A comparative transcriptome analysis identifying FGF23 regulated genes in the kidney of a mouse CKD model

Bing Dai, Valentin David, Aline Martin, Jinsong Huang, Hua Li, Yan Jiao, Weikuan Gu, L Darryl Quarles, Bing Dai, Valentin David, Aline Martin, Jinsong Huang, Hua Li, Yan Jiao, Weikuan Gu, L Darryl Quarles

Abstract

Elevations of circulating Fibroblast growth factor 23 (FGF23) are associated with adverse cardiovascular outcomes and progression of renal failure in chronic kidney disease (CKD). Efforts to identify gene products whose transcription is directly regulated by FGF23 stimulation of fibroblast growth factor receptors (FGFR)/α-Klotho complexes in the kidney is confounded by both systemic alterations in calcium, phosphorus and vitamin D metabolism and intrinsic alterations caused by the underlying renal pathology in CKD. To identify FGF23 responsive genes in the kidney that might explain the association between FGF23 and adverse outcomes in CKD, we performed comparative genome wide analysis of gene expression profiles in the kidney of the Collagen 4 alpha 3 null mice (Col4a3(-/-)) model of progressive kidney disease with kidney expression profiles of Hypophosphatemic (Hyp) and FGF23 transgenic mouse models of elevated FGF23. The different complement of potentially confounding factors in these models allowed us to identify genes that are directly targeted by FGF23. This analysis found that α-Klotho, an anti-aging hormone and FGF23 co-receptor, was decreased by FGF23. We also identified additional FGF23-responsive transcripts and activation of networks associated with renal damage and chronic inflammation, including lipocalin 2 (Lcn2), transforming growth factor beta (TGF-β) and tumor necrosis factor-alpha (TNF-α) signaling pathways. Finally, we found that FGF23 suppresses angiotensin-converting enzyme 2 (ACE2) expression in the kidney, thereby providing a pathway for FGF23 regulation of the renin-angiotensin system. These gene products provide a possible mechanistic links between elevated FGF23 and pathways responsible for renal failure progression and cardiovascular diseases.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. (A) Gross appearance and (B)…
Figure 1. (A) Gross appearance and (B) body weight of 12 week-old wild-type (WT), and Col4a3−/− mice.
(C) Kidney morphology showing reduced perfusion and (D) H&E renal histology showing glomerulosclerosis in the Col4a3−/− animals. Values are expressed as mean±SEM, P<0.05 vs: (*) WT, n≥13 mice/group.
Figure 2. (A) Cluster analysis of microarray…
Figure 2. (A) Cluster analysis of microarray performed on Kidneys from 12 week-old wild-type (WT), and Col4a3−/− mice.
Gene expression is represented on the heat map from the less expressed (blue) to the more expressed (red). (B) Graphic representation of transcripts expressed at least five fold in Col4a3−/− as compared to WT.
Figure 3. (A) Western blots and corresponding…
Figure 3. (A) Western blots and corresponding (B) quantification of the most upregulated and downregulated gene product in WT and Col4a3−/− mice.
Figure 4. Immunohistochemistry of Cyp27b1, Cyp24a1 and…
Figure 4. Immunohistochemistry of Cyp27b1, Cyp24a1 and α-klotho in the kidneys of WT and Col4a3−/− mice.
Figure 5. Venn diagram of (A) the…
Figure 5. Venn diagram of (A) the total number of genes detected in all 3 data sets and (B) significantly regulated genes in all 3 models.
Figure 6. Ingenuity Pathway analysis (IPA) performed…
Figure 6. Ingenuity Pathway analysis (IPA) performed on 31 listed genes.
The network is built according the identified interconnected pathways involving the highest majority of genes. Genes represented in pink color belong to the cluster. Genes represented in bold font are central regulators of the identified pathways that do not belong to the cluster. Genes represented in white color are other intermediary regulators that do not belong to the cluster.

References

    1. Martin A, David V, Quarles LD (2012) Regulation and function of the FGF23/klotho endocrine pathways. Physiol Rev 92: 131–155.
    1. Kurosu H, Ogawa Y, Miyoshi M, Yamamoto M, Nandi A, et al. (2006) Regulation of fibroblast growth factor-23 signaling by klotho. J Biol Chem 281: 6120–6123.
    1. Li H, Martin A, David V, Quarles LD (2011) Compound deletion of Fgfr3 and Fgfr4 partially rescues the Hyp mouse phenotype. Am J Physiol Endocrinol Metab 300: E508–517.
    1. Liu S, Quarles LD (2007) How fibroblast growth factor 23 works. J Am Soc Nephrol 18: 1637–1647.
    1. Liu S, Tang W, Zhou J, Stubbs JR, Luo Q, et al. (2006) Fibroblast growth factor 23 is a counter-regulatory phosphaturic hormone for vitamin D. J Am Soc Nephrol. 17: 1305–1315.
    1. Mirams M, Robinson BG, Mason RS, Nelson AE (2004) Bone as a source of FGF23: regulation by phosphate? Bone 35: 1192–1199.
    1. Liu S, Guo R, Simpson LG, Xiao ZS, Burnham CE, et al. (2003) Regulation of fibroblastic growth factor 23 expression but not degradation by PHEX. J Biol Chem 278: 37419–37426.
    1. Martin A, Liu S, David V, Li H, Karydis A, et al. (2011) Bone proteins PHEX and DMP1 regulate fibroblastic growth factor Fgf23 expression in osteocytes through a common pathway involving FGF receptor (FGFR) signaling. Faseb J 25: 2551–2562.
    1. Quarles LD (2008) Endocrine functions of bone in mineral metabolism regulation. J Clin Invest 118: 3820–3828.
    1. Hasegawa H, Nagano N, Urakawa I, Yamazaki Y, Iijima K, et al. (2010) Direct evidence for a causative role of FGF23 in the abnormal renal phosphate handling and vitamin D metabolism in rats with early-stage chronic kidney disease. Kidney Int 78: 975–980.
    1. Isakova T, Wolf MS (2010) FGF23 or PTH: which comes first in CKD ? Kidney Int 78: 947–949.
    1. Wetmore JB, Quarles LD (2009) Calcimimetics or vitamin D analogs for suppressing parathyroid hormone in end-stage renal disease: time for a paradigm shift? Nat Clin Pract Nephrol 5: 24–33.
    1. Weber TJ, Liu S, Indridason OS, Quarles LD (2003) Serum FGF23 levels in normal and disordered phosphorus homeostasis. J Bone Miner Res 18: 1227–1234.
    1. Imanishi Y, Inaba M, Nakatsuka K, Nagasue K, Okuno S, et al. (2004) FGF-23 in patients with end-stage renal disease on hemodialysis. Kidney Int 65: 1943–1946.
    1. Komaba H, Fukagawa M (2010) FGF23-parathyroid interaction: implications in chronic kidney disease. Kidney Int 77: 292–298.
    1. Nehgme R, Fahey JT, Smith C, Carpenter TO (1997) Cardiovascular abnormalities in patients with X-linked hypophosphatemia. J Clin Endocrinol Metab 82: 2450–2454.
    1. Hsu HJ, Wu MS (2009) Fibroblast growth factor 23: a possible cause of left ventricular hypertrophy in hemodialysis patients. Am J Med Sci 337: 116–122.
    1. Parker BD, Schurgers LJ, Brandenburg VM, Christenson RH, Vermeer C, et al. (2010) The associations of fibroblast growth factor 23 and uncarboxylated matrix Gla protein with mortality in coronary artery disease: the Heart and Soul Study. Ann Intern Med 152: 640–648.
    1. Stubbs JR, Quarles LD (2009) Fibroblast growth factor 23: uremic toxin or innocent bystander in chronic kidney disease? Nephrol News Issues 23: 33–34, 36–37.
    1. Gutierrez OM, Mannstadt M, Isakova T, Rauh-Hain JA, Tamez H, et al. (2008) Fibroblast growth factor 23 and mortality among patients undergoing hemodialysis. N Engl J Med 359: 584–592.
    1. Isakova T, Xie H, Yang W, Xie D, Anderson AH, et al. (2011) Fibroblast growth factor 23 and risks of mortality and end-stage renal disease in patients with chronic kidney disease. JAMA 305: 2432–2439.
    1. Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, et al. (2011) FGF23 induces left ventricular hypertrophy. J Clin Invest 121: 4393–4408.
    1. Fliser D, Kollerits B, Neyer U, Ankerst DP, Lhotta K, et al. (2007) Fibroblast growth factor 23 (FGF23) predicts progression of chronic kidney disease: the Mild to Moderate Kidney Disease (MMKD) Study. J Am Soc Nephrol 18: 2600–2608.
    1. Wolf M, Molnar MZ, Amaral AP, Czira ME, Rudas A, et al. (2011) Elevated fibroblast growth factor 23 is a risk factor for kidney transplant loss and mortality. J Am Soc Nephrol 22: 956–966.
    1. Liu S, Vierthaler L, Tang W, Zhou J, Quarles LD (2008) FGFR3 and FGFR4 do not mediate renal effects of FGF23. J Am Soc Nephrol 19: 2342–2350.
    1. Fukumoto S, Araya K, Backenroth R, Takeuchi Y, Nakayama K, et al. (2005) A novel mutation in fibroblast growth factor 23 gene as a cause of tumoral calcinosis. Journal of Clinical Endocrinology & Metabolism 90: 5523–5527.
    1. Stubbs JR, Liu S, Tang W, Zhou J, Wang Y, et al. (2007) Role of hyperphosphatemia and 1,25-dihydroxyvitamin D in vascular calcification and mortality in fibroblastic growth factor 23 null mice. J Am Soc Nephrol 18: 2116–2124.
    1. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, et al. (1997) Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 390: 45–51.
    1. Stubbs JR, He N, Idiculla A, Gillihan R, Liu S, et al. (2011) Longitudinal evaluation of FGF23 changes and mineral metabolism abnormalities in a mouse model of chronic kidney disease. J Bone Miner Res 27: 38–46.
    1. Liu S, Zhou J, Tang W, Jiang X, Rowe DW, et al. (2006) Pathogenic role of Fgf23 in Hyp mice. Am J Physiol Endocrinol Metab 291: E38–49.
    1. Marsell R, Krajisnik T, Goransson H, Ohlsson C, Ljunggren O, et al. (2008) Gene expression analysis of kidneys from transgenic mice expressing fibroblast growth factor-23. Nephrol Dial Transplant 23: 827–833.
    1. Stubbs JR, He N, Idiculla A, Gillihan R, Liu S, et al.. (2011) Longitudinal evaluation of FGF23 changes and mineral metabolism abnormalities in a mouse model of chronic kidney disease. J Bone Miner Res.
    1. Wolf DA, Zhou C, Wee S (2003) The COP9 signalosome: an assembly and maintenance platform for cullin ubiquitin ligases? Nat Cell Biol 5: 1029–1033.
    1. Boyden LM, Choi M, Choate KA, Nelson-Williams CJ, Farhi A, et al. (2012) Mutations in kelch-like 3 and cullin 3 cause hypertension and electrolyte abnormalities. Nature 482: 98–102.
    1. DeLozier TC, Tsao CC, Coulter SJ, Foley J, Bradbury JA, et al. (2004) CYP2C44, a new murine CYP2C that metabolizes arachidonic acid to unique stereospecific products. J Pharmacol Exp Ther 310: 845–854.
    1. Camargo SM, Singer D, Makrides V, Huggel K, Pos KM, et al. (2009) Tissue-specific amino acid transporter partners ACE2 and collectrin differentially interact with hartnup mutations. Gastroenterology 136: 872–882.
    1. Belge H, Devuyst O (2010) [Parvalbumin and regulation of ion transport in the distal convoluted tubule of the kidney]. Med Sci (Paris) 26: 566–568.
    1. Song YR, You SJ, Lee YM, Chin HJ, Chae DW, et al. (2010) Activation of hypoxia-inducible factor attenuates renal injury in rat remnant kidney. Nephrol Dial Transplant 25: 77–85.
    1. Wang W, Shen J, Cui Y, Jiang J, Chen S, et al. (2012) Impaired sodium excretion and salt-sensitive hypertension in corin-deficient mice. Kidney Int 82: 26–33.
    1. Meyer MH, Dulde E, Meyer RA Jr (2004) The genomic response of the mouse kidney to low-phosphate diet is altered in X-linked hypophosphatemia. Physiol Genomics 18: 4–11.
    1. Tu Z, Kelley VR, Collins T, Lee FS (2001) I kappa B kinase is critical for TNF-alpha-induced VCAM1 gene expression in renal tubular epithelial cells. J Immunol 166: 6839–6846.
    1. Machuca E, Benoit G, Antignac C (2009) Genetics of nephrotic syndrome: connecting molecular genetics to podocyte physiology. Hum Mol Genet 18: R185–194.
    1. Yoder BK, Mulroy S, Eustace H, Boucher C, Sandford R (2006) Molecular pathogenesis of autosomal dominant polycystic kidney disease. Expert Rev Mol Med 8: 1–22.
    1. Weinman EJ, Steplock D, Shenolikar S, Biswas R (2011) Fibroblast growth factor-23-mediated inhibition of renal phosphate transport in mice requires sodium-hydrogen exchanger regulatory factor-1 (NHERF-1) and synergizes with parathyroid hormone. J Biol Chem 286: 37216–37221.
    1. Yuan B, Xing Y, Horst RL, Drezner MK (2004) Evidence for abnormal translational regulation of renal 25-hydroxyvitamin D-1alpha-hydroxylase activity in the hyp-mouse. Endocrinology 145: 3804–3812.
    1. Yamazaki Y, Imura A, Urakawa I, Shimada T, Murakami J, et al. (2010) Establishment of sandwich ELISA for soluble alpha-Klotho measurement: Age-dependent change of soluble alpha-Klotho levels in healthy subjects. Biochem Biophys Res Commun 398: 513–518.
    1. Koh N, Fujimori T, Nishiguchi S, Tamori A, Shiomi S, et al. (2001) Severely reduced production of klotho in human chronic renal failure kidney. Biochem Biophys Res Commun 280: 1015–1020.
    1. Kuro-o M (2011) Phosphate and Klotho. Kidney Int Suppl 121: S20–23.
    1. Haussler MR, Haussler CA, Whitfield GK, Hsieh JC, Thompson PD, et al. (2010) The nuclear vitamin D receptor controls the expression of genes encoding factors which feed the "Fountain of Youth" to mediate healthful aging. J Steroid Biochem Mol Biol 121: 88–97.
    1. Freedman NJ, Kim LK, Murray JP, Exum ST, Brian L, et al. (2002) Phosphorylation of the platelet-derived growth factor receptor-beta and epidermal growth factor receptor by G protein-coupled receptor kinase-2. Mechanisms for selectivity of desensitization. J Biol Chem 277: 48261–48269.
    1. Haruna Y, Kashihara N, Satoh M, Tomita N, Namikoshi T, et al. (2007) Amelioration of progressive renal injury by genetic manipulation of Klotho gene. Proc Natl Acad Sci U S A 104: 2331–2336.
    1. Gurley SB, Allred A, Le TH, Griffiths R, Mao L, et al. (2006) Altered blood pressure responses and normal cardiac phenotype in ACE2-null mice. J Clin Invest 116: 2218–2225.
    1. de Borst MH, Vervloet MG, ter Wee PM, Navis G (2011) Cross talk between the renin-angiotensin-aldosterone system and vitamin D-FGF-23-klotho in chronic kidney disease. J Am Soc Nephrol 22: 1603–1609.
    1. Mitani H, Ishizaka N, Aizawa T, Ohno M, Usui S, et al. (2002) In vivo klotho gene transfer ameliorates angiotensin II-induced renal damage. Hypertension 39: 838–843.
    1. Chan MR, Thomas CP, Torrealba JR, Djamali A, Fernandez LA, et al. (2009) Recurrent atypical hemolytic uremic syndrome associated with factor I mutation in a living related renal transplant recipient. Am J Kidney Dis 53: 321–326.
    1. Thurman JM, Ljubanovic D, Royer PA, Kraus DM, Molina H, et al. (2006) Altered renal tubular expression of the complement inhibitor Crry permits complement activation after ischemia/reperfusion. J Clin Invest 116: 357–368.
    1. Hakkim A, Furnrohr BG, Amann K, Laube B, Abed UA, et al. (2010) Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A 107: 9813–9818.
    1. Yasutomo K, Horiuchi T, Kagami S, Tsukamoto H, Hashimura C, et al. (2001) Mutation of DNASE1 in people with systemic lupus erythematosus. Nat Genet 28: 313–314.
    1. Datta R, Shah GN, Rubbelke TS, Waheed A, Rauchman M, et al. (2010) Progressive renal injury from transgenic expression of human carbonic anhydrase IV folding mutants is enhanced by deficiency of p58IPK. Proc Natl Acad Sci U S A 107: 6448–6452.
    1. Kaunisto K, Parkkila S, Rajaniemi H, Waheed A, Grubb J, et al. (2002) Carbonic anhydrase XIV: luminal expression suggests key role in renal acidification. Kidney Int 61: 2111–2118.
    1. Santer R, Groth S, Kinner M, Dombrowski A, Berry GT, et al. (2002) The mutation spectrum of the facilitative glucose transporter gene SLC2A2 (GLUT2) in patients with Fanconi-Bickel syndrome. Hum Genet 110: 21–29.
    1. Voegele AF, Jerkovic L, Wellenzohn B, Eller P, Kronenberg F, et al. (2002) Characterization of the vitamin E-binding properties of human plasma afamin. Biochemistry 41: 14532–14538.
    1. Freue GV, Sasaki M, Meredith A, Gunther OP, Bergman A, et al. (2010) Proteomic signatures in plasma during early acute renal allograft rejection. Mol Cell Proteomics 9: 1954–1967.
    1. Cowland JB, Borregaard N (1997) Molecular characterization and pattern of tissue expression of the gene for neutrophil gelatinase-associated lipocalin from humans. Genomics 45: 17–23.
    1. Yang J, Goetz D, Li JY, Wang W, Mori K, et al. (2002) An iron delivery pathway mediated by a lipocalin. Mol Cell 10: 1045–1056.
    1. Yang J, Blum A, Novak T, Levinson R, Lai E, et al. (2002) An epithelial precursor is regulated by the ureteric bud and by the renal stroma. Dev Biol 246: 296–310.
    1. Meldrum KK, Hile K, Meldrum DR, Crone JA, Gearhart JP, et al. (2002) Simulated ischemia induces renal tubular cell apoptosis through a nuclear factor-kappaB dependent mechanism. J Urol 168: 248–252.
    1. Haussler U, von Wichert G, Schmid RM, Keller F, Schneider G (2005) Epidermal growth factor activates nuclear factor-kappaB in human proximal tubule cells. Am J Physiol Renal Physiol 289: F808–815.
    1. Mishra J, Mori K, Ma Q, Kelly C, Yang J, et al. (2004) Amelioration of ischemic acute renal injury by neutrophil gelatinase-associated lipocalin. J Am Soc Nephrol 15: 3073–3082.
    1. Viau A, El Karoui K, Laouari D, Burtin M, Nguyen C, et al. (2010) Lipocalin 2 is essential for chronic kidney disease progression in mice and humans. J Clin Invest 120: 4065–4076.
    1. Holman RR, Haffner SM, McMurray JJ, Bethel MA, Holzhauer B, et al. (2010) Effect of nateglinide on the incidence of diabetes and cardiovascular events. N Engl J Med 362: 1463–1476.
    1. Hunt MC, Rautanen A, Westin MA, Svensson LT, Alexson SE (2006) Analysis of the mouse and human acyl-CoA thioesterase (ACOT) gene clusters shows that convergent, functional evolution results in a reduced number of human peroxisomal ACOTs. FASEB J 20: 1855–1864.
    1. Ju W, Eichinger F, Bitzer M, Oh J, McWeeney S, et al. (2009) Renal gene and protein expression signatures for prediction of kidney disease progression. Am J Pathol 174: 2073–2085.
    1. Bolton CH, Downs LG, Victory JG, Dwight JF, Tomson CR, et al. (2001) Endothelial dysfunction in chronic renal failure: roles of lipoprotein oxidation and pro-inflammatory cytokines. Nephrol Dial Transplant 16: 1189–1197.
    1. Pereira BJ, Shapiro L, King AJ, Falagas ME, Strom JA, et al. (1994) Plasma levels of IL-1 beta, TNF alpha and their specific inhibitors in undialyzed chronic renal failure, CAPD and hemodialysis patients. Kidney Int 45: 890–896.
    1. Knight EL, Rimm EB, Pai JK, Rexrode KM, Cannuscio CC, et al. (2004) Kidney dysfunction, inflammation, and coronary events: a prospective study. J Am Soc Nephrol 15: 1897–1903.
    1. Koleganova N, Piecha G, Ritz E, Schirmacher P, Muller A, et al. (2009) Arterial calcification in patients with chronic kidney disease. Nephrol Dial Transplant 24: 2488–2496.
    1. Kresse A, Konermann C, Degrandi D, Beuter-Gunia C, Wuerthner J, et al. (2008) Analyses of murine GBP homology clusters based on in silico, in vitro and in vivo studies. BMC Genomics 9: 158.
    1. Feng JQ, Huang H, Lu Y, Ye L, Xie Y, et al. (2003) The Dentin matrix protein 1 (Dmp1) is specifically expressed in mineralized, but not soft, tissues during development. J Dent Res 82: 776–780.
    1. David V, Martin A, Hedge AM, Drezner MK, Rowe PS (2011) ASARM peptides: PHEX-dependent and -independent regulation of serum phosphate. Am J Physiol Renal Physiol 300: F783–791.
    1. Liu S, Zhou J, Tang W, Menard R, Feng JQ, et al. (2008) Pathogenic role of Fgf23 in Dmp1-null mice. Am J Physiol Endocrinol Metab 295: E254–261.
    1. David V, Martin A, Hedge AM, Rowe PS (2009) Matrix extracellular phosphoglycoprotein (MEPE) is a new bone renal hormone and vascularization modulator. Endocrinology 150: 4012–4023.
    1. Martin A, David V, Laurence JS, Schwarz PM, Lafer EM, et al. (2008) Degradation of MEPE, DMP1, and release of SIBLING ASARM-peptides (minhibins): ASARM-peptide(s) are directly responsible for defective mineralization in HYP. Endocrinology 149: 1757–1772.
    1. Gesek FA, Friedman PA (1995) Sodium entry mechanisms in distal convoluted tubule cells. Am J Physiol 268: F89–98.

Source: PubMed

Подписаться