+ All Categories
Home > Documents > Targeted Deletion of Klotho in Kidney Distal Tubule Disrupts Mineral Metabolism

Targeted Deletion of Klotho in Kidney Distal Tubule Disrupts Mineral Metabolism

Date post: 10-Oct-2016
Category:
Upload: t-e
View: 212 times
Download: 0 times
Share this document with a friend
11
BASIC RESEARCH www.jasn.org Targeted Deletion of Klotho in Kidney Distal Tubule Disrupts Mineral Metabolism Hannes Olauson,* Karolina Lindberg,* Risul Amin,* Ting Jia,* Annika Wernerson, Göran Andersson, and Tobias E. Larsson* *Division of Renal Medicine, Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden; Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden; and Department of Nephrology, Karolinska University Hospital, Stockholm, Sweden ABSTRACT Renal Klotho controls mineral metabolism by directly modulating tubular reabsorption of phosphate and calcium and by acting as a co-receptor for the phosphaturic and vitamin Dregulating hormone broblast growth factor-23 (FGF23). Klotho null mice have a markedly abnormal phenotype. We sought to determine effects of renal-specic and partial deletion of Klotho to facilitate investigation of its roles in health and dis- ease. We generated a mouse model with partial deletion of Klotho in distal tubular segments (Ksp-KL 2/2 ). In contrast to Klotho null mice, Ksp-KL 2/2 mice were fertile, had a normal gross phenotype, and did not have vascular or tubular calcication on renal histology. However, Ksp-KL 2/2 mice were hyperphos- phatemic with elevated FGF23 levels and abundant expression of the sodium-phosphate cotransporter Npt2a at the brush border membrane. Serum calcium and 1,25-dihydroxyvitamin D 3 levels were normal but parathyroid hormone levels were decreased. TRPV5 protein was reduced with a parallel mild increase in urinary calcium excretion. Renal expression of vitamin D regulatory enzymes and vitamin D receptor was higher in Ksp-KL 2/2 mice than controls, suggesting increased turnover of vitamin D metabolites and a functional increase in vitamin D signaling. There was a threshold effect of residual renal Klotho expression on FGF23: deletion of .70% of Klotho resulted in FGF23 levels 30250 times higher than in wild-type mice. A subgroup of Ksp-KL 2/2 mice with normal phosphate levels had elevated FGF23, suggesting a Klotho- derived renal-bone feedback loop. Taken together, renal FGF23-Klotho signaling, which is disrupted in CKD, is essential for homeostatic control of mineral metabolism. J Am Soc Nephrol 23: cccccc, 2012. doi: 10.1681/ASN.2012010048 Type I membrane-bound a-Klotho (Klotho) is ex- pressed in tissues requiring abundant calcium transport, such as the kidneys and parathyroid glands. It was originally described as a senescence- related protein, because mice lacking functional Klotho protein develop a syndrome resembling hu- man aging, including shortened life span, atheroscle- rosis, vascular calcication, and osteoporosis. 1 Klotho regulates mineral metabolism by pro- moting renal calcium reabsorption through stabi- lization of the transient receptor potential vanilloid-5 (TRPV5) channel in distal tubules 2 and inhibits in- organic phosphate reabsorption through decreased expression and activity of the sodium-dependent phosphate cotransporter type 2a and c (Npt2a, Npt2c) in proximal tubules. 3 Furthermore, Klotho forms a specic receptor complex with broblast growth factor receptor 1c (FGFR1c) that transmits signaling of the circulating hormone FGF23. 4 Renal FGF23-Klotho signaling leads to internalization of Npt2a and Npt2c 57 and alterations in Cyp27B1 and Cyp24A1 transcripts encoding regulatory Received January 19, 2012. Accepted June 25, 2012. Published online ahead of print. Publication date available at www.jasn.org. Correspondence: Dr. Tobias E. Larsson, Clinical Research Cen- ter, Sixth Floor, Novumhuset, Karolinska University Hospital in Huddinge, SE-14186 Stockholm, Sweden. Email: tobias.larsson@ ki.se Copyright © 2012 by the American Society of Nephrology J Am Soc Nephrol 23: cccccc, 2012 ISSN : 1046-6673/2310-ccc 1
Transcript

BASIC RESEARCH www.jasn.org

Targeted Deletion of Klotho in Kidney Distal TubuleDisrupts Mineral Metabolism

Hannes Olauson,* Karolina Lindberg,* Risul Amin,* Ting Jia,* Annika Wernerson,†

Göran Andersson,† and Tobias E. Larsson*‡

*Division of Renal Medicine, Department of Clinical Science, Intervention, and Technology, Karolinska Institutet,Stockholm, Sweden; †Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet and KarolinskaUniversity Hospital, Stockholm, Sweden; and ‡Department of Nephrology, Karolinska University Hospital, Stockholm,Sweden

ABSTRACTRenal Klotho controls mineral metabolism by directly modulating tubular reabsorption of phosphate andcalcium and by acting as a co-receptor for the phosphaturic and vitamin D–regulating hormone fibroblastgrowth factor-23 (FGF23).Klotho nullmice have amarkedly abnormal phenotype.We sought to determineeffects of renal-specific and partial deletion of Klotho to facilitate investigation of its roles in health and dis-ease. We generated amousemodel with partial deletion of Klotho in distal tubular segments (Ksp-KL2/2).In contrast to Klotho null mice, Ksp-KL2/2 mice were fertile, had a normal gross phenotype, and did nothave vascular or tubular calcification on renal histology. However, Ksp-KL2/2 mice were hyperphos-phatemic with elevated FGF23 levels and abundant expression of the sodium-phosphate cotransporterNpt2a at the brush border membrane. Serum calcium and 1,25-dihydroxyvitamin D3 levels were normalbut parathyroid hormone levels were decreased. TRPV5 protein was reduced with a parallel mild increasein urinary calcium excretion. Renal expression of vitamin D regulatory enzymes and vitaminD receptor washigher in Ksp-KL2/2 mice than controls, suggesting increased turnover of vitamin D metabolites and afunctional increase in vitamin D signaling. There was a threshold effect of residual renal Klotho expressionon FGF23: deletion of.70%of Klotho resulted in FGF23 levels 30–250 times higher than inwild-typemice.A subgroup of Ksp-KL2/2 mice with normal phosphate levels had elevated FGF23, suggesting a Klotho-derived renal-bone feedback loop. Taken together, renal FGF23-Klotho signaling, which is disrupted inCKD, is essential for homeostatic control of mineral metabolism.

J Am Soc Nephrol 23: ccc–ccc, 2012. doi: 10.1681/ASN.2012010048

Type I membrane-bound a-Klotho (Klotho) is ex-pressed in tissues requiring abundant calciumtransport, such as the kidneys and parathyroidglands. It was originally described as a senescence-related protein, because mice lacking functionalKlotho protein develop a syndrome resembling hu-man aging, including shortened life span, atheroscle-rosis, vascular calcification, and osteoporosis.1

Klotho regulates mineral metabolism by pro-moting renal calcium reabsorption through stabi-lization of the transient receptor potential vanilloid-5(TRPV5) channel in distal tubules2 and inhibits in-organic phosphate reabsorption through decreasedexpression and activity of the sodium-dependentphosphate cotransporter type 2a and c (Npt2a,Npt2c) in proximal tubules.3 Furthermore, Klotho

forms a specific receptor complex with fibroblastgrowth factor receptor 1c (FGFR1c) that transmitssignaling of the circulating hormone FGF23.4 RenalFGF23-Klotho signaling leads to internalization ofNpt2a and Npt2c5–7 and alterations in Cyp27B1and Cyp24A1 transcripts encoding regulatory

Received January 19, 2012. Accepted June 25, 2012.

Published online ahead of print. Publication date available atwww.jasn.org.

Correspondence: Dr. Tobias E. Larsson, Clinical Research Cen-ter, Sixth Floor, Novumhuset, Karolinska University Hospital inHuddinge, SE-14186 Stockholm, Sweden. Email: [email protected]

Copyright © 2012 by the American Society of Nephrology

J Am Soc Nephrol 23: ccc–ccc, 2012 ISSN : 1046-6673/2310-ccc 1

enzymes in vitamin D metabolism.8 At the systemic level,this translates into a reduction of serum phosphate and 1,25-dihydroxyvitamin D3 (1,25(OH)2D).

The importance of Klotho as a renal coreceptor for FGF23is evidenced by Klotho and Fg23 null mice that share nearlyidentical biochemical phenotypes consistent with dismantledFGF23 signaling, including hyperphosphatemia, elevated 1,25(OH)2D, and hypercalcemia.1,9 Notably, the aging character-istics of Klotho and Fgf23 null mice are attenuated by dietaryrestrictions of phosphate and/or 1,25(OH)2D, or by eliminat-ing vitaminD effects through ablation of its receptor (VDR) orthe enzyme responsible for its activation (Cyp27B1).10–13 Col-lectively, this suggests that the aging phenotype is due to bothsystemic toxicity of mineral metabolites and perturbedFGF23-Klotho signaling.

The FGF23 level gradually increases during progressionof CKD, whereas expression of Klotho decreases.14,15 Distur-bances in FGF23-Klotho activity presumably contribute to theaccelerated aging process and cardiovascular disease evidentin CKD patients.16,17 Given the profound phenotype ofKlotho null mice, we wanted to determine the effect of a renal-specific and partial deletion of Klotho. Such a model wouldshed light on several issues, including dose-dependent ef-fects and kidney-derived systemic influence of Klotho ongrowth development and life span. Furthermore, it wouldallow the investigation of cell-specific autocrine or paracrineeffects of renal Klotho involved in the pathophysiology ofCKD-related complications such as interstitial fibrosis andectopic calcification.

Herein, we generated a novel mouse model with a kidney-specific Klotho deletion mainly in distal tubular segments andprovide genetic evidence favoring a pivotal role of FGF23-Klotho signaling in controlling mineral metabolism.

RESULTS

Generation of Ksp-KL2/2 MiceThe targeting vector of floxed Klotho mice and genotypingresults are shown in Figure 1A. To obtain the desired Klothodeletion, we used Ksp-cadherin-Cre mice, which express Creexclusively in tubular epithelial cells in the mature and devel-oping kidney and in the developing genitourinary tract.18 Apartial deletion of renal Klotho was confirmed with immuno-histochemistry (Figure 1B). The relative level of remnant me-dian Klotho transcripts in 15 screened kidney homogenateswas 0.69 (range, 0.26–1.0) compared with wild-type controls.Protein levels of Klotho quantified by Western blotting corre-lated highly to transcript levels (Figure 1C).

Gross Phenotype of Adult Ksp-KL2/2 MiceKsp-KL2/2mice were viable, fertile, and did not differ in size ordisplay any gross physical or behavioral abnormalities (Figure 2).There was no correlation between residual Klotho expressionand body size.

Generation of b-KL2/2 MiceTo confirm functionality of the floxed Klotho allele, we gen-erated a global deletion of Klotho (b-KL2/2) using miceexpressing Cre under the control of the human b-actin pro-moter. In stark contrast to Ksp-KL2/2 mice, b-KL2/2 micerecapitulated the phenotype of existing Klotho null mice,1 in-cluding severe growth retardation, kyphosis, lessened activity,and significantly reduced life span (Figure 2). In agreement,b-KL2/2mice (n=2, mean6 SEM) were hyperphosphatemic(5.360.06 mmol/L), hypercalcemic (2.8060.06 mmol/L),with extremely elevated FGF23 levels (234,630630,000 pg/ml)despite normal serum creatinine (44.861.9 mmol/L).

Biochemistries of Ksp-KL2/2 MiceSerum biochemistries were examined in adult mice at 8 weeksof age (Figure 3A). Ksp-KL2/2mice were hyperphosphatemicwith elevated FGF23. Parathyroid hormone (PTH) was de-creased in Ksp-KL2/2 mice, whereas 1,25(OH)2D, calcium,and creatinine levels were unaltered. Urinary calcium excre-tion was significantly increased as determined by calcium/creatinine ratio (Figure 3B) and fractional excretion of cal-cium (1.82 versus 0.67; P,0.05), but no difference was foundin urinary phosphate/creatinine ratio (Figure 3B) or fractionalexcretion of phosphate (28.4 versus 35.0; P=0.44).

Ksp-KL2/2 Mice and Dietary Phosphate LoadingSerum biochemistries of 8-week-old Ksp-KL2/2 and wild-type mice challenged with a high phosphate diet areshown in Figure 3C. Both groups developed hyperphosphate-mia, albeit more pronounced in Ksp-KL2 /2 mice. TheFGF23 response to phosphate loading was accentuated inKsp-KL2/2 mice. Ksp-KL2/2 mice had higher PTH, contrast-ing the reduced PTH level when fed a regular diet (Figure 3A).In linear regression analysis, phosphate was the only assessedbiochemical parameter that correlated to FGF23 in mice on aregular diet (r2=0.29; P,0.01), whereas phosphate (r2=0.29,P,0.05) and PTH (r2=0.41, P,0.005) correlated with FGF23in mice on a high phosphate diet.

Comparison of Ksp-KL2/2 and Wild-Type Mice withMatched Serum Phosphate LevelsIn this subgroup analysis, Ksp-KL2/2 mice had significantlyhigher FGF23 despite similar phosphate values (Figure 3D).No differences in calcium, phosphate, PTH, 1,25(OH)2D, orcreatinine were noted (Table 1). In the same mice, transcriptlevel of Cyp27B1was increased (P,0.001) with a similar trendfor Cyp24A1 (P=0.08).

Serum Biochemistries as a Function of Residual KlothoExpressionBecause Klotho was deleted with variable efficacy, we ana-lyzed the Klotho transcript level as a continuous variable.FGF23 was the only serum parameter significantly correlat-ing to Klotho (r2=0.23; P,0.05), whereas no correlations werefound for phosphate (P=0.09), calcium (P=0.13), PTH (P=0.13),

2 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BASIC RESEARCH www.jasn.org

or 1,25(OH)2D (P=0.72). To determine possible thresholdeffects, we screened 15 Ksp-KL2/2 mice and arbitrarily cate-gorized them into the following three groups based ontheir residual Klotho level: Klotho level .70% of wild-typecounterparts (mean 88%; n=7); Klotho level of 30%–70%(mean 59%; n=6); and Klotho ,30% (mean 28%; n=2).Phosphate and calcium levels gradually increased, whereasPTH levels decreased with lower Klotho expression (Table2). In contrast, a marked threshold effect was establishedfor FGF23 when relative Klotho expression was ,30%,

leading to levels 30–250 times higher than in wild-type mice(Figure 4).

HistologyNo differences were found with regard to general renalmorphology, calcifications, or fibrosis in Ksp-KL2/2 mice(Figure 5). The parathyroid glands were grossly normal withsimilar Klotho expression as in wild-type mice (data notshown). In contrast, b-KL2/2 mice had reduced kidney sizeand cortex height, higher cell density, extensive vascular and

Figure 1. Generation of floxed Klotho allele and targeted deletion of Klotho in Ksp-KL2/2 and b-KL2/2 mice. (A) Left panel showsschematic representation of wild-type allele (top), targeting vector (middle), and floxed allele with deleted Neo cassette (bottom).LoxP sites are flanking exon 2, thus enabling targeted deletion of the Klotho gene by Cre recombination. Right panel shows ge-notyping of mice. Representative PCR products from Klothoflox/flox (0.47 kB), Klothoflox/+ (0.47 and 0.37 kB), and wild-type (0.37 kB)mice. (B) Immunohistochemical staining of kidneys revealed partial deletion of Klotho in Ksp-KL2/2 mice, and complete deletion ofKlotho in b-KL2/2 mice. (C) Western blotting of whole kidney extracts from wild-type, Ksp-KL2/2, and b-KL2/2 mice. Numbers in-dicate relative transcript levels of Klotho measured by real-time qPCR, and for Ksp-KL2/2 these were arbitrarily categorized in sep-arate groups based on degree on the efficacy of Klotho deletion. Klotho protein level quantified by Western blotting correlated highlyto its transcript level.

J Am Soc Nephrol 23: ccc–ccc, 2012 Renal Klotho and Mineral Metabolism 3

www.jasn.org BASIC RESEARCH

tubular calcifications, and slightly increased fibrosis comparedwith wild-type mice (Figure 5).

Renal Protein ExpressionImmunohistochemical analysis revealed abundant expressionof Npt2a at the brush border membrane in Ksp-KL2/2 micecompared with wild-type controls. The vitamin D receptor(VDR) had a more heterogeneous pattern in Ksp-KL2/2

mice. There was no difference in cell proliferation rate as de-termined by Ki67 index (1.0% versus 0.95%; P=0.66; n$5 ofeach genotype) (Figure 6A).

Dual immunofluorescence staining in wild-type miceshowed that Klotho predominantly colocalized with the distaltubuli marker TRPV5, and very weakly with the proximalmarkerNpt2a. A similar pattern of residual Klotho proteinwasfound inKsp-KL2/2mice. The signal intensity of Klotho in theproximal tubuli appeared unaltered in Ksp-KL2/2 comparedwith wild-type mice, confirming a distal rather than proximaldeletion of Klotho (Figure 6B).

Western blotting showed increased VDR and decreasedTRPV5 protein in Ksp-KL2/2 mice (Figure 6C). However,immunostaining did not show a cell-specific downregulationof TRPV5 in the tubular segments where Klotho was deleted(Figure 6B).

Renal Gene ExpressionTranscript level of renal Cyp27B1 was increased in Ksp-KL2/2

mice, whereas VDR, Npt2a, Npt2c, FGFR1, CaSR, and TRPV5were unaltered (Table 3). Significant correlations were foundbetween Klotho and Cyp27B1, VDR, Npt2a, and FGFR1 (Fig-ure 7A). Renal transcript levels of mice challenged with a highphosphate diet are also shown in Table 3. Similarly, Cyp27B1

expression was higher in Ksp-KL2/2 mice whereas VDR,Npt2a, and CaSR were reduced. The correlations betweenKlotho to VDR and Npt2a were strengthened by dietaryphosphate loading (Figure 7B).

DISCUSSION

We present a novel mouse model (Ksp-KL2/2) harboring a par-tial deletion of Klotho predominantly in distal tubular segmentsof the kidney, demonstrating a pivotal role of Klotho in regula-tion ofmineralmetabolism.Ksp-KL2/2mice provide additionalvaluable information compared with general Klotho null mice.First, the variable efficacy of Klotho deletion allowed testing forpotential dose-dependent effects, revealing a graded increase inserum phosphate and FGF23 levels with lower Klotho levels,corroborating with the established phosphaturic action ofFGF23-Klotho. There was also a trend toward an increase inserum calcium level that paralleled the Klotho reduction. Thereason for this remains unclear, especially given that fractionalexcretion of calciumwas increased and PTH somewhat lower inKsp-KL2/2 compared with wild-type mice. One explanationmay be a functional increase in VDR signaling because VDRprotein was quantitatively increased in the face of an unalteredsystemic 1,25(OH)2D level. We also found it rather surprisingthat the 1,25(OH)2D level remained normal given that FGF23-Klotho signaling is a counter-regulatory hormone system for1,25(OH)2D. Mechanistically, there was an increase inCyp27B1 and a borderline significant increase in Cyp24A1 atthe transcript level, which presumably translates into both anincreased synthesis and degradation of active vitaminD. The risein Cyp27B1 is in agreement with attenuated FGF23-Klotho

Figure 2. Weight and gross appearance of Ksp-KL2/2 and b-KL2/2 mice. (Left panel) Ksp-KL2/2 mice did not differ in body weightcompared with wild-type controls. Conversely, b-KL2/2 mice were markedly smaller than wild-type littermates already at 4 weeks ofage. (Right panel) Ksp-KL2/2 mice were viable, fertile, and had a normal gross phenotype. In contrast, b-KL2/2 mice displayed severegrowth retardation, kyphosis, and a reduced life span with spontaneous deaths occurring around 7 weeks of age. †††Wild-type versusb-KL2/2 males; #Wild-type versus b-KL2/2 females. #P,0.05; †††###P,0.001.

4 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BASIC RESEARCH www.jasn.org

signaling, whereas the rise in Cyp24A1 con-tradicts the established physiology of FGF23.The exact cause remains elusive but mayspeculatively be due to differences in distalversus proximal tubuli signaling of FGF23-Klotho.

The combination of hyperphosphatemiaand high FGF23 levels in Ksp-KL2/2 micestrongly indicates a partial renal FGF23 re-sistance, supported by the fact that FGF23was the only biochemical variable related toKlotho expression and the observed expo-nential increase in FGF23when renal Klothodrop below approximately 30% of normallevels. Although not entirely surprising,this is an important in vivo proof of prin-ciple suggesting that initial onset of FGF23-Klotho dysregulation in CKD at least in partcould be attributed to a renal FGF23 resis-tance. It may also explain the large interin-dividual FGF23 variation observed in ESRDpatients,16 which simply cannot be attrib-uted to differences in the circulating min-eral metabolites. However, this study isclearly limited in terms of investigating thedynamics of FGF23-Klotho in early CKD,and this important question should be ad-dressed in future studies.

A fundamental question is how a renalFGF23 resistance due to Klotho deficiencytranslates into increased FGF23 productionin bone. Secondary changes inmineral meta-bolism, including hyperphosphatemia, areplausible. Indeed, we found a gradual rise inserumphosphate and FGF23 with decliningKlotho levels, anddietary phosphate loading

Figure 3. Serum and urine biochemistry in adult Ksp-KL2/2 mice. (A) At 8 weeks ofage, Ksp-KL2/2 mice displayed hyperphosphatemia and elevated FGF23 levels.Conversely, serum PTH was lower in Ksp-KL2/2 mice than in controls. Calcium, cre-atinine, and 1,25(OH)2D were similar in the two groups (*P,0.05; n=10 in each group

for all analysis except 1,25(OH)2D wheren$5). (B) Analysis of urine biochemistry re-vealed hypercalciuria in Ksp-KL2/2 mice. Uri-nary phosphate excretion was unchanged(*P,0.05; n=9–10 per group). (C) Serum bio-chemistry in Ksp-KL2/2 and wild-type miceon a high phosphate diet (1.65% phosphatefor 10 days). Both groups developed hyper-phosphatemia and elevated FGF23, althoughmorepronounced inKsp-KL2/2mice.Ksp-KL2/2

mice on a high phosphate diet had higher se-rum PTH than wild-type controls (*P,0.05;**P,0.01; n$5 per group). (D) In a subgroupanalysis of Ksp-KL2/2 and wild-type mice withmatched serum phosphate levels (2.56 versus2.60 mmol/L; P=0.90; n=5 in each group),FGF23 was significantly higher in Ksp-KL2/2

mice (140.4 versus 80.6 pg/ml; P,0.05).

J Am Soc Nephrol 23: ccc–ccc, 2012 Renal Klotho and Mineral Metabolism 5

www.jasn.org BASIC RESEARCH

further increased FGF23. However, FGF23 was higher in Ksp-KL2/2 mice compared with wild-type mice with similar serumphosphate levels, suggesting that additional mechanisms arepresent. Speculatively, soluble Klotho may antagonize FGF re-ceptor signaling in bone, which has been implied as an impor-tant transcriptional regulator of FGF23,19 or interact with otheryet unidentified FGF23 regulatory elements. These possibilitiesneed to be explored in additional experimental studies.

In contrast to general Klotho nullmice, Ksp-KL2/2mice hada normal growth development and did not differ in body sizecompared with wild-type littermates. This contradicts that re-nal Klotho insufficiency causes systemic toxicity, although itmay also be explained by the incomplete knockout efficacy (i.e.,the residual Klotho amounts are sufficient to maintain a nor-mal growth status and body weight). Similarly, the biochemicalabnormalities in Ksp-KL2/2 mice may be too subtle to evoke

systemic toxicity as in general Klotho nullmice, favored by pre-vious studies showing that a low calcium- and phosphate-con-taining rescue diet attenuates the phenotype in animalmodelsof absent FGF23-Klotho signaling11

Another distinct difference compared with general Klothonull mice is the absence of overt renal abnormalities such asinterstitial fibrosis and ectopic calcifications in Ksp-KL2/2

mice. This was unexpected given that Klotho has been impli-cated as an antifibrotic agent by blocking TGF-b signaling20

and by acting as an endogenous inhibitor of vascular calcifi-cation.15 This argues against dominant cell-specific autocrineor paracrine effects of renal tubular Klotho, at least in theshort term. Additional long-term studies are warranted todetermine whether renal-specific Klotho deletion affectsthese phenotypes, especially in CKD.

Both FGF23 and PTH are phosphaturic hormones, actingon the same phosphate transporters in thekidney, namely Npt2a and Npt2c.7,21,22 Inthe face of hyperphosphatemia, serumPTH was suppressed, whereas FGF23 wasincreased in Ksp-KL2/2 mice on a regulardiet, suggesting that FGF23 may either re-spond earlier or play a more dominant rolethan PTH in regulating phosphate transportduring chronic hyperphosphatemia. The re-duction in PTH found in Ksp-KL2/2 micecould could either be caused by higher sys-temic calcium levels or the fact that FGF23inhibits PTH synthesis and secretion by di-rectly targeting the parathyroid glands.23,24

In contrast, Ksp-KL2/2 mice challengedwith a high-phosphate diet had higherPTH thanwild-type littermates. The underly-ing mechanisms remain unclear. One possi-bility is that parathyroid Klotho expressionis differentially regulated inKsp-KL2/2miceand that dietary phosphate loading evokes aKlotho-dependent parathyroid FGF23 resis-tance. Irrespectively, this phenomenon mayrepresent a rescue mechanism against phos-phate toxicity when FGF23 signaling is in-appropriately low in relation to the degreeof phosphate challenge.

Table 1. Serum biochemistry and gene expression in wild-type and Ksp-KL2/2

mice with matched serum phosphate levels

Parameters WT (n=5) Ksp-KL2/2 (n=5) P Value

Serum biochemistriesFGF23 (pg/mL) 79.6 (43.3–110–7) 133.5 (74.3–193.3) ,0.05Phosphate(mmol/L)

2.73 (1.81–3.02) 2.65 (1.80–2.91) 0.90

Calcium(mmol/L)

1.76 (1.70–1.89) 1.80 (1.67–1.92) 0.80

PTH (pg/mL) 78.4 (65.8–93.0) 79.4 (67.8–185.1) 0.261,25(OH)2D(pmol/L)

132.2 (102.0–204.9) 215.3 (115.3–382.4) 0.34

Creatinine(mmol/L)

36.0 (28.5–40.5) 39.0 (36.0–44.5) 0.20

Gene expressionKlotho 1.12 (0.86–1.17) 0.83 (0.51–1.13) 0.13Cyp27B1 1.00 (0.44–1.38) 2.45 (1.64–2.76) ,0.001Cyp24A1 0.73 (0.27–2.36) 2.32 (1.02–3.60) 0.08VDR 0.97 (0.69–1.39) 1.21 (0.85–1.57) 0.45Npt2a 1.01 (0.67–1.35) 0.98 (0.79–1.11) 0.63Npt2c 1.01 (0.53–1.79) 0.82 (0.78–1.45) 0.86FGFR1 1.06 (0.91–1.16) 1.02 (0.74–1.34) 0.73CaSR 0.97 (0.62–1.33) 0.87 (0.53–1.40) 0.86TRPV5 0.98 (0.79–1.54) 0.94 (0.75–1.21) 0.72

FGF23 was the only serum variable differing between wild-type and Ksp-KL2/2 mice in a subgroupanalysis ofmicewithmatched serumphosphate levels. In gene expression analysis, therewas amarkedincrease of Cyp27B1 in the Ksp-KL2/2 mice. Values are median (interquartile range).

Table 2. Serum biochemistry as a function of residual Klotho expression

Serum Biochemistries WT (KL=1) Ksp-KL2/2 (KL >0.7) Ksp-KL2/2 (KL 0.7–0.3) Ksp-KL2/2 (KL ,0.3)

FGF23 (pg/mL) 83.3 (43.3–122.8) 115.3 (74.3–193.3) 160.4 (78.9–182.0) 12,076 (3234–20,917)Phosphate (mmol/L) 2.71 (1.16–3.02) 2.63 (1.80–3.20) 3.09 (2.61–3.46) 3.69 (3.07–4.32)Calcium (mmol/L) 1.74 (1.67–1.89) 1.80 (1.67–1.92) 1.86 (1.65–1.93) 1.97 (1.84–2.10)PTH (pg/mL) 87.2 (65.8–188.9) 76.5 (54.2–185.1) 63.0 (53.2–79.4) 55.2 (53.2–57.1)1,25(OH)2D (pmol/L) 176.8 (102.0–168.9) 134.8 (81.1–215.3) 161.0 (66.5–382.4) 130.9 (n=1)Creatinine (mmol/L) 39.5 (28.5–41.0) 37.5 (33.5–44.5) 37.3 (32.5–41.0) 36.5 (34.5–38.5)In wild-type and Ksp-KL2/2 mice, categorized into groups based on residual Klotho level, there was a gradual increase in serum FGF23, phosphate, and calciumlevels and a reciprocal reduction in PTH level as Klotho expression declined.

6 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BASIC RESEARCH www.jasn.org

The proximal tubule is the principal sitefor renal phosphate reabsorption.22 Inagreement with a previous report, wefound that Klotho expression is largelyconfined to the distal tubules3 and thatthe Cre recombination driven by the Ksp-cadherin promoter is functional mainly indistal tubular cells. Although immunoflu-orescence staining suggested a low Klothoexpression also in proximal segments, itis still likely that distal Klotho plays animportant role in proximal phosphatetransport.

Hyperphosphatemic Ksp-KL2/2 micehad increased Npt2a protein expression atthe brush-border membrane. In contrast,the Npt2a transcript level was positivelycorrelated to the renal Klotho, indicatinguncoupling of Npt2a transcription totranslation. Because the observed reduc-tion in the Npt2a transcript level is anadequate compensatory response to hyper-phosphatemia, our data suggest that dis-mantling of renal FGF23-Klotho signalingmay result in aberrant Npt2a protein process-ing or degradation rather than transcrip-tional dysregulation. A similar uncoupling,possibly an adaptive response, was observedfor VDR.

Urinary calcium excretionwas increasedin Ksp-KL2/2 mice consistent with theknown action of renal Klotho from previ-ous in vitro studies2 and the observed re-duction in TRPV5 protein. In contrast, wedid not detect any difference in urinaryphosphate excretion despite hyperphos-phatemia and increased abundance ofNpt2a in Ksp-KL2/2 mice. We speculatethat this may be due to intermittent periodsof hypophosphaturia that were not cap-tured by our spot urinary analysis.

In summary, deletion of Klotho in dis-tal tubular segments reveals a fundamentalrole of Klotho in renal homeostatic con-trol of mineral metabolism and as a de-terminant of circulating FGF23.

CONCISE METHODS

Generation of Kidney-Specific KlothoKnockout MiceMice with a kidney-specific Klotho deletion

were generated using Cre-Lox recombination.

Briefly, the sequence of mouse chromosome 5

Figure 4. Serum phosphate and FGF23 as a function of renal Klotho expression.Klotho transcript levels were normalized to 1 in wild-type mice, and Ksp-KL2/2 micewere arbitrarily divided into three groups based on relative Klotho level: .0.7 (meanlevel 0.88; n=7); 0.30–0.7 (mean 0.59; n=6); and ,0.3 (mean 0.28; n=2). Serumphosphate level increased linearly after the decreased Klotho expression. In contrast,a threshold was found for FGF23 when relative Klotho expression was below 0.3,leading to levels 30–250 times higher than in control mice.

Figure 5. Renal histology in Ksp-KL2/2 and b-KL2/2 mice. Hematoxylin and eosinstaining (top) showed a normal renal morphology in Ksp-KL2/2 mice contrastingb-KL2/2 mice, which had reduced cortex height and higher cell density. In von Kossastaining (middle), b-KL2/2 mice displayed vascular and tubular calcifications (arrows Aand B, respectively). No calcifications were seen in wild-type or Ksp-KL2/2 mice. Asubtle increase in fibrosis was noted in kidneys from b-KL2/2 mice when stained withSirius red for collagen (bottom). Sirius red staining was similar in Ksp-KL2/2 mice andwild-type littermates. Number of kidneys examined was n=5 (wild-type); n=6 (Ksp-KL2/2); n=2 (b-KL2/2).

J Am Soc Nephrol 23: ccc–ccc, 2012 Renal Klotho and Mineral Metabolism 7

www.jasn.org BASIC RESEARCH

Figure 6. Renal protein expression of Ki67, VDR, TRPV5 and Npt2a in Ksp-KL2/2 mice. (A) Proliferation rate as determined by im-munostaining for Ki67 did not differ between Ksp-KL2/2 and wild-type mice (top) (1.0% versus 0.95%; P=0.66). Staining for VDR(middle) revealed a more heterogeneous expression pattern in Ksp-KL2/2 mice compared with wild-type littermates. Npt2a proteinwas abundantly expressed at the brush border membrane in Ksp-KL2/2 mice compared with wild-type controls (bottom). Number ofkidneys examined was n=5 (wild-type); n=6 (Ksp-KL2/2); n=2 (b-KL2/2). (B) Dual immunofluorescence staining for Klotho and the distaltubuli specific marker TRPV5 revealed complete colocalization in wild-type mice, and partial colocalization in Ksp-KL2/2 mice. Therewas no apparent difference in TRPV5 staining intensity between distal segments with or without Klotho expression. Klotho was onlyweakly expressed in the proximal tubuli, and dual immunofluorescence staining for Klotho and the proximal tubuli specific markerNpt2a showed almost no colocalization. Npt2a expression at the brush border membrane was markedly higher in Ksp-KL2/2 micecompared with wild-type controls. (C) Western blotting of whole kidney extracts from wild-type and Ksp-KL2/2 mice revealed a gradualdecrease of TRPV5 (left panel), whereas VDR protein (right panel) level increased with declining Klotho level. Original magnification,340 in top panel of B; 310 in bottom panel of B.

8 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BASIC RESEARCH www.jasn.org

was retrieved from the Ensembl database (http://www.ensembl.org).

The RP23–434H9 BAC clone was used for generation of homology

arms and the conditional knockout region of the targeting vector.

The fragments were cloned in the LoxFtNwCD or pCR4.0 vector

and electroporated into C57BL/6 embryonic stem cells. Male chi-

meras were generated and subsequently bred with wild-type females

to generate Klotho-LoxP heterozygotes (Klothoflox/+). Klothoflox/+

were crossed with mice expressing Cre recombinase under the

Ksp-cadherin promoter (B6.Cg-Tg(Cdh16-cre)91Igr/J; Jackson

Laboratory, Bar Harbor, ME). Homozygous mice without Cre

(Klothoflox/flox) served as wild-type controls. To minimize the intra-

litter variability, mice were intercrossed, making 50% of the offspring

kidney-specific Klotho null mice and 50% controls that were

subsequently analyzed. Mice with a systemic Klotho deletion were gen-

erated using mice expressing Cre under the human b-actin promotor

(FVB/N-Tg(ACTB-cre)2Mrt/J, Jackson Laboratory). Generation of the

targeting vector, cloning, and electroporation into embryonic stem cells

was carried out by Caliper Life Sciences (Hopkinton, MA).

GenotypingTotal DNA was extracted from tail biopsies using DirectPCR Lysis

Reagent (Viagen, Los Angeles, CA). PCR amplification was carried

out on a 2720 Thermal Cycler (Applied Biosystems, Carlsbad, CA)

using HotStarTaq DNA Polymerase (Qiagen, Venlo, Netherlands).

The PCR products were visualized on a 1% agarose gel with GelRed

Nucleic Acid Gel Stain (Biotium, Hayward, CA). Sequences of the

primers used for genotyping are listed in Sup-

plemental Table 1.

Animal HousingMice were fed standard rodent chow (RM1;

SDS, Essex, UK) containing 0.73% calcium and

0.52% phosphate. In the high phosphate exper-

iment mice were fed a diet containing 1.0% cal-

cium and 1.65% phosphate (TD.88345; Harlan

Laboratories, Indianapolis, IN) for 10 days, start-

ing at 8 weeks of age. Blood sampling was per-

formed by tail vein incision at intermediate time

points and by bleeding the axillary artery

at sacrifice. All experiments were conducted in

compliance with the guidelines of animal experi-

ments at Karolinska Institutet and approved by

the regional ethical board.

Figure 7. Gene expression in kidneys of Ksp-KL2/2 mice. (A) Klotho transcript level correlated significantly to Cyp27B1, VDR, Npt2a,and FGFR1. (B) The correlations between Klotho, VDR, and Npt2a were accentuated in mice on a high phosphate diet.

Table 3. Renal gene expression in Ksp-KL2/2 mice on a regular and highphosphate diet

Gene TranscriptRegular Diet High Phosphate Diet

WT Ksp-KL2/2 P Value WT Ksp-KL2/2 P Value

Klotho 160.06 0.5160.06 ,0.0001 160.08 0.5660.06 ,0.001Cyp27B1 160.13 4.0360.78 ,0.01 160.29 3.7760.86 ,0.01Cyp24A1 160.26 5.2062.24 0.10 160.20 0.9360.43 0.87VDR 160.10 0.8660.05 0.23 160.10 0.5760.07 ,0.01Npt2a 160.08 0.8160.09 0.14 160.06 0.4860.06 ,0.0001Npt2c 160.16 0.9560.22 0.85 160.20 0.95614 0.87FGFR1 160.05 0.8460.11 0.21 160.06 0.8460.04 0.07CaSR 160.11 0.9560.14 0.78 160.07 0.7360.06 ,0.05TRPV5 160.10 1.1060.11 0.51 160.07 1.0060.09 0.97

Data are mean 6 SD. Transcript level of Cyp27B1 was significantly higher in Ksp-KL2/2 mice ona regular diet. Cyp27B1, VDR,Npt2a, andCaSRwere significantly changed inKsp-KL2/2mice on a highphosphate diet in Ksp-KL2/2 mice.

J Am Soc Nephrol 23: ccc–ccc, 2012 Renal Klotho and Mineral Metabolism 9

www.jasn.org BASIC RESEARCH

BiochemistriesSerum and urine calcium, phosphate, and creatinine were measured

on a Konelab 20XTi (Thermo Scientific, Vantaa, Finland). Intact

FGF23 was measured using an intact FGF23 ELISA kit (Kainos

Laboratories, Tokyo, Japan). SerumPTHwasmeasuredusing aMouse

Intact PTH ELISA kit (Immutopics, Carlsbad, CA) and serum 1,25

(OH)2D using a RIA kit (IDS, Scottsdale, AZ). Fractional excretion of

calcium and phosphate was calculated using the formula: [urine cal-

cium or phosphate] 3 [serum creatinine] 3 100/[urine creatinine]

3 [serum calcium or phosphate].

RNA Isolation, cDNA Synthesis, and Real-Time qPCRKidneys were homogenized using a TissueLyzer LT (Qiagen) and total

RNA was extracted using E.Z.N.A. Total RNA Kit I (Omega Bio-tek,

Norcross, GA). DNAwas removed with E.Z.N.A. RNase-Free DNase

Set (Omega Bio-tek). First-strand cDNA synthesis was carried out

using the iScript cDNA Synthesis Kit (Bio-Rad, Hercules, CA). For

real-timeqPCRanalysis, theCFX96Real-TimePCRDetection System

and iQ SYBRGreen Supermix (Bio-Rad) were used. The relative gene

expression was calculated with the 2-DD Cq method normalizing the

gene of interest to b-actin in the same sample. Data are presented as

relative fold-change compared with wild-type mice. Sequences of

primers used are listed in Supplemental Table 1.

Immunohistochemistry and ImmunofluorescenceKidneys were dissected, fixed in 4% formalin overnight, and sub-

sequently embedded in paraffin. Immunohistochemical analyseswere

performed according to standard protocols on 4-mm sections using

the Vector ABC Reagent kit and developed with DAB substrate (Vec-

tor Laboratories). For immunofluorescence, Alexa Fluor conjugated

secondary antibodies were used for visualization (Invitrogen, Carls-

bad, CA, US). The primary antibodies usedwere ratmonoclonal anti-

Klotho (KM2076, kindly provided by Kyowa Hakko Kogyo Co. Ltd,

Japan), mousemonoclonal anti-VDR (sc-13133; Santa Cruz Biotech-

nology, Santa Cruz, CA), rabbit monoclonal anti-Ki67 (SP6; Thermo

Scientific, Fremont, CA), rabbit polyclonal anti-TRPV5 (SC-30187;

Santa Cruz Biotechnology), and rabbit polyclonal anti-Npt2a

(NPT27; Alpha Diagnostics, San Antonio, TX). Proliferation index

(%) was calculated as Ki67 positive cells/total number of cells in at

least four viewing fields (at 320 magnification) from two sections

of each assessed mouse.

HistologyParaffin sections from wild-type, Ksp-KL2/2, and b-KL2/2 kidneys

were stained with hematoxylin and eosin, periodic acid–Schiff , von

Kossa, and Sirius red according to standard histologic protocol. Sec-

tions were examined blinded by an experienced renal pathologist

(A.W.). Glomeruli, tubuli, interstitium, and vessels were systemati-

cally evaluated for all samples.

Western BlottingKidney extracts were homogenized in PBS-TDS buffer (Sigma-

Aldrich Co., St Louis, MO) using a Tissue Lyzer LT. Extracts were

incubated on ice for 30 minutes, and centrifuged at 10,000 rpm for

10 minutes at 4°C. Supernatants were collected for further analysis.

Protein quantification was carried out using a BCA protein assay kit

(Thermo Scientific, Rockford, IL). Forty micrograms of protein was

separated on a 7.5% SDS-PAGE and electrotransferred to a polyvi-

nylidene fluoride membrane (Bio-Rad). After blocking with Casein

blocking solution (Thermo Scientific), membranes were sequentially

incubated with primary and secondary antibodies. Primary antibod-

ies were anti-Klotho (KM2076), anti-VDR (sc-13133), anti-TRPV5

(SC-30187), and anti-b-tubulin (Santa Cruz). The secondary anti-

body was IRDyes 680LT (LI-COR Biosciences, Lincoln, NE). Visual-

ization was carried out using the ODYSSEY Infrared imaging system

(LI-COR Biosciences). The amount of Klotho protein was nor-

malized to the amount of b-tubulin in each sample. Image studio

2.0 (LI-COR Biosciences) was used for densitometry and results are

presented as arbitrary densitometric units.

Statistical AnalysesIn comparisons between Ksp-KL2/2 and wild-type mice, Ksp-KL2/2

mice with relative Klotho levels .70% were excluded. In correla-

tion tests, all mice were included. GraphPad Prism 5.0 software

(GraphPad Software Inc, La Jolla, CA) was used for statistical analy-

sis. Gaussian distribution was tested using the D’Agostino and Pear-

son omnibus normality test. Variables fulfilling the criteria for normal

distribution were tested with the two-tailed t test. Non-normally dis-

tributed variables were compared using the Mann–Whitney test. Cor-

relationswere testedwith linear regression analysis.P values,0.05were

considered statistically significant. A minimum of five mice of each

genotype was analyzed in all experiments, unless otherwise stated.

ACKNOWLEDGMENTS

This study was funded by grants from the Swedish Foundation for

Strategic Research, Swedish Research Council, Swedish Kidney Foun-

dation, and Karolinska Institutet.

DISCLOSUREST.E.L. served as a consultant and/or received honoraria from Genzyme,

Sanofi-Aventis, Shire, Amgen, Abbott, Astellas, Fresenius, and AstraZeneca.

REFERENCES

1. Kuro-oM,MatsumuraY,AizawaH, KawaguchiH, SugaT,Utsugi T,OhyamaY, Kurabayashi M, Kaname T, Kume E, Iwasaki H, Iida A, Shiraki-Iida T,Nishikawa S, Nagai R, Nabeshima YI: Mutation of the mouse klotho geneleads to a syndrome resembling ageing. Nature 390: 45–51, 1997

2. Chang Q, Hoefs S, van der Kemp AW, Topala CN, Bindels RJ,Hoenderop JG: The beta-glucuronidase klotho hydrolyzes and acti-vates the TRPV5 channel. Science 310: 490–493, 2005

3. Hu MC, Shi M, Zhang J, Pastor J, Nakatani T, Lanske B, Razzaque MS,Rosenblatt KP, Baum MG, Kuro-o M, Moe OW: Klotho: A novel phos-phaturic substance acting as an autocrine enzyme in the renal proximaltubule. FASEB J 24: 3438–3450, 2010

4. Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K,Fujita T, Fukumoto S, Yamashita T: Klotho converts canonical FGF re-ceptor into a specific receptor for FGF23. Nature 444: 770–774, 2006

10 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BASIC RESEARCH www.jasn.org

5. Larsson T, Marsell R, Schipani E, Ohlsson C, Ljunggren O, TenenhouseHS, Jüppner H, Jonsson KB: Transgenic mice expressing fibroblastgrowth factor 23 under the control of the alpha1(I) collagen promoterexhibit growth retardation, osteomalacia, and disturbed phosphatehomeostasis. Endocrinology 145: 3087–3094, 2004

6. Bai X, Miao D, Li J, Goltzman D, Karaplis AC: Transgenic mice over-expressing human fibroblast growth factor 23 (R176Q) delineate aputative role for parathyroid hormone in renal phosphate wasting dis-orders. Endocrinology 145: 5269–5279, 2004

7. Baum M, Schiavi S, Dwarakanath V, Quigley R: Effect of fibroblastgrowth factor-23 on phosphate transport in proximal tubules. KidneyInt 68: 1148–1153, 2005

8. Bai XY, Miao D, Goltzman D, Karaplis AC: The autosomal dominanthypophosphatemic rickets R176Q mutation in fibroblast growth factor23 resists proteolytic cleavage and enhances in vivo biological potency.J Biol Chem 278: 9843–9849, 2003

9. Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T,Fukumoto S, Tomizuka K, Yamashita T: Targeted ablation of Fgf23demonstrates an essential physiological role of FGF23 in phosphateand vitamin D metabolism. J Clin Invest 113: 561–568, 2004

10. Ohnishi M, Nakatani T, Lanske B, RazzaqueMS: Reversal of mineral ionhomeostasis and soft-tissue calcification of klotho knockout mice bydeletion of vitamin D 1alpha-hydroxylase. Kidney Int 75: 1166–1172,2009

11. Ohnishi M, Razzaque MS: Dietary and genetic evidence for phosphatetoxicity accelerating mammalian aging. FASEB J 24: 3562–3571, 2010

12. Sitara D, Kim S, Razzaque MS, Bergwitz C, Taguchi T, Schüler C, ErbenRG, Lanske B: Genetic evidence of serum phosphate-independentfunctions of FGF-23 on bone. PLoS Genet 4: e1000154, 2008

13. HesseM, Fröhlich LF, Zeitz U, Lanske B, Erben RG: Ablation of vitaminDsignaling rescues bone, mineral, and glucose homeostasis in Fgf-23deficient mice. Matrix Biol 26: 75–84, 2007

14. Larsson T, Nisbeth U, LjunggrenO, Jüppner H, Jonsson KB: Circulatingconcentration of FGF-23 increases as renal function declines in patientswith chronic kidney disease, but does not change in response to vari-ation in phosphate intake in healthy volunteers. Kidney Int 64: 2272–2279, 2003

15. Hu MC, Shi M, Zhang J, Quiñones H, Griffith C, Kuro-o M, Moe OW:Klotho deficiency causes vascular calcification in chronic kidney dis-ease. J Am Soc Nephrol 22: 124–136, 2011

16. Gutiérrez OM, Mannstadt M, Isakova T, Rauh-Hain JA, Tamez H, ShahA, Smith K, Lee H, Thadhani R, Jüppner H, Wolf M: Fibroblast growthfactor 23 and mortality among patients undergoing hemodialysis. NEngl J Med 359: 584–592, 2008

17. Isakova T, Xie H, Yang W, Xie D, Anderson AH, Scialla J, Wahl P,Gutiérrez OM, Steigerwalt S, He J, Schwartz S, Lo J, Ojo A, SondheimerJ, Hsu CY, Lash J, Leonard M, Kusek JW, Feldman HI, Wolf M; ChronicRenal Insufficiency Cohort (CRIC) Study Group: Fibroblast growth fac-tor 23 and risks of mortality and end-stage renal disease in patients withchronic kidney disease. JAMA 305: 2432–2439, 2011

18. Shao X, Somlo S, Igarashi P: Epithelial-specific Cre/lox recombinationin the developing kidney and genitourinary tract. J AmSocNephrol 13:1837–1846, 2002

19. Martin A, Liu S, David V, Li H, Karydis A, Feng JQ, Quarles LD: Boneproteins PHEX and DMP1 regulate fibroblastic growth factor Fgf23expression in osteocytes through a common pathway involving FGFreceptor (FGFR) signaling. FASEB J 25: 2551–2562, 2011

20. Doi S, Zou Y, Togao O, Pastor JV, John GB, Wang L, Shiizaki K,Gotschall R, Schiavi S, Yorioka N, Takahashi M, Boothman DA, Kuro-oM: Klotho inhibits transforming growth factor-beta1 (TGF-beta1) sig-naling and suppresses renal fibrosis and cancer metastasis in mice. JBiol Chem 286: 8655–8665, 2011

21. Shimada T, HasegawaH, Yamazaki Y, Muto T, Hino R, Takeuchi Y, FujitaT, Nakahara K, Fukumoto S, Yamashita T: FGF-23 is a potent regulatorof vitamin D metabolism and phosphate homeostasis. J Bone MinerRes 19: 429–435, 2004

22. Forster IC, Hernando N, Biber J, Murer H: Proximal tubular handling ofphosphate: A molecular perspective. Kidney Int 70: 1548–1559, 2006

23. Krajisnik T, Björklund P, Marsell R, Ljunggren O, Akerström G, JonssonKB, Westin G, Larsson TE: Fibroblast growth factor-23 regulates para-thyroid hormone and 1alpha-hydroxylase expression in cultured bo-vine parathyroid cells. J Endocrinol 195: 125–131, 2007

24. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, Goetz R, Kuro-o M,Mohammadi M, Sirkis R, Naveh-Many T, Silver J: The parathyroid is atarget organ for FGF23 in rats. J Clin Invest 117: 4003–4008, 2007

This article contains supplemental material online at http://jasn.asnjournals.org/lookup/suppl/doi:10.1681/ASN.2012010048/-/DCSupplemental.

J Am Soc Nephrol 23: ccc–ccc, 2012 Renal Klotho and Mineral Metabolism 11

www.jasn.org BASIC RESEARCH


Recommended