FGF23 regulates renal sodium handling and blood pressure

Olena Andrukhova, Svetlana Slavic, Alina Smorodchenko, Ute Zeitz, Victoria Shalhoub, Beate Lanske, Elena E Pohl, Reinhold G Erben, Olena Andrukhova, Svetlana Slavic, Alina Smorodchenko, Ute Zeitz, Victoria Shalhoub, Beate Lanske, Elena E Pohl, Reinhold G Erben

Abstract

Fibroblast growth factor-23 (FGF23) is a bone-derived hormone regulating renal phosphate reabsorption and vitamin D synthesis in renal proximal tubules. Here, we show that FGF23 directly regulates the membrane abundance of the Na(+):Cl(-) co-transporter NCC in distal renal tubules by a signaling mechanism involving the FGF receptor/αKlotho complex, extracellular signal-regulated kinase 1/2 (ERK1/2), serum/glucocorticoid-regulated kinase 1 (SGK1), and with-no lysine kinase-4 (WNK4). Renal sodium (Na(+)) reabsorption and distal tubular membrane expression of NCC are reduced in mouse models of Fgf23 and αKlotho deficiency. Conversely, gain of FGF23 function by injection of wild-type mice with recombinant FGF23 or by elevated circulating levels of endogenous Fgf23 in Hyp mice increases distal tubular Na(+) uptake and membrane abundance of NCC, leading to volume expansion, hypertension, and heart hypertrophy in a αKlotho and dietary Na(+)-dependent fashion. The NCC inhibitor chlorothiazide abrogates FGF23-induced volume expansion and heart hypertrophy. Our findings suggest that FGF23 is a key regulator of renal Na(+) reabsorption and plasma volume, and may explain the association of FGF23 with cardiovascular risk in chronic kidney disease patients.

Keywords: aldosterone; blood pressure; fibroblast growth factor‐23; heart hypertrophy; sodium homeostasis.

© 2014 The Authors. Published under the terms of the CC BY 4.0 license.

Figures

Figure 1. Fgf23 or Klotho deficiency induces…
Figure 1. Fgf23 or Klotho deficiency induces renal sodium wasting caused by reduced expression of the Na+:Cl− co-transporter NCC
  1. A, B (A) Urinary Na+ excretion corrected by urinary creatinine (Crea) (n = 10–12, one-way ANOVA followed by SNK test, *P = 0.0114 versus WT, #P = 0.0325 versus VDRΔ/Δ for Fgf23−/−/VDRΔ/Δ, *P = 0.0218 versus WT, #P = 0.0185 versus VDRΔ/Δ for Kl−/−/VDRΔ/Δ) and (B) urinary aldosterone concentration corrected by urinary creatinine and serum aldosterone concentration measured by ELISA (n = 8–10, one-way ANOVA followed by SNK test, *P < 0.05 versus WT, #P < 0.05 versus VDRΔ/Δ), in 9-month-old male wild-type (WT), VDRΔ/Δ, Fgf23−/−/VDRΔ/Δ, or Kl−/−/VDRΔ/Δ compound mutant mice on the rescue diet.

  2. C, D Western blotting analysis of NCC and α-ENaC protein expression in renal cortical total membrane fractions (n = 7–9, one-way ANOVA followed by SNK test, *P < 0.005 versus WT, #P < 0.005 versus VDRΔ/Δ), and immunohistochemical detection of NCC and α-ENaC protein expression in paraffin sections of paraformaldehyde-fixed kidneys (n = 3–5) in 9-month-old male wild-type (WT), VDRΔ/Δ, Fgf23−/−/VDRΔ/Δ, or Kl−/−/VDRΔ/Δ compound mutant mice on the rescue diet. Data represent mean ± s.e.m.

Source data are available for this figure.
Figure 2. Decreased mean arterial pressure and…
Figure 2. Decreased mean arterial pressure and blood volume in Fgf23- or Klotho-deficient mice
  1. A, B Mean arterial pressure (A) and blood volume (B) (n = 6–8, one-way ANOVA followed by SNK test, *P < 0.05 versus WT, #P < 0.05 versus VDRΔ/Δ) in 9-month-old male wild-type (WT), VDRΔ/Δ, Fgf23−/−/VDRΔ/Δ, or Kl−/−/VDRΔ/Δ compound mutant mice on the rescue diet. Data represent mean ± s.e.m.

Figure 3. Gain of FGF23 function induces…
Figure 3. Gain of FGF23 function induces renal Na+ retention through increased renal NCC expression and channel activation
  1. A Urine volume (n = 15–17), urinary Na+ excretion per 12 h (n = 15–17, Student's t-test, *P = 0.0085), urinary Na+ excretion corrected by urinary creatinine (Crea) (n = 15–17, Student's t-test, *P = 0.0251), serum Na+ concentration (n = 15–17, Student's t-test, *P = 0.0308), serum and urinary aldosterone concentrations corrected by urinary creatinine (n = 4–5, Student's t-test, * serum P = 0.0040, urine P = 0.0156), and plasma renin activity (RPA) (n = 4–5) after 5 days of treatment of 3-month-old male wild-type mice with vehicle (Veh) or recombinant FGF23 (10 μg per mouse per day).

  2. B, C Western blotting quantification (B) of NCC, α-ENaC, β-ENaC, and γ-ENaC protein expression in renal cortical total membrane fractions (n = 4–5, Student's t-test, *NCC P = 0.0014, α-ENaC P = 0.0007, β-ENaC P = 0.0251, γ-ENaC P = 0.0344), and immunohistochemical detection (C) of NCC and α-ENaC protein expression in kidney sections of 3-month-old wild-type mice treated for 5 days with vehicle or rFGF23 (n = 3–4).

  3. D Western blotting quantification of NCC phosphorylation at Ser71, Ser91, and Thr58 (pNCC S71, pNCC S91, pNCC T55) in total kidney homogenates of 3-month-old wild-type mice treated for 5 days with vehicle or rFGF23 (n = 4–5, Student's t-test, *pNCC S71 P = 0.0001, pNCC S91 P = 0.0182, pNCC T55 P = 0.0056).

  4. E Reciprocal immunoprecipitation (IP) of serine-phosphorylated (P-Ser) proteins, followed by Western blot (WB) analysis of WNK4 or vice versa from homogenized renal cortex protein samples of 3-month-old male wild-type mice treated for 5 days with vehicle or rFGF23 (n = 5–6, Student's t-test, *WNK4-P-Ser P = 0.0057). For co-immunoprecipitation of NCC/WNK4 complexes, WNK4 or NCC were immunoprecipitated with specific antibodies (anti-NCC and anti-WNK4) from homogenized renal cortex protein samples of 3-month-old wild-type mice treated for 5 days with vehicle or rFGF23. Western blot analysis was performed with corresponding anti-NCC or anti-WNK4 antibodies to identify co-precipitated NCC and WNK4 protein, respectively (n = 4–6, Student's t-test, WNK4-P-NCC *P = 0.0116).

  5. F Quantification and original images of intracellular Na+ levels in renal distal tubular cells in live 300-μm-thick kidney slices of 3-month-old WT mice treated with vehicle or rFGF23 (10 μg/mouse), 8 h before necropsy (n = 4, one-way ANOVA followed by SNK test, *P = 0.0026 versus vehicle-treated mice, #P = 0.0175 versus rFGF23-treated mice). Kidney slices were stained with the sodium-sensitive dye SBFI. Chlorothiazide (CTZ, 10 μM) was used as NCC inhibitor.

  6. G Time-dependent changes in intracellular Na+ levels in renal distal tubules in SBFI-loaded, 300-μm-thick, live kidney slices of 3-month-old WT mice treated in vitro at time 0 with rFGF23 (100 ng/ml) or vehicle (n = 3–6). After 105 min, 10 μM CTZ or vehicle was added. Fluorescence intensity in G and H was quantified in 4–9 regions of interest per image, sample, and time point from 2–3 independent experiments. Student's t-test, *P < 0.05 versus vehicle-treated or versus vehicle- + CTZ vehicle-treated (past 105 min). Data represent mean ± s.e.m.

Source data are available for this figure.
Figure 4. FGF23 administration induces hypertension and…
Figure 4. FGF23 administration induces hypertension and heart hypertrophy in a Klotho-dependent fashion
  1. Representative aortic blood pressure curves, heart weight/body weight ratios, and H&E-stained paraffin cross-sections of hearts from 3-month-old male wild-type mice treated for 5 days with vehicle or 10 μg rFGF23 per mouse per day (n = 5–6, Student's t-test, *P = 0.0216). Insets show systolic (BPs), diastolic (BPd), and mean arterial pressure (MAP).

  2. Urinary Na+ excretion per 12 h and heart weight/body weight ratio in 3-month-old male wild-type, VDRΔ/Δ, and Kl−/−/VDRΔ/Δ compound mutant mice treated for 5 days with vehicle or 10 μg rFGF23 per mouse per day (n = 4–6, Student's t-test, *P < 0.05 versus vehicle). Data represent mean ± s.e.m.

Figure 5. FGF23 regulates NCC expression and…
Figure 5. FGF23 regulates NCC expression and phosphorylation in isolated distal tubular segments in a Klotho-dependent manner
  1. Western blotting quantification of NCC expression in isolated distal tubular segments from wild-type (WT) and Fgf23−/− mice treated for 2 h in vitro with vehicle or rFGF23 alone or in combination with specific ERK1/2 (iERK1/2) or SGK1 inhibitors (iSGK1) (n = 4–6, one-way ANOVA followed by SNK test, *P < 0.05 versus vehicle, #P < 0.05 versus rFGF23 alone).

  2. Western blotting quantification of NCC phosphorylation at Ser71 (pNCC S71) in isolated distal tubular segments from WT and Kl−/− mice treated for 2 h in vitro with vehicle or rFGF23 alone or in combination with specific ERK1/2 or SGK1 inhibitors (n = 4, one-way ANOVA followed by SNK test, *P = 0.0002 versus vehicle, #P < 0.005 versus rFGF23 alone).

  3. Western blotting quantification of α-ENaC protein expression in isolated distal tubular segments from WT mice treated for 2 h in vitro with vehicle (Veh) or rFGF23 (n = 3–4). Data represent mean ± s.e.m.

Source data are available for this figure.
Figure 6. Co-treatment of mice with rFGF23…
Figure 6. Co-treatment of mice with rFGF23 and chlorothiazide abrogates the untoward cardiovascular effects of rFGF23
Urinary Na+ excretion per 12 h, urine volume, blood volume, central venous pressure, mean arterial pressure, and heart/body weight ratio in 3-month-old male wild-type mice treated for 5 days with vehicle (Veh), recombinant FGF23 (10 μg per mouse per day), or chlorothiazide (CTZ, 25 mg/kg) alone or in combination (n = 8–10). One-way ANOVA followed by SNK test, *P < 0.05 versus vehicle, #P < 0.05 versus rFGF23. Data represent mean ± s.e.m.
Figure 7. Dietary Na + modulates the…
Figure 7. Dietary Na+ modulates the effects of rFGF23 on blood pressure and renal Na+ handling
  1. A Mean arterial blood pressure (MAP) of 3-month-old male wild-type mice treated for 5 days with vehicle (Veh) or 10 μg rFGF23 per mouse per day on high (High Na), normal (Normal Na), and low (Low Na) sodium diets (n = 6–7, Student's t-test, * P < 0.05 versus vehicle). Inset shows results of two-way ANOVA.

  2. B, C Urinary Na+ excretion corrected by urinary creatinine (Crea), serum Na+ concentration (n = 6–7, Student's t-test * P < 0.05 versus vehicle) and urinary aldosterone corrected by urinary creatinine (Crea) and serum aldosterone concentrations (n = 6–7, Student's t-test, * urine P < 0.0005, serum P < 0.05 versus vehicle) after 5 days of treatment of 3-month-old male wild-type mice with vehicle or rFGF23 (10 μg per mouse per day) on high, normal and low sodium diets.

  3. D, E Western blotting quantification of NCC, α-ENaC, β-ENaC and γ-ENaC protein expression in renal cortical total membrane fractions (n = 4–5, Student's t-test, *P < 0.05 versus vehicle), and ratio of phospho-SGK1 versus total-SGK1 protein expression in kidney total homogenates of 3-month-old wild-type mice on high, normal, and low sodium diets treated for 5 days with vehicle or rFGF23 (n = 4–5, Student's t-test, *P < 0.01 versus vehicle). Data represent mean ± s.e.m.

Source data are available for this figure.
Figure 8. Hyp mice show hypertension and…
Figure 8. Hyp mice show hypertension and increased NCC expression and phosphorylation
  1. A–E Serum intact Fgf23 concentration (A, n = 8–9, Student's t-test, *P = 0.0001 versus vehicle); heart-to-body weight ratio, serum Na+ and urinary Na+ excretion corrected by urinary creatinine (Crea) (B); mean arterial blood pressure (C, n = 8–9, Student's t-test, *P < 0.05 versus vehicle); Western blotting quantification of renal NCC membrane expression and NCC phosphorylation at Ser71, Ser91, and Thr55 (pNCC S71, pNCC S91, pNCC T55) (n = 8–9) (D); and urinary aldosterone corrected by urinary creatinine and serum aldosterone concentrations in 3-month-old male wild-type (WT) and Hyp mice (n = 8–9, Student's t-test, *P < 0.05 versus vehicle) (E). Data represent mean ± s.e.m.

Source data are available for this figure.
Figure 9. Proposed model of FGF23-mediated bone-kidney-heart…
Figure 9. Proposed model of FGF23-mediated bone-kidney-heart axis
Increased circulating FGF23 augments distal renal tubular NCC expression and activity which leads to renal Na+ retention, volume expansion, hypertension, and heart hypertrophy. As a counter-regulatory mechanism, hypernatremia and increased blood volume decrease aldosterone secretion from adrenal glands, leading to a downregulation of renal α-ENaC expression. A low sodium diet augments the hypertensive effect of increased FGF23 signaling in this model, because it interferes with the counter-regulatory downregulation of aldosterone. Similarly, in chronic kidney failure FGF23 and aldosterone signaling pathways are concurrently activated, potentially leading to a stimulation of both NCC and α-ENaC-driven Na+ reabsorption mechanisms in renal distal tubules, and subsequent augmentation of the FGF23-induced volume expansion, hypertension, and heart hypertrophy.

References

    1. Andrukhova O, Zeitz U, Goetz R, Mohammadi M, Lanske B, Erben RG. FGF23 acts directly on renal proximal tubules to induce phosphaturia through activation of the ERK1/2-SGK1 signaling pathway. Bone. 2012;51:621–628.
    1. Andrukhova O, Smorodchenko A, Egerbacher M, Streicher C, Zeitz U, Goetz R, Shalhoub V, Mohammadi M, Pohl EE, Lanske B, et al. FGF23 promotes renal calcium reabsorption through the TRPV5 channel. EMBO J. 2014;33:229–246.
    1. Anour R, Andrukhova O, Ritter E, Zeitz U, Erben RG. Klotho lacks a vitamin D independent physiological role in glucose homeostasis, bone turnover, and steady-state PTH secretion in vivo. PLoS ONE. 2012;7:e31376.
    1. Barron WM, Stamoutsos BA, Lindheimer MD. Role of volume in the regulation of vasopressin secretion during pregnancy in the rat. J Clin Invest. 1984;73:923–932.
    1. Barros NM, Hoac B, Neves RL, Addison WN, Assis DM, Murshed M, Carmona AK, McKee MD. Proteolytic processing of osteopontin by PHEX and accumulation of osteopontin fragments in Hyp mouse bone, the murine model of X-linked hypophosphatemia. J Bone Miner Res. 2013;28:688–699.
    1. Cai H, Cebotaru V, Wang YH, Zhang XM, Cebotaru L, Guggino SE, Guggino WB. WNK4 kinase regulates surface expression of the human sodium chloride cotransporter in mammalian cells. Kidney Int. 2006;69:2162–2170.
    1. Castaneda-Bueno M, Cervantes-Perez LG, Vazquez N, Uribe N, Kantesaria S, Morla L, Bobadilla NA, Doucet A, Alessi DR, Gamba G. Activation of the renal Na+:Cl- cotransporter by angiotensin II is a WNK4-dependent process. Proc Natl Acad Sci USA. 2012;109:7929–7934.
    1. Chen SY, Bhargava A, Mastroberardino L, Meijer OC, Wang J, Buse P, Firestone GL, Verrey F, Pearce D. Epithelial sodium channel regulated by aldosterone-induced protein sgk. Proc Natl Acad Sci USA. 1999;96:2514–2519.
    1. Dhingra R, Sullivan LM, Fox CS, Wang TJ, D'Agostino RB, Sr, Gaziano JM, Vasan RS. Relations of serum phosphorus and calcium levels to the incidence of cardiovascular disease in the community. Arch Intern Med. 2007;167:879–885.
    1. Erben RG, Soegiarto DW, Weber K, Zeitz U, Lieberherr M, Gniadecki R, Möller G, Adamski J, Balling R. Deletion of deoxyribonucleic acid binding domain of the vitamin D receptor abrogates genomic and nongenomic functions of vitamin D. Mol Endocrinol. 2002;16:1524–1537.
    1. Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, Gutierrez OM, Aguillon-Prada R, Lincoln J, Hare JM, et al. FGF23 induces left ventricular hypertrophy. J Clin Invest. 2011;121:4393–4408.
    1. Fischer SS, Kempe DS, Leibrock CB, Rexhepaj R, Siraskar B, Boini KM, Ackermann TF, Foller M, Hocher B, Rosenblatt KP, et al. Hyperaldosteronism in Klotho-deficient mice. Am J Physiol Renal Physiol. 2010;299:F1171–F1177.
    1. Flores SY, Debonneville C, Staub O. The role of Nedd4/Nedd4-like dependant ubiquitylation in epithelial transport processes. Pflugers Arch. 2003;446:334–338.
    1. Harootunian AT, Kao JP, Eckert BK, Tsien RY. Fluorescence ratio imaging of cytosolic free Na+ in individual fibroblasts and lymphocytes. J Biol Chem. 1989;264:19458–19467.
    1. Hesse M, Frohlich LF, Zeitz U, Lanske B, Erben RG. Ablation of vitamin D signaling rescues bone, mineral, and glucose homeostasis in Fgf-23 deficient mice. Matrix Biol. 2007;26:75–84.
    1. Hu MC, Shi M, Zhang J, Pastor J, Nakatani T, Lanske B, Razzaque MS, Rosenblatt KP, Baum MG, Kuro-o M, et al. Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. FASEB J. 2010;24:3438–3450.
    1. Juppner H, Wolf M, Salusky IB. FGF-23: More than a regulator of renal phosphate handling? J Bone Miner Res. 2010;25:2091–2097.
    1. Kahle KT, Wilson FH, Leng Q, Lalioti MD, O'Connell AD, Dong K, Rapson AK, MacGregor GG, Giebisch G, Hebert SC, et al. WNK4 regulates the balance between renal NaCl reabsorption and K+ secretion. Nat Genet. 2003;35:372–376.
    1. Ko B, Joshi LM, Cooke LL, Vazquez N, Musch MW, Hebert SC, Gamba G, Hoover RS. Phorbol ester stimulation of RasGRP1 regulates the sodium-chloride cotransporter by a PKC-independent pathway. Proc Natl Acad Sci USA. 2007;104:20120–20125.
    1. Ko B, Kamsteeg EJ, Cooke LL, Moddes LN, Deen PM, Hoover RS. RasGRP1 stimulation enhances ubiquitination and endocytosis of the sodium-chloride cotransporter. Am J Physiol Renal Physiol. 2010;299:F300–F309.
    1. Ko B, Mistry AC, Hanson L, Mallick R, Wynne BM, Thai TL, Bailey JL, Klein JD, Hoover RS. Aldosterone acutely stimulates NCC activity via a SPAK-mediated pathway. Am J Physiol Renal Physiol. 2013;305:F645–F652.
    1. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, Ohyama Y, Kurabayashi M, Kaname T, Kume E, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51.
    1. Lang F, Artunc F, Vallon V. The physiological impact of the serum and glucocorticoid-inducible kinase SGK1. Curr Opin Nephrol Hypertens. 2009;18:439–448.
    1. Lattanzio MR, Weir MR. Does blockade of the Renin-Angiotensin-aldosterone system slow progression of all forms of kidney disease? Curr Hypertens Rep. 2010;12:369–377.
    1. Lee HB, Blaufox MD. Blood volume in the rat. J Nucl Med. 1985;26:72–76.
    1. Lee IH, Campbell CR, Song SH, Day ML, Kumar S, Cook DI, Dinudom A. The activity of the epithelial sodium channels is regulated by caveolin-1 via a Nedd4-2-dependent mechanism. J Biol Chem. 2009;284:12663–12669.
    1. Li YC, Amling M, Pirro AE, Priemel M, Meuse J, Baron R, Delling G, Demay MB. Normalization of mineral ion homeostasis by dietary means prevents hyperparathyroidism, rickets, and osteomalacia, but not alopecia in vitamin D receptor-ablated mice. Endocrinology. 1998;139:4391–4396.
    1. Liu S, Guo R, Simpson LG, Xiao ZS, Burnham CE, Quarles LD. Regulation of fibroblastic growth factor 23 expression but not degradation by PHEX. J Biol Chem. 2003;278:37419–37426.
    1. van der Lubbe N, Lim CH, Meima ME, van VR, Rosenbaek LL, Mutig K, Danser AH, Fenton RA, Zietse R, Hoorn EJ. Aldosterone does not require angiotensin II to activate NCC through a WNK4-SPAK-dependent pathway. Pflugers Arch. 2012;463:853–863.
    1. Martin A, David V, Quarles LD. Regulation and function of the FGF23/klotho endocrine pathways. Physiol Rev. 2012;92:131–155.
    1. Masilamani S, Kim GH, Mitchell C, Wade JB, Knepper MA. Aldosterone-mediated regulation of ENaC alpha, beta, and gamma subunit proteins in rat kidney. J Clin Invest. 1999;104:R19–R23.
    1. May A, Puoti A, Gaeggeler HP, Horisberger JD, Rossier BC. Early effect of aldosterone on the rate of synthesis of the epithelial sodium channel alpha subunit in A6 renal cells. J Am Soc Nephrol. 1997;8:1813–1822.
    1. Monroy A, Plata C, Hebert SC, Gamba G. Characterization of the thiazide-sensitive Na(+)-Cl(-) cotransporter: a new model for ions and diuretics interaction. Am J Physiol Renal Physiol. 2000;279:F161–F169.
    1. Naesens M, Steels P, Verberckmoes R, Vanrenterghem Y, Kuypers D. Bartter's and Gitelman's syndromes: from gene to clinic. Nephron Physiol. 2004;96:65–78.
    1. Nehgme R, Fahey JT, Smith C, Carpenter TO. Cardiovascular abnormalities in patients with X-linked hypophosphatemia. J Clin Endocrinol Metab. 1997;82:2450–2454.
    1. Nesterov V, Dahlmann A, Krueger B, Bertog M, Loffing J, Korbmacher C. Aldosterone-dependent and -independent regulation of the epithelial sodium channel (ENaC) in mouse distal nephron. Am J Physiol Renal Physiol. 2012;303:F1289–F1299.
    1. Ohta A, Rai T, Yui N, Chiga M, Yang SS, Lin SH, Sohara E, Sasaki S, Uchida S. Targeted disruption of the Wnk4 gene decreases phosphorylation of Na-Cl cotransporter, increases Na excretion and lowers blood pressure. Hum Mol Genet. 2009;18:3978–3986.
    1. Pacheco-Alvarez D, Cristobal PS, Meade P, Moreno E, Vazquez N, Munoz E, Diaz A, Juarez ME, Gimenez I, Gamba G. The Na+:Cl- cotransporter is activated and phosphorylated at the amino-terminal domain upon intracellular chloride depletion. J Biol Chem. 2006;281:28755–28763.
    1. Ring AM, Leng Q, Rinehart J, Wilson FH, Kahle KT, Hebert SC, Lifton RP. An SGK1 site in WNK4 regulates Na+ channel and K+ channel activity and has implications for aldosterone signaling and K+ homeostasis. Proc Natl Acad Sci USA. 2007;104:4025–4029.
    1. Rozansky DJ, Cornwall T, Subramanya AR, Rogers S, Yang YF, David LL, Zhu X, Yang CL, Ellison DH. Aldosterone mediates activation of the thiazide-sensitive Na-Cl cotransporter through an SGK1 and WNK4 signaling pathway. J Clin Invest. 2009;119:2601–2612.
    1. Saito H, Maeda A, Ohtomo S, Hirata M, Kusano K, Kato S, Ogata E, Segawa H, Miyamoto K, Fukushima N. Circulating FGF-23 is regulated by 1alpha,25-dihydroxyvitamin D3 and phosphorus in vivo. J Biol Chem. 2005;280:2543–2549.
    1. Scialla JJ, Lau WL, Reilly MP, Isakova T, Yang HY, Crouthamel MH, Chavkin NW, Rahman M, Wahl P, Amaral AP, et al. Fibroblast growth factor 23 is not associated with and does not induce arterial calcification. Kidney Int. 2013;83:1159–1168.
    1. Sharma S, McFann K, Chonchol M, de Boer IH, Kendrick J. Association between dietary sodium and potassium intake with chronic kidney disease in US adults: a cross-sectional study. Am J Nephrol. 2013;37:526–533.
    1. Shimada T, Mizutani S, Muto T, Yoneya T, Hino R, Takeda S, Takeuchi Y, Fujita T, Fukumoto S, Yamashita T. Cloning and characterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proc Natl Acad Sci USA. 2001;98:6500–6505.
    1. Shimada T, Hasegawa H, Yamazaki Y, Muto T, Hino R, Takeuchi Y, Fujita T, Nakahara K, Fukumoto S, Yamashita T. FGF-23 is a potent regulator of vitamin D metabolism and phosphate homeostasis. J Bone Miner Res. 2004a;19:429–435.
    1. Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T, Fukumoto S, Tomizuka K, Yamashita T. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest. 2004b;113:561–568.
    1. Shimada T, Yamazaki Y, Takahashi M, Hasegawa H, Urakawa I, Oshima T, Ono K, Kakitani M, Tomizuka K, Fujita T, et al. Vitamin D receptor-independent FGF23 actions in regulating phosphate and vitamin D metabolism. Am J Physiol Renal Physiol. 2005;289:F1088–F1095.
    1. Sitara D, Razzaque MS, Hesse M, Yoganathan S, Taguchi T, Erben RG, Juppner H, Lanske B. Homozygous ablation of fibroblast growth factor-23 results in hyperphosphatemia and impaired skeletogenesis, and reverses hypophosphatemia in Phex-deficient mice. Matrix Biol. 2004;23:421–432.
    1. Streicher C, Zeitz U, Andrukhova O, Rupprecht A, Pohl E, Larsson TE, Windisch W, Lanske B, Erben RG. Long-term Fgf23 deficiency does not influence aging, glucose homeostasis, or fat metabolism in mice with a nonfunctioning vitamin D receptor. Endocrinology. 2012;153:1795–1805.
    1. The ADHR Consortium. Autosomal dominant hypophosphataemic rickets is associated with mutations in FGF23. Nat Genet. 2000;26:345–348.
    1. Tsujikawa H, Kurotaki Y, Fujimori T, Fukuda K, Nabeshima Y. Klotho, a gene related to a syndrome resembling human premature aging, functions in a negative regulatory circuit of vitamin D endocrine system. Mol Endocrinol. 2003;17:2393–2403.
    1. Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K, Fujita T, Fukumoto S, Yamashita T. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature. 2006;444:770–774.
    1. Weber TJ, Liu S, Indridason OS, Quarles LD. Serum FGF23 levels in normal and disordered phosphorus homeostasis. J Bone Miner Res. 2003;18:1227–1234.
    1. Wilson FH, Disse-Nicodeme S, Choate KA, Ishikawa K, Nelson-Williams C, Desitter I, Gunel M, Milford DV, Lipkin GW, Achard JM, et al. Human hypertension caused by mutations in WNK kinases. Science. 2001;293:1107–1112.
    1. Xie J, Cha SK, An SW, Kuro O, Birnbaumer L, Huang CL. Cardioprotection by Klotho through downregulation of TRPC6 channels in the mouse heart. Nat Commun. 2012;3:1238.
    1. Yang CL, Angell J, Mitchell R, Ellison DH. WNK kinases regulate thiazide-sensitive Na-Cl cotransport. J Clin Invest. 2003;111:1039–1045.

Source: PubMed

3
購読する