Laboratory Abnormalities in CKD-MBD: Markers, Predictors, or Mediators of Disease? Pieter Evenepoel, MD,* Mariano Rodriguez, MD,† and Markus Ketteler, MD‡

Summary: Chronic kidney disease–mineral and bone disorder (CKD-MBD) is characterized by bone abnormalities, vascular calcification, and an array of laboratory abnormalities. The latter classically include disturbances in the parathyroid hormone/vitamin D axis. More recently, fibroblast growth factor 23 (FGF23) and klotho also have been identified as important regulators of mineral metabolism. Klotho deficiency and high circulating FGF23 levels precede secondary hyperparathyroidism in CKD patients. Levels of FGF23 and parathyroid hormone increase along the progression of CKD to maintain mineral homeostasis and to overcome end-organ resistance. It is hard to define when the increase of both hormones becomes maladaptive. CKD-MBD is associated with adverse outcomes including cardiovascular disease and mortality. This review summarizes the complex pathophysiology of CKD-MBD and outlines which laboratory abnormalities represent biomarkers of disease severity, which laboratory abnormalities are predictors of cardiovascular disease, and which laboratory abnormalities should be considered (direct) uremic toxins exerting organ damage. This information may help to streamline current and future therapeutic efforts. Semin Nephrol 34:151-163 C 2014 Elsevier Inc. All rights reserved. Keywords: Calcium, hyperphosphatemia, vitamin D, uremic toxins, FGF23, klotho, PTH

C

alcium and phosphate are essential to many vital physiological processes. Consequently, the maintenance of Ca and P homeostasis is essential to a healthy existence. Several organs contribute to the exquisite regulation of calcium and phosphate homeostasis by facilitating intestinal absorption, bone (de)mineralization, and renal excretion/ reabsorption of both ions. Regulation of these processes occurs by a number of hormones. The biologically active forms of vitamin D (1,25‐dihydroxyvitamin D3, further referred to as calcitriol), parathyroid hormone (PTH), and calcitonin have been studied extensively in this regard. More recently, fibroblast growth factor 23 (FGF23) and klotho were

*

Department of Nephrology, University Hospitals Leuven, Leuven, Belgium. Department of Nephrology, IMIBIC, University Hospital Reina Sofia, Cordoba, Spain. ‡ Division of Nephrology, Klinikum Coburg GmbH, Coburg, Germany. Financial support: none. Conflict of interest statements: Pieter Evenepoel has served on the advisory board, received research grants, and/or speaker’s fees from Amgen, Genzyme/Sanofi, and Shire; Mariano Rodriguez has received financial support from Syskid and the Government of Spain, and has received regional research support from Amgen, Fresenius, and AbbVie; and Markus Ketteler has served on the advisory board, received research grants, and/or speaker’s fees from Amgen, AbbVie, Fresenius Medical Care, Medice, Mitsubishi, Sanofi, Shire, and Vifor. Address reprint requests to Markus Ketteler, MD, Fellow of the European Renal Association, Division of Nephrology, Klinikum Coburg GmbH, Ketschendorfer Str. 33, D-96450 Coburg, Germany. E-mail: [email protected] 0270-9295/ - see front matter & 2014 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.semnephrol.2014.02.007 †

Seminars in Nephrology, Vol 34, No 2, March 2014, pp 151–163

identified as new players essential to the regulation of calcium and phosphate homeostasis (Fig. 1).1 Acknowledging the important role of the kidneys in the homeostasis of calcium and phosphate, it is not surprising that disturbances in mineral metabolism occur early in the course of chronic kidney disease (CKD). Current data postulate that increased FGF23 may be the earliest alteration in mineral metabolism in CKD.2,3 Gradually increasing FGF23 levels cause the early decrease in calcitriol levels.4,5 This frees PTH from feedback inhibition, leading to secondary hyperparathyroidism. All of these changes occur long before changes in serum phosphate and calcium levels become evident. The primary stimulus for enhanced FGF23 secretion in early CKD remains obscure. On the basis of scant human and experimental data, it has been proposed that decreased expression of klotho, which functions as a co-receptor for FGF23, may be one step further upstream from FGF23 excess.6 The pattern of metabolic changes, and especially the observation of a modest reduction in serum phosphate during early CKD, however, argues against the latter hypothesis. Additional work must be performed to determine if phosphate plays any direct role in the early increase in FGF23. Certainly, early experimental work has been able to show that strict reduction of phosphate intake prevents PTH increase in early CKD.7 To emphasize the close association and interaction between laboratory abnormalities, bone disease, and vascular calcification, KDIGO in 2006 coined the term CKD-mineral and bone disorder (CKD-MBD).8 Bone disease starts in patients with CKD stage 2, and becomes almost universal in patients with CKD stage 5.9,10 There is significant heterogeneity of histologic abnormalities and patients may go through phases with 151

152

P. Evenepoel, M. Rodriguez, and M. Ketteler FGFR / Klotho

FGFR / Klotho

filtration rate (GFR) decreases to less than 30 mL/ min/1.73 m2 (ie, when compensatory mechanisms [mediated to a large extent by high PTH and FGF23] start to fail). Thus, hyperphosphatemia is a late marker of a disturbed phosphate metabolism.

Parathyroid glands kidneys • Suppression of PTH secreon / synthesis

FGF23

• Phosphaturia inducon • Suppression of calcitriol synthesis

bone

Figure 1. FGF23 and klotho effects on the parathyroid glands and on the kidneys.

different, partly treatment-associated abnormalities, or the initial changes may progress with deterioration of kidney function. Strikingly, there has been a marked increase of the prevalence of low bone turnover disease over the past 2 decades.11 This most probably reflects demographic changes (ageing population with high prevalence of diabetes) and overzealous suppression of PTH through the use of calcium-based phosphate binders and pharmacologic doses of calcitriol.12 Extraskeletal and especially vascular calcifications are extremely common in CKD patients. Prevalence rates reported in the literature vary according to the vascular bed examined (coronary versus aortic) and the imaging technique used, but overall range from 40% in stage 3, to 80% to 90% in patients with CKD stage 5D.13,14 Biochemical abnormalities in bone and mineral metabolism, bone disease, and vascular calcification all have been associated with poor outcomes in CKD patients.15

Phosphate as a Uremic Toxin Phosphate and cardiovascular disease

CKD patients suffer from a high cardiovascular mortality rate. It was not until the late 1990s when nephrologists became aware of phosphate as an independent predictor of cardiovascular mortality.18–20 Phosphate is a main factor in the development of vascular calcification and therefore it affects coronary pathobiology as well as myocardial performance by inducing arterial stiffness, valvular rigidity, and left ventricular hypertrophy. Epidemiologic observations are corroborated by abundant experimental data. Phosphate, in synergism with calcium, drives accelerated calcification by its distinct effect on vascular smooth muscle cells (VSMCs), in particular through hydroxyapatite nanocrystal formation. Key processes include osteochondrocytic differentiation, apoptosis, and vesicle release, and perturbation of calcification inhibitor levels (Fig. 2).21–28 Osteochondrocytic differentiation is characterized by active cellular phosphate uptake, downregulation of VSMC-specific genes, de novo induction of the osteoblast transcription factor cbfa-1 (runx2), release of precalcified matrix vesicles, and local expression of bone proteins including alkaline phosphatase, osteocalcin, osteopontin, and collagen I, all within the vessel wall. pre-calcified matrix vesicles

PHOSPHATE Phosphate Balance Inorganic phosphorus is essential for multiple biological functions such as intracellular signal transduction, the production and function of cell membranes, and energy exchange. In the healthy state, serum phosphate levels are maintained within the physiological range by the regulation of dietary absorption, bone formation and resorption, and renal excretion, as well as by equilibration with intracellular stores. Given the important role of the kidneys, it is not surprising that phosphate metabolism is disturbed early in CKD. Although it is common to indicate that phosphate retention is typical for CKD, there are no data to suggest that total-body phosphate load is actually increased, at least not in predialysis CKD.16,17 It should be emphasized that hyperphosphatemia only occurs once the estimated glomerular

PO

Na

Hydroxylapatite NPC

Pi Cbfa-1

SMC-genes Human aortic smooth muscle cell (SMC)

alkaline phosphatase Ca -binding proteins (osteocalcin, osteopontin) Collagen-rich extracellular matrix

Cbfa-1 = core-binding factor-1 (central transcription factor for osteogenic differentiation)

Figure 2. Phosphate as a uremic toxin. Hyperphosphatemia leads to active uptake of phosphate into human aortic smooth muscle cells (SMCs). The consecutive increases in intracellular phosphate concentrations cause down-regulation of SMC-specific genes and up-regulation of the osteoblast transcription factor cbfa -1 (or runx2). Subsequently, the human aortic SMCs act similar to bone cell–producing precalcified matrix vesicles and a variety of bone-specific proteins (alkaline phosphatase, osteocalcin, osteopontin, collagen I). This specific process of a phenotypic cell change is termed osteochondrogenic transdifferentiation. Cbfa-1, core-binding factor-1; NPC, sodium-phosphate co-transporter PIT-1.

CKD-MBD abnormalities

Independent of the deleterious effect of high phosphate on VSMCs, recent studies have shown that high phosphate levels directly affect endothelial function both in vitro and in vivo. For example, di Marco et al29 showed annexin II down-regulation and inhibition of endothelial-dependent angiogenesis after phosphate exposure in vitro. Finally, high phosphate may contribute to the accelerated aging observed in CKD patients; phosphate appears to be a key element in defining the Klotho-/phenotype, and reducing serum phosphate levels in Klotho-/- mice ameliorated premature aging-like features and prolonged survival.30

153

Calcitriol METABOLIC BONE DISEASE

FGF-23

PTH

LEFT VENTRICULAR HYPERTROPHY VASCULAR CALCIFICATION ENDOTHELIAL DYSFUNCTION

Phosphate and renal disease progression

Several studies have reported an association between a disturbed phosphate metabolism and accelerated loss of renal function.31–37 The negative effect of phosphate on cardiovascular status may condition an accelerated progression of renal disease. In addition, according to the precipitation-calcification hypothesis, this accelerated loss may be explained (at least partly) by deposition of calcium phosphate crystals in the renal parenchyma, resulting in nephrocalcinosis,38 which in its turn leads to inflammatory response and renal damage, inciting a vicious circle.39 Dietary phosphorus restriction or phosphate binders reduce the occurrence of nephrocalcinosis and attenuate the progressive loss of renal function in animal studies.31,38,40–44 Phosphate as a Therapeutic Target High phosphate levels may be blamed for being a key contributor to cardiovascular mortality in CKD. This defines phosphate as a very relevant uremic toxin and therefore as a main target for therapy (Fig. 3). Current guidelines suggest maintaining serum phosphate levels in the normal range in CKD stages 3 to 5 and decreasing serum phosphate levels toward the normal range in dialysis patients. Dietary restriction of phosphate has long been the cornerstone of therapy for disordered mineral metabolism. Recent data point to phosphate additives as an important and theoretically avoidable source of dietary phosphorous.45–47 Dietary measures, overall, are not sufficient to control hyperphosphatemia. Therefore, adjunctive therapy with oral phosphate binders is used in most patients with advanced CKD. Numerous studies have investigated the safety and efficacy of oral phosphate binder therapy in patients on dialysis.48 Recent large observational studies have supported a beneficial effect of oral phosphate binder therapy in dialysis patients.49 Definite evidence from a randomized controlled trial, however, is still awaited. Because phosphate metabolism is disrupted early in the course of CKD, a

Cardiovascular Mortality Figure 3. Excess of phosphate leads to high FGF23, which in turn reduces calcitriol production, which in its turn causes hyperparathyroidism. High phosphate by itself also is responsible for the increase in PTH levels. Each of these factors has been associated with increased mortality and is involved directly in some of the cardiovascular abnormalities observed in CKD. In addition, metabolic bone disease is the consequence of abnormal PTH and vitamin D (calcitriol) metabolism.

rationale may exist for early treatment of this disorder. To date, however, few studies have investigated the role of oral phosphate binder therapy in patients with CKD not yet on dialysis, and most studies yielded disappointing results, both with regard to biochemical and intermediate cardiovascular end points.50,51 In aggregate, these data question the appropriateness of phosphate binder therapy in patients who have moderate to advanced CKD and normal or near-normal serum phosphorus concentrations.

CALCIUM Calcium Balance Given the crucial role of calcium in many metabolic processes, serum calcium concentrations are tightly regulated, mainly by the actions of PTH and calcitriol. Serum calcium concentrations are maintained in the normal range until very late in CKD when they decrease slightly. The key issue when considering calcium as a potential uremic toxin is that serum calcium levels poorly reflect calcium balance in CKD.52 Recently, rigorous calcium balance studies were performed by two research groups in CKD patients not yet on dialysis.16,53 Although participants of both studies were in neutral or slightly negative balance when consuming the normal calcium diet, they went into markedly positive calcium balance when treated with calcium supplements for up to 3 weeks. Additional long-term experiments are required to

154

P. Evenepoel, M. Rodriguez, and M. Ketteler

investigate whether prolonged exposure yields similar results and to elucidate the fate of the retained calcium.17,54 Calcium as Uremic Toxin Although the subject of ongoing controversy, several lines of evidence have suggested that calcium loading is an important culprit of cardiovascular disease in CKD. The poor relationship between serum calcium levels and calcium balance52 probably explains the observations from epidemiologic trials that even high calcium levels are usually only weak predictors of adverse outcomes, but that calcium exposure may be a cardiovascular risk factor.19,55,56 One of the key studies pointing to calcium toxicity in uremia was published in 2010.57 The investigators used arterial samples obtained during abdominal surgery in children with normal renal function, with impaired renal function, and on dialysis, respectively. These intact arteries, free of any signs of atherosclerosis, were first exposed ex vivo to hyperphosphatemia to induce osteochondrocytic differentiation and calcification of VSMC. Although samples from healthy children were resistant to such phenotypic changes, there was some moderate calcification induction already in CKD arteries, and a severe calcification induction in vessels from children on dialysis. When the ex vivo calcium concentrations were increased, osteochondrocytic differentiation and calcification became synergistically accelerated—an experimental proof of the concept that calcium may not just be a passive compound in the process of intravascular hydroxyapatite formation.

A recent clinical pilot study was supportive for the concept of calcium toxicity in the context of cardiovascular calcification. Block et al50 performed a placebo-controlled trial on the effects of three different phosphate binders (sevelamer carbonate, lanthanum carbonate, and calcium acetate) on serum phosphate levels and parameters of phosphate metabolism in patients with CKD stages 3 to 4. Binders were all capable of modestly decreasing serum phosphate levels, and urinary phosphate excretion was reduced by 22%. Unexpectedly, there was an increase in coronary calcification scores with binder therapy over 9 months, and no progression in the placebo group. Looking at the 3 binders separately, this progressive calcification was observed exclusively in the group treated with the calcium-containing binder. In addition, 11 randomized clinical studies on the comparison of calcium-free versus calcium-containing phosphate binders in hyperphosphatemic patients recently were reviewed together by means of a metaanalysis (n ¼ 4,622 patients in different CKD stages).58 This systematic review showed a 22% survival benefit with calcium-free binders, associated with inhibition of cardiovascular calcification progression (Fig. 4). The remaining open question now is whether calcium-free binders are the superior choice in hyperphosphatemia treatment, or whether calciumcontaining binders are primarily harmful. Data from the recent Current Management of Secondary Hyperparathyroidism: a Multicentre Observational Study (COSMOS) trial seem to favor the first explanation, showing beneficial outcome associations of dialysis patients on calcium-containing binders versus those patients without phosphate binder treatment.59 Calcium

Figure 4. Calcium as a uremic toxin. Data from a recent systematic review showed a significant 22% survival disadvantage if hyperphosphatemic CKD patients were treated with calcium-containing versus calcium-free phosphate binders. Accelerated cardiovascular calcification is suspected as the major pathophysiologic mechanism behind this observation. Modified with permission.58

CKD-MBD abnormalities

still may qualify as a uremic toxin, but in this context high calcium loads or a markedly positive calcium balance may be the essential harmful culprits rather than the immediate serum calcium concentrations. However, one should always take into account the limitations with regard to the proper determination of calcium load and balance. Calcium as a Therapeutic Target Defining the optimal calcium intake in CKD is challenging given ongoing uncertainties concerning calcium balance in the different stages of disease, as mentioned earlier. Current guidelines suggest limiting elemental calcium intake in the presence of hypercalcemia, arterial calcification, and/or adynamic bone disease, and if serum PTH levels are persistently low.15 FGF23 FGF23 is a bone-derived hormone that plays an important function in regulating mineral metabolism.4 The physiological actions of FGF23 are to promote phosphaturia, decrease production of calcitriol, and suppress secretion of PTH. These effects are mediated through activation of the FGF receptor (FGFR) and its co-receptor klotho. FGF23 levels increase early in the course of CKD to reach levels in CKD stage 5D exceeding normal values by a factor of up to 1,000. In predialysis CKD, high FGF23 values may represent a compensatory mechanism to overcome FGF23 resistance and to maintain normophosphatemia. FGF23 Resistance It increasingly is recognized that CKD is a state of FGFR and especially klotho deficiency.60,61 In the human kidney, decreased klotho expression occurs as early as in CKD stage 2.60 In the parathyroid gland, the klotho/FGFR complex is down-regulated, particularly in nodular areas of the hyperplastic glands.62 The mechanisms contributing to klotho deficiency in CKD remain incompletely understood, but ischemia, angiotensin II, inflammation, and oxidative stress are all thought to be involved.63 Recent experimental evidence also points to the protein-bound uremic toxins indoxyl sulfate (IS) and p-cresyl sulfate as potential culprits. Administration of IS to hypertensive rats reduced renal expression of klotho, promoted cell senescence, and increased renal fibrosis. Cell signaling pathways involve induction of reactive oxygen and nuclear factor-κB activation, and subsequent interference with RNA transcription.64 Recently, Sun et al65 showed that IS and p-cresyl sulfate cause epigenetic modification and silencing of the klotho gene, resulting from increased DNA methyltransferase expression and DNA hypermethylation. The

155

toxicity of p-cresyl sulfate is discussed in more detail in the article by Sirich et al of this issue.66 Another possible mechanism of FGF23 resistance is impaired FGF23 signaling caused by accumulation of the C-terminal tail of FGF23, which competes with full-length ligand for binding to the klotho/FGFR complex,67 although this scenario may not be the case in patients undergoing dialysis.68,69 Parathyroid resistance to FGF23 may explain why high PTH levels may co-exist with high FGF23 levels in patients with advanced CKD.62 FGF23 as Uremic Toxin Recent epidemiologic studies have shown strong associations between increased FGF23 levels and poor renal37,70 and cardiovascular outcomes both in CKD patients and in the general population.71–74 Of note, adjustment for important covariates including phosphate, calcitriol, and vitamin D therapy did not affect the result substantially. Several explanations can be raised for these findings. First, FGF23 may be a kidney function-dependent surrogate for another risk factor. High on the list concerning this hypothesis is klotho deficiency because both factors have been shown to be related. It is well established that high FGF23 levels correlate with klotho deficiency.6 Klotho deficiency may potentiate the development of arterial calcification and thereby confer an increased risk of cardiovascular morbidity and mortality.75 This line of reasoning is supported by a recent study in older communitydwelling adults, linking low soluble klotho levels to all-cause mortality, even after adjustment for age, classic cardiovascular risk factors, and traditional markers of calcium-phosphate metabolism.76 The latter findings, however, could not be reproduced in a cohort of 312 CKD patients.77 Second, FGF23 may directly exert toxic effects. It has been shown in a series of elegant clinical and experimental studies that left ventricular hypertrophy (LVH) may be triggered by FGF23 in a klotho-independent manner and can be reduced by nonspecific FGF-receptor blockade.78,79 Intriguingly, Seiler et al80 showed the same LVH association in a cardiovascular cohort with no or only slightly impaired renal function. It is unlikely that in these individuals LVH is mediated by activation of nonspecific FGF receptors. This again fuels the thesis that FGF23 is a surrogate of another risk factor. FGF23 as a Therapeutic Target At present, it remains unclear whether FGF23 is a marker, predictor, or mediator of disease. However, directly antagonizing FGF23 might not be the right therapeutic approach, at least not in CKD patients not yet on dialysis because antagonizing FGF23 in these patients means antagonizing the phosphaturic defenses

156

of the body. This may confer important health risks as was shown recently in a study by Shalhoub et al.81 In the latter study, the use of specific FGF23 antibodies in an animal model of CKD led to increased mortality, despite some favorable effects on the majority of classic parameters of CKD-MBD and particularly on the development of secondary hyperparathyroidism. Hyperphosphatemia was assumed to be the mediator of mortality leading to accelerated calcification in animals after FGF23 antagonism. Treatment strategies that indirectly target FGF23 such as dietary phosphate restriction and phosphate binder therapy might be a better alternative. However, it should be emphasized that the efficacy, potency, and even safety of these treatment strategies remain to be shown.50 PTH PTH is a single-chain polypeptide of 84 amino acids that is secreted by parathyroid cells at a constant, low rate. PTH secretion is stimulated primarily in response to reductions in serum calcium concentration. The entire PTH molecule (1-84) is also called whole or biointact PTH; this is to differentiate it from other smallermolecular-weight carboxy-terminal fragments (CPTH), which also are normally present in plasma. The hormonal fragments arise from metabolism of 1–84 PTH by peripheral organs as well as from secretion of C-PTH fragments from the parathyroid glands.82 Cleavage of PTH (1–84) by liver cells generates C-PTH fragments that subsequently are removed from the circulation by the kidneys.83 Amino-terminal fragments (N-PTH) also are produced by Kupffer cells during cleavage of I-PTH but rapidly are degraded. Parathyroid cells secrete both PTH (1–84) and C-PTH.82 Although hypercalcemia reduces the secretion of both PTH (1–84) and C-PTH, the reduction of PTH (1–84) secretion is more marked than that of C-PTH.77 Cleavage of PTH (1–84) by liver cells generates C-PTH fragments that subsequently are removed from the circulation by the kidneys.83 Thus, impairment of renal function causes accumulation of C-PTH. The primary physiological actions of PTH are to increase calcium levels and to induce phosphaturia. PTH exerts its effects by binding the parathyroid hormone/parathyroid hormone–related peptide receptor (PTHR1). Major target organs of PTH include the bone and the kidney.84 PTH Resistance The response to PTH has been shown to be decreased and variable in uremic conditions.85–87 The underlying pathophysiology and causative agents remain incompletely understood.88 Suppressed expression of PTHR1 has been shown by several investigators both in uremic kidney tissue and bone,89–91 but data with regard to

P. Evenepoel, M. Rodriguez, and M. Ketteler

bone have not been unequivocal.92 A circulating factor is considered responsible for the decreased expression of the PTHR1 gene in uremia.93 In this regard, in vitro and animal studies point to the protein-bound uremic retention molecule IS as the potential culprit.94 IS and the other protein-bound uremic toxins are discussed in more detail in the article by Sirich et al of this issue.66 Also, PTH fragments may contribute to resistance to PTH, either through mechanisms involving receptor internalization and/or through mechanisms not involving the PTHR1.95,96 Large amino-terminal truncated C-PTH fragments may constitute up to 50% or more of the total PTH immunoreactivity in CKD. Administration of a four-fold molar excess of PTH 7-84 fragments approximately halved the phosphaturic effect of biointact PTH.97,98 In experimental animals, the administration of PTH (7-84) prevented the increase in serum calcium otherwise induced by PTH (1–84). The decrease in calcium serum levels induced by administration of PTH (7–84) may be owing to a decrease in osteoclast precursors, which express C-PTH receptors.99 Thus, PTH (7–84) can inhibit bone resorption, also in part by reducing the rate of formation of new osteoclasts. Furthermore, CKD is a state of inflammation and oxidative stress,100 and PTH modified by oxidative stress has no end organ activity.101 Finally, bone morphogenetic protein-7 produced in the kidney is a strong osteoblast differentiation factor, and low serum bone morphogenetic protein-7 levels have been reported in renal failure. It is speculated that low bone morphogenetic protein-7 levels may lead to a reduced number of osteoblasts and decreased PTH response.102 The clinical relevance of all these resistance factors remains to be shown. PTH as Uremic Toxin PTH has been implicated in the pathogenesis of several complications of CKD, including peripheral neuropathy, carbohydrate intolerance, hypertension, and dyslipidemia.103 PTH produces an increase in intracellular calcium104,105 in many cell types with the exception of VSMCs. The increase in cytosolic calcium levels seems to be caused by both an increased influx and a decreased efflux from the cell. High PTH levels inhibit mitochondrial respiration, reduce phosphorylation, and uncouple oxidative phosphorylation in isolated heart mitochondria, which causes adverse effects on the myocardium.106 An inhibitory effect of PTH on energy production and utilization also was observed in skeletal muscle.107 In hemodialysis patients, high PTH levels have been associated with abnormalities of left ventricular myocardial function and cardiac hypertrophy cardiac fibrosis.108–110 In addition, both high111 and low PTH112 levels may foster vascular calcification. Last but not least, PTH is the master regulator of bone remodeling. Bone disease

CKD-MBD abnormalities

is almost universal in CKD. Bone turnover, mineralization, and volume may be abnormal. Inappropriate PTH levels may cause high bone turnover disease, low bone turnover disease, and mixed bone disease. Osteitis fibrosa is observed in patients with high PTH levels; in that case, the bone turnover is abnormally high with evidence of marrow fibrosis. A portion of the calcified bone loses its lamellar structure and appears as woven bone. In patients with high PTH, it also is characteristic to observe an increased resorption of cortical bone. Osteomalacia and adynamic bone disease represent low bone turnover. In osteomalacia, the bone surface is covered with uncalcified osteoid, and cell activity (osteoblasts and osteoclasts) is decreased. Bone formation rate is reduced. Adynamic bone disease also is characterized by low bone turnover, but in this case osteoid is almost absent. Mixed bone disorder possesses characteristics of both high and low turnover.113 PTH level overall correlates poorly with bone turnover at least partly owing to skeletal resistance to the action of PTH being highly variable. Observational studies mostly showed U-shaped associations between PTH serum levels and mortality, with moderate relationships at the high end of PTH concentration, and strong effects at the low end.55 PTH as a Therapeutic Target The optimal PTH level in the individual patient remains a black box. Current guidelines suggest maintaining PTH levels in the broad range of approximately 2 to 9 times the upper normal limit.15 Calcimimetics have been proven to be potent drugs to control PTH levels. Two high-quality, prospective, placebocontrolled treatment studies with intermediate and hard end points in dialysis patients with secondary hyperparathyroidism were completed recently with this drug. In the ADVANCE (A randomizeD VAscular calcificatioN study to evaluate the effects of CinacalcEt) trial (n ¼ 360 hemodialysis patients), the effect of the calcimimetic cinacalcet on cardiovascular calcification progression was tested, with coronary artery calcification progression analyzed by Agatston score as the defined primary end point.114 This end point was not reached, but coronary artery calcification progression analyzed by volume score and aortic valve calcification progression, both predefined as secondary end points, were inhibited significantly by cinacalcet. The larger EVOLVE (EValuation Of Cinacalcet HCl Therapy to Lower CardioVascular Events) trial (n ¼ 3,883 hemodialysis patients) used the same intervention, but a different primary composite end point of time until death, myocardial infarction, hospitalization for unstable angina, heart failure, or a peripheral vascular event.115 This primary end point was not reached in the intention-to-treat analysis, however, predefined

157

adjustments for age or time on study drug (lag censoring at 6 months) showed a treatment benefit for cinacalcet. This study suffered from a disproportionally high drop-out rate of patients from the placebo arm, who subsequently were switched to cinacalcet. Clinical outcome studies related to uremia are discussed more extensively in by Liabeuf et al of this issue.116

VITAMIN D When discussing vitamin D in the context of CKDMBD, it is of outmost importance to distinguish between inactive (vitamin D2 and D3, calcidiol) and active (calcitriol and its synthetic analogues) compounds. Calcitriol is an important modulator of calcium and phosphate absorption in intestine, calcium re-absorption in kidney, and calcium mobilization in bone. In addition to maintaining calcium/phosphate homeostasis, calcitriol regulates cellular proliferation and differentiation of certain cells, modulates the immune response, and affects the cardiovascular system (Fig. 5). Calcitriol exerts its functions mainly by binding to a nuclear vitamin D receptor (VDR), a member of the steroid hormone receptor superfamily, leading to transcriptional regulation of target genes. The VDR is expressed in many tissues, but most prominently in the intestine, kidney, parathyroid gland, and bone.117 CKD goes along with low circulating calcitriol levels. This is not surprising because the kidney produces the bulk of this hormone. Mechanisms governing renal calcitriol metabolism in CKD, however, are complex and incompletely understood.118,119 Vitamin D metabolism is facilitated by hydroxylation, and recent evidence indicates that calcitriol synthesis by CYP27B1 and catabolism by CYP24A1, as well as tubular reabsorption of the precursor calcidiol, all may be disturbed in CKD.120,121 Extrarenal vitamin D metabolism has VITAMIN D EFFECTS AND VASCULAR CALCIFICATION Calcium and Phosphate.

PTH.

Bone metabolism / turnover Cell proliferaon and differenaon Macrophage funcon Immune modulaon

Protects against Inflammaon

VASCULAR CALCIFICATION

Insulin producon Renin producon

Figure 5. Active vitamin D analogues such as calcitriol play an essential role in the regulation of many cell functions in the body. Bone turnover is regulated by vitamin D. Vitamin D also regulates proliferation and differentiation of various cell types as well as certain immune responses. Some of these vitamin D effects such as an increase in calcium and phosphate will favor vascular calcifications whereas other functions such as modulation of an inflammatory response may prevent calcification.

158

P. Evenepoel, M. Rodriguez, and M. Ketteler

gained much interest in recent years. The existence of CYP27B1 has been documented in many tissues and cells including the vasculature, placenta, and monocytes. Data on the impact of CKD on extrarenal vitamin D metabolism are limited and mainly relate to monocytes. Available evidence points to impaired uptake of calcidiol and a blunted immune signal-induced CYP27B1 expression, the latter probably mediated by high FGF23 levels.122–124 Calcitriol Resistance CKD also is characterized by a state of calcitriol resistance.125 Vitamin D hyporesponsiveness may be explained by decreased expression of the VDR or by a decreased binding affinity of VDR with vitamin D– responsive elements.126,127 Responsible culprits remain to be identified, but most probably also involve uremic toxins.125 Importantly, vitamin D hyporesponsiveness may occur early in the course of CKD and explain why low intestinal calcium absorption and high PTH levels precede low calcitriol levels.128,129 Vitamin D Deficiency as a Uremic Toxin Calcidiol and calcitriol deficiency are frequent in CKD, and both have been associated with poor outcomes.130–133 The underlying mechanisms are complex and ill-defined.

Vitamin D as a Therapeutic Target Cholecalciferol or ergocalciferol supplementation, in doses to correct calcidiol deficiency, seem to be harmless and without any side effects, however, there are no randomized controlled data yet about its benefits. There is, however, great ambiguity with regard to active vitamin D (analogues). On the one hand, a significant body of observational trials quite consistently suggests that active vitamin D treatment is associated with improved outcomes, mostly independent of other parameters of CKD-MBD and possibly related to some pleiotropic effect of the hormone.20,134 On the other hand, active vitamin D analogues increase intestinal calcium absorption and may—in pharmacologic doses—oversuppress bone turnover, thus creating a risk scenario for cardiovascular calcification. No prospective trials are currently available that systematically evaluate the role of active vitamin D analogues on hard outcomes. One randomized controlled study addressed the effect of paricalcitol on the surrogate end point LVH in CKD stages 3 and 4 patients, but showed no benefit versus the placebo arm.135 Another recent randomized controlled study addressed the effects of paricalcitol on albuminuria in diabetic CKD stages 3 and 4 patients and found significant reductions with a daily dose of 2 mcg of the compound.136 Preclinical studies, however, may provide some helpful advice. Mizobuchi et al137 studied 5/6nephrectomized rats and showed that calcitriol and

Figure 6. Active vitamin D as a uremic toxin. Experimental data by Mathew et al139 suggest that active vitamin D analogues in low (physiological?) doses may protect against vascular calcification by inhibition of osteogenic transdifferentiation, whereas they may induce this process at high (supraphysiological/pharmacologic?) doses.

CKD-MBD abnormalities

doxercalciferol, but not paricalcitol, induced media calcification, despite similar effects on PTH levels. The suggested difference was that the former two compounds induced osteochondrocytic differentiation in the vessel wall, but paricalcitol did not. Lopez et al138 used the same model, but compared calcitriol, paricalcitol, a calcimimetic (AMG-641), and combinations of these compounds. Treatment with the calcimimetic did not induce any cardiovascular calcification, whereas calcitriol and paricalcitol induced severe and moderate, respectively, media calcification. Those calcifications were ameliorated by co-treatment with the calcimimetic. A key article in this context was published by Mathew et al,139 who treated uremic lowdensity lipoprotein–receptor knockout mice with different doses of calcitriol or paricalcitol (Fig. 6). This model represents a combined phenotype of atherosclerosis, CKD, and metabolic syndrome. Moderate doses of both vitamin D analogues blunted plaque calcification, whereas high doses caused significant calcification progression. In summary, the effects of exogenous calcitriol and its synthetic active analogues are dose-dependent, with adverse effects on the high (pharmacologic) end, and possible benefits on the low (physiological) end of the dose spectrum. The modality of a hormone replacement therapy thus may be a feasible description of the optimal use of these compounds as steroid hormones. However, as with many of the other CKD-MBD–related parameters and therapeutics, there is no randomized controlled proof for these assumptions.

CONCLUSIONS Laboratory parameters of CKD-MBD are markers of disease severity and predictors of outcome. Circulating levels of PTH, FGF23, and calcitriol are tightly regulated by several feedback mechanisms to maintain bone and mineral homeostasis both in health and disease. All these hormones exert their biological effects by binding their cognate receptors in target organs. One should acknowledge that target organs in CKD may be resistant to the action of these hormones as a consequence of impaired binding to the receptor, decreased expression of the receptor, or postreceptor disturbances. Thus, increasing hormone levels are to some extent required in CKD to overcome end-organ resistance to their action. Hitherto, it has been very difficult, if not impossible, at least on the individual level, to define when the increase turns maladaptive and the hormone becomes a uremic toxin. At present, assessment of the end-organ response (eg, by bone histomorphometry) may be the only means to judge the appropriateness of the PTH exposure in the individual patient accurately. Hyperphosphatemia occurs late in

159

the course of CKD, when compensatory mechanisms start to fail. Despite overwhelming experimental and clinical observational evidence linking high phosphate levels to poor outcomes, randomized controlled intervention trials with hard end points have not been conducted to date. Several aspects of interventions targeting high phosphate levels remain vague. Although several lines of evidence indicate that calcium loading may be harmful, uncertainties also remain with regard to calcium balance in CKD, especially during prolonged loading and with regard to the fate of retained calcium. Current evidence, however, dictates that calcium, either as a phosphate binder or as a supplement, should be prescribed with caution, especially if adynamic bone disease or vascular calcification is present. During the past 2 decades it has become clear that laboratory parameters of CKDMBD eventually may turn into direct uremic toxins, mediating renal, cardiovascular, and/or bone disease. Additional research is required urgently to prove the benefit of therapeutic approaches targeting these laboratory parameters.

REFERENCES 1. Renkema KY, Alexander RT, Bindels RJ, Hoenderop JG. Calcium and phosphate homeostasis: concerted interplay of new regulators. Ann Med. 2008;40:82-91. 2. Isakova T, Wahl P, Vargas GS, Gutierrez OM, Scialla J, Xie H, et al. Fibroblast growth factor 23 is elevated before parathyroid hormone and phosphate in chronic kidney disease. Kidney Int. 2011;79:1370-8. 3. Evenepoel P, Meijers B, Viaene L, Bammens B, Claes K, Kuypers D, et al. Fibroblast growth factor-23 in early chronic kidney disease: additional support in favor of a phosphatecentric paradigm for the pathogenesis of secondary hyperparathyroidism. Clin J Am Soc Nephrol. 2010;5:1268-76. 4. Wolf M. Update on fibroblast growth factor 23 in chronic kidney disease. Kidney Int. 2012;82:737-47. 5. Silver J, Rodriguez M, Slatopolsky E. FGF23 and PTH– double agents at the heart of CKD. Nephrol Dial Transplant. 2012;27:1715-20. 6. Pavik I, Jaeger P, Ebner L, Wagner CA, Petzold K, Spichtig D, et al. Secreted Klotho and FGF23 in chronic kidney disease stage 1 to 5: a sequence suggested from a cross-sectional study. Nephrol Dial Transplant. 2013;28:352-9. 7. Slatopolsky E, Caglar S, Gradowska L, Canterbury J, Reiss E, Bricker NS. On the prevention of secondary hyperparathyroidism in experimental chronic renal disease using “proportional reduction” of dietary phosphorus intake. Kidney Int. 1972;2:147-51. 8. Moe S, Drueke T, Cunningham J, Goodman W, Martin K, Olgaard K, et al. Definition, evaluation, and classification of renal osteodystrophy: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int. 2006;69:1945-53. 9. Hamdy NA, Kanis JA, Beneton MN, Juttmann JR, Jordans JG, Josse S, et al. Effect of alfacalcidol on natural course of renal bone disease in mild to moderate renal failure. Br Med J.. 1995;310:358-63. 10. Coen G, Ballanti P, Bonucci E, Calabria S, Costantini S, Ferrannini M, et al. Renal osteodystrophy in predialysis and hemodialysis patients: comparison of histologic patterns and

160

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

22. 23.

24.

25.

26.

P. Evenepoel, M. Rodriguez, and M. Ketteler diagnostic predictivity of intact PTH. Nephron. 2002;91: 103-11. Malluche HH, Mawad HW, Monier-Faugere MC. Renal osteodystrophy in the first decade of the new millennium: analysis of 630 bone biopsies in black and white patients. J Bone Miner Res. 2011;26:1368-76. Malluche HH, Mawad H, Monier-Faugere MC. The importance of bone health in end-stage renal disease: out of the frying pan, into the fire? Nephrol Dial Transplant. 2004;19 (Suppl 1):i9-13. Budoff MJ, Rader DJ, Reilly MP, Mohler ER III, Lash J, Yang W, et al. Relationship of estimated GFR and coronary artery calcification in the CRIC (Chronic Renal Insufficiency Cohort) study. Am J Kidney Dis. 2011;58:519-26. Toussaint ND, Lau KK, Strauss BJ, Polkinghorne KR, Kerr PG. Associations between vascular calcification, arterial stiffness and bone mineral density in chronic kidney disease. Nephrol Dial Transplant. 2008;23:586-93. Kidney Disease: Improving Global Outcomes (KDIGO) CKD-MBD Work Group. KDIGO clinical practice guideline for the diagnosis, evaluation, prevention, and treatment of chronic kidney disease-mineral and bone disorder (CKDMBD). Kidney Int Suppl. 2009;113:S1-130. Hill KM, Martin BR, Wastney ME, McCabe GP, Moe SM, Weaver CM, et al. Oral calcium carbonate affects calcium but not phosphorus balance in stage 3-4 chronic kidney disease. Kidney Int. 2013;83:959-66. Evenepoel P, Wolf M. A balanced view of calcium and phosphate homeostasis in chronic kidney disease. Kidney Int. 2013;83:789-91. Block G, Hulbert-Shearon T, Levin NW, Port F. Association of serum phosphorus and calcium x phosphate product with mortality risk in chronic hemodialysis patients: a national study. Am J Kidney Dis. 1998;31:607-17. Block GA, Klassen PS, Lazarus JM, Ofsthun N, Lowrie EG, Chertow GM. Mineral metabolism, mortality, and morbidity in maintenance hemodialysis. J Am Soc Nephrol. 2004;15: 2208-18. Kalantar-Zadeh K, Kuwae N, Regidor DL, Kovesdy CP, Kilpatrick RD, Shinaberger CS, et al. Survival predictability of time-varying indicators of bone disease in maintenance hemodialysis patients. Kidney Int. 2006;70:771-80. Jono S, McKee MD, Murry CE, Shioi A, Nishizawa Y, Mori K, et al. Phosphate regulation of vascular smooth muscle cell calcification. Circ Res. 2000;87:10e-17. Giachelli CM. The emerging role of phosphate in vascular calcification. Kidney Int. 2009;75:890-7. Villa-Bellosta R, Sorribas V. Phosphonoformic acid prevents vascular smooth muscle cell calcification by inhibiting calcium-phosphate deposition. Arterioscler Thromb Vasc Biol. 2009;29:761-6. Zhao MM, Xu MJ, Cai Y, Zhao G, Guan Y, Kong W, et al. Mitochondrial reactive oxygen species promote p65 nuclear translocation mediating high-phosphate-induced vascular calcification in vitro and in vivo. Kidney Int. 2011;79:1071-9. Reynolds JL, Joannides AJ, Skepper JN, McNair R, Schurgers LJ, Proudfoot D, et al. Human vascular smooth muscle cells undergo vesicle-mediated calcification in response to changes in extracellular calcium and phosphate concentrations: a potential mechanism for accelerated vascular calcification in ESRD. J Am Soc Nephrol. 2004;15:2857-67. Ewence AE, Bootman M, Roderick HL, Skepper JN, McCarthy G, Epple M, et al. Calcium phosphate crystals induce cell death in human vascular smooth muscle cells: a potential mechanism in atherosclerotic plaque destabilization. Circ Res. 2008;103:e28-34.

27. Sage AP, Lu J, Tintut Y, Demer LL. Hyperphosphatemiainduced nanocrystals upregulate the expression of bone morphogenetic protein-2 and osteopontin genes in mouse smooth muscle cells in vitro. Kidney Int. 2011;79:414-22. 28. Lau WL, Pai A, Moe SM, Giachelli CM. Direct effects of phosphate on vascular cell function. Adv Chronic Kidney Dis. 2011;18:105-12. 29. Di Marco GS, Konig M, Stock C, Wiesinger A, Hillebrand U, Reiermann S, et al. High phosphate directly affects endothelial function by downregulating annexin II. Kidney Int. 2013;83:213-22. 30. Kuro O. Klotho, phosphate and FGF-23 in ageing and disturbed mineral metabolism. Nat Rev Nephrol. 2013;9: 650-60. 31. Zoccali C, Ruggenenti P, Perna A, Leonardis D, Tripepi R, Tripepi G, et al. Phosphate may promote CKD progression and attenuate renoprotective effect of ACE inhibition. J Am Soc Nephrol. 2011;22:1923-30. 32. Bellasi A, Mandreoli M, Baldrati L, Corradini M, Di NP, Malmusi G, et al. Chronic kidney disease progression and outcome according to serum phosphorus in mild-to-moderate kidney dysfunction. Clin J Am Soc Nephrol. 2011;6:883-91. 33. Chue CD, Edwards NC, Davis LJ, Steeds RP, Townend JN, Ferro CJ. Serum phosphate but not pulse wave velocity predicts decline in renal function in patients with early chronic kidney disease. Nephrol Dial Transplant. 2011;26: 2576-82. 34. Scialla JJ, Astor BC, Isakova T, Xie H, Appel LJ, Wolf M. Mineral metabolites and CKD progression in African Americans. J Am Soc Nephrol. 2013;24:125-35. 35. Lezaic V, Tirmenstajn-Jankovic B, Bukvic D, Vujisic B, Perovic M, Novakovic N, et al. Efficacy of hyperphosphatemia control in the progression of chronic renal failure and the prevalence of cardiovascular calcification. Clin Nephrol. 2009;71:21-9. 36. Schwarz S, Trivedi BK, Kalantar-Zadeh K, Kovesdy CP. Association of disorders in mineral metabolism with progression of chronic kidney disease. Clin J Am Soc Nephrol. 2006;1:825-31. 37. Fliser D, Kollerits B, Neyer U, Ankerst DP, Lhotta K, Lingenhel A, et al. Fibroblast growth factor 23 (FGF23) predicts progression of chronic kidney disease: the mild to moderate kidney disease (MMKD) study. J Am Soc Nephrol. 2007;18:2601-8. 38. Lau K. Phosphate excess and progressive renal failure: the precipitation-calcification hypothesis. Kidney Int. 1989;36: 918-37. 39. Sayer JA, Carr G, Simmons NL. Nephrocalcinosis: molecular insights into calcium precipitation within the kidney. Clin Sci. 2004;106:549-61. 40. Brown SA, Crowell WA, Barsanti JA, White JV, Finco DR. Beneficial effects of dietary mineral restriction in dogs with marked reduction of functional renal mass. J Am Soc Nephrol. 1991;1:1169-79. 41. Lumlertgul D, Burke TJ, Gillum DM, Alfrey AC, Harris DC, Hammond WS, et al. Phosphate depletion arrests progression of chronic renal failure independent of protein intake. Kidney Int. 1986;29:658-66. 42. Cozzolino M, Dusso AS, Liapis H, Finch J, Lu Y, Burke SK, et al. The effects of sevelamer hydrochloride and calcium carbonate on kidney calcification in uremic rats. J Am Soc Nephrol. 2002;13:2299-308. 43. Cozzolino M, Staniforth ME, Liapis H, Finch J, Burke SK, Dusso AS, et al. Sevelamer hydrochloride attenuates kidney and cardiovascular calcifications in long-term experimental uremia. Kidney Int. 2003;64:1653-61.

CKD-MBD abnormalities 44. Haut LL, Alfrey AC, Guggenheim S, Buddington B, Schrier N. Renal toxicity of phosphate in rats. Kidney Int. 1980;17: 722-31. 45. Kalantar-Zadeh K, Gutekunst L, Mehrotra R, Kovesdy CP, Bross R, Shinaberger CS, et al. Understanding sources of dietary phosphorus in the treatment of patients with chronic kidney disease. Clin J Am Soc Nephrol. 2010;5:519-30. 46. Uribarri J. Phosphorus additives in food and their effect in dialysis patients. Clin J Am Soc Nephrol. 2009;4:1290-2. 47. Sullivan C, Sayre SS, Leon JB, Machekano R, Love TE, Porter D, et al. Effect of food additives on hyperphosphatemia among patients with end-stage renal disease: a randomized controlled trial. JAMA. 2009;301:629-35. 48. Tonelli M, Pannu N, Manns B. Oral phosphate binders in patients with kidney failure. N Engl J Med. 2010;362: 1312-24. 49. Isakova T, Gutierrez OM, Chang Y, Shah A, Tamez H, Smith K, et al. Phosphorus binders and survival on hemodialysis. J Am Soc Nephrol. 2009;20:388-96. 50. Block GA, Wheeler DC, Persky MS, Kestenbaum B, Ketteler M, Spiegel DM, et al. Effects of phosphate binders in moderate CKD. J Am Soc Nephrol. 2012;23:1407-15. 51. Seifert ME, de Las FL, Rothstein M, Dietzen DJ, Bierhals AJ, Cheng SC, et al. Effects of phosphate binder therapy on vascular stiffness in early-stage chronic kidney disease. Am J Nephrol. 2013;38:158-67. 52. Houillier P, Froissart M, Maruani G, Blanchard A. What serum calcium can tell us and what it can’t. Nephrol Dial Transplant. 2006;21:29-32. 53. Spiegel DM, Brady K. Calcium balance in normal individuals and in patients with chronic kidney disease on low- and highcalcium diets. Kidney Int. 2012;81:1116-22. 54. Evenepoel P, Viaene L, Meijers B. Calcium balance in chronic kidney disease: walking the tightrope. Kidney Int. 2012;81: 1057-9. 55. Floege J, Kim J, Ireland E, Chazot C, Drueke T, de Francisco A, et al. Serum iPTH, calcium and phosphate, and the risk of mortality in a European haemodialysis population. Nephrol Dial Transplant. 2011;26:1948-55. 56. Goodman WG, Goldin J, Kuizon BD, Yoon C, Gales B, Sider D, et al. Coronary-artery calcification in young adults with end-stage renal disease who are undergoing dialysis. N Engl J Med. 2000;342:1478-83. 57. Shroff RC, McNair R, Skepper JN, Figg N, Schurgers LJ, Deanfield J, et al. Chronic mineral dysregulation promotes vascular smooth muscle cell adaptation and extracellular matrix calcification. J Am Soc Nephrol. 2010;21:103-12. 58. Jamal SA, Vandermeer B, Raggi P, Mendelssohn DC, Chatterley T, Dorgan M, et al. Effect of calcium-based versus non-calcium-based phosphate binders on mortality in patients with chronic kidney disease: an updated systematic review and meta-analysis. Lancet. 2013;382:1268-77. 59. Cannata-Andia JB, Fernandez-Martin JL, Locatelli F, London G, Gorriz JL, Floege J, et al. Use of phosphate-binding agents is associated with a lower risk of mortality. Kidney Int. 2013;84:998-1008. 60. Koh N, Fujimori T, Nishiguchi S, Tamori A, Shiomi S, Nakatani T, et al. Severely reduced production of klotho in human chronic renal failure kidney. Biochem Biophys Res Commun. 2001;280:1015-20. 61. Hu MC, Shi M, Zhang J, Quiñones H, Griffith C, Kuro-o M, et al. Klotho deficiency causes vascular calcification in chronic kidney disease. J Am Soc Nephrol. 2011;22:124-36. 62. Komaba H, Goto S, Fujii H, Hamada Y, Kobayashi A, Shibuya K, et al. Depressed expression of Klotho and FGF receptor 1 in hyperplastic parathyroid glands from uremic patients. Kidney Int. 2010;77:232-8.

161 63. Kuro-o M. Klotho in health and disease. Curr Opin Nephrol Hypertens. 2012;21:362-8. 64. Shimizu H, Bolati D, Adijiang A, Adelibieke Y, Muteliefu G, Enomoto A, et al. Indoxyl sulfate downregulates renal expression of Klotho through production of ROS and activation of nuclear factor-kB. Am J Nephrol. 2011;33:319-24. 65. Sun CY, Chang SC, Wu MS. Suppression of Klotho expression by protein-bound uremic toxins is associated with increased DNA methyltransferase expression and DNA hypermethylation. Kidney Int. 2012;81:640-50. 66. Sirich TL, Meyer TW, Gondouin G, Brunet P, Niwa T. Protein-bound molecules: a large family with a bad character. Semin Nephrol. 2014;34:106-17. 67. Goetz R, Nakada Y, Hu MC, Kurosu H, Wang L, Nakatani T, et al. Isolated C-terminal tail of FGF23 alleviates hypophosphatemia by inhibiting FGF23-FGFR-Klotho complex formation. Proc Natl Acad Sci U S A. 2010;107:407-12. 68. Smith ER, Cai MM, McMahon LP, Holt SG. Biological variability of plasma intact and C-terminal FGF23 measurements. J Clin Endocrinol Metab. 2012;97:3357-65. 69. Shimada T, Urakawa I, Isakova T, Yamazaki Y, Epstein M, Wesseling-Perry K, et al. Circulating fibroblast growth factor 23 in patients with end-stage renal disease treated by peritoneal dialysis is intact and biologically active. J Clin Endocrinol Metab. 2010;95:578-85. 70. Wolf M, Molnar MZ, Amaral AP, Czira ME, Rudas A, Ujszaszi A, et al. Elevated fibroblast growth factor 23 is a risk factor for kidney transplant loss and mortality. J Am Soc Nephrol. 2011;22:956-66. 71. Isakova T, Xie H, Yang W, Xie D, Anderson AH, Scialla J, et al. Fibroblast growth factor 23 and risks of mortality and end-stage renal disease in patients with chronic kidney disease. JAMA. 2011;305:2432-9. 72. Gutierrez OM, Mannstadt M, Isakova T, Rauh-Hain JA, Tamez H, Shah A, et al. Fibroblast growth factor 23 and mortality among patients undergoing hemodialysis. N Engl J Med. 2008;359:584-92. 73. Jean G, Terrat JC, Vanel T, Hurot JM, Lorriaux C, Mayor B, et al. High levels of serum fibroblast growth factor (FGF)-23 are associated with increased mortality in long haemodialysis patients. Nephrol Dial Transplant. 2009;24:2792-6. 74. Parker BD, Schurgers LJ, Brandenburg VM, Christenson RH, Vermeer C, Ketteler M, et al. The associations of fibroblast growth factor 23 and uncarboxylated matrix Gla protein with mortality in coronary artery disease: the Heart and Soul Study. Ann Intern Med. 2010;152:640-8. 75. Lim K, Lu TS, Molostvov G, Lee C, Lam F, Zehnder D, et al. Vascular klotho deficiency potentiates the development of human artery calcification and mediates resistance to FGF-23. Circulation. 2012;125:2243-55. 76. Semba RD, Cappola AR, Sun K, Bandinelli S, Dalal M, Crasto C, et al. Plasma klotho and mortality risk in older community-dwelling adults. J Gerontol A Biol Sci Med Sci. 2011;66:794-800. 77. Seiler S, Wen M, Roth HJ, Fehrenz M, Flugge F, Herath E, et al. Plasma Klotho is not related to kidney function and does not predict adverse outcome in patients with chronic kidney disease. Kidney Int. 2013;83:121-8. 78. Gutierrez OM, Januzzi JL, Isakova T, Laliberte K, Smith K, Collerone G, et al. Fibroblast growth factor 23 and left ventricular hypertrophy in chronic kidney disease. Circulation. 2009;119:2545-52. 79. Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, et al. FGF23 induces left ventricular hypertrophy. J Clin Invest. 2011;121:4393-408. 80. Seiler S, Cremers B, Rebling NM, Hornof F, Jeken J, Kersting S, et al. The phosphatonin fibroblast growth factor 23 links

162

81.

82.

83.

84.

85.

86.

87.

88.

89.

90.

91.

92.

93.

94.

95.

96.

P. Evenepoel, M. Rodriguez, and M. Ketteler calcium-phosphate metabolism with left-ventricular dysfunction and atrial fibrillation. Eur Heart J. 2011;32:2688-96. Shalhoub V, Shatzen EM, Ward SC, Davis J, Stevens J, Bi V, et al. FGF23 neutralization improves chronic kidney diseaseassociated hyperparathyroidism yet increases mortality. J Clin Invest. 2012;122:2543-53. Santamaria R, Almaden Y, Felsenfeld A, Martin-Malo A, Gao P, Cantor T, et al. Dynamics of PTH secretion in hemodialysis patients as determined by the intact and whole PTH assays. Kidney Int. 2003;64:1867-73. D’Amour P, Lazure C. Metabolism of radioiodinated carboxyterminal fragments of bovine parathyroid hormone in normal and anephric rats. Endocrinology. 1985;117:127-34. Mannstadt M, Juppner H, Gardella TJ. Receptors for PTH and PTHrP: their biological importance and functional properties. Am J Physiol. 1999;277:F665-75. Berdud I, Martin-Malo A, Almaden Y, Tallon S, Concepcion MT, Torres A, et al. Abnormal calcaemic response to PTH in the uraemic rat without secondary hyperparathyroidism. Nephrol Dial Transplant. 1996;11:1292-8. Wesseling-Perry K, Harkins GC, Wang HJ, Elashoff R, Gales B, Horwitz MJ, et al. The calcemic response to continuous parathyroid hormone (PTH)(1-34) infusion in end-stage kidney disease varies according to bone turnover: a potential role for PTH(7-84). J Clin Endocrinol Metab. 2010;95:2772-80. Massry SG, Coburn JW, Lee DB, Jowsey J, Kleeman CR. Skeletal resistance to parathyroid hormone in renal failure. Studies in 105 human subjects. Ann Intern Med. 1973;78: 357-64. Iwasaki Y, Yamato H, Nii-Kono T, Fujieda A, Uchida M, Hosokawa A, et al. Insufficiency of PTH action on bone in uremia. Kidney Int Suppl. 2006;102:S34-6. Urena P, Mannstadt M, Hruby M, Ferreira A, Schmitt F, Silve C, et al. Parathyroidectomy does not prevent the renal PTH/ PTHrP receptor down-regulation in uremic rats. Kidney Int. 1995;47:1797-805. Picton ML, Moore PR, Mawer EB, Houghton D, Freemont AJ, Hutchison AJ, et al. Down-regulation of human osteoblast PTH/PTHrP receptor mRNA in end-stage renal failure. Kidney Int. 2000;58:1440-9. Iwasaki-Ishizuka Y, Yamato H, Nii-Kono T, Kurokawa K, Fukagawa M. Downregulation of parathyroid hormone receptor gene expression and osteoblastic dysfunction associated with skeletal resistance to parathyroid hormone in a rat model of renal failure with low turnover bone. Nephrol Dial Transplant. 2005;20:1904-11. Langub MC, Monier-Faugere MC, Qi Q, Geng Z, Koszewski NJ, Malluche HH. Parathyroid hormone/parathyroid hormonerelated peptide type 1 receptor in human bone. J Bone Miner Res. 2001;16:448-56. Disthabanchong S, Hassan H, McConkey CL, Martin KJ, Gonzalez EA. Regulation of PTH1 receptor expression by uremic ultrafiltrate in UMR 106-01 osteoblast-like cells. Kidney Int. 2004;65:897-903. Nii-Kono T, Iwasaki Y, Uchida M, Fujieda A, Hosokawa A, Motojima M, et al. Indoxyl sulfate induces skeletal resistance to parathyroid hormone in cultured osteoblastic cells. Kidney Int. 2007;71:738-43. Sneddon WB, Magyar CE, Willick GE, Syme CA, Galbiati F, Bisello A, et al. Ligand-selective dissociation of activation and internalization of the parathyroid hormone (PTH) receptor: conditional efficacy of PTH peptide fragments. Endocrinology. 2004;145:2815-23. Divieti P, John MR, Juppner H, Bringhurst FR. Human PTH(7-84) inhibits bone resorption in vitro via actions independent of the type 1 PTH/PTHrP receptor. Endocrinology. 2002;143: 171-6.

97. Slatopolsky E, Finch J, Clay P, Martin D, Sicard G, Singer G, et al. A novel mechanism for skeletal resistance in uremia. Kidney Int. 2000;58:753-61. 98. Nguyen-Yamamoto L, Rousseau L, Brossard JH, Lepage R, D’Amour P. Synthetic carboxyl-terminal fragments of parathyroid hormone (PTH) decrease ionized calcium concentration in rats by acting on a receptor different from the PTH/ PTH-related peptide receptor. Endocrinology. 2001;142: 1386-92. 99. Liu BY, Guo J, Lanske B, Divieti P, Kronenberg HM, Bringhurst FR. Conditionally immortalized murine bone marrow stromal cells mediate parathyroid hormonedependent osteoclastogenesis in vitro. Endocrinology. 1998;139:1952-64. 100. Himmelfarb J. Uremic toxicity, oxidative stress, and hemodialysis as renal replacement therapy. Semin Dial. 2009;22: 636-43. 101. Hocher B, Armbruster FP, Stoeva S, Reichetzeder C, Gron HJ, Lieker I, et al. Measuring parathyroid hormone (PTH) in patients with oxidative stress–do we need a fourth generation parathyroid hormone assay? PLoS One. 2012;7:e40242. 102. Lund RJ, Davies MR, Brown AJ, Hruska KA. Successful treatment of an adynamic bone disorder with bone morphogenetic protein-7 in a renal ablation model. J Am Soc Nephrol. 2004;15:359-69. 103. Klahr S, Slatopolsky E. Toxicity of parathyroid hormone in uremia. Annu Rev Med. 1986;37:71-8. 104. Slatopolsky E, Martin K, Hruska K. Parathyroid hormone metabolism and its potential as a uremic toxin. Am J Physiol. 1980;239:F1-12. 105. Massry SG, Smogorzewski M. The mechanisms responsible for the PTH-induced rise in cytosolic calcium in various cells are not uniform. Miner Electrolyte Metab. 1995;21:13-28. 106. Bogin E, Levi J, Harary I, Massry SG. Effects of parathyroid hormone on oxidative phosphorylation of heart mitochondria. Miner Electrolyte Metab. 1982;7:151-6. 107. Baczynski R, Massry SG, Kohan R, Magott M, Saglikes Y, Brautbar N. Effect of parathyroid hormone on myocardial energy metabolism in the rat. Kidney Int. 1985;27: 718-25. 108. Drueke T, Fauchet M, Fleury J, Lesourd P, Toure Y, Le PC, et al. Effect of parathyroidectomy on left-ventricular function in haemodialysis patients. Lancet. 1980;1:112-4. 109. London GM, de Vernejoul MC, Fabiani F, Marchais SJ, Guerin AP, Metivier F, et al. Secondary hyperparathyroidism and cardiac hypertrophy in hemodialysis patients. Kidney Int. 1987;32:900-7. 110. Amann K, Ritz E, Wiest G, Klaus G, Mall G. A role of parathyroid hormone for the activation of cardiac fibroblasts in uremia. J Am Soc Nephrol. 1994;4:1814-9. 111. Neves KR, Graciolli FG, dos Reis LM, Graciolli RG, Neves CL, Magalhaes AO, et al. Vascular calcification: contribution of parathyroid hormone in renal failure. Kidney Int. 2007;71: 1262-70. 112. Shao JS, Cheng SL, Pingsterhaus JM, Charlton-Kachigian N, Loewy AP, Towler DA. Msx2 promotes cardiovascular calcification by activating paracrine Wnt signals. J Clin Invest. 2005;115:1210-20. 113. Sherrard DJ, Hercz G, Pei Y, Maloney NA, Greenwood C, Manuel A, et al. The spectrum of bone disease in end-stage renal failure–an evolving disorder. Kidney Int. 1993;43: 436-42. 114. Raggi P, Chertow GM, Torres PU, Csiky B, Naso A, Nossuli K, et al. The ADVANCE study: a randomized study to evaluate the effects of cinacalcet plus low-dose vitamin D on vascular calcification in patients on hemodialysis. Nephrol Dial Transplant. 2011;26:1327-39.

CKD-MBD abnormalities 115. Chertow GM, Block GA, Correa-Rotter R, Drueke TB, Floege J, Goodman WG, et al. Effect of cinacalcet on cardiovascular disease in patients undergoing dialysis. N Engl J Med. 2012;367:2482-94. 116. Liabeuf S, Neirynck N, Drueke T, Vanholder R, Massy Z. Clinical studies and chronic kidney disease: what did we learn recently? Sem Nephrol. 2014 117. Wang Y, Zhu J, DeLuca HF. Where is the vitamin D receptor? Arch Biochem Biophys. 2012;523:123-33. 118. Dusso AS, Rodriguez M. Enhanced induction of Cyp24a1 by FGF23 but low serum 24,25-dihydroxyvitamin D in CKD: implications for therapy. Kidney Int. 2012;82:1046-9. 119. Helvig CF, Cuerrier D, Hosfield CM, Ireland B, Kharebov AZ, Kim JW, et al. Dysregulation of renal vitamin D metabolism in the uremic rat. Kidney Int. 2010;78:463-72. 120. Dai B, David V, Alshayeb HM, Showkat A, Gyamlani G, Horst RL, et al. Assessment of 24,25(OH)2D levels does not support FGF23-mediated catabolism of vitamin D metabolites. Kidney Int. 2012;82:1061-70. 121. Bosworth CR, Levin G, Robinson-Cohen C, Hoofnagle AN, Ruzinski J, Young B, et al. The serum 24,25-dihydroxyvitamin D concentration, a marker of vitamin D catabolism, is reduced in chronic kidney disease. Kidney Int. 2012;82: 693-700. 122. Gallieni M, Kamimura S, Ahmed A, Bravo E, Delmez J, Slatopolsky E, et al. Kinetics of monocyte 1 alphahydroxylase in renal failure. Am J Physiol Renal Physiol. 1995;268:F746-53. 123. Bacchetta J, Sea JL, Chun RF, Lisse TS, Wesseling-Perry K, Gales B, et al. Fibroblast growth factor 23 inhibits extrarenal synthesis of 1,25-dihydroxyvitamin D in human monocytes. J Bone Miner Res. 2013;28:46-55. 124. Viaene L, Evenepoel P, Meijers B, Vanderschueren D, Overbergh L, Mathieu C. Uremia suppresses immune signalinduced CYP27B1 expression in human monocytes. Am J Nephrol. 2012;36:497-508. 125. Patel SR, Ke H-Q, Vanholder R, Koenig RJ, Hsu CH. Inhibition of calcitriol receptor binding to vitamin D response elements by uremic toxins. J Clin Invest. 1995;96:50-9. 126. Vanholder R, Glorieux G. Molecular mechanisms of vitamin D hyporesponsiveness in renal failure. Nephrol Dial Transplant. 1999;14:16-9. 127. Sawaya BP, Koszewski NJ, Qi Q, Langub MC, MonierFaugere MC, Malluche HH. Secondary hyperparathyroidism and vitamin D receptor binding to vitamin D response

163

128.

129.

130.

131.

132.

133.

134.

135.

136.

137.

138.

139.

elements in rats with incipient renal failure. J Am Soc Nephrol. 1997;8:271-8. Cremer B, Lubbers E, Klooker P, Schmidt-Gayk H, Ritz E. Calciuric response to 1,25-(OH)2D3 in early renal failure. Miner Electrolyte Metab. 1985;11:182-5. Viaene L, Meijers BK, Vanrenterghem Y, Evenepoel P. Evidence in favor of a severely impaired net intestinal calcium absorption in patients with (early-stage) chronic kidney disease. Am J Nephrol. 2012;35:434-41. Mehrotra R, Kermah D, Budoff M, Salusky IB, Mao SS, Gao YL, et al. Hypovitaminosis D in chronic kidney disease. Clin J Am Soc Nephrol. 2008;3:1144-51. Ravani P, Malberti F, Tripepi G, Pecchini P, Cutrupi S, Pizzini P, et al. Vitamin D levels and patient outcome in chronic kidney disease. Kidney Int. 2009;75:88-95. Drechsler C, Verduijn M, Pilz S, Dekker FW, Krediet RT, Ritz E, et al. Vitamin D status and clinical outcomes in incident dialysis patients: results from the NECOSAD study. Nephrol Dial Transplant. 2011;26:1024-32. Wolf M, Shah A, Gutierrez O, Ankers E, Monroy M, Tamez H, et al. Vitamin D levels and early mortality among incident hemodialysis patients. Kidney Int. 2007;72:1004-13. Teng M, Wolf M, Ofsthun MN, Lazarus JM, Hernan MA, Camargo CA Jr, et al. Activated injectable vitamin D and hemodialysis survival: a historical cohort study. J Am Soc Nephrol. 2005;16:1115-25. Tamez H, Zoccali C, Packham D, Wenger J, Bhan I, Appelbaum E, et al. Vitamin D reduces left atrial volume in patients with left ventricular hypertrophy and chronic kidney disease. Am Heart J. 2012;164:902-9. de Zeeuw D, Agarwal R, Amdahl M, Audhya P, Coyne D, Garimella T, et al. Selective vitamin D receptor activation with paricalcitol for reduction of albuminuria in patients with type 2 diabetes (VITAL study): a randomised controlled trial. Lancet. 2010;376:1543-51. Mizobuchi M, Finch JL, Martin DR, Slatopolsky E. Differential effects of vitamin D receptor activators on vascular calcification in uremic rats. Kidney Int. 2007;72:709-15. Lopez I, Mendoza FJ, Guilera-Tejero E, Perez J, Guerrero F, Martin D, et al. The effect of calcitriol, paricalcitol, and a calcimimetic on extraosseous calcifications in uremic rats. Kidney Int. 2007;73:300-7. Mathew S, Lund RJ, Chaudhary LR, Geurs T, Hruska KA. Vitamin D receptor activators can protect against vascular calcification. J Am Soc Nephrol. 2008;19:1509-19.

Laboratory abnormalities in CKD-MBD: markers, predictors, or mediators of disease?

Chronic kidney disease-mineral and bone disorder (CKD-MBD) is characterized by bone abnormalities, vascular calcification, and an array of laboratory ...
908KB Sizes 0 Downloads 0 Views