eph1497.pdf;jsessionid=AEB50AF8A0F2451036D47F9956C20E77.f03t01

download eph1497.pdf;jsessionid=AEB50AF8A0F2451036D47F9956C20E77.f03t01

of 6

Transcript of eph1497.pdf;jsessionid=AEB50AF8A0F2451036D47F9956C20E77.f03t01

  • 8/17/2019 eph1497.pdf;jsessionid=AEB50AF8A0F2451036D47F9956C20E77.f03t01

    1/6

    p

    y

    gy

    1140   Exp Physiol  99.9 (2014) pp 1140–1145

    Symposium ReportSymposium Report

    Intestinal regulation of urinary sodium excretion and the

    pathophysiology of diabetic kidney disease: a focus onglucagon-like peptide 1 and dipeptidyl peptidase 4

    Volker Vallon1,2,3 and Neil G. Docherty 4

    1Department of Medicine, University of California San Diego, La Jolla, CA, USA 2 Department of Pharmacology, University of California San Diego, La Jolla, CA, USA 3Department of Veterans Affairs, San Diego Healthcare System, San Diego, CA, USA 4 Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences,

    University College Dublin, Belfield, Dublin 4, Ireland 

    New Findings    What is the topic of this review?

    This review describes how hyperglycaemia and attendant increases in proximal tubular

    growth and sodium reclamation form the mechanistic basis for the tubular hypothesis of 

    hyperfiltrationandnephropathyinthekidneysofpatientswithdiabetes.Thereviewhighlights

    how signals arising from the gastrointestinal tract may be capable of modulating this response

    and how pharmacological enhancement of these signals may ameliorate the progression of 

    diabetic kidney disease.    What advances does it highlight?

    The review highlights the potential of glucagon-like peptide 1 analogues, glucagon-like

    peptide 1 receptor agonists and dipeptidyl peptidase 4 inhibitors to exercise renoprotective

    effects in diabetes independent of their classical effects on endocrine pancreas.

    The tubular hypothesis of glomerular filtration and nephropathy in diabetes is a

    pathophysiological concept that assigns a critical role to the tubular system, including proximal

    tubular hyper-reabsorption and growth, which is relevant for early glomerular hyperfiltration

    and later chronic kidney disease. Here we focus on how harnessing the bioactivity of hormones

    released from the gut may ameliorate the early effects of diabetes on the kidney in part by 

    attenuating proximal tubular hyper-reabsorption and growth. The endogenous tone of the

    glucagon-like peptide 1 (GLP-1)/GLP-1 receptor (GLP-1R) system and its pharmacological

    activation are nephroprotective in diabetes independent of changes in blood glucose. This is

    associated with suppression of increases in kidney weight and glomerular hyperfiltration, which

    may reflect, at least in part, its inhibitory effects on tubular hyper-reabsorption and growth.Inhibition of dipeptidyl peptidase 4 (DPP-4) is also nephroprotective independent of changes

    in blood glucose and involves GLP-1/GLP-1R-dependent and -independent mechanisms. The

    GLP-1R agonist exendin-4 induces natriuresis via activation of the GLP-1R. In contrast, DPP4

    inhibition increases circulating GLP-1, but drives a GLP-1R-independent natriuretic response,

    implying a role for other DPP-4 substrates. The extent to which the intrarenal DPP-4/GLP-1

    DOI: 10.1113/expphysiol.2014.078766   C 2014 The Authors. Experimental Physiology   C 2014 The Physiological Society

  • 8/17/2019 eph1497.pdf;jsessionid=AEB50AF8A0F2451036D47F9956C20E77.f03t01

    2/6

    Exp Physiol  99.9 (2014) pp 1140–1145   1141Glucagon-like peptide 1 and dipeptidyl peptidase 4 in diabetic kidney disease

    receptor system contributes to all these changes remains to be established, as does the direct

    impact of the system on renal inflammation.

    (Received 15 May 2014; accepted after revision 23 July 2014)

    Corresponding author V. Vallon: University of California and VA San Diego Healthcare System, 3350 La Jolla Village

    Drive (9151), San Diego, CA 92161, USA. Email: [email protected]

    Introduction

    Diabetes is the single largest contributor to the growingprevalence of chronic kidney disease (CKD; Collins  et al.2009). In a subset of diabetic patients, the kidneysgrow large and glomerular filtration rate (GFR) becomessupranormal early during the disease, both of which arerisk factors for the later development of CKD. The tubularhypothesis of glomerular filtration and nephropathy isa pathophysiological concept to explain these changes(Fig. 1).

    The tubular hypothesis of glomerular hyperfiltration

    and nephropathy in diabetes

    The early distal tubule of every nephron makes contactwith the vascular pole of its own glomerulus. At thissite, the macula densa cells sense the luminal Na+,Cl− and K+ concentrations and adjust single-nephronGFR to stabilize delivery to the further distal nephron,where fine regulation of salt and fluid homeostasis isestablished. Diabetescauses a primary hyper-reabsorptionin the tubular segments upstream of the macula densa,especially in the proximal tubule (Vallon, 2011; Vallon

    & Thomson, 2012). Through the physiology of thedescribed tubuloglomerular feedback, single-nephronGFR is increased in diabetes in order to normalize saltdelivery to the distal nephron. The hyper-reabsorptionin the diabetic kidney is due to increased filtered glucoseload, which enhances glucose and sodium uptake throughsodium–glucose cotransport (SGLT; Vallon, 2011; Vallon& Thomson, 2012). As a consequence, genetic andpharmacological inhibition of SGLT2 blunts diabetichyperfiltration independent of changes in blood glucose(Vallon  et al.  2013, 2014). The second mechanism thatenhances tubular reabsorption upstream of the maculadensa in diabetes is tubular growth (Vallon, 2011; Vallon

    & Thomson, 2012).The diabetic milieu triggers early tubular cell

    proliferation, but the induction of transforming growthfactor-β and cyclin-dependent kinase inhibitors causes aG1   cell cycle arrest and switch to tubular hypertrophy and a senescence-like phenotype (Vallon, 2011; Vallon &Thomson, 2012). While this growth phenotype explainsunusual responses, such as the salt paradox of the early diabetic kidney (an inverse relationship between NaClintake and GFR;Vallon etal. 2002),the activatedmolecularpathways may set the stage for tubulointerstitial injury and

    diabetic nephropathy (Vallon, 2011; Vallon & Thomson,2012). In other words, we hypothesize that the tubulargrowth response to hyperglycaemia differs from patient topatient and explains early changes in the diabetic kidney,such as glomerular hyperfiltration or the salt paradox,but also sets the stage for later diabetic nephropathy andCKD (Vallon, 2011; Vallon & Thomson, 2012). Complex interactions of glucose and growth factors act on thetubular cells to promote inflammation, fibrosis, oxidativestress and hypoxia (Vallon, 2011; Vallon & Thomson,2012).

    Evidence exists to suggest that the processes describedabove may be amenable to inhibition via the tonic andpostprandial actions of hormonal signals derived fromthe gut, which link sensing of nutrient intake in thegastrointestinal lumen to modulation of renal tubularfunction.

    The gut–kidney natriuretic axis

    Seminal studies in humans and in experimental animalmodels have indicated that oral ingestion of sodiumchloride (NaCl) has a greater natriuretic effect than

    intravenous administration and that these effects appearto be independent of changes in circulating aldosteroneor atrial natriuretic peptide, as reviewed by Michell et al.(2008).

    Many gut-derived hormones are natriuretic, includingglucagon, glucagon-like peptide 1 (GLP-1), amylin, secr-etin, vasointestinal polypeptide, gastrin/cholecystokinin,peptide YY and uroguanylin (Forte, 2004; Michell  et al.2008). While some of these hormones activate adenylylcyclase, others inhibit this enzyme (or the local isoform).Given that inhibition of reabsorption can be induced by activation of adenylyl cyclase in the proximal tubule butalso by inhibition of this enzyme in the distal nephron

    and collecting duct, we hypothesize that gut-derivednatriuretic hormones act on different sites in the kidney.To speculate further, dietary composition (including Na+,Cl−, K+, Ca2+, phosphate, glucose, amino acids andprotons) may also determine the qualitative make-up of gut hormone ‘cocktails’ in order for adjustments in renalexcretion to track the specific composition of dietary intake closely. Very little is known about this issue, andin the following section we focus the discussion ononly one of the natriuretic gut-derived peptides, namely GLP-1.

    C 2014 The Authors. Experimental Physiology   C 2014 The Physiological Society

  • 8/17/2019 eph1497.pdf;jsessionid=AEB50AF8A0F2451036D47F9956C20E77.f03t01

    3/6

    1142   Exp Physiol  99.9 (2014) pp 1140–1145V. Vallon and N. G. Docherty

    Natriuretic responses to GLP-1 receptor agonists and

    dipeptidyl peptidase 4 (DPP-4) inhibitors

    Glucagon-like peptide 1 is an incretin hormone secretedfrom enteroendocrine L cells in the intestine duringthe postprandial phase and stimulates glucose-dependentinsulin release (Drucker & Nauck, 2006). After secretion,

    active GLP-1 is rapidly cleaved by membrane-boundDPP-4 in capillaries, thereby limiting systemic levels of bioactive GLP-1 and reducing its half-life to  

  • 8/17/2019 eph1497.pdf;jsessionid=AEB50AF8A0F2451036D47F9956C20E77.f03t01

    4/6

    Exp Physiol  99.9 (2014) pp 1140–1145   1143Glucagon-like peptide 1 and dipeptidyl peptidase 4 in diabetic kidney disease

    Glucagon-like peptide 1 is one attractive candidate in thisregard.

    In order to define the role of the GLP-1/GLP-1R system further, we performed studies in mice lackingthe GLP-1R (Glp1r −/−). These   Glp1r −/− mice have anincreased GFR (Rieg  et al.  2012); one explanation couldbe that GLP-1R knockout removes its tonic inhibitory influence on proximal tubular reabsorption, which lowersthe NaCl concentration at the macula densa and increasesGFR via tubuloglomerular feedback. We showed that thepresence of the GLP-1R is critical for exendin-4-inducednatriuresis and phosphorylation of NHE3. Natriureticresponses to acute oral NaCl loading and the natriureticeffects of the DPP-4 inhibitor alogliptin are, however,surprisingly both independent of the GLP-1R. Moreover,the alogliptin-induced natriuresis is not associated withchanges in NHE3 phosphorylation, arguing that NHE3phosphorylation may notbe a critical step in the inductionof natriuresis (Rieg et al. 2012).

    In the   Glp1r −/− mice as well as in response tothe application of GLP-1 or other GLP-1R agonists,such as extendin-4, the integrated GFR response willbe determined by the magnitude of the effects onproximal reabsorption and tubuloglomerular feedback versus a potential direct renal vasodilator effect of GLP-1R activation (Green   et al.   2008; Crajoinas   et al.   2011).Similar to studies in obese humans (Gutzwiller   et al.2004), we found that exendin-4 lowered GFR in obesedb /db   mice (Rieg   et al.   2012). The observed decreasein GFR in diabetes in response to GLP-1/exendin-4may relate to a stronger influence of proximal transport

    inhibition on GFR via tubuloglomerular feedback. Thetubular hypothesis outlined above proposes that aprimary proximal tubular hyper-reabsorption contributesto glomerular hyperfiltration in the diabetic kidney.The GFR-lowering effect of GLP-1/exendin-4 in diabetesmay indicate that this tubular hyper-reabsorption isparticularly sensitive to inhibition by GLP-1R activation;as a consequence, with regard to the net GFR response,the tubular effect of GLP-1R activation may dominateany potential direct renal vascular effect in diabetes.Alternatively or in addition, the renal vasodilator responseto GLP-1R activation could be blunted in diabetes. We alsonoted a blunted natriuretic response to alogliptin in db /db 

    mice, indicating that diabetes may affect the availability of the natriuretic factor regulated by DPP-4 inhibition (Rieget al. 2012).

    Does GLP-1 R activation have direct effects

    on kidney injury?

    Kodera and colleagues demonstrated a glucose-independent anti-inflammatory and protective role of GLP-1R signalling in type 1 diabetic rats (Kodera  et al.2011, 2014); the beneficial effects of exendin-4 in diabetic

    rats included a reduction in glomerular hypertrophy,mesangial matrix expansion, albuminuria, macrophageinfiltration and markers of fibrosis and oxidative stress.The authors reported GLP-1 receptor expression onmonocytes/macrophages and glomerular endothelial cellsand the capacity for GLP-1 to oppose pro-inflammatory activation of both cell types   in vitro . While exendin-4did not affect kidney weight or tubular hypertrophy, itprevented the increase in creatinine clearance in diabeticrats. This last effect is consistent with the above-describedGFR-lowering response to GLP-1/exendin-4 observed inobese humans (Gutzwiller et al. 2004) and in obese db /db mice (Rieg et al. 2012).

    Fujita  et al.  (2014) reported that the GLP-1R agonistliraglutide attenuated diabetes-induced renal changesin wild-type Akita mice (a genetic model of type 1diabetes), including attenuation of the increase inkidney weight and GFR, without significantly impactingupon glycaemic control. On the contrary, GLP-1R 

    knockout Akita mice showed increased albuminuria,mesangial expansion, markers of glomerular and renaloxidative stress, and further increases in kidney weight and GFR. Inhibition of DPP-4 also inducedglucose-independent nephroprotective effects in type 1diabetes that involved anti-inflammatory actions (Koderaet al.  2014) and suppression of the advanced glycationend-products–receptor axis (Nakashima et al. 2014).

    This provides evidence to support the contentionthat the endogenous GLP-1/GLP-1R system and itstherapeutic activation suppress diabetic kidney growthand injury and glomerular hyperfiltration, which

    may be due to it inhibiting tubular growth andhyper-reabsorption and affording protection againstthe cytotoxic effects of hyperglycaemia and throughenhancement of anti-inflammatory signalling (Fig. 1).

    The argument for a more direct nephroprotective effectfor DPP-4 inhibition and GLP-1 action is supportedby findings in euglycaemic renal injury models, withrecent studies showing that DPP-4 inhibition protects thekidney against nephrotoxins through GLP-1-independent(indoxylsulfate; Wang etal. 2014) and GLP-1R-dependentmechanisms (cisplatin; Katagiri et al. 2013).

    Summary and perspectivesGut hormones can regulate urinary sodium and fluidexcretion, but thephysiological relevance and their specificroles remain unclear. The antidiabetic drug extendin-4induces natriuresis via activation of the GLP-1R. Incontrast, the natriuretic effect of the DPP4 inhibitoralogliptin is independent of the GLP-1R. The GLP-1R is not critical to the ability to excrete an oral NaClload, but the observed increase in GFR in   Glp1r −/−

    mice may reflect a loss of tonic inhibition of proximalreabsorption by the endogenous GLP-1/GLP-1R system,

    C 2014 The Authors. Experimental Physiology   C 2014 The Physiological Society

  • 8/17/2019 eph1497.pdf;jsessionid=AEB50AF8A0F2451036D47F9956C20E77.f03t01

    5/6

    1144   Exp Physiol  99.9 (2014) pp 1140–1145V. Vallon and N. G. Docherty

    which normally suppresses GFR through the physiology of tubuloglomerular feedback. Inhibition of DPP-4 can benephroprotectiveindependent of changes in blood glucosethrough GLP-1/GLP-1R-dependent and -independentmechanisms. Preclinical evidence points to a role foran endogenous tone within the GLP-1/GLP-1R systemacting to limit inflammation and attenuate diabetic kidney growth and glomerular hyperfiltration. Therapeuticactivation of theGLP-1R canfurther enhance these effects.Further studies are needed to provide better definition of the effects of GLP-1 on the vasculature and tubular systemin the diabetic kidney, including the role of the intrarenalDPP-4/GLP-1R system.

    These data support a future focus on the efficacy of GLP-1- and DPP-4-based antidiabetic regimensin relationship to renal end-points in large cohortstudies. As GLP-1 levels are also augmented followingRoux-en-Y gastric bypass, sleeve gastrectomy andendoluminal sleeve delivery, the establishment of a

    potential blood pressure-lowering and nephroprotectivegut–kidney axis after these bariatric procedures warrantsfurther investigation.

    Call for comments

    Readers are invited to give their opinion on this article.To submit a comment, go to: http://ep.physoc.org/letters/submit/expphysiol;99/9/1140

    References

    Collins AJ, Foley RN, Herzog C, Chavers B, Gilbertson D,

    Ishani A, Kasiske B, Liu J, Mau LW, McBean M, Murray A, StPeter W, Guo H, Li Q, Li S, Li S, Peng Y, Qiu Y, Roberts T,Skeans M, Snyder J, Solid C, Wang C, Weinhandl E, Zaun D,Arko C, Chen SC, Dalleska F, Daniels F, Dunning S, Ebben J,Frazier E, Hanzlik C, Johnson R, Sheets D, Wang X, ForrestB, Constantini E, Everson S, Eggers P & Agodoa L (2009).United States Renal Data System 2008 Annual Data Report.Am J Kidney Dis  53, S1–S374.

    Crajoinas RO, Oricchio FT, Pessoa TD, Pacheco BP, Lessa LM,Malnic G & Girardi AC (2011). Mechanisms mediating thediuretic and natriuretic actions of the incretin hormoneglucagon-like peptide-1. Am J Physiol Renal Physiol  301,F355–F363.

    Drucker DJ & Nauck MA (2006). The incretin system:glucagon-like peptide-1 receptor agonists and dipeptidylpeptidase-4 inhibitors in type 2 diabetes. Lancet  368,1696–1705.

    Forte LR Jr (2004). Uroguanylin and guanylin peptides:pharmacology and experimental therapeutics. Pharmacol Ther  104, 137–162.

    Fujita H, Morii T, Fujishima H, Sato T, Shimizu T, Hosoba M,Tsukiyama K, Narita T, Takahashi T, Drucker DJ, Seino Y &Yamada Y (2014). The protective roles of GLP-1R signalingin diabetic nephropathy: possible mechanism andtherapeutic potential. Kidney Int  85, 579–589.

    Girardi AC, Degray BC, Nagy T, Biemesderfer D & Aronson PS(2001). Association of Na+-H+ exchanger isoform NHE3and dipeptidyl peptidase IV in the renal proximal tubule. J Biol Chem 276, 46671–46677.

    Girardi AC, Fukuda LE, Rossoni LV, Malnic G & Rebouças NA(2008). Dipeptidyl peptidase IV inhibition downregulatesNa+–H+ exchanger NHE3 in rat renal proximal tubule. Am J 

    Physiol Renal Physiol  294, F414–F422.Girardi AC, Knauf F, Demuth HU & Aronson PS (2004). Role

    of dipeptidyl peptidase IV in regulating activity of Na+/H+

    exchanger isoform NHE3 in proximal tubule cells.  Am J Physiol Cell Physiol  287, C1238–C1245.

    Green BD, Hand KV, Dougan JE, McDonnell BM, Cassidy RS& Grieve DJ (2008). GLP-1 and related peptides causeconcentration-dependent relaxation of rat aorta through apathway involving KATP and cAMP. Arch Biochem Biophys 478, 136–142.

    Gutzwiller JP, Hruz P, Huber AR, Hamel C, Zehnder C, DreweJ, Gutmann H, Stanga Z, Vogel D & Beglinger C (2006).Glucagon-like peptide-1 is involved in sodium and waterhomeostasis in humans. Digestion  73, 142–150.

    Gutzwiller JP, Tschopp S, Bock A, Zehnder CE, Huber AR,Kreyenbuehl M, Gutmann H, Drewe J, Henzen C, Goeke B &Beglinger C (2004). Glucagon-like peptide 1 inducesnatriuresis in healthy subjects and in insulin-resistant obesemen. J Clin Endocrinol Metab  89, 3055–3061.

    Katagiri D, Hamasaki Y, Doi K, Okamoto K, Negishi K,Nangaku M & Noiri E (2013). Protection of glucagon-likepeptide-1 in cisplatin-induced renal injury elucidates gut-kidney connection. J Am Soc Nephrol  24,2034–2043.

    Kenny AJ, Booth AG, George SG, Ingram J, Kershaw D, WoodEJ & Young AR (1976). Dipeptidyl peptidase IV,a kidney brush-border serine peptidase. Biochem J  157,

    169–182.Kim M, Platt MJ, Shibasaki T, Quaggin SE, Backx PH, Seino S,Simpson JA & Drucker DJ (2013). GLP-1 receptor activationand Epac2 link atrial natriuretic peptide secretion to controlof blood pressure. Nat Med  19, 567–575.

    Kodera R, Shikata K, Kataoka HU, Takatsuka T, Miyamoto S,Sasaki M, Kajitani N, Nishishita S, Sarai K, Hirota D, Sato C,Ogawa D & Makino H (2011). Glucagon-like peptide-1receptor agonist ameliorates renal injury through itsanti-inflammatory action without lowering blood glucoselevel in a rat model of type 1 diabetes.  Diabetologia  54,965–978.

    Kodera R, Shikata K, Takatsuka T, Oda K, Miyamoto S, KajitaniN, Hirota D, Ono T, Usui HK & Makino H (2014).

    Dipeptidyl peptidase-4 inhibitor ameliorates early renalinjury through its anti-inflammatory action in a rat modelof type 1 diabetes. Biochem Biophys Res Commun  443,828–833.

    Michell AR, Debnam ES & Unwin RJ (2008). Regulation of renal function by the gastrointestinal tract: potential role of gut-derived peptides and hormones. Annu Rev Physiol  70,379–403.

    Moreno C, Mistry M & Roman RJ (2002). Renal effects of glucagon-like peptide in rats. Eur J Pharmacol  434,163–167.

    C 2014 The Authors. Experimental Physiology   C 2014 The Physiological Society

  • 8/17/2019 eph1497.pdf;jsessionid=AEB50AF8A0F2451036D47F9956C20E77.f03t01

    6/6

    Exp Physiol  99.9 (2014) pp 1140–1145   1145Glucagon-like peptide 1 and dipeptidyl peptidase 4 in diabetic kidney disease

    Nakashima S, Matsui T, Takeuchi M & Yamagishi SI (2014).Linagliptin blocks renal damage in type 1 diabetic rats by suppressing advanced glycation end products-receptor axis.Horm Metab Res , DOI: 10.1055/s-0034-1371892

    Rieg T, Gerasimova M, Murray F, Masuda T, Tang T, Rose M,Drucker DJ & Vallon V (2012). Natriuretic effect by exendin-4, but not the DPP-4 inhibitor alogliptin, is

    mediated via the GLP-1 receptor and preserved in obesetype 2 diabetic mice. Am J Physiol Renal Physiol  303,F963–F971.

    Schlatter P, Beglinger C, Drewe J & Gutmann H (2007).Glucagon-like peptide 1 receptor expression inprimary porcine proximal tubular cells.  Regul Pept  141,120–128.

    Skov J, Holst JJ, Gøtze JP, Frøkiær J & Christiansen JS (2014).Glucagon-like peptide-1: effect on pro-atrialnatriuretic peptide in healthy males. Endocr Connect  3,11–16.

    Thomson SC, Kashkouli A & Singh P (2013). Glucagon-likepeptide-1 receptor stimulation increases GFR and suppressesproximal reabsorption in the rat. Am J Physiol Renal Physiol 304, F137–F144.

    Vallon V (2011). The proximal tubule in the pathophysiology of the diabetic kidney. Am J Physiol Regul Integr Comp Physiol  300, R1009–R1022.

    Vallon V, Gerasimova M, Rose MA, Masuda T, Satriano J,Mayoux E, Koepsell H, Thomson SC & Rieg T (2014).SGLT2 inhibitor empagliflozin reduces renal growth andalbuminuria in proportion to hyperglycemia and preventsglomerular hyperfiltration in diabetic Akita mice. Am J Physiol Renal Physiol  306, F194–F204.

    Vallon V, Huang DY, Deng A, Richter K, Blantz RC &Thomson S (2002). Salt-sensitivity of proximal reabsorptionalters macula densa salt and explains the paradoxical effectof dietary salt on glomerular filtration rate in diabetesmellitus. J Am Soc Nephrol  13, 1865–1871.

    Vallon V, Rose M, Gerasimova M, Satriano J, Platt KA, KoepsellH, Cunard R, Sharma K, Thomson SC & Rieg T (2013).

    Knockout of Na-glucose transporter SGLT2 attenuateshyperglycemia and glomerular hyperfiltration but notkidney growth or injury in diabetes mellitus. Am J Physiol Renal Physiol  304, F156–F167.

    Vallon V & Thomson SC (2012). Renal function in diabeticdisease models: the tubular system in the pathophysiology of the diabetic kidney. Annu Rev Physiol  74, 351–375.

    Wang WJ, Chang CH, Sun MF, Hsu SF & Weng CS (2014).DPP-4 inhibitor attenuates toxic effects of indoxyl sulfate onkidney tubular cells. PloS ONE  9, e93447.

    Additional Information

    Competing interests

    V.V. has received within the past 12 months researchgrant support for basic science studies from BoehringerIngelheim Pharma GmbH & Co.KG and Amylin/Brystol-MyersSquibb/Astra Zeneca.

    Funding

    This work was supported by NIH grants R01DK56248,R01HL94728, the UAB/UCSD O’Brien Center of Acute Kidney Injury, NIH-P30DK079337 and the Department of VeteransAffairs.

    C 2014 The Authors. Experimental Physiology   C 2014 The Physiological Society