<<

REVIEW www.jasn.org

A Land of Controversy: Fibroblast Growth Factor-23 and Uremic Cardiac Hypertrophy

Jing-Fu Bao ,1 Pan-Pan Hu,1 Qin-Ying She ,2 and Aiqing Li1

1State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China 2National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China

ABSTRACT Cardiac hypertrophy is a common feature in patients with CKD. Recent studies amino-terminal and approximately revealed that two phosphate regulators, fibroblast growth factor-23 and 12 kDa carboxy-terminal (cFGF-23) a-Klotho, are highly involved in the pathophysiologic process of CKD-induced car- fragments, and the latter can compete diac hypertrophy. With decreasing renal function, elevated fibroblast growth factor- with iFGF-23 for binding to the FGF re- 23 and decreased a-Klotho may contribute to cardiac hypertrophy by targeting the ceptor (FGFR), thus exerting certain bi- heart directly or by inducing systemic changes, such as vascular injury, hemody- ologic functions (Figure 1).6,7 Unlike namic disorders, and inflammation. However, several studies have demonstrated other FGF subfamilies, the FGF-19 sub- that disturbances in the fibroblast growth factor-23/a-Klotho axis do not lead to family lacks a heparin binding domain, cardiac hypertrophy. In this review, we describe the cardiac effects of the fibroblast which is replaced by a Klotho binding growth factor-23/a-Klotho axis and summarize recent progress in this field. In ad- domain. Hence, low affinity to heparin dition, we present not only the main controversies in this field but also provide sulfate enables FGF-19 subfamily mem- possible directions to resolve these disputes. bers to enter the bloodstream and func- tion as endocrine factors.8–10 On the JASN 31: 1423–1434, 2020. doi: https://doi.org/10.1681/ASN.2020010081 other hand, the lack of a heparin binding domain renders these incapable of forming the FGF-FGFR-heparin sul- Cardiovascular complications of CKD FGF-23/a-Klotho axis is unable to in- fate complex and activating downstream seriously affect the prognosis of patients duce cardiac hypertrophy. This review signaling.11 Klotho, a single-pass trans- with CKD.1 Cardiac hypertrophy is the will focus on the FGF-23/a-Klotho axis membrane , interacts with FGF- most common of such complications in uremic cardiac hypertrophy, present 19 subfamily members and FGFR to and underlies the pathology for other controversies about their roles in cardio- form the FGF-FGFR-Klotho complex, heart conditions. However, there are still vascular abnormalities, and provide di- and then activates FGFR signaling.12,13 many unknowns regarding the patho- rection for future research. physiology of CKD-induced cardiac hy- a-Klotho pertrophy.2 To date, several factors are Klotho is a membrane protein that dem- onstrates antiaging effects.14 It can be di- considered to be involved in the patho- BRIEF INTRODUCTION TO THE genesis of uremic cardiac hypertrophy, vided into three subtypes: a-Klotho, FGF-23/a-KLOTHO AXIS 15–17 including hemodynamic overload, sys- b-Klotho, and g-Klotho. Among fl temic in ammation, accumulation of FGF-23 3 uremic toxins, and other factors. Recent FGF-23, a newly discovered FGF-19 sub- fi Published online ahead of print. Publication date ndings suggest that two proteins that are family member, is an approximately available at www.jasn.org. highly involved in phosphate metabolism, 32 kDa glycoprotein that is mainly fi Correspondence: Dr. Aiqing Li, State Key Labora- broblast growth factor-23 (FGF-23) and secreted by osteocytes or osteoblasts.4 tory of Organ Failure Research, National Clinical its coreceptor a-Klotho play pivotal roles After removal of the signal peptide and Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, 1838 North in cardiovascular abnormalities (espe- glycosylation, intact FGF-23 (iFGF-23) Guangzhou Avenue, Guangzhou 510515, China. cially cardiac hypertrophy) in the setting is secreted into the blood (Figure 1).5 Email: [email protected] 176 179 of CKD. However, some researchers dis- iFGF-23 can be cleaved at R XXR / Copyright © 2020 by the American Society of 180 pute this, arguing that disturbance in the S into inactive approximately 18 kDa Nephrology

JASN 31: 1423–1434, 2020 ISSN : 1046-6673/3107-1423 1423 REVIEW www.jasn.org

roidism lead to increased circulating levels of FGF-23. Often, worse kidney function is associated with higher circu- lating FGF-23 concentrations.27–29 Al- though the increase in FGF-23 helps maintain a normal serum phosphate level, epidemiologic studies revealed that a high circulating level of FGF-23 is an independent risk factor for cardiovas- cular disease and may increase mortality in patients with CKD.30–35

Uremic Cardiac Hypertrophy and FGF-23 The primary link between FGF-23 and cardiovascular complications in CKD is left ventricular hypertrophy (LVH).33,36 The detailed mechanism of FGF-23– induced LVH was first revealed by Faul et al.33 A series of studies confirmed that hypertrophic growth of cardiomyocytes Figure 1. The maturation and disintegration of FGF-23. FGF-23 precursor contains 251 can be induced by FGF-23, and that this amino acids (aa) and can be divided into a signal sequence (24 aa), a FGFR binding domain effect does not require FGFR-1/Klotho (155 aa), and an a-Klotho binding domain (72 aa). After removal of the signal sequence, iFGF-23 is secreted into the blood or cleaved between aa 179 and 180 into inactive amino- but does need the presence of FGFR- 26,33,37 fi terminal FGF-23 and active cFGF-23. 4. Speci cally, FGF-23 binds to FGFR-4 in cardiomyocytes and results in stimulation of the phospholipase Cg these, a-Klotho and b-Klotho interact and inhibits the activation of 1,25-dihy- (PLCg)/calcineurin pathway,37 which 23 with FGF-23 and FGF-19/FGF-21, re- droxyvitamin D3. In the parathyroid is an important mediator of cardiac hy- spectively, and assist the binding of gland, FGF-23 restricts the synthesis of pertrophy in response to a majority of FGF and FGFR.15,17,18 However, the bi- parathyroid hormone.24 In the heart, pathologic stimuli (including CKD con- ologic function of the third member, Klotho is expressed in the sinoatrial ditions).38,39 FGFR-4 null mice fed a g-Klotho, remains unclear. In mam- node, but the mRNA level of Klotho is high-phosphate diet37 and 5/6 nephrec- mals, a-Klotho (abbreviated as Klotho fairly low or even undetectable in the tomized rats treated with an FGFR-4 an- below) is considered to exist in two myocardium.25 Studies using either tagonist37 or a calcineurin inhibitor40 all forms in vivo: membranous Klotho PCR or RNA sequencing confirmed low demonstrated no obvious cardiac hyper- (mKlotho) and secreted Klotho (sKlotho). levels or even a lack of Klotho mRNA in trophy. These findings further indicate The secreted A disintegrin and mouse and human heart,22 whereas Klo- an important role of FGFR-4/PLCg/cal- metalloproteinase-10 (ADAM-10), tho protein can be detected by Western cineurin signaling in FGF-23–induced ADAM-17, and b-site amyloid precur- blotting.26 Such results may indicate that cardiac hypertrophy. Our studies also re- sor protein cleaving -1 can incise the actions of FGF-23 and Klotho in car- vealed that microRNA-30, which is an en- the extracellular domain of mKlotho, diac tissue are different from those in the dogenous post-transcriptional inhibitor of which then generates sKlotho.19,20 Al- kidney and parathyroid gland. calcineurin,41 attenuates CKD-induced ternative splicing of Klotho mRNA gen- LVH (unpublished data). In addition, ex- erates sKlotho as well.21 ogenous microRNA-30 supplementation FGF-23 exerts its functions mainly CARDIAC EFFECTS OF FGF-23 IN also inhibits FGF-23–induced cardio- through FGFR-1; however, FGF-23 CKD myocyte hypertrophy (unpublished needs the assistance of mKlotho to ac- data). Recently, Han et al.42 found tivate FGFR-1.12 The expression of CKD is a common result of various kid- that mice with cardiomyocyte FGFR-4 mKlotho is tissue specific, predomi- ney and urologic diseases, with a gradual deletion exhibit resistance to FGF-23– nantly in distal convoluted tubules of decrease in the number of functional induced cardiac hypertrophy, which pro- the kidney and chief cells in the parathy- nephrons and excretory function. As vides further evidence for the vital role of roid gland.22 In the kidney, FGF-23 sup- a result of impaired excretory capacity, cardiac FGFR-4 in FGF-23–induced car- presses the reabsorption of phosphate hyperphosphatemia and hyperparathy- diac hypertrophy.

1424 JASN JASN 31: 1423–1434, 2020 www.jasn.org REVIEW

The myocardium of patients with can be normalized by a high-phosphate contribute to cardiac hypertrophy.22,33 CKD26 and CKD rats43 can secrete diet. Leifheit-Nestler et al.26 revealed the FGF-23 and exert a paracrine effect, sug- Remarkably, there are few studies that presence of Klotho protein in cardiac gesting there may be a local regulatory seem to refute or support the prohyper- muscle, which suggests that the heart system of FGF-23. Leifheit-Nestler trophic effect of FGF-23. Shalhoub can take up sKlotho from the circula- et al.26 confirmed that FGF-23 promotes et al.52 found that administration of tion. A series of studies found that cardiac hypertrophy from multiple per- FGF-23–neutralizing antibody in CKD sKlotho inhibits the prohypertrophic spectives and, in a follow-up study, rats did not attenuate cardiac hypertro- signaling pathway and exerts unique Leifheit-Nestler et al.43 further revealed phy. On the other hand, ferric citrate ad- biologic functions that are indepen- that myocardial FGF-23 significantly ministration from 6 to 10 weeks of age dent of FGF-23.64–67 For example, promotes LVH in patients with CKD in reduces iFGF-23 and significantly im- sKlotho directly binds to the type II a paracrine manner. In addition, FGF-23 proves cardiac function in Col4a3 TGF-b receptor,68 blocking TGF-b1 released by cardiomyocytes also stimu- knockout mice (a model of progressive signaling, and TGF-b169 is highly in- lates fibroblasts to secrete TGF-b, con- CKD).53 These two seemingly contradic- volved in cardiac hypertrophy. nective tissue growth factor, and other tory results may be due to interference of Intriguingly, sKlotho suppresses car- profibrotic factors.44 These studies indi- other factors related to cardiac hypertro- diac hypertrophy that is induced by Ang- cate that the paracrine role of FGF-23 in phy. For example, FGF-23 neutralization II,58 TGF-b,58,67 or isoprenaline58,67,70; cardiac tissue may not be ignored in facilitates aortic calcification and severe thus, sKlotho demonstrates a broad CKD-induced cardiac hypertrophy. hyperphosphatemia,52 which are prohy- spectrum antihypertrophic effects. sKlo- pertrophic stimuli. In addition, it is pos- tho inhibits the production of reactive Controversies sible that ferric citrate improves cardiac oxygen species (ROS), and sKlotho de- Although a number of studies33,36 indi- function by alleviating iron deficiency ficiency may lead to an increase in ROS cate that FGF-23 increases the incidence and anemia rather than by lowering in myocardial tissue and induce cardiac of cardiovascular abnormalities, there FGF-23.53 Therefore, these two studies hypertrophy.71 In addition, several sig- also are studies that do not seem to sup- were unable to prove or exclude the di- naling pathways that may be involved port this conclusion. X-linked hypo- rect cardiac role of FGF-23. in CKD-induced cardiac injury, includ- phosphatemia (XLH), which causes an ing the PI3K,65 WNT,72,73 and NF-kB increased concentration of FGF-23, can pathways,74 can be regulated by sKlotho. be used as a convenient model to inves- CARDIAC EFFECTS OF KLOTHO IN Recently, a specific receptor for sKlotho, tigate the role of FGF-23 in the myocar- CKD monosialoganglioside, was identified; dium. Recently, Pastor-Arroyo et al.45 sKlotho binds to it and inhibits raft- found that the XLH mouse model, The kidney’s distal tubules are a main dependent PI3K signaling.75 This illus- which has normal kidney function, ex- source of Klotho and sKlotho.22 In the trates that sKlotho can inhibit the hibits high circulating FGF-23 and hy- setting of CKD, deficiency in Klotho and translocation of transient receptor po- pophosphatemia and has no activation sKlotho can be caused by a reduction in tential cation channel, subfamily C, of the calcineurin pathway in the myo- functional nephrons and inhibition of member 6 (TRPC6), an ion channel cardium; they also did not observe LVH Klotho expression. CKD-related factors that is involved in the formation of car- or abnormal cardiac function in these such as angiotensin II (Ang-II),54,55 in- diac hypertrophy.65,76,77 mice. Although this result is consistent flammatory cytokines,56,57 phosphate,58 with the results from some groups,46,47 deficiencies in vitamin D59 and erythro- Controversies other studies have observed significant poietin,60,61 or even FGF-2362 can trig- Although animal studies have estab- cardiac hypertrophy in patients with ger the suppression of Klotho. lished a clear relationship between XLH or XLH models.48–50 The reasons Klotho and the cardiovascular system, for these differences may depend on fac- Uremic Cardiac Hypertrophy and some clinical studies have failed to detect tors other than FGF-23, including age, Klotho an independent association between diet, and genetic background (Table 1). Animal experiments established a clear sKlotho level and left ventricular mass In addition, XLH and CKD exert different relationship between Klotho and the index in patients on hemodialysis.78–80 physiologic effects, such as hypophospha- cardiovascular system. Klotho-null mice In patients with CKD from stage 1 to 5 temia in XLH versus hyperphosphatemia develop CKD-like cardiovascular phe- (predialysis), however, sKlotho demon- in CKD, and these factors may signifi- notypes, such as vascular calcification, strates a significant inverse association cantly affect the myocardial effects of cardiac hypertrophy, and fibrosis.14,33,58,63 with left ventricular mass index.81 An- FGF-23 (Figure 2). For example, Liu However, Klotho mRNA is not de- other parameter of cardiac hypertrophy, et al.51 showed that overexpression of tected in the myocardium, aside from the wall thickness of the left ventricle, FGF-23 leads to hypotension and cardiac the sinoatrial node25; thus, loss of Klotho has an inverse association with sKlotho hypertrophy, and that these abnormalities in the myocardium may not directly in patients on dialysis, whereas such an

JASN 31: 1423–1434, 2020 FGF-23 and Cardiac Hypertrophy 1425 REVIEW www.jasn.org

Table 1. The comparison of the different high circulating FGF-23 models Model (genetic background) Circulating FGF-23 (model:control)a Blood-Related Parameters Cardiac Changes Reference 80 mg/kg per d FGF-23 IOCV for 5 d (c 1 i) FGF-23 ↑ (3.2:1) / LVH 33 (C57BL/6J) Klotho heterozygous (/) (c 1 i) FGF-23 ↑ (3.0:1) Phosphate ↑ LVH 33

Active VitD3 ↑ 5/6 nephrectomy (Sprague Dawley) (c 1 i) FGF-23 ↑ (12.5:1) Creatinine ↑ LVH 33

CCreatinine ↓ BUN ↑ Systolic pressure ↑ 5/6 nephrectomy (Sprague Dawley) (c 1 i) FGF-23 ↑ (3.5:1) Creatinine ↑ LVH 37

CCreatinine ↓ BUN ↑ Systolic pressure ↑ Diastolic pressure ↑ Phosphate → 2.0% phosphate diet for 12 wk (C57BL/6) (c 1 i) FGF-23 ↑ (6.0:1) BUN → LVH 37 75 mg/kg per d FGF-23 IP for 5 d (C57BL/6) FGF-23 ↑ (/) Systolic pressure ↑ LVH 42 2.0% phosphate diet for 3 mo (C57BL/6) iFGF-23 ↑ (5.6:1) / LVH 121 121 5/6 nephrectomy (Sprague Dawley) iFGF-23 ↑ (/) CCreatinine ↓ LVH BUN ↑ Systolic pressure ↑ Diastolic pressure ↑ Phosphate ↑ PhexC733R (C3Heb/FeJ) iFGF-23 ↑ (12.8:1) Phosphate ↓ No changes 45 Calcium → sKlotho ↓

Active VitD3 ↓ PTH ↑ BUN → Creatinine → Systolic pressure ↓ Cholesterol → Hyp (C57BL/6J) / Systolic pressure → Smaller heart 47 Hyp (C57BL/6) iFGF-23 ↑ (14.4:1) Aldosterone ↓ LVH 50 Transgenic human FGF-23 in liver / Systolic pressure ↓ LVH 51 (C57BL/6J) Phosphate ↓ Epinephrine ↑ Norepinephrine ↑ Dopamine ↑ Ang-II ↑ Cortisol ↑ Glucose ↓

IOCV, injection of caudal vein; (c 1 i) FGF-23, C-terminal and intact FGF-23; /, not mentioned; VitD3, vitamin D3;CCreatinine, creatinine clearance; →,nosignificant difference between model group and control group; IP, intraperitoneal injection; PTH, parathyroid hormone; Hyp, X-linked semidominant mutation of Phex that causes hypophosphatemia and high circulating FGF-23, whereas does not affect kidney functions. aModel:control is calculated by average value of each group.

associationdoesnotexistinpredialysis changes in the heart or predict cardiovas- the principal reason for these unexpected patients.79,80 As mentioned by Zhang cular risk in patients with heart disease results.84 Indeed, a high concentration of et al.,80 such discrepancies may be who have normal renal function.82 bilirubin (250 mg/L) can significantly re- caused by dialysis, which is associated Surprisingly,astudybySugiura duce the measured value of sKlotho.84 In with such factors as interdialytic weight et al.83 found increased sKlotho levels addition, different kits contain different gain, ultrafiltration, dialysate, and dia- in patients with CKD. Authors of a sub- sKlotho antibodies, which are directed to lyzers that do not apply to nondialysis pa- sequent study that found a significant different epitopes, and such differences tients with CKD. Furthermore, sKlotho increase in sKlotho level in CKD hypoth- may obviously influence the measure- also does not associate with pathologic esized that the reliability of assay kits is ments. Currently, the best way to measure

1426 JASN JASN 31: 1423–1434, 2020 www.jasn.org REVIEW

to uremic cardiac hypertrophy. Accord- ing to this hypothesis, the paracrine effect of FGF-23 may further lead to an increase in the ratio of FGF-23 to sKlotho locally and aggravate cardiac hypertrophy.26,43 To our knowledge, Klotho is not only a coreceptor of FGF-23 but also a con- verter of FGF-23/FGFR interaction. Mechanistically, FGF-23 has a higher af- finity for FGFR-4 than for FGFR-1 in the absence of Klotho, but it shows a nearly 20-fold increase in affinity for FGFR-1 when Klotho is present.90 Recent studies have revealed that sKlotho acts as a cor- eceptor for FGF-2391 and a protective factor for the myocardium through “pathway conversion,” and these were previously considered as essential func- tions of mKlotho (Figure 3).12,42 Specific cardiac ablation of Fgfr-4 blocks FGF- 23–induced cardiac hypertrophy, and sKlotho administration converts FGF- 23–induced PLCg/calcineurin signaling into mitogen-activated protein kinase fi Figure 2. The cardiac effects of elevated FGF-23 is signi cantly different in CKD and (MAPK) signaling.42 In addition, sKlo- XLH. Increased FGF-23 is observed in both CKD and XLH. However, the increase in FGF-23 tho leads to only a small increase in ac- occurs secondary to hyperphosphatemia, whereas it is idiopathic and induces hypo- tivation of MAPK activity in response to phosphatemia in XLH. Elevated FGF-23 induces cardiac hypertrophy in CKD, whereas no 92 cardiac changes are observed in XLH. In CKD, perhaps the relative levels of circulating FGF-23, which means that this path- FGF-23 and phosphate determine the prohypertrophic effects of FGF-23. Moreover, it is way conversion may not induce robust unclear whether any other factors might affect the prohypertrophic effect of FGF-23. activation of MAPK signaling and result in cardiac hypertrophy. Unfortunately, however, these studies were not repro- sKlotho in human samples remains con- low-phosphate diet containing only 0.2% duced in CKD models. Theoretically, troversial. Thus, the association between inorganic phosphate does not block the high FGF-23 and low sKlotho levels in sKlotho and cardiovascular risk needs exacerbation of cardiac hypertrophy in CKD may lead to the interaction of FGF- careful consideration. CKD mice with heterozygous Klotho de- 23 and FGFR-4 and activate the calci- letion.66 Hence, the reduction in sKlotho neurin pathway, which can cause cardiac may be involved in CKD-induced cardiac hypertrophy.37 COMBINED CARDIAC EFFECTS OF hypertrophy regardless of FGF-23 level. FGF-23 AND KLOTHO IN CKD Six et al.89 found that Klotho deficiency is detrimental to the endothelium and that INDIRECT CARDIAC EFFECTS OF As a known scenario of cardiac hypertro- Klotho sufficiency prevents the damag- FGF-23 AND a-KLOTHO IN CKD phy, CKD is accompanied by a low level ing effects of FGF-23 and hyperphos- of sKlotho and a high level of FGF-23 in phate. In addition, Hu et al.58 revealed Effects on Vessels circulation.85,86 Although FGF-23 in- that a positive correlation between FGF- Vascular lesions, another pathologic fea- duces cardiac hypertrophy via FGFR-4 23 levels and cardiac hypertrophy and fi- ture of patients with CKD, are closely in the absence of Klotho,33,37 sKlotho brosis occurs only with relatively low related to cardiac impairment. Animal may exert cardioprotective effects with- sKlotho levels. Han et al.42 found that models or patients with genetic Klotho out the presence of FGF-23.68,77,87 sKlotho injection inhibits FGF-23– deficiency develop extensive arteriosclero- Whether an increased ratio of circulating induced cardiac hypertrophy, providing sis and vascular calcification, which are FGF-23 to sKlotho in CKD results in further evidence for the antihypertrophic similar to CKD-related vascular injury.14,93 more serious cardiac hypertrophy is effect of sKlotho. Collectively, we may in- In CKD mice, Klotho preservation not unknown.88 fer that beyond the levels of FGF-23 only attenuates hyperphosphatemia but It seems that normalization of cir- or sKlotho individually, the ratio of also suppresses the activity of the NaPi-3 culating FGF-23 and phosphate via a these two proteins is a key contributor groupofsodium-coupledtransporters

JASN 31: 1423–1434, 2020 FGF-23 and Cardiac Hypertrophy 1427 REVIEW www.jasn.org

In contrast, other work demonstrated that FGF-23 overexpression results in hypotension, and hypophosphatemia caused by high levels of circulating FGF-23 may mediate such BP de- creases.51 To illustrate this inconsistency, the balance in circulating levels of FGF- 23 and phosphate should be considered.

Effects on Immunocytes In recent years, inflammation has been considered as a consequence of distur- bance of the FGF-23/Klotho axis,105 sug- gesting another way FGF-23 and Klotho can indirectly affect the heart. Elevated FGF-23 acts on FGFR-2 on neutrophils in the absence of Klotho; this deactivates b2-integrin and impairs recruitment of neutrophils to infection sites,106–108 which may partially explain the im- paired host defense resulting from neu- trophil dysfunction in patients with CKD. Studies also have demonstrated Figure 3. FGF-23 activates MAPK signaling in the presence of sKlotho, whereas acti- that FGF-23 inhibits the production of vates calcineurin signaling in the absence of sKlotho. (A) Under physiologic conditions, 1,25-dihydroxyvitamin D3 and results sKLOTHO functions as a coreceptor of FGF-23 and leads to MAPK signaling activation. in monocyte dysfunction (increasing sus- Importantly, sKlotho only leads to a small increase in activation of MAPK activity in re- ceptibility to bacterial infection)109 and sponse to FGF-23, which means it may not induce robust activation of MAPK signaling and macrophage activation,110 which are result in cardiac hypertrophy. (B) In CKD, decreased sKlotho blunts the interaction between closely related to chronic inflammation. FGF-23 and FGFR-1, and FGF-23 preferentially binds to FGFR-4, thus resulting in calci- In addition, FGF-23 promotes the release neurin cascade activation and pathologic cardiac hypertrophy. of proinflammatory factors in hepatocytes under CKD conditions and may partici- (Pit-1 and Pit-2), thus inhibiting calcifi- both FGFR-dependent ROS formation pate in chronic inflammation.111 cation and dedifferentiation of vascular via activation of NADPH oxidase 2, as Given Klotho’sroleasananti- smooth muscle cells.94 In addition, Klo- well as ROS degradation via activation inflammatory effector,112 its deficiency in tho deficiency is highly involved in endo- of SOD2 and catalase.96 However, the ab- CKD may also play a role in the generation thelial dysfunction, which is mediated sence of Klotho blunts FGF-23–induced of inflammation. The cardiovascular by increased apoptosis and reduced pro- eNOS, SOD2, and catalase generation, pathogenic role of chronic inflammation duction of endothelial nitric oxide synthase whereas FGF-23–induced ROS synthesis in CKD has been demonstrated113; hence, (eNOS).95–97 In vivo treatment with is unaffected.96 The balance between the the association between FGF-23 and in- Klotho leads to alleviation of endothelial generation and degradation of ROS may flammation may greatly enhance the sig- dysfunction, high BP, and perivascular be lost in CKD, thus leading to the suc- nificance of FGF-23 in uremic cardiac fibrosis.98 cessive production of ROS and vascular hypertrophy. It is not surprising that FGF-23 defi- injury. Therefore, FGF-23’s vascular ef- ciency results in a phenotype in blood fects, as they affect cardiomyocytes, are vessels similar to that of CKD-related vas- probably also determined by the ratio of SUMMARY AND FUTURE cular injury, given the disturbance in FGF-23 to sKlotho. PERSPECTIVES phosphate metabolism.99,100 Conversely, high circulating FGF-23 also results in vas- Effects on Kidney As endocrine factors, FGF-23 and Klo- cular injury. Studies in patients with CKD Elevated FGF-23 has been described as a tho exert effects on various tissues in the found that high FGF-23 is associated with hypertension-related factor, and studies absence or presence of one another. In vascular dysfunction.101–103 In endothelial suggest that prohypertensive effect may this review, we described the direct and cells, FGF-23/Klotho/FGFR-1 promotes partially be due to the increase in sodium indirect effects of FGF-23 and Klotho on the production of eNOS via the AKT- reabsorption via sodium chloride co- cardiac tissues, including hypertrophy- dependent pathway. FGF-23 stimulates transporters in the distal tubules.48,104 related effects, vessel-related effects,

1428 JASN JASN 31: 1423–1434, 2020 www.jasn.org REVIEW

Figure 4. Direct and indirect effects of FGF-23 and KLOTHO on myocardium. In CKD, high FGF-23 level and Klotho/sKLOTHO deficiency lead to several changes in blood vessels, inflammatory responses, and the kidney, including vascular injury, inflammation, hypertension, and active 1,25-dihydroxyvitamin D3 (1,25-[OH]2-VitD3)deficiency. These changes can induce cardiac hypertrophy directly and indirectly. Moreover, disturbances in FGF-23/sKLOTHO may induce cardiac hypertrophy and fibrosis directly. 25-OH-VitD3, 25-hydroxyvitamin D3; 2 1 Cl ,chlorideion;Na , sodium ion.

JASN 31: 1423–1434, 2020 FGF-23 and Cardiac Hypertrophy 1429 REVIEW www.jasn.org and inflammation-related effects (Fig- response and promotes cardiac hyper- be a consequence of promoting the stress ure 4). However, in light of continuing trophy as a compensatory response re- response, although it reduces the excre- controversies regarding the cardiac ef- quires further study. tion of phosphate. fects of FGF-23 and Klotho, further in- According to current understanding, Collectively, we hypothesize that se- vestigations are needed. the FGF-23/Klotho axis interacts with vere hyperphosphatemia, hypophospha- The discrepancy in cardiac changes in the RAS44,50 and ROS,96 which are all temia, and a high circulating level of XLH and CKD is the most controversial constituents of the stress response. It is FGF-23 trigger the stress response, thus aspect of FGF-23–induced cardiac hy- possible that elevated FGF-23 in CKD downregulating Klotho/sKlotho and ul- pertrophy, and several mechanisms can act as a stress hormone, similar to timately enhancing the stress response. may account for this discrepancy. The another member of the FGF-19 subfam- The balance of FGF-23, sKlotho, and process of FGF-23 cleavage may explain ily, FGF-21,118,119 a speculation that can phosphate actually represents the degree this discordance. Under physiologic be partially verified in cardiomyocytes. of the stress response. Under this hy- conditions, iFGF-23 and its cleavage Incubation of cardiomyocytes with pothesis, FGF-23 is not a detrimental are in dynamic equilibrium, but in FGF-23 results not only in cardiomyo- factor for the heart but rather a hormone CKD, iFGF-23 cleavage may be im- cyte hypertrophy, but also augments the to induce the stress response and protect paired, leading to iFGF-23 accumulation contractility of cardiomyocytes, and the the body from injurious factors. In some and several pathologic changes.114–116 hypertrophic condition seems to be a disease states, such as pathologic cardiac Mechanistically, cFGF-23, which is the compensatory response for increased hypertrophy, the secretion of FGF-23 cleavage product of iFGF-23, can bind to contractility.120 An in vivo study further from damaged organs may represent a FGFR-1/Klotho competitively, thereby in- revealed that FGF-23–induced cardiac/ protective response.123 Obviously, how- hibiting hypophosphatemia induced by a cardiomyocyte hypertrophy is revers- ever, the continuous stress response high circulating level of iFGF-23.117 Arecent ible.121 This suggests that FGF-23– leads to organ damage, including cardiac study found that cFGF-23 attenuates fibro- induced hypertrophy is a reversible re- hypertrophy and vascular injury.124 Under sis and inflammation in diabetic nephrop- sponse to increased contractility, and the this scenario, persistent FGF-23/sKlotho athy, although the underlying mechanisms disappearance of the stressor (FGF-23) disturbance converts protective effects are not clear.7 It is possible that cFGF-23 results in a decreased stress response into detrimental effects. Thus, we suspect interferes with the interaction between (mitigation of cardiac hypertrophy). that the FGF-23/Klotho axis is closely re- FGF-23 and FGFR-4. Notably, the cleavage Moreover, FGF-23 upregulates the intra- lated to the stress response, and that elu- of iFGF-23 seems normal in XLH.45 Hence, cellular expression of Ang-II in cardiomyo- cidating the detailed relationship of the abnormal cleavage of iFGF-23 may contrib- cytes and shares a common mechanism of stress response and FGF-23/Klotho in ute to uremic cardiac hypertrophy. calcium-dependent cardiomyocyte hyper- uremic cardiac hypertrophy may offer a The largest difference between CKD trophy with Ang-II. This further indicates a new research direction. and XLH is that XLH exhibits significant close interaction of FGF-23 and the stress hypophosphatemia instead of hyper- response in cardiomyocytes.122 phosphatemia. Interestingly, overex- XLH is also an abnormal condition DISCLOSURES pression of FGF-23 leads to hypotension and can trigger a stress response as and cardiac hypertrophy, with activation well. Several studies reported a high All authors have nothing to disclose. of the renin-angiotensin system (RAS) rate of LVH in patients with XLH or and the sympathetic nervous system. XLH models.48–50 One possibility is The latter phenomena (cardiac hyper- that severe hypotension and hypophos- trophy and activation of the RAS and phatemia in XLH trigger a stress re- FUNDING sympathetic nervous system) are consid- sponse and strengthen FGF-23–induced ered a stress response to hypotension hypertrophy; in turn, FGF-23 enhances A. Li was supported by National Natural Science and can be mitigated by a high- the stress response, forming a positive Foundation of China grants 81270825 and phosphate diet.51 However, Pastor- feedback loop and ultimately leading to 81770727, Guangdong Province Universities and Colleges Pearl River Scholar Funded Scheme grant 45 fi Arroyo et al. observed hypotension in signi cant cardiac hypertrophy. CKD 2017, Science and Technology Planning Project of XLH models, but there were no cardiac can be regarded as a stress state, Guangdong Province grant 2017A010103041, and changes in these models. This inconsis- wherein the stress response aims to cor- Key Project of Guangzhou Science Technology and tency may be caused by different degrees rect manifestations of the disorder such Innovation Commission grant 201804020054. of stress responses due to genetic back- as hyperphosphatemia and uremic toxin ground (Table 1). As mentioned above, accumulation. Klotho partially blunts REFERENCES FGF-23 results in hypotension, and hy- the stress response by inhibiting the potension induces a stress response and RAS activation, ROS generation, and 1. Parfrey PS, Foley RN: The clinical epidemiol- fl 71,112,118 leads to cardiac hypertrophy. However, even in ammation. Thus, ogy of cardiac disease in chronic renal failure. whether FGF-23 leads directly to a stress downregulation of Klotho in CKD may J Am Soc Nephrol 10: 1606–1615, 1999

1430 JASN JASN 31: 1423–1434, 2020 www.jasn.org REVIEW

2. Shimizu I, Minamino T: Physiological and 17. Ding X, Boney-Montoya J, Owen BM, 30. Wolf M, Molnar MZ, Amaral AP, Czira ME, pathological cardiac hypertrophy. JMol Bookout AL, Coate KC, Mangelsdorf DJ, Rudas A, Ujszaszi A, et al.: Elevated fibro- Cell Cardiol 97: 245–262, 2016 et al.: bKlotho is required for fibroblast blast growth factor 23 is a risk factor for 3. Wang X, Shapiro JI: Evolving concepts in growth factor 21 effects on growth and kidney transplant loss and mortality. JAm the pathogenesis of uraemic cardiomyopa- metabolism. Cell Metab 16: 387 –393, 2012 Soc Nephrol 22: 956–966, 2011 thy. Nat Rev Nephrol 15: 159–175, 2019 18.LinBC,WangM,BlackmoreC,Desnoyers 31. Isakova T, Xie H, Yang W, Xie D, Anderson 4. Quarles LD: Skeletal secretion of FGF-23 LR: Liver-specific activities of FGF19 re- AH, Scialla J, et al.: Chronic Renal In- regulates phosphate and vitamin D metab- quire Klotho beta. JBiolChem282: sufficiency Cohort (CRIC) Study Group: Fi- olism. Nat Rev Endocrinol 8: 276–286, 2012 27277–27284, 2007 broblast growth factor 23 and risks of 5. Kato K, Jeanneau C, Tarp MA, Benet-Pagès 19. Chen CD, Podvin S, Gillespie E, Leeman SE, mortality and end-stage renal disease in A, Lorenz-Depiereux B, Bennett EP, et al.: Abraham CR: Insulin stimulates the cleav- patients with chronic kidney disease. JAMA Polypeptide GalNAc- T3 and famil- age and release of the extracellular domain 305: 2432–2439, 2011 ial tumoral calcinosis. Secretion of fibroblast of Klotho by ADAM10 and ADAM17. Proc 32. Hsu HJ, Wu MS: Fibroblast growth factor growth factor 23 requires O-glycosylation. Natl Acad Sci U S A 104: 19796–19801, 23: A possible cause of left ventricular hy- J Biol Chem 281: 18370–18377, 2006 2007 pertrophy in hemodialysis patients. Am 6. Kocełak P, Olszanecka-Glinianowicz M, 20. Bloch L, Sineshchekova O, Reichenbach D, JMedSci337: 116–122, 2009 Chudek J: Fibroblast growth factor 23-- Reiss K, Saftig P, Kuro-o M, et al.: Klotho is a 33. Faul C, Amaral AP, Oskouei B, Hu MC, Sloan structure, function and role in kidney dis- substrate for alpha-, beta- and gamma- A, Isakova T, et al.: FGF23 induces left eases. Adv Clin Exp Med 21: 391–401, 2012 secretase. FEBS Lett 583: 3221–3224, 2009 ventricular hypertrophy. J Clin Invest 121: 7. Zhang X, Guo K, Xia F, Zhao X, Huang Z, Niu 21. Shiraki-Iida T, Aizawa H, Matsumura Y, 4393–4408, 2011 J: FGF23C-tail improves diabetic nephropa- Sekine S, Iida A, Anazawa H, et al.: Structure 34. Gutiérrez OM, Mannstadt M, Isakova T, thy by attenuating renal fibrosis and in- of the mouse klotho gene and its two tran- Rauh-HainJA,TamezH,ShahA,etal.:Fi- flammation. BMC Biotechnol 18: 33, 2018 scripts encoding membrane and secreted broblast growth factor 23 and mortality 8. Ornitz DM: FGFs, heparan sulfate and protein. FEBS Lett 424: 6–10, 1998 among patients undergoing hemodialysis. FGFRs: Complex interactions essential for 22. Olauson H, Mencke R, Hillebrands JL, N Engl J Med 359: 584–592, 2008 development. BioEssays 22: 108–112, 2000 Larsson TE: Tissue expression and source of 35. Seeherunvong W, Abitbol CL, Chandar J, 9. Shimada T, Mizutani S, Muto T, Yoneya T, circulating aKlotho. Bone 100: 19–35, 2017 Rusconi P, Zilleruelo GE, Freundlich M: Fi- Hino R, Takeda S, et al.: Cloning and char- 23. Shimada T, Hasegawa H, Yamazaki Y, Muto broblast growth factor 23 and left ventricu- acterization of FGF23 as a causative factor T, Hino R, Takeuchi Y, et al.: FGF-23 is a lar hypertrophy in children on dialysis. of tumor-induced osteomalacia. Proc Natl potent regulator of vitamin D metabolism Pediatr Nephrol 27: 2129–2136, 2012 Acad Sci U S A 98: 6500–6505, 2001 and phosphate homeostasis. JBoneMiner 36. Kirkpantur A, Balci M, Gurbuz OA, Afsar B, 10. Fukumoto S: Physiological regulation and Res 19: 429–435, 2004 Canbakan B, Akdemir R, et al.: Serum fi- disorders of phosphate metabolism--pivotal 24. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, broblast growth factor-23 (FGF-23) levels role of fibroblast growth factor 23. Intern Med Goetz R, Kuro-o M, Mohammadi M, et al.: are independently associated with left 47: 337–343, 2008 The parathyroid is a target organ for FGF23 ventricular mass and myocardial perfor- 11. Goetz R, Beenken A, Ibrahimi OA, Kalinina in rats. J Clin Invest 117: 4003–4008, 2007 mance index in maintenance haemodialysis J, Olsen SK, Eliseenkova AV, et al.: Molec- 25. Takeshita K, Fujimori T, Kurotaki Y, Honjo H, patients. Nephrol Dial Transplant 26: ular insights into the klotho-dependent, Tsujikawa H, Yasui K, et al.: Sinoatrial node 1346–1354, 2011 endocrine mode of action of fibroblast dysfunction and early unexpected death of 37. Grabner A, Amaral AP, Schramm K, Singh S, growth factor 19 subfamily members. Mol mice with a defect of klotho gene expres- Sloan A, Yanucil C, et al.: Activation of car- Cell Biol 27: 3417–3428, 2007 sion. Circulation 109: 1776–1782, 2004 diac fibroblast growth factor receptor 4 12. Urakawa I, Yamazaki Y, Shimada T, Iijima K, 26. Leifheit-Nestler M, Große Siemer R, causes left ventricular hypertrophy. Cell Hasegawa H, Okawa K, et al.: Klotho con- Flasbart K, Richter B, Kirchhoff F, Ziegler Metab 22: 1020–1032, 2015 verts canonical FGF receptor into a specific WH, et al.: Induction of cardiac FGF23/ 38. Molkentin JD, Lu JR, Antos CL, Markham B, receptor for FGF23. Nature 444: 770–774, FGFR4 expression is associated with left Richardson J, Robbins J, et al.: A calcineurin- 2006 ventricular hypertrophy in patients with dependent transcriptional pathway for 13. Suzuki M, Uehara Y, Motomura-Matsuzaka chronic kidney disease. Nephrol Dial cardiac hypertrophy. Cell 93: 215–228, K, Oki J, Koyama Y, Kimura M, et al.: beta- Transplant 31: 1088–1099, 2016 1998 Klotho is required for fibroblast growth 27. Gutierrez O, Isakova T, Rhee E, Shah A, 39. Wilkins BJ, Dai YS, Bueno OF, Parsons SA, factor (FGF) 21 signaling through FGF re- Holmes J, Collerone G, et al.: Fibroblast Xu J, Plank DM, et al.: Calcineurin/NFAT ceptor (FGFR) 1c and FGFR3c. Mol Endo- growth factor-23 mitigates hyper- coupling participates in pathological, but crinol 22: 1006–1014, 2008 phosphatemia but accentuates calcitriol not physiological, cardiac hypertrophy. Circ 14. Kuro-o M, Matsumura Y, Aizawa H, deficiency in chronic kidney disease. JAm Res 94: 110–118, 2004 Kawaguchi H, Suga T, Utsugi T, et al.: Mu- Soc Nephrol 16: 2205–2215, 2005 40. Di Marco GS, Reuter S, Kentrup D, Ting L, tation of the mouse klotho gene leads to a 28. Hasegawa H, Nagano N, Urakawa I, Ting L, Grabner A, et al.: Cardioprotective syndrome resembling ageing. Nature 390: Yamazaki Y, Iijima K, Fujita T, et al.: Direct effect of calcineurin inhibition in an animal 45–51, 1997 evidence for a causative role of FGF23 in the model of renal disease. Eur Heart J 32: 15. Kurosu H, Ogawa Y, Miyoshi M, Yamamoto abnormal renal phosphate handling and 1935–1945, 2011 M, Nandi A, Rosenblatt KP, et al.: Regulation vitamin D metabolism in rats with early- 41. Wu J, Zheng C, Wang X, Yun S, Zhao Y, Liu of fibroblast growth factor-23 signaling by stage chronic kidney disease. Kidney Int L, et al.: MicroRNA-30 family members klotho. JBiolChem281: 6120–6123, 2006 78: 975–980, 2010 regulate calcium/calcineurin signaling in 16. Fon Tacer K, Bookout AL, Ding X, Kurosu H, 29. Isakova T, Wahl P, Vargas GS, Gutiérrez OM, podocytes. JClinInvest125: 4091–4106, John GB, Wang L, et al.: Research resource: Scialla J, Xie H, et al.: Fibroblast growth factor 2015 Comprehensive expression atlas of the fi- 23 is elevated before parathyroid hormone 42. Han X, Cai C, Xiao Z, Quarles LD: FGF23 broblast growth factor system in adult mouse. and phosphate in chronic kidney disease. induced left ventricular hypertrophy medi- Mol Endocrinol 24: 2050–2064, 2010 Kidney Int 79: 1370–1378, 2011 ated by FGFR4 signaling in the myocardium

JASN 31: 1423–1434, 2020 FGF-23 and Cardiac Hypertrophy 1431 REVIEW www.jasn.org

is attenuated by soluble Klotho in mice. in rat renal tubular epithelial cells. Kidney 67. Yang K, Wang C, Nie L, Zhao X, Gu J, Guan J Mol Cell Cardiol 138: 66–74, 2020 Blood Press Res 33: 186–192, 2010 X, et al.: Klotho protects against indoxyl 43. Leifheit-Nestler M, Grabner A, Hermann L, 55.YoonHE,GheeJY,PiaoS,SongJ-H,Han sulphate-induced myocardial hypertrophy. Richter B, Schmitz K, Fischer DC, et al.: Vi- DH, Kim S, et al.: Angiotensin II blockade J Am Soc Nephrol 26: 2434–2446, 2015 tamin D treatment attenuates cardiac upregulates the expression of Klotho, the 68. Doi S, Zou Y, Togao O, Pastor JV, John GB, FGF23/FGFR4 signaling and hypertrophy in anti-ageing gene, in an experimental Wang L, et al.: Klotho inhibits transforming uremic rats. Nephrol Dial Transplant 32: model of chronic cyclosporine nephropa- growth factor-beta1 (TGF-beta1) signaling 1493–1503, 2017 thy. NephrolDialTransplant26: 800–813, and suppresses renal fibrosis and cancer 44. Leifheit-Nestler M, Kirchhoff F, Nespor J, 2011 metastasis in mice. JBiolChem286: Richter B, Soetje B, Klintschar M, et al.: 56. Thurston RD, Larmonier CB, Majewski PM, 8655–8665, 2011 Fibroblast growth factor 23 is induced by Ramalingam R, Midura-Kiela M, Laubitz D, 69. Dobaczewski M, Chen W, Frangogiannis an activated renin-angiotensin-aldosterone et al.: Tumor necrosis factor and interferon- NG: Transforming growth factor (TGF)-b system in cardiac myocytes and promotes Gamma down-regulate Klotho in mice with signaling in cardiac remodeling. J Mol Cell the pro-fibrotic crosstalk between cardiac colitis. Gastroenterology 138: 1384–1394, Cardiol 51: 600–606, 2011 myocytes and fibroblasts. Nephrol Dial 1394.e1–1394.e2, 2010 70. Song S, Si LY: Klotho ameliorated Transplant 33: 1722–1734, 2018 57. Moreno JA, Izquierdo MC, Sanchez-Niño isoproterenol-induced pathological changes 45. Pastor-Arroyo EM, Gehring N, Krudewig C, MD, Suárez-Alvarez B, Lopez-Larrea C, in cardiomyocytes via the regulation of oxida- Costantino S, Bettoni C, Knöpfel T, et al.: Jakubowski A, et al.: The inflammatory cy- tive stress. Life Sci 135: 118–123, 2015 The elevation of circulating fibroblast tokines TWEAK and TNFa reduce renal 71. Yamamoto M, Clark JD, Pastor JV, Gurnani growth factor 23 without kidney disease klotho expression through NFkB. JAmSoc P,NandiA,KurosuH,etal.:Regulationof does not increase cardiovascular disease Nephrol 22: 1315–1325, 2011 oxidative stress by the anti-aging hormone risk. Kidney Int 94: 49–59, 2018 58. Hu MC, Shi M, Cho HJ, Adams-Huet B, Paek klotho. JBiolChem280: 38029–38034, 46. Takashi Y, Kinoshita Y, Hori M, Ito N, J, Hill K, et al.: Klotho and phosphate are 2005 Taguchi M, Fukumoto S: Patients with modulators of pathologic uremic cardiac 72. van de Schans VA, van den Borne SW, FGF23-related hypophosphatemic rickets/ remodeling. JAmSocNephrol26: Strzelecka AE, Janssen BJ, van der Velden osteomalacia do not present with left ven- 1290–1302, 2015 JL, Langen RC, et al.: Interruption of Wnt tricular hypertrophy. Endocr Res 42: 59. Lau WL, Leaf EM, Hu MC, Takeno MM, signaling attenuates the onset of pressure 132–137, 2017 Kuro-o M, Moe OW, et al.: Vitamin D re- overload-induced cardiac hypertrophy. 47. Liu ES, Thoonen R, Petit E, Yu B, Buys ES, ceptor agonists increase klotho and osteo- Hypertension 49: 473–480, 2007 Scherrer-Crosbie M, et al.: Increased circu- pontin while decreasing aortic calcification 73. Zhou L, Li Y, Zhou D, Tan RJ, Liu Y: Loss of lating fgf23 does not lead to cardiac hy- in mice with chronic kidney disease fed a Klotho contributes to kidney injury by de- pertrophy in the male hyp mouse model of high phosphate diet. Kidney Int 82: repression of Wnt/b-catenin signaling. JAm XLH. Endocrinology 159: 2165–2172, 2018 1261–1270, 2012 Soc Nephrol 24: 771–785, 2013 48. Andrukhova O, Slavic S, Smorodchenko A, 60. Leone F, Lofaro D, Gigliotti P, Perri A, Vizza 74. Buendía P, Carracedo J, Soriano S, Zeitz U, Shalhoub V, Lanske B, et al.: FGF23 D, Toteda G, et al.: Soluble Klotho levels in Madueño JA, Ortiz A, Martín-Malo A, et al.: regulates renal sodium handling and blood adult renal transplant recipients are modu- Klotho prevents NFkB translocation and pressure. EMBO Mol Med 6: 744–759, 2014 lated by recombinant human erythropoie- protects endothelial cell from senescence 49. Nehgme R, Fahey JT, Smith C, Carpenter tin. JNephrol27: 577–585, 2014 induced by uremia. J Gerontol A Biol Sci TO: Cardiovascular abnormalities in pa- 61. Ritter CS, Zhang S, Delmez J, Finch JL, Med Sci 70: 1198–1209, 2015 tients with X-linked hypophosphatemia. Slatopolsky E: Differential expression and 75. Dalton G, An SW, Al-Juboori SI, Nischan N, J Clin Endocrinol Metab 82: 2450–2454, regulation of Klotho by paricalcitol in the Yoon J, Dobrinskikh E, et al.: Soluble klotho 1997 kidney, parathyroid, and aorta of uremic binds monosialoganglioside to regulate 50. Pi M, Ye R, Han X, Armstrong B, Liu X, Chen rats. Kidney Int 87: 1141–1152, 2015 membrane microdomains and growth fac- Y, et al.: Cardiovascular interactions be- 62. Dai B, David V, Martin A, Huang J, Li H, Jiao signaling. Proc Natl Acad Sci U S A 114: tween fibroblast growth factor-23 and an- Y, et al.: A comparative transcriptome 752–757, 2017 giotensin II. Sci Rep 8: 12398, 2018 analysis identifying FGF23 regulated 76. Kuwahara K, Wang Y, McAnally J, 51. Liu P, Bai X, Wang H, Karaplis A, Goltzman in the kidney of a mouse CKD model. PLoS Richardson JA, Bassel-Duby R, Hill JA, et al.: D, Miao D: Hypophosphatemia-mediated One 7: e44161, 2012 TRPC6 fulfi lls a calcineurin signaling circuit hypotension in transgenic mice over- 63. Hu MC, Kuro-o M, Moe OW: aKlotho and during pathologic cardiac remodeling. expressing human FGF-23. Am J Physiol vascular calcification: an evolving para- JClinInvest116: 3114–3126, 2006 Heart Circ Physiol 297: H1514–H1520, digm. Curr Opin Nephrol Hypertens 23: 77. Wright JD, An SW, Xie J, Lim C, Huang CL: 2009 331–339, 2014 Soluble klotho regulates TRPC6 calcium 52. Shalhoub V, Shatzen EM, Ward SC, Davis J, 64. Dalton GD, Xie J, An SW, Huang CL: New signaling via lipid rafts, independent of the Stevens J, Bi V, et al.: FGF23 neutralization insights into the mechanism of action of FGFR-FGF23 pathway. FASEB J 33: improves chronic kidney disease-associated soluble Klotho. Front Endocrinol (Lau- 9182–9193, 2019 hyperparathyroidism yet increases mortality. sanne) 8: 323–332, 2017 78. Buiten MS, de Bie MK, Bouma-de Krijger A, J Clin Invest 122: 2543–2553, 2012 65. Xie J, Cha SK, An SW, Kuro-O M, van Dam B, Dekker FW, Jukema JW, et al.: 53. Francis C, Courbon G, Gerber C, Neuburg Birnbaumer L, Huang C-L: Cardioprotection Soluble Klotho is not independently asso- S, Wang X, Dussold C, et al.: Ferric citrate by Klotho through downregulation of ciated with cardiovascular disease in a reduces fibroblast growth factor 23 levels TRPC6 channels in the mouse heart. Nat population of dialysis patients. BMC and improves renal and cardiac function in a Commun 3: 1238, 2012 Nephrol 15: 197, 2014 mouse model of chronic kidney disease. 66. Xie J, Yoon J, An SW, Kuro-o M, Huang CL: 79. Abdallah E, Mosbah O, Khalifa G, Metwaly Kidney Int 96: 1346–1358, 2019 Soluble klotho protects against uremic car- A, El-Bendary O: Assessment of the re- 54. Zhou Q, Lin S, Tang R, Veeraragoo P, Peng W, diomyopathy independently of fibroblast lationship between serum soluble Klotho Wu R: Role of fosinopril and valsartan on klo- growth factor 23 and phosphate. JAmSoc and carotid intima-media thickness and tho gene expression induced by angiotensin ii Nephrol 26: 1150–1160, 2015 left ventricular dysfunction in hemodialysis

1432 JASN JASN 31: 1423–1434, 2020 www.jasn.org REVIEW

patients. Kidney Res Clin Pract 35: 42–49, 93. McCullough PA, Agrawal V, Danielewicz E, 105. Rossaint J, Unruh M, Zarbock A: Fibroblast 2016 Abela GS: Accelerated atherosclerotic cal- growth factor 23 actions in inflammation: A 80. Zhang AH, Guo WK, Yu L, Liu WH: Re- cification and Monckeberg’s sclerosis: A key factor in CKD outcomes. Nephrol Dial lationship of serum soluble klotho levels continuum of advanced vascular pathology Transplant 32: 1448–1453, 2017 and echocardiographic parameters in in chronic kidney disease. Clin J Am Soc 106. Anding K, Gross P, Rost JM, Allgaier D, patients on maintenance hemodialysis. Nephrol 3: 1585–1598, 2008 Jacobs E: The influence of uraemia and Kidney Blood Press Res 44: 396–404, 94. Hu MC, Shi M, Zhang J, Quiñones H, Griffith haemodialysis on neutrophil phagocytosis 2019 C, Kuro-o M, et al.: Klotho deficiency causes and antimicrobial killing. Nephrol Dial 81. Kim HJ, Kang E, Oh YK, Kim YH, Han SH, vascular calcification in chronic kidney dis- Transplant 18: 2067–2073, 2003 Yoo TH, et al.: The association between ease. JAmSocNephrol22: 124–136, 2011 107. Anders HJ, Ninichuk V, Schlöndorff D: Pro- soluble klotho and cardiovascular parame- 95. Alesutan I, Feger M, Tuffaha R, Castor T, gression of kidney disease: Blocking leu- ters in chronic kidney disease: Results from Musculus K, Buehling SS, et al.: Augmen- kocyte recruitment with chemokine the KNOW-CKD study. BMC Nephrol 19: tation of phosphate-induced osteo-/chon- receptor CCR1 antagonists. Kidney Int 69: 51, 2018 drogenic transformation of vascular smooth 29–32, 2006 82. Brandenburg VM, Kleber ME, Vervloet MG, muscle cells by homoarginine. Cardiovasc 108. Rossaint J, Oehmichen J, Van Aken H, Larsson TE, Tomaschitz A, Pilz S, et al.: Res 110: 408–418, 2016 Reuter S, Pavenstädt HJ, Meersch M, et al.: Soluble klotho and mortality: The ludwig- 96. Richter B, Haller J, Haffner D, Leifheit- FGF23 signaling impairs neutrophil re- shafen risk and cardiovascular health study. Nestler M: Klotho modulates FGF23- cruitment and host defense during CKD. Atherosclerosis 242: 483–489, 2015 mediated NO synthesis and oxidative J Clin Invest 126: 962–974, 2016 83. Sugiura H, Tsuchiya K, Nitta K: Circulating stress in human coronary artery endothelial 109. Bacchetta J, Sea JL, Chun RF, Lisse TS, levels of soluble a-Klotho in patients with cells. Pflugers Arch 468: 1621–1635, 2016 Wesseling-Perry K, Gales B, et al.: Fibro- chronic kidney disease. Clin Exp Nephrol 97. Lim K, Lu TS, Molostvov G, Lee C, Lam FT, blast growth factor 23 inhibits extrarenal 15: 795–796, 2011 Zehnder D, et al.: Vascular Klotho de- synthesis of 1,25-dihydroxyvitamin D in 84. Devaraj S, Syed B, Chien A, Jialal I: Valida- ficiency potentiates the development of human monocytes. J Bone Miner Res 28: tion of an immunoassay for soluble Klotho human artery calcification and mediates 46–55, 2013 protein: Decreased levels in diabetes and resistance to fibroblast growth factor 23. 110.HanX,LiL,YangJ,KingG,XiaoZ,Quarles increased levels in chronic kidney disease. Circulation 125: 2243–2255, 2012 LD: Counter-regulatory paracrine actions of Am J Clin Pathol 137: 479–485, 2012 98. Lim K, Groen A, Molostvov G, Lu T, Lilley FGF-23 and 1,25(OH)2 D in macrophages. 85. Pavik I, Jaeger P, Ebner L, Wagner CA, KS, Snead D, et al.: a-Klotho expression in FEBS Lett 590: 53–67, 2016 PetzoldK,SpichtigD,etal.:SecretedKlo- human tissues. J Clin Endocrinol Metab 111. Singh S, Grabner A, Yanucil C, Schramm K, tho and FGF23 in chronic kidney disease 100: E1308–E1318, 2015 Czaya B, Krick S, et al.: Fibroblast growth Stage 1 to 5: A sequence suggested from a 99. Shimada T, Kakitani M, Yamazaki Y, factor 23 directly targets hepatocytes to cross-sectional study. Nephrol Dial Trans- Hasegawa H, Takeuchi Y, Fujita T, et al.: promote inflammation in chronic kidney plant 28: 352–359, 2013 Targeted ablation of Fgf23 demonstrates disease. Kidney Int 90: 985–996, 2016 86. Rotondi S, Pasquali M, Tartaglione L, Muci an essential physiological role of FGF23 in 112. Liu F, Wu S, Ren H, Gu J: Klotho suppresses ML, Mandanici G, Leonangeli C, et al.: Sol- phosphate and vitamin D metabolism. RIG-I-mediated senescence-associated in- uble a-Klotho serum levels in chronic kidney JClinInvest113: 561–568, 2004 flammation. Nat Cell Biol 13: 254–262, 2011 disease. Int J Endocrinol 2015: 872193, 100. DeLuca S, Sitara D, Kang K, Marsell R, 113. Kaysen GA: Association between inflamma- 2015 Jonsson K, Taguchi T, et al.: Amelioration of tion and malnutrition as risk factors of cardio- 87. Wolf I, Levanon-Cohen S, Bose S, Ligumsky the premature ageing-like features of Fgf- vascular disease. Blood Purif 24: 51–55, 2006 H, Sredni B, Kanety H, et al.: Klotho: A tumor 23 knockout mice by genetically restoring 114. Kinoshita Y, Fukumoto S: X-linked hypo- suppressor and a modulator of the IGF-1 the systemic actions of FGF-23. J Pathol phosphatemia and FGF23-related hypo- and FGF pathways in human breast cancer. 216: 345–355, 2008 phosphatemic diseases: Prospect for new Oncogene 27: 7094–7105, 2008 101. Yilmaz MI, Sonmez A, Saglam M, Yaman H, treatment. Endocr Rev 39: 274–291, 2018 88. Grabner A, Faul C: The role of fibroblast Kilic S, Demirkaya E, et al.: FGF-23 and 115. Shimada T, Urakawa I, Isakova T, Yamazaki growth factor 23 and Klotho in uremic car- vascular dysfunction in patients with stage 3 Y, Epstein M, Wesseling-Perry K, et al.: diomyopathy. Curr Opin Nephrol Hyper- and 4 chronic kidney disease. Kidney Int 78: Circulating fibroblast growth factor 23 in tens 25: 314–324, 2016 679–685, 2010 patients with end-stage renal disease 89. Six I, Okazaki H, Gross P, Cagnard J, Boudot 102. Tamei N, Ogawa T, Ishida H, Ando Y, Nitta treated by peritoneal dialysis is intact and C, Maizel J, et al.: Direct, acute effects of K: Serum fibroblast growth factor-23 levels biologically active. J Clin Endocrinol Metab Klotho and FGF23 on vascular smooth and progression of aortic arch calci fication 95: 578–585, 2010 muscle and endothelium. PLoS One 9: in non-diabetic patients on chronic hemo- 116. Smith ER, Cai MM, McMahon LP, Holt SG: e93423, 2014 dialysis. JAtherosclerThromb18: 217–223, Biological variability of plasma intact and 90. Yu X, Ibrahimi OA, Goetz R, Zhang F, Davis 2011 C-terminal FGF23 measurements. JClin SI, Garringer HJ, et al.: Analysis of the bio- 103. Turan MN, Kircelli F, Yaprak M, Sisman AR, Endocrinol Metab 97: 3357–3365, 2012 chemical mechanisms for the endocrine Gungor O, Bayraktaroglu S, et al.: FGF-23 117. Goetz R, Nakada Y, Hu MC, Kurosu H, Wang actions of fibroblast growth factor-23. En- levels are associated with vascular calcifi- L, Nakatani T, et al.: Isolated C-terminal tail docrinology 146: 4647–4656, 2005 cation, but not with atherosclerosis, in he- of FGF23 alleviates hypophosphatemia by 91. Chen G, Liu Y, Goetz R, Fu L, Jayaraman S, modialysis patients. Int Urol Nephrol 48: inhibiting FGF23-FGFR-Klotho complex Hu MC, et al.: a-Klotho is a non-enzymatic 609–617, 2016 formation. Proc Natl Acad Sci U S A 107: molecular scaffold for FGF23 hormone sig- 104. Fyfe-Johnson AL, Alonso A, Selvin E, Bower 407–412, 2010 nalling. Nature 553: 461–466, 2018 JK, Pankow JS, Agarwal SK, et al.: Serum 118. de Borst MH, Vervloet MG, ter Wee PM, 92. XiaoZ,KingG,MancarellaS,Munkhsaikhan fibroblast growth factor-23 and incident Navis G: Cross talk between the renin-an- U, Cao L, Cai C, et al.: FGF23 expression is hypertension: The Atherosclerosis Risk in giotensin-aldosterone system and vitamin stimulated in transgenic a-Klotho longevity Communities (ARIC) Study. J Hypertens 34: D-FGF-23-klotho in chronic kidney disease. mouse model. JCI Insight 4: e132820, 2019 1266–1272, 2016 J Am Soc Nephrol 22: 1603–1609, 2011

JASN 31: 1423–1434, 2020 FGF-23 and Cardiac Hypertrophy 1433 REVIEW www.jasn.org

119. Kim KH, Lee MS: FGF21 as a stress hormone: 121. Grabner A, Schramm K, Silswal N, Hendrix hypertrophy elevates serum levels of fi- The roles of FGF21 in stress adaptation and M, Yanucil C, Czaya B, et al.: FGF23/FGFR4- broblast growth factor 23. Kidney Int 94: the treatment of metabolic diseases. Di- mediated left ventricular hypertrophy is re- 60–71, 2018 abetes Metab J 38: 245–251, 2014 versible. Sci Rep 7: 1993, 2017 124. Zoccali C, Vanholder R, Massy ZA, Ortiz A, 120. Touchberry CD, Green TM, Tchikrizov V, 122. Mhatre KN, Wakula P, Klein O, Bisping E, Sarafidis P, Dekker FW, et al.: European Renal MannixJE,MaoTF,CarneyBW,etal.: Völkl J, Pieske B, et al.: Crosstalk between and Cardiovascular Medicine (EURECA-m) 1 FGF23 is a novel regulator of in- FGF23- and angiotensin II-mediated Ca2 Working Group of the European Renal tracellular calcium and cardiac contrac- signaling in pathological cardiac hypertro- Association – European Dialysis Trans- tility in addition to cardiac hypertrophy. phy. Cell Mol Life Sci 75: 4403–4416, 2018 plantation Association (ERA-EDTA): The Am J Physiol Endocrinol Metab 304: 123. Matsui I, Oka T, Kusunoki Y, Mori D, systemic nature of CKD. Nat Rev Nephrol E863–E873, 2013 Hashimoto N, Matsumoto A, et al.: Cardiac 13: 344–358, 2017

1434 JASN JASN 31: 1423–1434, 2020