university of groningen fibroblast growth factor 23: a ... · the role of extracellular ... in...

19
University of Groningen Fibroblast Growth Factor 23: a Bridge Between Bone Minerals and Renal Volume Handling Humalda, Jelmer Kor IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below. Document Version Publisher's PDF, also known as Version of record Publication date: 2016 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): Humalda, J. K. (2016). Fibroblast Growth Factor 23: a Bridge Between Bone Minerals and Renal Volume Handling. [Groningen]: Rijksuniversiteit Groningen. Copyright Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons). Take-down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum. Download date: 26-09-2020

Upload: others

Post on 25-Jul-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

University of Groningen

Fibroblast Growth Factor 23: a Bridge Between Bone Minerals and Renal Volume HandlingHumalda, Jelmer Kor

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite fromit. Please check the document version below.

Document VersionPublisher's PDF, also known as Version of record

Publication date:2016

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):Humalda, J. K. (2016). Fibroblast Growth Factor 23: a Bridge Between Bone Minerals and Renal VolumeHandling. [Groningen]: Rijksuniversiteit Groningen.

CopyrightOther than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of theauthor(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policyIf you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediatelyand investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons thenumber of authors shown on this cover page is limited to 10 maximum.

Download date: 26-09-2020

Page 2: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

Chapter 1Introduction and Aims of Thesis

Page 3: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several
Page 4: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

11

Introduction and Aims of Thesis

1Introduction

Chronic Kidney Disease – When Renal Function Sinks

The prevalence of chronic kidney disease (CKD) varies among European countries between 3 and 17 per 100 persons (1). In the Netherlands, CKD prevalence has been estimated between 7.6 and 10.4 patients per 100 persons, according to the large observational cohort studies LifeLines (1) and PREVEND (2). A reduced renal function, commonly expressed as the estimated glomerular filtration rate (eGFR), is associated with a 3‒19-fold increased mortality risk and an increased risk of progression to end stage renal disease (ESRD), depending on age and disease severity (3). Blockade of the renin-angiotensin-aldosterone system (RAAS) is the mainstay of CKD treatment. It aims at reduction of rate of renal function loss and of cardiovascular complica-tions by reduction of blood pressure and proteinuria (4, 5). RAAS blockade interferes with the hemodynamic effects of increased RAAS-activity, i.e. systemic and renal vasoconstriction, as well as with its effects on volume status, and its pro-fibrotic effects. Despite proven efficacy however, risk reduction is at best modest. Treatment with angiotensin receptor blockers (ARB) delays progression to ESRD with a mere four to eight months (6, 7). RAAS blockade was introduced in the 1980’s as antihypertensive treatment. Its renoprotective properties were investigated in the 90’s, and large RCTs corroborated use of RAAS blockade for reno- and cardioprotection in CKD around 2000. Unfortunately, from that time until now in 2016 little progress has been made to further retard renal function loss or protect from cardiovascular complications. Our understanding of renal disease, and the factors driving progressive renal function loss, is far from complete, and other mechanisms than the RAAS are likely also involved. In this thesis, we address the interaction of RAAS-activity, sodium- and volume homeostasis, with another major hormonal system, that governs mineral-bone homeostasis.

In CKD the RAAS is aberrantly activated. In line, the prevalence of hypertension increases as renal function declines: for instance from 18.1 per cent when eGFR is 90–119 mL/min/1.73m2 to 82.1 per cent when eGFR has dropped below 30 mL/min/1.73m2 (8). RAAS blockade (e.g. angiotensin-converting enzyme [ACE] inhibitors or ARBs) reduces blood pressure and protein-uria, and these effects are assumed to be main factors for cardiovascular and renal protection. However, ACE inhibitors or ARBs reduce proteinuria only by some 40% on the average (9). The more ‘residual’ proteinuria remains, the higher the residual risk for subsequent cardiovascular events (10) and for progressive renal function loss (11). Combination of ACE inhibition with ARBs was initially thought to enhance RAAS blockade efficacy and improve outcomes. However, large RCTs pointed out that dual-blockade does not provide additional benefits, and even may cause more harm (12-14), as discussed more elaborately in (→ Chapter 2). Therefore, additional strategies to improve efficacy of RAAS blockade are urgently needed. The role of extracellular volume overload has long since been identified as an important mediator of RAAS blockade efficacy, based on the consistent observation that high salt intake blunts the effect of RAAS

Page 5: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

Chapter 1

12

blockade in patients with hypertension (15) and with CKD (16). Moreover, volume overload correlates with proteinuria, and identifies patients that have greater therapeutic benefit from sodium restriction and/or diuretics on blood pressure and proteinuria (17, 18). The other way round, proteinuria in itself may induce sodium retention and thus promote volume overload (19). All in all, there is compelling evidence for an interaction of volume overload with the effects of RAAS blockade, with blunting of efficacy during volume-overload. More recent data suggest that phosphate status may interact with efficacy of RAAS blockade as well, and more-over, that phosphate status may interact with volume status. Intriguingly, nephrotic ‒and hence probably volume overloaded‒ pediatric patients have higher serum phosphate concentrations despite preserved eGFR (20). Moreover, higher serum phosphate levels are associated with impaired cardiorenal protective effect of ACE-inhibitors (21). This suggests interaction between bone-mineral and volume-hemodynamic pathways in pathophysiology, and in the therapeutic response to RAAS blockade in CKD. This placed phosphate and mineral bone homeostasis firmly in our sights.

Phosphate and Fibroblast Growth Factor 23 – Between Balancing and Capsizing

The kidney controls serum phosphate concentrations by regulating its excretion. Only in the setting of ESRD, hyperphosphatemia occurs and is strongly associated with increased mortality (22). However, in earlier stages of CKD, serum phosphate is regulated to remain within the normal range (23). Even within this normal range, higher serum phosphate concentrations are associated with a higher risk of mortality in patients with patients with CKD (24, 25) and the general population (26), suggesting that mild disturbances in the regulatory system already have clinical impact. Of note, in CKD patients with higher serum phosphate concentrations, cardiorenal protection by RAAS blockade is less effective (21). Notwithstanding the unequivocal beneficial effect of reducing serum phosphate concentrations in haemodialysis patients (27), strategies to reduce serum phosphate in earlier stages of CKD lack currently evidence for long-term cardiovascular protection.

Phosphate regulation is under control of three different hormones (28). Active, 1,25-dihydroxy-vitamin D3 (1,25D) increases serum calcium and phosphate levels, predominantly by increasing gastrointestinal and renal tubular reabsorption. Parathyroid hormone (PTH) decreases renal phosphate reabsorption –and stimulates the activation of vitamin D. In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several bone disorders that are characterized by excessive renal phosphate loss (29, 30). FGF-23 inhibits tubular phosphate reabsorption; in-hibits 1-alpha-hydroxylase, the enzyme that converts inactive 25-hydroxy vitamin D3 into 1,25D (31); and inhibits PTH secretion (32, 33). FGF-23 is able to bind to the FGF-receptor 1c only when the coreceptor α-klotho is present (34). Indeed, mice lacking αklotho or FGF-23 share the same phenotype of premature ageing and ectopic calcification, partly due to high calcium and phosphate concentrations (28, 35, 36). FGF-23 thus counterbalances the effects of vitamin

Page 6: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

13

Introduction and Aims of Thesis

1D on phosphate homeostasis. An additional layer of complexity is added by the interaction of FGF-23 and vitamin D with the RAAS (37). FGF-23 lowers 1,25D, and in this fashion attenuates the inhibiting effect of vitamin D on the RAAS (38). Moreover, FGF-23 may induce left ventricular hypertrophy (39), possibly by aberrant stimulation of the FGF-receptor 4 in the cardiac myocytes (40). The actions of FGF-23 are summarized in Figure 1.

1,25D

PTH

Phosphate1,25D

NaPi PO4 excretionFGF-23

Figure 1. Schematic representation of FGF-23 physiology. FGF-23 signals via the FGF receceptor 1c (FGFR1c) and its obligate coreceptor, α-klotho (Kl) the parathyroid gland to inhibit production of parathyroid hormone (PTH); inhibits expression of sodium-phosphate (NaPi) cotransporters in the kidney, thus stimulate phosphate excretion in the urine; inhibits conversion of inactive vitamin D to active, 1,25-dihydroxy-vitamin D3 (1,25D). Dashed red ar-row signifies pathophysiologic association of FGF-23 and left ventricular hypertrophy, possibly mediated by FGFR-4 signaling or FGF-23 induced volume overload.

The principal site of FGF-23 production is the osteocyte. FGF-23 production increases in re-sponse to 1,25D (31), PTH (41) and phosphate (42), effectively closing negative feedback loops. Also higher calcium concentrations may stimulate FGF-23 production (43), as may inflammatory cytokines or iron deficiency (44). The strongest determinant of FGF-23 concentrations, however, is renal function.

In persons with preserved renal function, the normal value of FGF-23 is typically <100 RU/mL (45). FGF-23 concentrations increase early in the course of CKD (46), long before disturbances in PTH and phosphate values are apparent (23). Allegedly, this increase serves to stimulate phosphate excretion in the remaining functioning nephrons. With the progression of CKD, FGF-23 rises exponentially from values in the hundreds in CKD stage 3–4 to reach extreme values in haemodialysis patients: 10 000 to even 100 000 RU/mL, as illustrated in Figure 2.

Page 7: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

Chapter 1

14

Figure 2. Summary of FGF-23 concentrations reported in this thesis. FGF-23 rises exponentially as renal function declines. This occurs earlier and to a more extreme extent than parathyroid hormone (PTH, left Y- axis) and serum phosphate concentrations (right Y-axis). Data points reflect patients with CKD (chapter 4, 7), haemodialysis patients (chapter 6) and renal transplant recipients (chapter 5).

Even the presence of minimal residual renal function in haemodialysis patients is associated with lower FGF-23 levels (47, 48). After renal transplantation and thus restoration of renal function, FGF-23 levels generally return to normal values within 3 months (49). Higher FGF-23 concentrations are associated with an increased mortality risk in haemodialysis patients (50, 51), predominantly cardiovascular mortality in predialysis CKD (52, 53), and even with cardiovascular and all-cause mortality in the general population (54, 55). Because the adverse consequences of high FGF-23 concentrations have repeatedly been demonstrated, FGF-23 has been hypothesized to be a target for intervention. Accordingly, known determinants of FGF-23 have been targeted relentlessly in efforts to lower FGF-23 levels.

A Barrage of Interventions Targeting Fibroblast Growth Factor-23

Efforts to reduce FGF-23 concentrations consist of strategies aimed at various elements, includ-ing dietary, immunological and hemodynamic interventions summarized in table 1.

Table 1. Strategies to reduce FGF-23 concentrations or limit its consequences. Strategies in italic are based on unpublished data.

Strategy Rationale

Dietary strategies

Phosphate restriction Reduce intake of a phosphate.

Phosphate binders Reduce intestinal absorption of phosphate

Sodium restriction May attenuate effects of FGF-23 on volume status

Potassium supplementation May lower FGF-23 and increase serum phosphate

Antibodies to FGF-23 Inhibit FGF-23 action in body

Intensified haemodialysis schedule Reduce phosphate and fluid accumulation, both determinants of FGF-23

Page 8: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

15

Introduction and Aims of Thesis

1Because FGF-23 regulates phosphate homeostasis, dietary phosphate was considered first. Net phosphate reabsorption can be reduced by limiting intake by a phosphate-restricted diet, and deployment of phosphate binders, which are commonly used in ESRD. High dietary phosphate intake results in higher FGF-23 concentrations in healthy volunteers (42, 56, 57). However, in patients with CKD, dietary phosphate restriction did not achieve a reduction of FGF-23 (58). The efficacy of phosphate binders on FGF-23 varies, with absent effects on FGF-23 by calcium-based phosphate binders (59); the effect of lanthanum carbonate that varied from none (60) to strong if combined with dietary phosphate restriction (58); and marked reductions by sevelamer treatment (59). Third, antibodies to FGF-23 have been tested, however full-blockade of FGF-23 function resulted in extreme calcifications and premature death of animals (61), underscoring the physiological necessity of FGF-23 for keeping calcium-phosphate balance in check. Fourth, in haemodialysis, better phosphate control can be achieved by more frequent, or longer dialy-sis sessions (62); or longer hemodiafiltration (63): both strategies also achieve lower FGF-23 concentrations in haemodialysis (63, 64), however both are obviously not applicable to the predialysis CKD population. A fifth strategy follows from the finding that FGF-23 is a particular strong predictor of heart failure rather than ischemic cardiovascular events (53, 65). These observational data have led to the hypothesis that FGF-23 may induce volume overload. Ex-perimental studies found that FGF-23 induces expression of the sodium-chloride cotransporter (NCC) to stimulate sodium retention and induce hypertension (66). This effect could be over-come by the diuretic hydrochlorothiazide. This emphasizes that FGF-23 and the renal regulation of the elements phosphorus (P) and sodium (Na) are possibly intertwined, and interventions that target sodium (diuretics, a low sodium diet) may thus influence phosphate regulation. Consequently, both elements may be identified as twin-targets for dietary restriction. A sixth strategy involves potassium supplementation. Potassium supplementation increases serum phosphate levels (67). This suggest that potassium is another dietary component capable of influencing phosphate regulation, possibly potassium could lower FGF-23 concentrations. If so, potassium would be another target for dietary intervention. However, achievement of dietary intake targets is difficult: notwithstanding dietary recommendations (4), few patients with CKD achieve adherence to dietary sodium restriction (68). Therefore, better strategies for sustained changes in dietary habits are direly needed.

Need for a Multidisciplinary Approach for Enhancing Elements of a Healthy Lifestyle

Restriction of phosphate uptake can be achieved by pharmacological and dietary interventions. Pharmacological phosphate binder therapy is costly, not fully effective and comes with a sub-stantial pill-burden for patients (69). Compliance to a phosphate-restricted diet is difficult and challenging, as phosphate is ubiquitously present in our food products (70), although avoidance of phosphate-rich additives is feasible and facilitates control of hyperphosphatemia.

Page 9: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

Chapter 1

16

Several dietary components affect outcome in CKD and the general population. A healthy lifestyle must thus address such dietary factors including, but not restricted to, phosphate. A higher intake of sodium puts patients with CKD at higher risk for progression to ESRD (71) and increased risk for cardiovascular complications despite treatment (72). Even in the general population, high sodium intake is associated with higher blood pressure (73) and increased car-diovascular mortality especially in overweight subjects (74), whereas higher potassium intake may be protective against development of hypertension (75) and mortality (76). Phosphate intake is not routinely analyzed, however the results are mixed between detrimental (77), absent (25), or protective effects (78) in the general, CKD and mixed population respectively. Conversely, the concentration of phosphate in the serum phosphate is a strong predictor of renal disease progression (21) and mortality (24). Sodium restriction has been assessed in several trials in our center (79-82), and elsewhere (83, 84). These studies typically consist of short-term (6 weeks) interventions, and many patients relapse in old habits after participation (68). Of course, a sustained change resulting from dietary interventions is needed to elicit true impact in clinical practice. A sustained change in health behavior however is notoriously dif-ficult to achieve. To achieve and maintain a successful change in health behavior, behavioral interventions can be deployed, for example based on the self-regulation therapy (85). Guidance in the attainment of goals throughout the phases of behavioral change in self-regulation theory however can be time-extensive ‒and hence costly. E-health technology has emerged rapidly over the last few years, and may have potential to support the process of behavioral change and self-management skills of patients with CKD, but experience so far is limited, and not up to the level of evidence-based medicine. All in all, there is an urgent need for novel approaches that can achieve long-term beneficial changes in dietary elements of a healthy lifestyle.

Outline of Thesis

RAAS blockade is the ship-of-the-line in prevention of progressive renal function loss. However, RAAS blockade only delays end stage renal disease by 4‒8 months and indiscriminate combina-tion of several RAAS blocking agents together may be harmful, so alternative approaches to enhance the protective effects of RAAS blockade are needed. These approaches could be phar-macological or dietary intervention, or their combination. This thesis addresses interactions between RAAS/volume status, and regulators of phosphate-, bone- and mineral homeostasis, to identify better treatment strategies based on combined modulation of RAAS/volume status, and of factors involved in phosphate‒bone-mineral homeostasis. We expand this introduction with a review of vitamin D as adjunct to improve antiproteinuric efficacy of RAAS blockade (→ Chapter 2). As became clear, sodium intake remains a treatment target in CKD. This deserves a more elaborate explanation, as we review in (→ Chapter 3). Because FGF-23 may interact with the renin-angiotensin-aldosterone system (37) and higher serum phosphate concentrations

Page 10: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

17

Introduction and Aims of Thesis

1reduce the protective effects of RAAS blockade (21), we set out to investigate whether higher FGF-23 concentrations may impair the efficacy of intensification of RAAS blockade by a low sodium diet (→ Chapter 4). FGF-23 emerged as a robust risk factor for cardiovascular mortality in haemodialysis patients (50, 51), but after renal transplantation FGF-23 concentrations return to near-normal values (49). We hypothesized that relatively higher FGF-23 concentrations in stable renal transplant recipients still herald a high risk of cardiovascular mortality (→Chapter 5). Because FGF-23 is correlated more strongly with volume overload rather than ischemic cardiovascular events (65), we suggested that volume overload itself may be a determinant of FGF-23 levels. Therefore, we assessed in haemodialysis patients the relation between ultrafil-tration volume – the amount of volume that needs to be withdrawn– during a haemodialysis session and FGF-23 concentrations (→ Chapter 6). We also investigated whether the steep drop in serum phosphate during haemodialysis may lower FGF-23 values (→ Chapter 6). To expand the hypothesis that FGF-23 may directly induce volume overload by effects on sodium handling (66), we assessed, the other way round, whether changes in volume status may influence FGF-23 concentrations in patients with preserved renal function (→ Chapter 7). The elements sodium and phosphorus are literally connected at the molecular level in many food products as sodium-phosphate salts (86), and a diet rich of these additives increases FGF-23 levels (87). In a broader perspective, we investigated if sodium intake and phosphate intake are correlated in different patient populations (→ Chapter 8). Another viewpoint arose from the observation that potassium supplementation increases serum phosphate levels (67). We suggested that potassium exerts this effect by reducing FGF-23 concentrations (→ Chapter 9).

This thesis addresses several dietary elements that affect renal and cardiovascular outcomes. However, to realize the therapeutic potential of dietary improvements in clinical practice, a sustained change in dietary behavior must be established. Individual behavioral counseling of patients is time-extensive and costly. E-health technology could facilitate counseling and sup-port behavioral approaches based on self-regulation theory. Therefore, we investigated the fea-sibility and cost-efficacy of a multidisciplinary e-health approach aimed at reduction of dietary sodium intake in renal patients in the randomized, multicenter SUBLIME trial (→ Chapter 10). A general discussion of all these chapters in the perspective of current literature will be provided in (→ Chapter 11).

Page 11: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

Chapter 1

18

References

1. Bruck K, Stel VS, Gambaro G, et al. CKD Prevalence Varies across the European General Population. J.Am.Soc.Nephrol. 2015; 27: 2135-47.

2. de Zeeuw D, Hillege HL, de Jong PE. The kidney, a cardiovascular risk marker, and a new target for therapy. Kidney Int.Suppl. 2005; (98): S25-9.

3. Hallan SI, Matsushita K, Sang Y, et al. Age and association of kidney measures with mortality and end-stage renal disease. JAMA 2012; 308: 2349-2360.

4. Werkgroep Richtlijn Chronische Nierschade, Rensma PL, Hagen EC, et al. Richtlijn voor de behandeling van atiënten met Chronische Nierschade (CNS). 2009.

5. Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group. KDIGO 2012 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney Int.Suppl. 2013; 3: 1-150.

6. Lewis EJ, Hunsicker LG, Clarke WR, et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N.Engl.J.Med. 2001; 345: 851-860.

7. Brenner BM, Cooper ME, de Zeeuw D, et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N.Engl.J.Med. 2001; 345: 861-869.

8. Inker LA, Coresh J, Levey AS, Tonelli M, Muntner P. Estimated GFR, Albuminuria, and Complications of Chronic Kidney Disease. Journal of the American Society of Nephrology 2011; 22: 2322-2331.

9. Kunz R, Friedrich C, Wolbers M, Mann JF. Meta-analysis: effect of monotherapy and combination therapy with inhibitors of the renin angiotensin system on proteinuria in renal disease. Ann.Intern.Med. 2008; 148: 30-48.

10. Holtkamp FA, de Zeeuw D, de Graeff PA, et al. Albuminuria and blood pressure, independent targets for cardioprotective therapy in patients with diabetes and nephropathy: a post hoc analysis of the combined RENAAL and IDNT trials. Eur.Heart J. 2011; 32: 1493-1499.

11. Ruggenenti P, Perna A, Remuzzi G, GISEN Group Investigators. Retarding progression of chronic renal disease: the neglected issue of residual proteinuria. Kidney Int. 2003; 63: 2254-2261.

12. Mann JF, Schmieder RE, McQueen M, et al. Renal outcomes with telmisartan, ramipril, or both, in people at high vascular risk (the ONTARGET study): a multicentre, randomised, double-blind, controlled trial. Lancet 2008; 372: 547-553.

13. Parving HH, Brenner BM, McMurray JJ, et al. Cardiorenal end points in a trial of aliskiren for type 2 diabe-tes. N.Engl.J.Med. 2012; 367: 2204-2213.

14. Fried LF, Emanuele N, Zhang JH, et al. Combined Angiotensin inhibition for the treatment of diabetic nephropathy. N.Engl.J.Med. 2013; 369: 1892-1903.

15. Navis G, de Jong PE, Donker AJ, van der Hem GK, de Zeeuw D. Moderate sodium restriction in hypertensive subjects: renal effects of ACE-inhibition. Kidney Int. 1987; 31: 815-819.

16. Heeg JE, de Jong PE, van der Hem GK, de Zeeuw D. Efficacy and variability of the antiproteinuric effect of ACE inhibition by lisinopril. Kidney Int. 1989; 36: 272-279.

17. Slagman MC, Waanders F, Vogt L, et al. Elevated N-terminal pro-brain natriuretic peptide levels predict an enhanced anti-hypertensive and anti-proteinuric benefit of dietary sodium restriction and diuretics, but not angiotensin receptor blockade, in proteinuric renal patients. Nephrol.Dial.Transplant. 2012; 27: 983-990.

18. Hung SC, Kuo KL, Peng CH, et al. Volume overload correlates with cardiovascular risk factors in patients with chronic kidney disease. Kidney Int. 2014; 85: 703-709.

19. Svenningsen P, Friis UG, Versland JB, et al. Mechanisms of renal NaCl retention in proteinuric disease. Acta Physiol.(Oxf) 2013; 207: 536-545.

Page 12: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

19

Introduction and Aims of Thesis

1 20. de Seigneux S, Courbebaisse M, Rutkowski JM, et al. Proteinuria Increases Plasma Phosphate by Altering

Its Tubular Handling. Journal of the American Society of Nephrology 2015; 26: 1608-1618. 21. Zoccali C, Ruggenenti P, Perna A, et al. Phosphate may promote CKD progression and attenuate renopro-

tective effect of ACE inhibition. J.Am.Soc.Nephrol. 2011; 22: 1923-1930. 22. Floege J, Kim J, Ireland E, et al. Serum iPTH, calcium and phosphate, and the risk of mortality in a European

haemodialysis population. Nephrol.Dial.Transplant. 2011; 26: 1948-1955. 23. Isakova T, Wahl P, Vargas GS, et al. Fibroblast growth factor 23 is elevated before parathyroid hormone and

phosphate in chronic kidney disease. Kidney Int. 2011; 79: 1370-1378. 24. Kestenbaum B, Sampson JN, Rudser KD, et al. Serum phosphate levels and mortality risk among people

with chronic kidney disease. J.Am.Soc.Nephrol. 2005; 16: 520-528. 25. Selamet U, Tighiouart H, Sarnak MJ, et al. Relationship of dietary phosphate intake with risk of end-stage

renal disease and mortality in chronic kidney disease stages 3-5: The Modification of Diet in Renal Disease Study. Kidney Int. 2015; 89: 176-84.

26. Chang AR, Grams ME. Serum phosphorus and mortality in the Third National Health and Nutrition Exami-nation Survey (NHANES III): effect modification by fasting. Am.J.Kidney Dis. 2014; 64: 567-573.

27. Fernández-Martín JL, Martínez-Camblor P, Dionisi MP, et al. Improvement of mineral and bone metabo-lism markers is associated with better survival in haemodialysis patients: the COSMOS study. Nephrology Dialysis Transplantation 2015; 30: 1542-1551.

28. Haussler MR, Whitfield GK, Kaneko I, et al. The role of vitamin D in the FGF23, klotho, and phosphate bone-kidney endocrine axis. Rev.Endocr Metab.Disord. 2012; 13: 57-69.

29. Shimada T, Mizutani S, Muto T, et al. Cloning and characterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proc.Natl.Acad.Sci.U.S.A. 2001; 98: 6500-6505.

30. Shimada T, Muto T, Urakawa I, et al. Mutant FGF-23 responsible for autosomal dominant hypophospha-temic rickets is resistant to proteolytic cleavage and causes hypophosphatemia in vivo. Endocrinology 2002; 143: 3179-3182.

31. Liu S, Tang W, Zhou J, et al. Fibroblast growth factor 23 is a counter-regulatory phosphaturic hormone for vitamin D. J.Am.Soc.Nephrol. 2006; 17: 1305-1315.

32. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, et al. The parathyroid is a target organ for FGF23 in rats. J.Clin.Invest. 2007; 117: 4003-4008.

33. Galitzer H, Ben-Dov IZ, Silver J, Naveh-Many T. Parathyroid cell resistance to fibroblast growth factor 23 in secondary hyperparathyroidism of chronic kidney disease. Kidney Int. 2010; 77: 211-218.

34. Goetz R, Beenken A, Ibrahimi OA, et al. Molecular insights into the klotho-dependent, endocrine mode of action of fibroblast growth factor 19 subfamily members. Mol.Cell.Biol. 2007; 27: 3417-3428.

35. Kuro-o M, Matsumura Y, Aizawa H, et al. Mutation of the mouse klotho gene leads to a syndrome resem-bling ageing. Nature 1997; 390: 45-51.

36. Shimada T, Kakitani M, Yamazaki Y, et al. Targeted ablation of Fgf23 demonstrates an essential physiologi-cal role of FGF23 in phosphate and vitamin D metabolism. J.Clin.Invest. 2004; 113: 561-568.

37. de Borst MH, Vervloet MG, ter Wee PM, Navis G. Cross talk between the renin-angiotensin-aldosterone system and vitamin D-FGF-23-klotho in chronic kidney disease. J.Am.Soc.Nephrol. 2011; 22: 1603-1609.

38. Li YC, Kong J, Wei M, Chen ZF, Liu SQ, Cao LP. 1,25-Dihydroxyvitamin D(3) is a negative endocrine regulator of the renin-angiotensin system. J.Clin.Invest. 2002; 110: 229-238.

39. Faul C, Amaral AP, Oskouei B, et al. FGF23 induces left ventricular hypertrophy. J.Clin.Invest. 2011; 121: 4393-4408.

40. Grabner A, Amaral A, Schramm K, et al. Activation of Cardiac Fibroblast Growth Factor Receptor 4 Causes Left Ventricular Hypertrophy. Cell Metabolism. 2015;22(6):1020-1032.

Page 13: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

Chapter 1

20

41. Lavi-Moshayoff V, Wasserman G, Meir T, Silver J, Naveh-Many T. PTH increases FGF23 gene expression and mediates the high-FGF23 levels of experimental kidney failure: a bone parathyroid feedback loop. Am.J.Physiol.Renal Physiol. 2010; 299: F882-9.

42. Ferrari SL, Bonjour JP, Rizzoli R. Fibroblast growth factor-23 relationship to dietary phosphate and renal phosphate handling in healthy young men. J.Clin.Endocrinol.Metab. 2005; 90: 1519-1524.

43. Quinn SJ, Thomsen AR, Pang JL, et al. Interactions between calcium and phosphorus in the regulation of the production of fibroblast growth factor 23 in vivo. Am.J.Physiol.Endocrinol.Metab. 2013; 304: E310-20.

44. David V, Martin A, Isakova T, et al. Inflammation and functional iron deficiency regulate fibroblast growth factor 23 production. Kidney Int. 2015; 89: 135-46.

45. Jonsson KB, Zahradnik R, Larsson T, et al. Fibroblast growth factor 23 in oncogenic osteomalacia and X-linked hypophosphatemia. N.Engl.J.Med. 2003; 348: 1656-1663.

46. Larsson T, Nisbeth U, Ljunggren O, Juppner H, Jonsson KB. Circulating concentration of FGF-23 increases as renal function declines in patients with chronic kidney disease, but does not change in response to variation in phosphate intake in healthy volunteers. Kidney Int. 2003; 64: 2272-2279.

47. Wang M, You L, Li H, et al. Association of circulating fibroblast growth factor-23 with renal phosphate excretion among hemodialysis patients with residual renal function. Clin.J.Am.Soc.Nephrol. 2013; 8: 116-125.

48. Viaene L, Bammens B, Meijers BK, Vanrenterghem Y, Vanderschueren D, Evenepoel P. Residual renal func-tion is an independent determinant of serum FGF-23 levels in dialysis patients. Nephrol.Dial.Transplant. 2012; 27: 2017-2022.

49. Wolf M, Weir MR, Kopyt N, et al. A Prospective Cohort Study of Mineral Metabolism After Kidney Trans-plantation. Transplantation 2015; 100: 184-93..

50. Jean G, Terrat J, Vanel T, et al. High levels of serum fibroblast growth factor (FGF)-23 are associated with increased mortality in long haemodialysis patients. Nephrology Dialysis Transplantation 2009; 24: 2792-2796.

51. Gutierrez OM, Mannstadt M, Isakova T, et al. Fibroblast growth factor 23 and mortality among patients undergoing hemodialysis. N.Engl.J.Med. 2008; 359: 584-592.

52. Isakova T, Xie H, Yang W, et al. Fibroblast growth factor 23 and risks of mortality and end-stage renal disease in patients with chronic kidney disease. JAMA 2011; 305: 2432-2439.

53. Seiler S, Rogacev KS, Roth HJ, et al. Associations of FGF-23 and sKlotho with cardiovascular outcomes among patients with CKD stages 2-4. Clin.J.Am.Soc.Nephrol. 2014; 9: 1049-1058.

54. Lutsey PL, Alonso A, Selvin E, et al. Fibroblast growth factor-23 and incident coronary heart disease, heart failure, and cardiovascular mortality: the Atherosclerosis Risk in Communities study. J.Am.Heart Assoc. 2014; 3: e000936.

55. Ix JH, Katz R, Kestenbaum BR, et al. Fibroblast growth factor-23 and death, heart failure, and cardiovascu-lar events in community-living individuals: CHS (Cardiovascular Health Study). J.Am.Coll.Cardiol. 2012; 60: 200-207.

56. Antoniucci DM, Yamashita T, Portale AA. Dietary phosphorus regulates serum fibroblast growth factor-23 concentrations in healthy men. J.Clin.Endocrinol.Metab. 2006; 91: 3144-3149.

57. Vervloet MG, van Ittersum FJ, Buttler RM, Heijboer AC, Blankenstein MA, ter Wee PM. Effects of dietary phosphate and calcium intake on fibroblast growth factor-23. Clin.J.Am.Soc.Nephrol. 2011; 6: 383-389.

58. Isakova T, Barchi-Chung A, Enfield G, et al. Effects of dietary phosphate restriction and phosphate binders on FGF23 levels in CKD. Clin.J.Am.Soc.Nephrol. 2013; 8: 1009-1018.

59. Yilmaz MI, Sonmez A, Saglam M, et al. Comparison of calcium acetate and sevelamer on vascular function and fibroblast growth factor 23 in CKD patients: a randomized clinical trial. Am.J.Kidney Dis. 2012; 59: 177-185.

Page 14: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

21

Introduction and Aims of Thesis

1 60. Urena-Torres P, Prie D, Keddad K, et al. Changes in fibroblast growth factor 23 levels in normophospha-

temic patients with chronic kidney disease stage 3 treated with lanthanum carbonate: results of the PREFECT study, a phase 2a, double blind, randomized, placebo-controlled trial. BMC Nephrol. 2014; 15: 71-2369-15-71.

61. Shalhoub V, Shatzen EM, Ward SC, et al. FGF23 neutralization improves chronic kidney disease-associated hyperparathyroidism yet increases mortality. J.Clin.Invest. 2012; 122: 2543-2553.

62. Daugirdas JT, Chertow GM, Larive B, et al. Effects of frequent hemodialysis on measures of CKD mineral and bone disorder. J.Am.Soc.Nephrol. 2012; 23: 727-738.

63. Cornelis T, van der Sande FM, Eloot S, et al. Acute hemodynamic response and uremic toxin removal in con-ventional and extended hemodialysis and hemodiafiltration: a randomized crossover study. Am.J.Kidney Dis. 2014; 64: 247-256.

64. Zaritsky J, Rastogi A, Fischmann G, et al. Short daily hemodialysis is associated with lower plasma FGF23 levels when compared with conventional hemodialysis. Nephrol.Dial.Transplant. 2014; 29: 437-441.

65. Scialla JJ, Xie H, Rahman M, et al. Fibroblast growth factor-23 and cardiovascular events in CKD. J.Am.Soc.Nephrol. 2014; 25: 349-360.

66. Andrukhova O, Slavic S, Smorodchenko A, et al. FGF23 regulates renal sodium handling and blood pres-sure. EMBO Mol.Med. 2014; 6: 744-759.

67. Sebastian A, Hernandez RE, Portale AA, Colman J, Tatsuno J, Morris RC,Jr. Dietary potassium influences kidney maintenance of serum phosphorus concentration. Kidney Int. 1990; 37: 1341-1349.

68. de Borst MH, Navis G. Sodium intake, RAAS-blockade and progressive renal disease. Pharmacol Res Avail-able online 31 March 2016, ISSN 1043-6618, http://dx.doi.org/10.1016/j.phrs.2016.03.037.

69. Tonelli M, Pannu N, Manns B. Oral phosphate binders in patients with kidney failure. N.Engl.J.Med. 2010; 362: 1312-1324.

70. Kalantar-Zadeh K, Gutekunst L, Mehrotra R, et al. Understanding sources of dietary phosphorus in the treatment of patients with chronic kidney disease. Clin.J.Am.Soc.Nephrol. 2010; 5: 519-530.

71. Vegter S, Perna A, Postma MJ, Navis G, Remuzzi G, Ruggenenti P. Sodium intake, ACE inhibition, and progression to ESRD. J.Am.Soc.Nephrol. 2012; 23: 165-173.

72. Lambers Heerspink HJ, Holtkamp FA, Parving HH, et al. Moderation of dietary sodium potentiates the renal and cardiovascular protective effects of angiotensin receptor blockers. Kidney Int. 2012; 82: 330-337.

73. Geleijnse JM, Kok FJ, Grobbee DE. Blood pressure response to changes in sodium and potassium intake: a metaregression analysis of randomised trials. J.Hum.Hypertens. 2003; 17: 471-480.

74. Tuomilehto J, Jousilahti P, Rastenyte D, et al. Urinary sodium excretion and cardiovascular mortality in Finland: a prospective study. The Lancet 2001; 357: 848-851.

75. Kieneker LM, Gansevoort RT, Mukamal KJ, et al. Urinary potassium excretion and risk of developing hyper-tension: the prevention of renal and vascular end-stage disease study. Hypertension 2014; 64: 769-776.

76. O’Donnell M, Mente A, Rangarajan S, et al. Urinary sodium and potassium excretion, mortality, and cardiovascular events. N.Engl.J.Med. 2014; 371: 612-623.

77. Chang AR, Lazo M, Appel LJ, Gutierrez OM, Grams ME. High dietary phosphorus intake is associated with all-cause mortality: results from NHANES III. Am.J.Clin.Nutr. 2014; 99: 320-327.

78. Palomino HL, Rifkin DE, Anderson C, Criqui MH, Whooley MA, Ix JH. 24-Hour Urine Phosphorus Excretion and Mortality and Cardiovascular Events. Clinical Journal of the American Society of Nephrology 2013; 8: 1202-1210.

79. Vogt L, Waanders F, Boomsma F, de Zeeuw D, Navis G. Effects of dietary sodium and hydrochlorothiazide on the antiproteinuric efficacy of losartan. J.Am.Soc.Nephrol. 2008; 19: 999-1007.

Page 15: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

Chapter 1

22

80. Slagman MC, Waanders F, Hemmelder MH, et al. Moderate dietary sodium restriction added to angio-tensin converting enzyme inhibition compared with dual blockade in lowering proteinuria and blood pressure: randomised controlled trial. BMJ 2011; 343: d4366.

81. Kwakernaak AJ, Krikken JA, Binnenmars SH, et al. Effects of sodium restriction and hydrochlorothiazide on RAAS blockade efficacy in diabetic nephropathy: a randomised clinical trial. Lancet Diabetes Endocrinol. 2014; 2: 385-395.

82. de Vries LV, Dobrowolski LC, van den Bosch JJ, et al. Effects of Dietary Sodium Restriction in Kidney Trans-plant Recipients Treated With Renin-Angiotensin-Aldosterone System Blockade: A Randomized Clinical Trial. Am.J.Kidney Dis. 2016; 67: 936-44.

83. McMahon EJ, Bauer JD, Hawley CM, et al. A randomized trial of dietary sodium restriction in CKD. J.Am.Soc.Nephrol. 2013; 24: 2096-2103.

84. Ekinci EI, Thomas G, Thomas D, et al. Effects of salt supplementation on the albuminuric response to telmisartan with or without hydrochlorothiazide therapy in hypertensive patients with type 2 diabetes are modulated by habitual dietary salt intake. Diabetes Care 2009; 32: 1398-1403.

85. Maes S, Karoly P. Self-Regulation Assessment and Intervention in Physical Health and Illness: A Review. Appl.Psychol. 2005; 54: 267-299.

86. Lampila LE. Applications and functions of food-grade phosphates. Ann.N.Y.Acad.Sci. 2013; 1301: 37-44. 87. Gutierrez OM, Luzuriaga-McPherson A, Lin Y, Gilbert LC, Ha SW, Beck GR,Jr. Impact of phosphorus-based

food additives on bone and mineral metabolism. J.Clin.Endocrinol.Metab. 2015; jc20152279.

Page 16: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several
Page 17: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several
Page 18: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several

Part IMinerals, Volume and

Fibroblast Growth Factor 23

Page 19: University of Groningen Fibroblast Growth Factor 23: a ... · The role of extracellular ... In 2001, fibroblast growth factor 23 (FGF-23) was discovered as the culprit of several