markedly lower vs. WT mice, irrespective of the dietary regimen. The protein abundances of other EP-Rs were not altered across the groups irrespective of the dietary regimen. Ex vivo stimulation of mCD with PGE(2) generated significantly more cAMP in Li-fed KO mice (130%) vs. Li-fed WT mice (100%). Taken together, these data suggest 1) genetic deletion of P2Y(2)-R offers significant resistance to the development of Li-induced polyuria; and 2) this resistance is apparently due to altered PGE(2) signaling mediated by a marked decrease in EP3-R protein abundance in the medulla, thus attenuating the EP3-mediated decrease in cAMP levels in mCD.
Collapse
Affiliation(s)
- Yue Zhang
- Nephrology Research (151M VA SLC Health Care System, 500 Foothill Dr., Salt Lake City, UT 84148, USA
| | | | | | | |
Collapse
|
33
|
|
34
|
Purinergic activation of Ca2+-permeable TRPV4 channels is essential for mechano-sensitivity in the aldosterone-sensitive distal nephron. PLoS One 2011; 6:e22824. [PMID: 21850238 PMCID: PMC3151261 DOI: 10.1371/journal.pone.0022824] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 07/03/2011] [Indexed: 11/19/2022] Open
Abstract
Mechanical forces are known to induce increases of [Ca2+]i in the aldosterone-sensitive distal nephron (ASDN) cells to regulate epithelial transport. At the same time, mechanical stress stimulates ATP release from ASDN cells. In this study, we combined ratiometric Fura-2 based monitoring of [Ca2+]i in freshly isolated split-opened ASDN with targeted deletion of P2Y2 and TRPV4 in mice to probe a role for purinergic signaling in mediating mechano-sensitive responses in ASDN cells. ATP application causes a reproducible transient Ca2+ peak followed by a sustained plateau. Individual cells of the cortical collecting duct (CCD) and the connecting tubule (CNT) respond to purinergic stimulation with comparative elevations of [Ca2+]i. Furthermore, ATP-induced Ca2+-responses are nearly identical in both principal (AQP2-positive) and intercalated (AQP2-negative) cells as was confirmed using immunohistochemistry in split-opened ASDN. UTP application produces elevations of [Ca2+]i similar to that observed with ATP suggesting a dominant role of P2Y2-like receptors in generation of [Ca2+]i response. Indeed, genetic deletion of P2Y2 receptors decreases the magnitude of ATP-induced and UTP-induced Ca2+ responses by more than 70% and 90%, respectively. Both intracellular and extracellular sources of Ca2+ appeared to contribute to the generation of ATP-induced Ca2+ response in ASDN cells. Importantly, flow- and hypotonic-induced Ca2+ elevations are markedly blunted in P2Y2 −/− mice. We further demonstrated that activation of mechano-sensitive TRPV4 channel plays a major role in the sustained [Ca2+]i elevation during purinergic stimulation. Consistent with this, ATP-induced Ca2+ plateau are dramatically attenuated in TRV4 −/− mice. Inhibition of TRPC channels with 10 µM BTP2 also decreased ATP-induced Ca2+ plateau whilst to a lower degree than that observed with TRPV4 inhibition/genetic deletion. We conclude that stimulation of purinergic signaling by mechanical stimuli leads to activation of TRPV4 and, to a lesser extent, TRPCs channels, and this is an important component of mechano-sensitive response of the ASDN.
Collapse
|
35
|
Sinke AP, Deen PMT. The physiological implication of novel proteins in systemic osmoregulation. FASEB J 2011; 25:3279-89. [PMID: 21737616 DOI: 10.1096/fj.11-188433] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Maintenance of the osmobalance is important for life. In this process, in which brain and kidney act in concert, mammals have to cope with significant deviations as drinking water reduces plasma osmolality, whereas salty food increases it. To restore homeostasis, specialized nuclei within the hypothalamus play a pivotal role in detecting changes in plasma osmolality and initiating appropriate responses. These responses are accomplished by either changing the intake of water or the excretion of water by the kidney. In the past decade, several novel findings have made significant contributions to our insights in the process of systemic osmoregulation. Novel proteins have been identified in the brain as well as in the kidney that are fulfilling important roles in the process of systemic osmoregulation. In this review, recent evidence of the involvement of TRPV channels (TRPV1, TRPV2, and TRPV4) and proteins, such as sodium channels NALCN and Na(x), in neuronal osmoregulation, as well as; e.g., the purinergic P2Y2 receptor in renal osmoregulation, are discussed, and integrated with existing knowledge of systemic osmoregulation.
Collapse
Affiliation(s)
- Anne P Sinke
- Department of Physiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | |
Collapse
|
36
|
Vallon V, Rieg T. Regulation of renal NaCl and water transport by the ATP/UTP/P2Y2 receptor system. Am J Physiol Renal Physiol 2011; 301:F463-75. [PMID: 21715471 DOI: 10.1152/ajprenal.00236.2011] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Extracellular nucleotides (e.g., ATP) activate ionotropic P2X and metabotropic P2Y receptors in the plasma membrane to regulate and maintain cell function and integrity. This includes the renal tubular and collecting duct system, where the locally released nucleotides act in a paracrine and autocrine way to regulate transport of electrolytes and water and maintain cell volume. A prominent role has been assigned to Gq-coupled P2Y(2) receptors, which are typically activated by both ATP and UTP. Studies in gene knockout mice revealed an antihypertensive activity of P2Y(2) receptors that is linked to vasodilation and an inhibitory influence on renal salt reabsorption. Flow induces apical ATP release in the thick ascending limb, and first evidence indicates an inhibitory influence of P2Y(2) receptor tone on the expression and activity of the Na-K-2Cl cotransporter NKCC2 in this segment. The apical ATP/UTP/P2Y(2) receptor system in the connecting tubule/cortical collecting duct mediates the inhibitory effect of dietary salt on the open probability of the epithelial sodium channel ENaC and inhibits ENaC activity during aldosterone escape. Connexin 30 has been implicated in the luminal release of the ATP involved in the regulation of ENaC. An increase in collecting duct cell volume in response to manipulating water homeostasis increases ATP release. The subsequent activation of P2Y(2) receptors inhibits vasopressin-induced cAMP formation and water reabsorption, which facilitates water excretion and stabilizes cell volume. Thus recent studies have established the ATP/UTP/P2Y(2) receptor system as a relevant regulator of renal salt and water homeostasis and blood pressure regulation. The pathophysiological relevance and therapeutic potential remains to be determined, but dual effects of P2Y(2) receptor activation on both the vasculature and renal salt reabsorption implicate these receptors as potential therapeutic targets in hypertension.
Collapse
Affiliation(s)
- Volker Vallon
- Dept. of Medicine, Univ. of California San Diego, 92161, USA.
| | | |
Collapse
|
37
|
Abstract
Urea transport proteins were initially proposed to exist in the kidney in the late 1980s when studies of urea permeability revealed values in excess of those predicted by simple lipid-phase diffusion and paracellular transport. Less than a decade later, the first urea transporter was cloned. Currently, the SLC14A family of urea transporters contains two major subgroups: SLC14A1, the UT-B urea transporter originally isolated from erythrocytes; and SLC14A2, the UT-A group with six distinct isoforms described to date. In the kidney, UT-A1 and UT-A3 are found in the inner medullary collecting duct; UT-A2 is located in the thin descending limb, and UT-B is located primarily in the descending vasa recta; all are glycoproteins. These transporters are crucial to the kidney's ability to concentrate urine. UT-A1 and UT-A3 are acutely regulated by vasopressin. UT-A1 has also been shown to be regulated by hypertonicity, angiotensin II, and oxytocin. Acute regulation of these transporters is through phosphorylation. Both UT-A1 and UT-A3 rapidly accumulate in the plasma membrane in response to stimulation by vasopressin or hypertonicity. Long-term regulation involves altering protein abundance in response to changes in hydration status, low protein diets, adrenal steroids, sustained diuresis, or antidiuresis. Urea transporters have been studied using animal models of disease including diabetes mellitus, lithium intoxication, hypertension, and nephrotoxic drug responses. Exciting new animal models are being developed to study these transporters and search for active urea transporters. Here we introduce urea and describe the current knowledge of the urea transporter proteins, their regulation, and their role in the kidney.
Collapse
Affiliation(s)
- Janet D Klein
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|
38
|
Zhang Y, Listhrop R, Ecelbarger CM, Kishore BK. Renal sodium transporter/channel expression and sodium excretion in P2Y2 receptor knockout mice fed a high-NaCl diet with/without aldosterone infusion. Am J Physiol Renal Physiol 2011; 300:F657-68. [PMID: 21190950 PMCID: PMC4068121 DOI: 10.1152/ajprenal.00549.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 12/23/2010] [Indexed: 12/11/2022] Open
Abstract
The P2Y(2) receptor (P2Y2-R) antagonizes sodium reabsorption in the kidney. Apart from its effect in distal nephron, hypothetically, P2Y(2)-R may modulate activity/abundances of sodium transporters/channel subunits along the nephron via antagonism of aldosterone or vasopressin or interaction with mediators such as nitric oxide (NO), and prostaglandin E(2) (PGE(2)) or oxidative stress (OS). To determine the extent of the regulatory role of P2Y(2)-R in renal sodium reabsorption, in study 1, we fed P2Y(2)-R knockout (KO; n = 5) and wild-type (WT; n = 5) mice a high (3.15%)-sodium diet (HSD) for 14 days. Western blotting revealed significantly higher protein abundances for cortical and medullary bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2), medullary α-1-subunit of Na-K-ATPase, and medullary α-subunit of the epithelial sodium channel (ENaC) in KO vs. WT mice. Molecular analysis of urine showed increased excretion of nitrates plus nitrites (NOx), PGE(2), and 8-isoprostane in the KO, relative to WT mice, supporting a putative role for these molecules in determining alterations of proteins involved in sodium transport along the nephron. To determine whether genotype differences in response to aldosterone might have played a role in these differences due to HSD, in study 2 aldosterone levels were clamped (by osmotic minipump infusion). Clamping aldosterone (with HSD) led to significantly impaired natriuresis with elevated Na/H exchanger isoform 3 in the cortex, and NKCC2 in the medulla, and modest but significantly lower levels of NKCC2, and α- and β-ENaC in the cortex of KO vs. WT mice. This was associated with significantly reduced urinary NOx in the KO, although PGE(2) and 8-isoprostane remained significantly elevated vs. WT mice. Taken together, our results suggest that P2Y(2)-R is an important regulator of sodium transporters along the nephron. Pre- or postreceptor differences in the response to aldosterone, perhaps mediated via prostaglandins or changes in NOS activity or OS, likely play a role.
Collapse
Affiliation(s)
- Yue Zhang
- Nephrology Research, Department of Veterans Administration Salt Lake City Health Care System, Departments of Medicine Georgetown University, Washington, District of Columbia, USA
| | | | | | | |
Collapse
|
39
|
Boone M, Kortenoeven MLA, Robben JH, Tamma G, Deen PMT. Counteracting vasopressin-mediated water reabsorption by ATP, dopamine, and phorbol esters: mechanisms of action. Am J Physiol Renal Physiol 2011; 300:F761-71. [PMID: 21209006 DOI: 10.1152/ajprenal.00247.2010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Water homeostasis is regulated by a wide variety of hormones. When in need for water conservation, vasopressin, released from the brain, binds renal principal cells and initiates a signaling cascade resulting in the insertion of aquaporin-2 (AQP2) water channels in the apical membrane and water reabsorption. Conversely, hormones, including extracellular purines and dopamine, antagonize AVP-induced water permeability, but their mechanism of action is largely unknown, which was investigated here. Addition of these hormones to mpkCCD cells decreased total and plasma membrane abundance of AVP-induced AQP2, partly by increasing its internalization to vesicles and lysosomal degradation. This internalization was ubiquitin dependent, because the hormones increased AQP2 ubiquitination, and the plasma membrane localization of AQP2-K270R, which cannot be monoubiquitinated, was unaffected by these hormones. Both hormones also increased AQP2 phosphorylation at S261, which followed ubiquitination, but was not essential for hormone-induced AQP2 degradation. A similar process occurs in vivo, as incubation of dDAVP-treated kidney slices with both hormones also resulted in the internalization and S261 phosphorylation of AQP2. Both hormones also reduced cAMP and AQP2 mRNA levels, suggesting an additional effect on AQP2 gene transcription. Interestingly, phorbol esters only reduced AQP2 through the first pathway. Together, our results indicate that ATP and dopamine counteract AVP-induced water permeability by increasing AQP2 degradation in lysosomes, preceded by ubiquitin-dependent internalization, and by decreasing AQP2 gene transcription by reducing the AVP-induced cAMP levels.
Collapse
Affiliation(s)
- Michelle Boone
- Department of General and Environmental Physiology, University of Bari, Bari, Italy
| | | | | | | | | |
Collapse
|
40
|
Sinke AP, Caputo C, Tsaih SW, Yuan R, Ren D, Deen PMT, Korstanje R. Genetic analysis of mouse strains with variable serum sodium concentrations identifies the Nalcn sodium channel as a novel player in osmoregulation. Physiol Genomics 2010; 43:265-70. [PMID: 21177381 DOI: 10.1152/physiolgenomics.00188.2010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In central osmoregulation, a 1-2% rise in plasma osmolality is detected by specialized osmoreceptors located in the circumventricular organs of the hypothalamus. A disturbance in this tightly regulated balance will result in either hyponatremia or hypernatremia, which are both common electrolyte disorders in hospitalized patients. Despite the high clinical importance of hypo- and hypernatremia and the fact that this vital process has been studied for many years, the genes and corresponding proteins involved in this process are just beginning to be identified. To identify novel genes involved in the (patho-)physiology of osmoregulation, we therefore employed haplotype association mapping on an aging group of 27 inbred mouse strains. Serum sodium concentrations were determined in all strains at 6, 12, and 18 mo of age, and high-resolution mapping was performed for males and females separately. We identified a total of five loci associated with the serum sodium concentration of which the locus on chromosome 14, containing only one known gene (Nalcn), showed the strongest correlation. Within this locus three different haplotypes could be distinguished, which associated with different average serum sodium levels. The association of Nalcn with sodium levels was confirmed by analysis of heterozygous Nalcn knockout mice, which displayed hypernatremia compared with wild-type littermates. Our study demonstrates that Nalcn associates with serum sodium concentrations in mice and indicates that Nalcn is an important novel player in osmoregulation.
Collapse
Affiliation(s)
- Anne P Sinke
- Department of Physiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
41
|
Praetorius HA, Leipziger J. Intrarenal purinergic signaling in the control of renal tubular transport. Annu Rev Physiol 2010; 72:377-93. [PMID: 20148681 DOI: 10.1146/annurev-physiol-021909-135825] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Renal tubular epithelial cells receive hormonal input that regulates volume and electrolyte homeostasis. In addition, numerous intrarenal, local signaling agonists have appeared on the stage of renal physiology. One such system is that of intrarenal purinergic signaling. This system involves all the elements necessary for agonist-mediated intercellular communication. ATP is released from epithelial cells, which activates P2 receptors in the apical and basolateral membrane and thereby modulates tubular transport. Termination of the signal is conducted via the breakdown of ATP to adenosine. Recent far-reaching advances indicate that ATP is often used as a local transmitter for classical sensory transduction. This transmission apparently also applies to sensory functions in the kidney. Locally released ATP is involved in sensing of renal tubular flow or in detecting the distal tubular load of NaCl at the macula densa. This review describes the relevant aspects of local, intrarenal purinergic signaling and outlines its integrative concepts.
Collapse
Affiliation(s)
- Helle A Praetorius
- Department of Physiology and Biophysics, The Water and Salt Research Center, Aarhus University, Aarhus C, Denmark
| | | |
Collapse
|
42
|
The P2Y2 receptor sensitizes mouse bladder sensory neurons and facilitates purinergic currents. J Neurosci 2010; 30:2365-72. [PMID: 20147562 DOI: 10.1523/jneurosci.5462-09.2010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Sensitization of bladder afferents is an underlying contributor to the development and maintenance of painful bladder syndrome/interstitial cystitis. Extracellular purines and pyrimidines (e.g., ATP and UTP), released during bladder distension or from damaged cells after tissue insult, are thought to play an important role in bladder physiological and pathological states by actions at ionotropic P2X and metabotropic P2Y receptors. In the present study, we examined the ability of P2Y receptors to sensitize and modulate P2X-mediated responses in mouse bladder sensory neurons. UTP (a P2Y(2) and P2Y(4) agonist) increased excitability of bladder neurons by depolarizing resting membrane potential, increasing action potential firing, and facilitating responses to suprathreshold current injection as well as to P2X agonist application. These effects of UTP on bladder neuron excitability were blocked by the P2Y(2) receptor antagonist suramin. UTP also facilitated bladder neuron homomeric P2X(2) sustained currents and homomeric P2X(3) fast currents. The facilitatory effect of UTP on P2X(2) sustained currents was mediated by a G-protein-coupled P2Y(2) receptor/PKC pathway, whereas the effect of UTP on P2X(3) fast currents was G-protein independent. We also examined P2X and P2Y receptor expression in bladder neurons. P2Y(2) and P2Y(4) transcripts were detected in approximately 50 and approximately 20% of bladder neurons, respectively. Approximately 50% of P2X(2)- and P2X(3)-positive bladder neurons expressed P2Y(2) transcripts, whereas < or =25% of the same bladder neurons expressed P2Y(4) transcripts. These results support involvement of P2Y(2) receptors in bladder sensation, suggesting an important contribution to bladder neuron excitability and hypersensitivity.
Collapse
|
43
|
Corriden R, Insel PA. Basal release of ATP: an autocrine-paracrine mechanism for cell regulation. Sci Signal 2010; 3:re1. [PMID: 20068232 DOI: 10.1126/scisignal.3104re1] [Citation(s) in RCA: 268] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cells release adenosine triphosphate (ATP), which activates plasma membrane-localized P2X and P2Y receptors and thereby modulates cellular function in an autocrine or paracrine manner. Release of ATP and the subsequent activation of P2 receptors help establish the basal level of activation (sometimes termed "the set point") for signal transduction pathways and regulate a wide array of responses that include tissue blood flow, ion transport, cell volume regulation, neuronal signaling, and host-pathogen interactions. Basal release and autocrine or paracrine responses to ATP are multifunctional, evolutionarily conserved, and provide an economical means for the modulation of cell, tissue, and organismal biology.
Collapse
Affiliation(s)
- Ross Corriden
- Departments of Pharmacology and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
44
|
Zhang Y, Kohan DE, Nelson RD, Carlson NG, Kishore BK. Potential involvement of P2Y2 receptor in diuresis of postobstructive uropathy in rats. Am J Physiol Renal Physiol 2009; 298:F634-42. [PMID: 20007349 DOI: 10.1152/ajprenal.00382.2009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
AVP resistance of the medullary collecting duct (mCD) in postobstructive uropathy (POU) has been attributed to increased production of PGE2. P2Y2 receptor activation causes production of PGE2 by the mCD. We hypothesize that increased P2Y2 receptor expression and/or activity may contribute to the diuresis of POU. Sprague-Dawley rats were subjected to bilateral ureteral obstruction for 24 h followed by release (BUO/R, n = 17) or sham operation (SHM/O, n = 15) and euthanized after 1 wk or 12 days. BUO/R rats developed significant polydipsia, polyuria, urinary concentration defect, and increased urinary PGE2 and decreased aquaporin-2 protein abundance in the inner medulla compared with SHM/O rats. After BUO/R, the relative mRNA expression of P2Y2 and P2Y6 receptors was increased by 2.7- and 4.9-fold, respectively, without significant changes in mRNA expression of P2Y1 or P2Y4 receptor. This was associated with a significant 3.5-fold higher protein abundance of the P2Y2 receptor in BUO/R than SHM/O rats. When freshly isolated mCD fractions were challenged with different types of nucleotides (ATPgammaS, ADP, UTP, or UDP), BUO/R and SHM/O rats responded to only ATPgammaS and UTP and released PGE2, consistent with involvement of the P2Y2, but not P2Y6, receptor. ATPgammaS- or UTP-stimulated increases in PGE2 were much higher in BUO/R (3.20- and 2.28-fold, respectively, vs. vehicle controls) than SHM/O (1.68- and 1.30-fold, respectively, vs. vehicle controls) rats. In addition, there were significant 2.4- and 2.1-fold increases in relative mRNA expression of prostanoid EP1 and EP3 receptors, respectively, in the inner medulla of BUO/R vs. SHM/O rats. Taken together, these data suggest that increased production of PGE2 by the mCD in POU may be due to increased expression and activity of the P2Y2 receptor. Increased mRNA expression of EP1 and EP3 receptors in POU may also help accentuate PGE2-induced signaling in the mCD.
Collapse
Affiliation(s)
- Yue Zhang
- Nephrology Research, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah 84148, USA
| | | | | | | | | |
Collapse
|
45
|
Odgaard E, Praetorius HA, Leipziger J. AVP-stimulated nucleotide secretion in perfused mouse medullary thick ascending limb and cortical collecting duct. Am J Physiol Renal Physiol 2009; 297:F341-9. [PMID: 19515810 DOI: 10.1152/ajprenal.00190.2009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Extracellular nucleotides are local, short-lived signaling molecules that inhibit renal tubular transport via both luminal and basolateral P2 receptors. Apparently, the renal epithelium itself is able to release nucleotides. The mechanism and circumstances under which nucleotide release is stimulated remain elusive. Here, we investigate the phenomenon of nucleotide secretion in intact, perfused mouse medullary thick ascending limb (mTAL) and cortical collecting duct (CCD). The nucleotide secretion was monitored by a biosensor adapted to register nucleotides in the tubular outflow. Intracellular Ca(2+) concentration ([Ca(2+)](i)) was measured simultaneously in the biosensor cells and the renal tubule with fluo 4. We were able to identify spontaneous tubular nucleotide secretion in resting perfused mTAL. In this preparation, 10 nM AVP and 1-desamino-8-D-arginine vasopressin (dDAVP) induced robust [Ca(2+)](i) oscillations, whereas AVP in the CCD induced large, slow, and transient [Ca(2+)](i) elevations. Importantly, we identify that AVP/dDAVP triggers tubular secretion of nucleotides in the mTAL. After addition of AVP/dDAVP, the biosensor registered bursts of nucleotides in the tubular perfusate, corresponding to a tubular nucleotide concentration of approximately 0.2-0.3 microM. A very similar response was observed after AVP stimulation of CCDs. Thus AVP stimulated tubular secretion of nucleotides in a burst-like pattern with peak tubular nucleotide concentrations in the low-micromolar range. We speculate that local nucleotide signaling is an intrinsic feedback element of hormonal control of renal tubular transport.
Collapse
Affiliation(s)
- Elvin Odgaard
- Department of Physiology and Biophysics, The Water and Salt Research Center, Aarhus University, Aarhus C, Denmark
| | | | | |
Collapse
|
46
|
Zhang Y, Nelson RD, Carlson NG, Kamerath CD, Kohan DE, Kishore BK. Potential role of purinergic signaling in lithium-induced nephrogenic diabetes insipidus. Am J Physiol Renal Physiol 2009; 296:F1194-201. [PMID: 19244398 DOI: 10.1152/ajprenal.90774.2008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Lithium (Li)-induced nephrogenic diabetes insipidus (NDI) has been attributed to the increased production of renal prostaglandin (PG)E(2). Previously we reported that extracellular nucleotides (ATP/UTP), acting through P(2y2) receptor in rat medullary collecting duct (mCD), produce and release PGE(2). Hence we hypothesized that increased production of PGE(2) in Li-induced NDI may be mediated by enhanced purinergic signaling in the mCD. Sprague-Dawley rats were fed either control or Li-added diet for 14 or 21 days. Li feeding resulted in marked polyuria and polydipsia associated with a decrease in aquaporin (AQP)2 protein abundance in inner medulla ( approximately 20% of controls) and a twofold increase in urinary PGE(2). When acutely challenged ex vivo with adenosine 5'-O-(3-thiotriphosphate) (ATPgammaS), UTP, or ADP, mCD of Li-fed rats showed significantly higher increases (50-130% over control diet-fed rats) in PGE(2) production, indicating that more than one subtype of P(2y) receptor is involved. This was associated with a 3.4-fold increase in P(2y4), but not P(2y2), receptor mRNA expression in the inner medulla of Li-fed rats compared with control diet-fed rats. Confocal laser immunofluorescence microscopy revealed predominant localization of both P(2y2) and P(2y4) receptors in the mCD of control or Li diet-fed rats. Together, these data indicate that in Li-induced NDI 1) purinergic signaling in the mCD is sensitized with increased production of PGE(2) and 2) P(2y2) and/or P(2y4) receptors may be involved in the enhanced purinergic signaling. Our study also reveals the potential beneficial effects of P(2y) receptor antagonists in the treatment and/or prevention of Li-induced NDI.
Collapse
Affiliation(s)
- Yue Zhang
- Nephrology Research (151M VA SCL Health Care System, 500 Foothill Dr., Salt Lake City, UT 84148. )
| | | | | | | | | | | |
Collapse
|