You are on page 1of 11

www.kidney-international.

org review

Phosphate—a poison for humans?


Hirotaka Komaba1,2,3 and Masafumi Fukagawa1
1
Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan; 2Interactive Translational
Research Center for Kidney Diseases, Tokai University School of Medicine, Isehara, Japan; and 3The Institute of Medical Sciences, Tokai
University, Isehara, Japan

S
Maintenance of phosphate balance is essential for life, and ince the discovery of phosphate by Hennig Brand in
mammals have developed a sophisticated system to 1669 using a preparation from urine,1 the essential role
regulate phosphate homeostasis over the course of of phosphate in living organisms has been well estab-
evolution. However, due to the dependence of phosphate lished. Phosphate serves as a structural component of nucleic
elimination on the kidney, humans with decreased kidney acids, adenosine triphosphate, and the phospholipids of
function are likely to be in a positive phosphate balance. membranes. Phosphate also plays a critical role in cellular
Phosphate excess has been well recognized as a critical signaling through phosphorylation reactions. For these rea-
factor in the pathogenesis of mineral and bone disorders sons, phosphate is essential for life, and the intake of
associated with chronic kidney disease, but recent phosphate-containing products is crucial for all animals.
investigations have also uncovered toxic effects of During the course of evolution, animals have acquired
phosphate on the cardiovascular system and the aging “bone” that serves as a reservoir for life-essential phosphate
process. Compelling evidence also suggests that increased and calcium. This evolutionary event has likely eliminated the
fibroblastic growth factor 23 and parathyroid hormone need to store phosphate in extracellular fluids. Because
levels in response to a positive phosphate balance extremely high phosphate concentrations can have toxic
contribute to adverse clinical outcomes. These insights effects,2,3 animals have developed a system to excrete excess
support the current practice of managing serum phosphate phosphate through the kidneys and thereby maintain extra-
in patients with advanced chronic kidney disease, although cellular phosphate concentrations within physiologically
definitive evidence of these effects is lacking. Given the acceptable ranges. In mammals, parathyroid hormone (PTH)
potential toxicity of excess phosphate, the general and fibroblastic growth factor 23 (FGF23) both play an
population may also be viewed as a target for phosphate important role in regulating extracellular phosphate concen-
management. However, the widespread implementation of trations by stimulating urinary phosphate excretion.4
dietary phosphate intervention in the general population In this way, mammals have developed a sophisticated
may not be warranted due to the limited impact of system to tightly regulate phosphate homeostasis. However,
increased phosphate intake on mineral metabolism and due to the dependence of phosphate elimination on urinary
clinical outcomes. Nonetheless, the increasing incidence of excretion by the kidneys, patients with decreased kidney
kidney disease or injury in our aging society emphasizes function are likely to be in a positive phosphate balance.
the potential importance of this issue. Further work is The resulting hyperphosphatemia has been shown to be
needed to more completely characterize phosphate toxicity independently associated with increased risks of death and
and to establish the optimal therapeutic strategy for cardiovascular complications in patients with chronic kidney
managing phosphate in patients with chronic kidney disease (CKD),5,6 with the most pronounced associations being
disease and in the general population. observed in patients undergoing dialysis.7–9 Phosphate excess
Kidney International (2016) 90, 753–763; http://dx.doi.org/10.1016/ has also been shown to be associated with vascular calcification
j.kint.2016.03.039 and cardiovascular events in individuals with normal kidney
KEYWORDS: chronic kidney disease; fibroblastic growth factor 23; function.10–12 Of importance, individuals consume large
phosphate; parathyroid hormone amounts of phosphate through processed foods that are rich
Copyright ª 2016, International Society of Nephrology. Published by in phosphate additives,13,14 and there is a growing interest
Elsevier Inc. All rights reserved.
in restricting dietary phosphate as a matter of public health.
In this review, we outline disorders of phosphate meta-
bolism and the adverse effects of hyperphosphatemia with
a primary focus on CKD, address more general aspects of
phosphate toxicity, and discuss future directions for the
management of phosphate balance in individuals with normal
and impaired kidney function.
Correspondence: Masafumi Fukagawa, Division of Nephrology, Endocri-
nology and Metabolism, Tokai University School of Medicine, 143
Shimo-Kasuya, Isehara 259-1193, Japan. E-mail: fukagawa@tokai-u.jp Phospate omeostatis
Received 18 January 2016; revised 3 March 2016; accepted 24 March In humans, w85% of the total phosphate is in the bone and
2016; published online 7 June 2016 teeth, 10% to 15% is in soft tissues, and <1% is in extracellular

Kidney International (2016) 90, 753–763 753


review H Komaba and M Fukagawa: Phosphate—a poison for humans?

fluids. Phosphate homeostasis is maintained by a delicate Npt2c.19 The amount of Npt2a expressed on the apical
balance between intestinal absorption, renal excretion, and membrane of the tubule cells is primarily regulated by
influx to and efflux from bone (Figure 1). PTH20,21 and FGF23.22,23 PTH decreases Npt2a expression in
Regulation of intestinal absorption. Phosphate is abundant the proximal tubule by driving internalization of Npt2a from
in the human diet, with daily dietary phosphate intake the apical membrane and by decreasing Npt2a gene tran-
reaching $1500 mg. Approximately 40% to 80% of total scription. On the other hand, the detailed mechanisms by
ingested phosphate is absorbed through the intestine. There are which FGF23 regulates Npt2a expression in the proximal
the following 2 mechanisms of intestinal phosphate absorp- tubule are not clear. FGF23 exerts its biological functions by
tion: a passive paracellular pathway through tight junctions and binding to its cognate FGF receptor in the presence of Klo-
an active transport pathway through the sodium-dependent tho, which acts as an obligate cofactor.24,25 However, it is
phosphate cotransporter Npt2b.15 Active transport of phos- unknown why FGF23-mediated phosphate excretion takes
phate is regulated by calcitriol (1,25-dihydroxyvitamin D), place in the proximal tubules even though Klotho is pre-
which induces the expression of Npt2b on the apical mem- dominantly expressed in the distal tubular cells.26 One pre-
brane of intestinal epithelial cells.16 Low-phosphate diets also vious study detected a robust induction of phosphorylated
upregulate Npt2b expression independent of calcitriol,17 ERK1 only within Klotho-expressing distal tubule cells
although the detailed mechanism of this regulation remains following an FGF23 injection,27 suggesting that FGF23-
unclear. mediated intracellular signaling is initiated in the distal tu-
In addition to these intrinsic factors, the type of the bule. However, the targeted deletion of Klotho in the distal
diet also has a considerable impact on intestinal phosphate tubule28 yielded a significant but subtle change in phenotype
absorption. The modern diet often contains high levels of relative to that of kidney-specific Klotho-deficient mice.29
phosphate in the form of preservatives and additive salts that Furthermore, a recent study showed that Klotho is also
are readily absorbed by the intestine and can contribute to an expressed in proximal tubular cells and that FGF23 directly
individual’s phosphate burden.13,14 In contrast, phosphate in downregulates Npt2a in the proximal tubule.30 Further work
plants is primarily in the form of phytic acid and is generally is needed to completely elucidate the independent or inter-
not bioavailable to humans because humans do not have the active roles of proximal and distal tubular cells in mediating
digestive enzyme phytase that degrades phytic acid.18 the actions of FGF23.
Regulation of urinary excretion. In humans with normal In addition to its role as a coreceptor for FGF23, the
kidney function, urinary excretion of excess phosphate is extracellular domain of Klotho can be cleaved and released
primarily responsible for maintaining phosphate balance. into circulation where it may function as an endocrine fac-
After glomerular filtration, the majority of phosphate is tor.31 One possible action of this secreted Klotho is to
reabsorbed in the proximal tubule. Phosphate transport promote urinary phosphate excretion by suppressing the
across the proximal tubule cells is mediated by Npt2a and expression of Npt2a.32 However, the physiological importance

Parathyroid
Dietary glands
phosphate intake
1500 mg/day PTH

Kidney

Stimulates
PTH
secretion
Npt2a/2c
Filtration
Extracellular
Phosphate phosphate Reabsorption
absorption
Intestine 900 mg/day
Stimulates
Formation FGF23
secretion Urinary
phosphate
Fecal excretion
phosphate 900 mg/day
600 mg/day
FGF23
Resorption

Bone

Figure 1 | Phosphate homeostasis in normal physiology. In healthy adults, phosphate homeostasis is maintained by a delicate balance
between intestinal absorption, renal excretion, and influx to and efflux from bone. Fibroblast growth factor 23 (FGF23) and parathyroid
hormone (PTH) play a central role in the regulation of phosphate homeostasis.

754 Kidney International (2016) 90, 753–763


H Komaba and M Fukagawa: Phosphate—a poison for humans? review

of this effect remains to be elucidated and deserves further work is needed to determine the primary organs that sense
investigation. the phosphate balance in the body and the biological nature
Phosphate-sensing mechanisms. Although putative of these sensing mechanisms.
phosphate-sensing mechanisms have been shown in Escher-
ichia coli and yeast, it is unclear how mammals sense changes Hyperphosphatemia in CKD and other diseases
in extracellular phosphate. Given that FGF23 and PTH are the Phosphate metabolism in early to moderate CKD. Hyper-
2 major hormones that regulate phosphate metabolism, bone phosphatemia occurs in a variety of disorders, among which
cells and parathyroid cells are the most likely candidates for the most common is CKD. Urinary excretion of excess
sensing extracellular phosphate. Indeed, a series of studies phosphate is the primary mechanism for maintaining a
have shown that high extracellular phosphate levels increase neutral phosphate balance; however, serum phosphate levels
PTH gene expression and secretion.33,34 However, it remains are normal in the vast majority of patients in the early and
unclear how parathyroid cells sense changes in extracellular intermediate stages of CKD, presumably due to a compen-
phosphate. Several studies have also shown that FGF23 pro- satory reduction in tubular resorption mediated by increased
duction is increased by prolonged dietary phosphate levels of FGF23 and PTH42,43 (Figure 2). Emerging data
loading35–37; however, such findings were not evident in a suggest that increased FGF23 develops earlier than the in-
study of the effects of acute phosphate infusion38 and were crease in PTH and that it plays a more important role in
not reliably reproducible in cell culture experiments.39,40 maintaining phosphate balance in patients with CKD.44 The
Thus, it is unknown whether bone cells per se sense changes precise mechanism by which FGF23 levels are increased in
in extracellular phosphate or whether there are other organs this stage of the disease is unclear. A recent study found that
involved in this phosphate-sensing mechanism. the kidney plays a key role in FGF23 metabolism,45 but
Of interest, a recent study performed in rats showed that additional research is required to clarify the mechanisms
infusion of phosphate into the duodenum generated a rapid involved in the regulation of circulating FGF23 levels for
increase in urinary phosphate excretion without changes in maintaining a neutral phosphate balance.
PTH or FGF23. Furthermore, the infusion of extracts from Several factors have been identified that modify the rela-
duodenal homogenates increased urinary phosphate excre- tive importance and roles of FGF23 and PTH in patients with
tion.41 These data led to the hypothesis that the intestine has CKD. A recent study found that in patients with vitamin D
a phosphate sensor that regulates the secretion of an as-yet deficiency, PTH levels were markedly elevated relative to
undefined intestine-derived phosphaturic factor. Further those of FGF23.46 This suggests that the phosphaturic role of

Kidney
Decreased
Liver kidney function
Npt2a/2c

Insufficient
phosphate excretion
Fetuin A
Compensation by
stimulating
Positive phosphate excretion
phosphate
balance

Parathyroid
Removal glands
by reticulo- Stimulates
Fetuin-mineral Stimulates
endothelial FGF23
complex PTH
system secretion
(calciprotein secretion
particles)

PTH
Bone FGF23

Figure 2 | Compensatory mechanisms to maintain phosphate balance in early to moderate chronic kidney disease (CKD). In patients
with early to moderate CKD, increased fibroblast growth factor 23 (FGF23) and parathyroid hormone (PTH) stimulate urinary phosphate
excretion to maintain phosphate balance. In addition to the increased phosphate excretion induced by FGF23 and PTH, fetuin-A also plays an
important role in the prevention of ectopic calcification. Fetuin-A inhibits the precipitation of calcium phosphate by forming soluble colloidal
protein-mineral nanoparticles called fetuin-mineral complexes or calcitropin particles. These particles are removed by phagocytic cells of the
reticuloendothelial system.

Kidney International (2016) 90, 753–763 755


review H Komaba and M Fukagawa: Phosphate—a poison for humans?

FGF23 is less pronounced when PTH secretion is stimulated maintaining a normal phosphate balance.61 After the iden-
in response to vitamin D deficiency. Another recent study tification of FGF23, it is now thought that FGF23 is
showed that proteinuria impairs urinary phosphate excretion increased in early CKD, presumably to maintain a neutral
by inhibiting the effects of FGF23, which is likely caused by phosphate balance, which in turn results in reductions in the
decreased Klotho expression.47 Therefore, patients with renal production of calcitriol, thereby stimulating PTH
proteinuria have increased levels of both FGF23 and PTH, secretion.62 FGF23 is also known to inhibit PTH secretion,63
presumably to maintain a normal phosphate balance. but this effect is less pronounced in ESRD because of the
In addition to maintaining phosphate balance, the body downregulation of the Klotho-FGF receptor 1 complex in
uses a variety of mechanisms to prevent ectopic mineral the parathyroid.64,65 Studies performed before the identifi-
precipitation. Emerging research has identified fetuin-A, a cation of FGF23 also suggested that phosphate directly
hepatocyte-derived glycoprotein, as a major systemic inhibi- suppresses renal calcitriol production, thereby causing sec-
tor of calcification.48 Fetuin-A inhibits the precipitation of ondary hyperparathyroidism.66 The validity of this hypoth-
calcium phosphate by forming soluble colloidal protein- esis remains to be determined because a recent study has
mineral nanoparticles, which have been called fetuin- shown that a phosphate-deficient diet did not increase but
mineral complexes or calcitropin particles (CPPs).49 If rather decreased serum calcitriol levels in FGF23-knockout
supersaturated conditions persist even after primary CPP mice.67
formation, the particles develop crystalline structures to form In addition to these indirect effects, several studies have
elongated particles called secondary CPPs,50 through which shown that high phosphate levels also directly stimulate PTH
continuous inhibition of ectopic mineral deposition may be secretion.33,34 This effect has been shown to be mediated by a
achieved. Removal of CPPs from the circulation has recently posttranscriptional mechanism that regulates the stability of
been shown to be predominantly mediated by phagocytic cells PTH mRNA.68 High phosphate levels have also been shown
of the reticuloendothelial system51 (Figure 2). to stimulate parathyroid cell proliferation, which involves the
Phosphate metabolism in advanced CKD. In patients with enhanced expression of transforming growth factor-a.69
advanced stages of CKD, progressive reductions in the However, as mentioned earlier, there is no evidence suggest-
number of functional nephrons leads to insufficient phos- ing the existence of a phosphate sensor on parathyroid cells,
phate excretion even with the compensatory mechanisms of and little is known regarding the initial steps involved in these
FGF23 and PTH.44 This results in a positive phosphate bal- phosphate-induced effects.
ance and almost universally produces hyperphosphatemia in Hyperphosphatemia in other diseases. Hyperphosphatemia
patients with end-stage renal disease. also occurs in several other diseases (Table 1). Among these,
In addition to a reduction in urinary phosphate excre- familial tumoral calcinosis is clinically uncommon but
tion, abnormal bone metabolism can also contribute to informative for understanding the critical role of the FGF23-
hyperphosphatemia in patients with CKD through the Klotho system in regulating phosphate homeostasis. This
effects of disordered bone remodeling characterized by genetic disorder is caused by an inactivating mutation in the
excess bone resorption relative to the rate of bone forma- GALNT3,70 FGF23,71–73 or Klotho74 genes, any of which can
tion. This theory is supported by a recent observational lead to the functional impairment of FGF23. The GALNT3
study showing that higher serum PTH levels are associated gene encodes a glycosyltransferase that likely prevents the
with a higher risk of the development of hyper- degradation of FGF23 through a mucin-type O-glycosylation
phosphatemia.52 In addition, clinical studies have shown at the processing site; thus, an inactivating mutation in the
that the treatment of secondary hyperparathyroidism, using GALNT3 gene can cause enhanced processing of FGF23,
either cinacalcet hydrochloride53,54 or surgical para- thereby decreasing circulating levels of biologically active full-
thyroidectomy,55,56 leads to sustained reductions in serum length FGF23.75,76
phosphate and FGF23 levels, which may in part explain the
potential survival benefit of these interventions.57–59 A Table 1 | Major causes of hyperphosphatemia
more recent study also showed that velcalcetide, a novel
Phosphate overload
long-acting calcimimetic agent, attenuated the rise in Phosphate-containing enemas
serum phosphate levels during the interdialytic period.60 In Vitamin D toxicity
future research, it would be interesting to examine whether Excess bone resorption
treatment of high-turnover bone disease leads to improved Immobilization
Extracellular sift
survival through better control of hyperphosphatemia in Tumor lysis syndrome
patients with CKD. Rhabdomyolysis
Secondary hyperparathyroidism. The role of phosphate Acidosis
retention in the development and progression of secondary Decreased renal excretion
Kidney disease
hyperparathyroidism in patients with CKD has been well Hypoparathyroidism
established. Traditionally, it has been thought that phos- Pseudohypoparathyroidism (renal resistance to PTH)
phate retention causes a reciprocal decrease in serum cal- Familial tumoral calcinosis
cium, which in turn stimulates PTH secretion, thereby PTH, parathyroid hormone.

756 Kidney International (2016) 90, 753–763


H Komaba and M Fukagawa: Phosphate—a poison for humans? review

Toxicity of excess phosphate concentrations impair endothelium-dependent vasodilation


Vascular calcification. A number of observational studies by inhibiting nitric oxide production.92–94 High phosphate
have shown associations between hyperphosphatemia and an levels also induce apoptosis in endothelial cells, thereby
increased risk of death in patients with CKD.5–9 One of the impairing endothelial integrity.92,93 Supporting these experi-
most likely mechanisms linking hyperphosphatemia to higher mental data, recent clinical studies have shown that dietary
risks of mortality is vascular calcification. A growing body of phosphate loading lowered flow-mediated dilation in healthy
in vitro studies has shown that the process of vascular men,94 whereas treatment with the phosphate binder seve-
calcification is not merely a passive deposition of calcium- lamer improved flow-mediated dilation in predialysis CKD
phosphate crystals but an actively regulated process patients.95 The effects of high phosphate levels on endothelial
resembling osteogenesis in bone.77,78 In support of these cells appear to be direct and mediated via phosphate uptake
findings, a key osteogenic factor, core binding factor a-1, through Pit1.92,93 It is unknown whether calcium phosphate
which is also known as runt-related transcription factor 2, nanocrystals induce endothelial cell dysfunction, as has been
was shown to be expressed in calcified arteries from end-stage shown in the experiments on vascular smooth muscle cells.
renal disease patients.79 A more recent study showed that the Left ventricular hypertrophy. Left ventricular hypertrophy
targeted deletion of core binding factor a-1/runt-related (LVH) is another common manifestation of cardiovascular
transcription factor 2 in arterial smooth muscle cells in mice disease in patients with CKD, which may also mediate the
prevented the vascular calcification induced by high-dose association between hyperphosphatemia and the higher risk
calcitriol treatment,80 indicating that core binding factor of death in this population. Observational studies96,97 and
a-1/runt-related transcription factor 2 plays a critical role in animal studies98,99 have shown associations between hyper-
the transition of vascular smooth muscle cells to an osteo- phosphatemia and LVH. However, the direct causal role of
genic phenotype. High phosphate levels also induce apoptosis phosphate in the pathogenesis of LVH is still not completely
in vascular smooth muscle cells, which in turn triggers understood.
vascular calcification through the release of apoptotic Recently, increasing attention has been devoted to the
bodies.81,82 In addition, a recent experimental study showed possibility that FGF23 can cause pathologic LVH. This pos-
that in rats with adenine-induced CKD, high-phosphate diets sibility was first raised based on clinical observations showing
induced inflammation both locally in the artery and sys- associations between elevations in FGF23 levels and the
temically,83 which may also play a role in the pathogenesis of prevalence of LVH100 and the future risk of new-onset
the vascular calcification associated with hyperphosphatemia. LVH.101 These associations were subsequently proven in cell
Although the effects of high phosphate levels on vascular culture experiments and animal studies.101 As cardiomyocytes
smooth muscle cells are considered to be mediated via the do not express Klotho, it was suggested that the hypertrophic
uptake of phosphate through the sodium-dependent phos- effects of FGF23 are mediated via Klotho-independent
phate cotransporter Pit1,84 recent experimental data suggest signaling through FGF receptors. A follow-up study from
the possibility that calcium phosphate crystals, rather than the same group further demonstrated that FGF23 exerts hy-
increases in free phosphate ions, contribute to the patho- pertrophic effects by activating FGF receptor 4 and that a
genesis of vascular calcification.85,86 However, it is unknown specific inhibition of this receptor inhibits the FGF23-induced
whether these nanocrystals are present in the human circu- hypertrophy of isolated cardiac myocytes and attenuates LVH
latory system where fetuin-A serves as a systemic inhibitor of in rats with CKD.102 Another recent study showed that FGF23
calcium phosphate precipitation. It is also unknown whether increases renal sodium reabsorption by upregulating the
CPPs, which are produced during this process, contribute to Naþ:Cl cotransporter in the distal tubules via a Klotho-
the pathogenesis of vascular calcification.50 Although clinical dependent pathway.103 This could also induce LVH through
studies have identified associations between serum CPP levels volume expansion and hypertension. In addition to the
and vascular calcification87 and aortic stiffness,88 it remains to pathogenic roles of FGF23 in LVH, previous studies have
be determined whether serum CPP level is merely a marker of shown that PTH also exerts hypertrophic effects on car-
the propensity for vascular calcification or whether they are diomyocytes.104 Collectively, it is likely that LVH associated
directly involved in the pathogenesis of this process. with hyperphosphatemia in patients with CKD is, at least in
Several investigators have also explored the possibility that part, mediated through elevated FGF23 and PTH levels.
vascular calcification associated with hyperphosphatemia Kidney disease progression. Acute hyperphosphatemia, as
might, in part, be mediated by FGF23. However, there are encountered in patients treated with phosphate-containing
conflicting reports on whether vascular calcification can enemas, can cause acute or subacute kidney injury, which is
either be potentiated or inhibited by FGF23.89–91 These referred to as acute phosphate nephropathy.105 Although the
inconsistent results may suggest the relative importance of detailed mechanisms of this process are not fully understood,
phosphate per se rather than FGF23 in vascular calcification. it is generally accepted that an elevated intratubular phos-
Endothelial dysfunction. Endothelial dysfunction is phate concentration results in the precipitation and tissue
another consequence of hyperphosphatemia, and it can also deposition of calcium phosphate salts, which causes luminal
lead to an increased risk of cardiovascular disease. Several obstructions and tubular epithelial injury. Similar pathologic
experimental studies have shown that high phosphate processes can occur in CKD rats,106 and several animal107,108

Kidney International (2016) 90, 753–763 757


review H Komaba and M Fukagawa: Phosphate—a poison for humans?

and human109 studies have suggested that a low phosphate phosphate overload has also been shown to shorten life spans
diet slows the progression of CKD. Epidemiologic studies also in nonmammalian species such as Drosophila,127 suggesting
suggested that higher serum phosphate110 or FGF23111–113 that the toxic effect of excess phosphate is evolutionarily
levels predict a more rapid decline in kidney function in the conserved across diverse species.
CKD population. These data raise the possibility that high The mechanisms by which hyperphosphatemia accelerates
phosphate intake together with a decreased number of aging are largely unknown, but several possibilities exist.
nephrons can result in an elevated intratubular phosphate Probably one of the most reasonable mechanisms is that the
concentration, which induces tubular damage and renal vascular calcification induced by hyperphosphatemia is
fibrosis. Of note, a recent cohort study of the general popu- responsible for the observed age-related effects. Although
lation showed that higher FGF23 levels were independently there is no direct evidence, alterations in systemic or regional
associated with higher risks of the development of end-stage hemodynamics due to vascular calcification may induce
renal disease, but that it was not as predictive of the incidence cellular damage and stress in the perfused tissue, which may
of CKD.114 These findings may suggest that enhanced phos- accelerate the aging process. From the perspective of cellular
phaturia, predominantly mediated by elevated FGF23 levels, metabolism, it is possible that excess phosphate, if taken up
exposes tubules to intraluminal phosphate concentrations intracellularly, might cause dysregulation of the
that are sufficiently high to induce tubular toxicity in those phosphorylation-dependent signaling pathways.2 In addition,
with established CKD but not in those with normal kidney recent studies have shown that elevated extracellular phos-
function.115 phate concentrations cause mitochondrial oxidative stress by
Bone disease. Hyperphosphatemia is one of the major increasing the mitochondrial membrane potential, leading to
contributors to the development of secondary hyperparathy- caspase activation and the subsequent induction of
roidism; thus, excess phosphate can cause high-turnover bone apoptosis.128 Another recent study showed that high phos-
disease through the stimulation of PTH secretion. Indeed, in phate levels directly induce systemic inflammation,83 which
rodent models of CKD, a high-phosphate diet is known to may cause accelerated aging and vascular calcification. Finally,
induce high-turnover bone disease, and treatment of hyper- a high-phosphate diet has also been shown to stimulate the
phosphatemia by sevelamer hydrochloride has been shown to growth of lung tumors,129 suggesting that excess phosphate
improve high-turnover bone disease.116 This may also be the may induce tumorigenesis in the aging process.
case for humans, although previous studies focused on Possible toxicity of compensatory mechanisms. Observa-
comparisons between different phosphate binder clas- tional studies have shown associations between higher serum
ses,117,118 and no placebo-controlled trials have evaluated the phosphate levels and poor clinical outcomes in patients with
impact of phosphate-lowering therapy on bone histology. predialysis CKD.5,6 Epidemiologic studies of the general
Several lines of experimental evidence also suggest that FGF23 population have also shown associations of serum phosphate
directly affects bone metabolism,119,120 but conflicting results with cardiovascular events and vascular calcification.10–12
have been reported regarding the associations of serum However, it should be noted that serum phosphate levels
FGF23 levels with fracture risk in older individuals.121,122 associated with adverse outcomes are within or only slightly
Aging and cytotoxicity. The toxic effects of excess phos- above the normal range and are much lower than those used
phate have been well characterized in genetically modified to induce pathologic calcification in experimental models.
animal models. Klotho-deficient (kl/kl) mice26 and FGF23- There are 3 possibilities that could explain these findings.
knockout mice23 are 2 major mouse models that have pro- First, the slight increase in serum phosphate level may reflect
vided instructive information on the effects of phosphate an elevated intratubular phosphate concentration, which can
toxicity. Klotho was originally identified as an antiaging gene, cause kidney injury, as described previously. This may be
as complex phenotypes resembling those observed in human clinically relevant because impaired kidney function is a
aging were shown to develop in kl/kl mice.26 An identical powerful predictor of mortality and cardiovascular events.130
phenotype was also observed in FGF23-knockout mice.23 It is Second, it is possible that a single measurement of serum
currently accepted that, for the most part, these premature- phosphate provides only a partial assessment of total phos-
aging features are caused by severe hyperphosphatemia as a phate balance and its associated calcification risk. This pos-
result of impaired function of the FGF23-Klotho system. sibility is supported by the observation that serum CPP levels,
Indeed, it has been shown that the lethal phenotypic effects a potential marker of calcification propensity, begin to rise
observed in kl/kl mice and FGF23-knockout mice were before the appearance of hyperphosphatemia in those
attenuated by feeding them a low-phosphate diet.67,123 In with CKD.87,88 Third, the homeostatic response to maintain
addition, genetic ablation of Npt2a has also been shown to phosphate balance may be responsible for the associations
rescue the phenotype of Klotho-knockout mice124 and between higher serum phosphate levels and adverse out-
FGF23-knockout mice.125 More importantly, in Klotho/ comes. As discussed previously, a growing body of evidence
Npt2a double knockout mice, the aging phenotype was suggests causal roles for FGF23 and PTH in the pathologic
restored when the mice were fed a high-phosphate diet.126 development of LVH.100–104 Previous and recent studies have
These data suggest the hypothesis that phosphate toxicity also suggested a link between FGF23 or PTH and renal
can accelerate the mammalian aging process. Of interest, anemia131,132 and between these molecules and immune

758 Kidney International (2016) 90, 753–763


H Komaba and M Fukagawa: Phosphate—a poison for humans? review

dysfunction.133,134 Furthermore, a very recent study showed mechanisms is clinically impracticable because it leads to
that in 5/6 nephrectomized mice, PTH induced adipose tissue detrimental alterations in mineral metabolism.140,141 Thus, an
browning and cachexia and, thereby, presumably induced adequate approach may be dietary phosphate restriction and
skeletal muscle atrophy.135 Thus, there are multiple mecha- the administration of phosphate binders. Testing this
nisms linking the physiological response for maintaining approach, several recent studies compared the effects of
phosphate homeostasis with adverse clinical outcomes. A phosphate binders versus those of placebos on intermediate
schematic representation of the toxic effects of phosphate per endpoints for cardiovascular disease.142,143 However, these
se and the compensatory mechanisms involved are provided studies found no significant benefit of phosphate binders.
in Figure 3. One possible reason for these null findings may be a
compensatory upregulation of Npt2b in the intestine, which
Future directions for phosphate management could counteract the effects of phosphate binders. This pos-
The importance of controlling hyperphosphatemia has sibility has recently been demonstrated in an experimental
already been well recognized for the management of dialysis study using Npt2b-deficient mice144 and will be addressed by
patients.136 However, it should be stressed that the current the COMBINE study.145 This clinical study will examine the
approach is not based on definitive evidence. Although recent effects of lanthanum carbonate plus nicotinamide, which
observational studies have shown independent associations of decreases Npt2b expression, on serum phosphate and FGF23
phosphate binder use with improved survival,137–139 no ran- levels in patients with stage 3-4 CKD.
domized trials have yet proved whether the management of Emerging data on the general toxicity of excess phosphate
serum phosphate actually improves clinical outcomes. The also highlight the importance of subjects with normal kidney
ideal target level of serum phosphate also remains to be function as potential targets for phosphate management.
determined. In this regard, clinical trials comparing the sur- However, several caveats should be addressed. First, if kidney
vival of intensive versus conventional serum phosphate con- function is normal, dietary phosphate overload causes only a
trol are urgently needed and would be appropriate as the next slight increase in serum phosphate levels, if any at all, and this
step for research in this field of study. is unlikely to exert any cytotoxic effects. Second, because
Observational data linking higher serum phosphate levels healthy subjects have a sufficient number of nephrons, in-
with adverse outcomes in predialysis CKD patients suggest creases in intratubular phosphate concentration that are high
the potential benefit of managing phosphate in this popula- enough to induce kidney injury may not occur. Third, the
tion.5,6 Several lines of evidence suggest that elevations in increases in serum FGF23 or PTH in response to phosphate
FGF23 and PTH levels are responsible for the adverse effects overload are much smaller in the general population than
of excess phosphate, but the inhibition of these compensatory they are in patients with CKD.35–37 Therefore, a physiological

Vascular calcification • Dysregulation of cell signaling


Endothelial dysfunction • Oxidative stress
Direct cell • Apoptosis
toxicity • Inflammation
Induces osteogenic
differentiation
Phosphate High intratubular
?
phosphate

Stimulates PTH
Fetuin-mineral Stimulates secretion Kidney disease
complex FGF23 progression
(calciprotein particles) secretion

FGF23 Stimulates bone Secondary


resorption PTH hyperparathyroidism
Induces hypertrophy
of cardiomyocytes

Browning of
Inhibits adipose tissue
Left ventricular sodium excretion
hypertrophy
Volume Immune Muscle atrophy
expansion dysfunction
Inhibits Renal
calcitriol production anemia

Figure 3 | Schematic representation of phosphate toxicity. Excess phosphate exerts toxic effects through a variety of pathways. High
phosphate levels directly potentiate vascular calcification and endothelial dysfunction, promote the progression of kidney disease, and induce
cell stress and apoptosis. High phosphate levels also contribute to adverse outcomes through increases in the levels of fibroblast growth factor
23 (FGF23) and parathyroid hormone (PTH), including left ventricular hypertrophy, renal anemia, immune dysfunction, adipose tissue browning,
and skeletal muscle atrophy.

Kidney International (2016) 90, 753–763 759


review H Komaba and M Fukagawa: Phosphate—a poison for humans?

increase in FGF23 or PTH would not generate harmful effects 6. Eddington H, Hoefield R, Sinha S, et al. Serum phosphate and mortality
in patients with chronic kidney disease. Clin J Am Soc Nephrol. 2010;5:
because the off-target effects of FGF23 and PTH require 2251–2257.
marked elevations in their circulating levels.100–104 Taken 7. Block GA, Klassen PS, Lazarus JM, et al. Mineral metabolism, mortality,
together, our current knowledge does not convincingly sup- and morbidity in maintenance hemodialysis. J Am Soc Nephrol. 2004;15:
2208–2218.
port the idea that increased phosphate intake within the usual 8. Tentori F, Blayney MJ, Albert JM, et al. Mortality risk for dialysis patients
dietary range causes adverse outcomes. One possible excep- with different levels of serum calcium, phosphorus, and PTH: the
tion is in cases in which de novo kidney disease or injury has Dialysis Outcomes and Practice Patterns Study (DOPPS). Am J Kidney
Dis. 2008;52:519–530.
occurred. In this situation, dietary phosphate overload, as 9. Taniguchi M, Fukagawa M, Fujii N, et al. Serum phosphate and calcium
reflected by elevated FGF23 levels, may accelerate the pro- should be primarily and consistently controlled in prevalent
gression of kidney damage until the disease is identified. hemodialysis patients. Ther Apher Dial. 2013;17:221–228.
10. Tonelli M, Sacks F, Pfeffer M, et al. Relation between serum phosphate
level and cardiovascular event rate in people with coronary disease.
CONCLUSIONS Circulation. 2005;112:2627–2633.
11. Dhingra R, Sullivan LM, Fox CS, et al. Relations of serum phosphorus
Recent investigations have uncovered the toxic effects of and calcium levels to the incidence of cardiovascular disease in the
excess phosphate on the cardiovascular system and the aging community. Arch Intern Med. 2007;167:879–885.
process. Compelling evidence also suggests that elevated 12. Foley RN, Collins AJ, Herzog CA, et al. Serum phosphorus levels
associate with coronary atherosclerosis in young adults. J Am Soc
FGF23 and PTH levels in response to a positive phosphate Nephrol. 2009;20:397–404.
balance contribute to these adverse clinical outcomes. These 13. Uribarri J. Phosphorus additives in food and their effect in dialysis
insights support the current practice of managing serum patients. Clin J Am Soc Nephrol. 2009;4:1290–1292.
14. Kalantar-Zadeh K, Gutekunst L, Mehrotra R, et al. Understanding
phosphate in patients with advanced CKD. However, defini-
sources of dietary phosphorus in the treatment of patients with chronic
tive evidence is lacking, and clinical trials to determine the kidney disease. Clin J Am Soc Nephrol. 2010;5:519–530.
benefit of phosphate management are desperately needed and 15. Sabbagh Y, O’Brien SP, Song W, et al. Intestinal npt2b plays a major role
should be strongly encouraged. in phosphate absorption and homeostasis. J Am Soc Nephrol. 2009;20:
2348–2358.
Given the potential toxicity of excess phosphate, the gen- 16. Xu H, Bai L, Collins JF, et al. Age-dependent regulation of rat intestinal
eral population may also be viewed as a target for phosphate type IIb sodium-phosphate cotransporter by 1,25-(OH)2 vitamin D3. Am
management. However, a widespread implementation of di- J Physiol Cell Physiol. 2002;282:C487–C493.
17. Segawa H, Kaneko I, Yamanaka S, et al. Intestinal Na-P(i) cotransporter
etary phosphate intervention in the general population may adaptation to dietary P(i) content in vitamin D receptor null mice. Am J
not be warranted because the impact of increased phosphate Physiol Renal Physiol. 2004;287:F39–F47.
intake on mineral metabolism and clinical outcomes is much 18. Moe SM, Zidehsarai MP, Chambers MA, et al. Vegetarian compared with
meat dietary protein source and phosphorus homeostasis in chronic
less pronounced in individuals with normal kidney function. kidney disease. Clin J Am Soc Nephrol. 2011;6:257–264.
Nonetheless, the increasing incidence of kidney disease or 19. Segawa H, Onitsuka A, Furutani J, et al. Npt2a and Npt2c in mice play
injury in our aging society146 emphasizes the potential distinct and synergistic roles in inorganic phosphate metabolism and
skeletal development. Am J Physiol Renal Physiol. 2009;297:F671–F678.
importance of this issue. Further work is needed to more 20. Déliot N, Hernando N, Horst-Liu Z, et al. Parathyroid hormone treatment
completely characterize phosphate toxicity and to establish induces dissociation of type IIa Naþ-P(i) cotransporter-Naþ/Hþ
the optimal therapeutic strategy for managing phosphate in exchanger regulatory factor-1 complexes. Am J Physiol Cell Physiol.
2005;289:C159–C167.
patients with CKD and in the general population. 21. Weinman EJ, Biswas RS, Peng G, et al. Parathyroid hormone inhibits renal
phosphate transport by phosphorylation of serine 77 of sodium-hydrogen
exchanger regulatory factor-1. J Clin Invest. 2007;117:3412–3420.
DISCLOSURE
22. Shimada T, Hasegawa H, Yamazaki Y, et al. FGF-23 is a potent regulator
HK has received honoraria, consulting fees, and grant/research of vitamin D metabolism and phosphate homeostasis. J Bone Miner Res.
support from Kyowa Hakko Kirin. MF has received honoraria, 2004;19:429–435.
consulting fees, and/or grant/research support from Bayer Yakuhin, 23. Shimada T, Kakitani M, Yamazaki Y, et al. Targeted ablation of Fgf23
Kyowa Hakko Kirin, and Torii Pharmaceutical. demonstrates an essential physiological role of FGF23 in phosphate
and vitamin D metabolism. J Clin Invest. 2004;113:561–568.
24. Urakawa I, Yamazaki Y, Shimada T, et al. Klotho converts canonical FGF
ACKNOWLEDGMENTS receptor into a specific receptor for FGF23. Nature. 2006;444:770–774.
This work was supported in part by a Grant-in-Aid for Scientific 25. Kurosu H, Ogawa Y, Miyoshi M, et al. Regulation of fibroblast growth
Research (C) (25461257) from the Ministry of Education, Culture, factor-23 signaling by Klotho. J Biol Chem. 2006;281:6120–6123.
Sports, Science and Technology, Japan. 26. Kuro-o M, Matsumura Y, Aizawa H, et al. Mutation of the mouse Klotho
gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51.
27. Farrow EG, Davis SI, Summers LJ, et al. Initial FGF23-mediated signaling
REFERENCES occurs in the distal convoluted tubule. J Am Soc Nephrol. 2009;20:955–960.
1. Krafft F. Phosphorus. From elemental light to chemical element. Angew 28. Olauson H, Lindberg K, Amin R, et al. Targeted deletion of Klotho in
Chem Int Ed Engl. 1969;8:660–671. kidney distal tubule disrupts mineral metabolism. J Am Soc Nephrol.
2. Razzaque MS. Phosphate toxicity: new insights into an old problem. Clin 2012;23:1641–1651.
Sci (Lond). 2011;120:91–97. 29. Lindberg K, Amin R, Moe OW, et al. The kidney is the principal organ
3. Kuro-o M. A phosphate-centric paradigm for pathophysiology and mediating klotho effects. J Am Soc Nephrol. 2014;25:2169–2175.
therapy of chronic kidney disease. Kidney Int Suppl. 2013;3:420–426. 30. Andrukhova O, Zeitz U, Goetz R, et al. FGF23 acts directly on renal
4. Komaba H, Fukagawa M. FGF23-parathyroid interaction: implications in proximal tubules to induce phosphaturia through activation of the
chronic kidney disease. Kidney Int. 2010;77:292–298. ERK1/2-SGK1 signaling pathway. Bone. 2012;51:621–628.
5. Kestenbaum B, Sampson JN, Rudser KD, et al. Serum phosphate levels 31. Hu MC, Shiizaki K, Kuro-o M, et al. Fibroblast growth factor 23 and
and mortality risk among people with chronic kidney disease. J Am Soc Klotho: physiology and pathophysiology of an endocrine network of
Nephrol. 2005;16:520–528. mineral metabolism. Annu Rev Physiol. 2013;75:503–533.

760 Kidney International (2016) 90, 753–763


H Komaba and M Fukagawa: Phosphate—a poison for humans? review

32. Hu MC, Shi M, Zhang J, Pastor J, et al. Klotho: a novel phosphaturic severe secondary hyperparathyroidism. J Clin Endocrinol Metab. 2014;99:
substance acting as an autocrine enzyme in the renal proximal tubule. E652–E658.
FASEB J. 2010;24:3438–3450. 57. Kestenbaum B, Andress DL, Schwartz SM, et al. Survival following
33. Slatopolsky E, Finch J, Denda M, et al. Phosphorus restriction prevents parathyroidectomy among United States dialysis patients. Kidney Int.
parathyroid gland growth. High phosphorus directly stimulates PTH 2004;66:2010–2016.
secretion in vitro. J Clin Invest. 1996;97:2534–2540. 58. Komaba H, Taniguchi M, Wada A, et al. Parathyroidectomy and survival
34. Almaden Y, Canalejo A, Hernandez A, et al. Direct effect of phosphorus among Japanese hemodialysis patients with secondary
on PTH secretion from whole rat parathyroid glands in vitro. J Bone hyperparathyroidism. Kidney Int. 2015;88:350–359.
Miner Res. 1996;11:970–976. 59. EVOLVE Trial Investigators. Effect of cinacalcet on cardiovascular disease
35. Ferrari SL, Bonjour JP, Rizzoli R. Fibroblast growth factor-23 relationship in patients undergoing dialysis. N Engl J Med. 2012;367:2482–2494.
to dietary phosphate and renal phosphate handling in healthy young 60. Martin KJ, Pickthorn K, Huang S, et al. AMG 416 (velcalcetide) is a novel
men. J Clin Endocrinol Metab. 2005;90:1519–1524. peptide for the treatment of secondary hyperparathyroidism in a
36. Burnett SM, Gunawardene SC, Bringhurst FR, et al. Regulation of single-dose study in hemodialysis patients. Kidney Int. 2014;85:191–197.
C-terminal and intact FGF-23 by dietary phosphate in men and women. 61. Bricker NS. On the pathogenesis of the uremic state. An exposition of
J Bone Miner Res. 2006;21:1187–1196. the ‘trade-off hypothesis’. N Engl J Med. 1972;286:1093–1099.
37. Antoniucci DM, Yamashita T, Portale AA. Dietary phosphorus regulates 62. Hasegawa H, Nagano N, Urakawa I, et al. Direct evidence for a causative
serum fibroblast growth factor-23 concentrations in healthy men. J Clin role of FGF23 in the abnormal renal phosphate handling and vitamin D
Endocrinol Metab. 2006;91:3144–3149. metabolism in rats with early-stage chronic kidney disease. Kidney Int.
38. Ito N, Fukumoto S, Takeuchi Y, et al. Effect of acute changes of serum 2010;78:975–980.
phosphate on fibroblast growth factor (FGF)23 levels in humans. J Bone 63. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, et al. The parathyroid is a
Miner Metab. 2007;25:419–422. target organ for FGF23 in rats. J Clin Invest. 2007;117:4003–4008.
39. Liu S, Tang W, Zhou J, et al. Fibroblast growth factor 23 is a counter- 64. Komaba H, Goto S, Fujii H, et al. Depressed expression of Klotho and
regulatory phosphaturic hormone for vitamin D. J Am Soc Nephrol. FGF receptor 1 in hyperplastic parathyroid glands from uremic patients.
2006;17:1305–1315. Kidney Int. 2010;77:232–238.
40. Hori M, Kinoshita Y, Taguchi M, et al. Phosphate enhances Fgf23 65. Galitzer H, Ben-Dov IZ, Silver J, et al. Parathyroid cell resistance to
expression through reactive oxygen species in UMR-106 cells. J Bone fibroblast growth factor 23 in secondary hyperparathyroidism of
Miner Metab. 2016;34:132–139. chronic kidney disease. Kidney Int. 2010;77:211–218.
41. Berndt T, Thomas LF, Craig TA, et al. Evidence for a signaling axis by 66. Portale AA, Booth BE, Halloran BP, et al. Effect of dietary phosphorus on
which intestinal phosphate rapidly modulates renal phosphate circulating concentrations of 1,25-dihydroxyvitamin D and
reabsorption. Proc Natl Acad Sci U S A. 2007;104:11085–11090. immunoreactive parathyroid hormone in children with moderate renal
42. Shigematsu T, Kazama JJ, Yamashita T, et al. Possible involvement of insufficiency. J Clin Invest. 1984;73:1580–1589.
circulating fibroblast growth factor 23 in the development of secondary 67. Stubbs JR, Liu S, Tang W, et al. Role of hyperphosphatemia and 1,25-
hyperparathyroidism associated with renal insufficiency. Am J Kidney dihydroxyvitamin D in vascular calcification and mortality in fibroblastic
Dis. 2004;44:250–256. growth factor 23 null mice. J Am Soc Nephrol. 2007;18:2116–2124.
43. Gutierrez O, Isakova T, Rhee E, et al. Fibroblast growth factor-23 68. Moallem E, Kilav R, Silver J, et al. RNA-Protein binding and post-
mitigates hyperphosphatemia but accentuates calcitriol deficiency in transcriptional regulation of parathyroid hormone gene expression by
chronic kidney disease. J Am Soc Nephrol. 2005;16:2205–2215. calcium and phosphate. J Biol Chem. 1998;273:5253–5259.
44. Isakova T, Wahl P, Vargas GS, et al. Fibroblast growth factor 23 is 69. Cozzolino M, Lu Y, Sato TJ, et al. A critical role for enhanced TGF-alpha
elevated before parathyroid hormone and phosphate in chronic kidney and EGFR expression in the initiation of parathyroid hyperplasia in
disease. Kidney Int. 2011;79:1370–1378. experimental kidney disease. Am J Physiol Renal Physiol. 2005;289:
45. Mace ML, Gravesen E, Hofman-Bang J, et al. Key role of the kidney in the F1096–F1102.
regulation of fibroblast growth factor 23. Kidney Int. 2015;88:1304–1313. 70. Topaz O, Shurman DL, Bergman R, et al. Mutations in GALNT3 encoding
46. Taal MW, Thurston V, McIntyre NJ, et al. The impact of vitamin D status a protein involved in O-linked glycosylation, cause familial tumoral
on the relative increase in fibroblast growth factor 23 and parathyroid calcinosis. Nat Genet. 2004;36:579–581.
hormone in chronic kidney disease. Kidney Int. 2014;86:407–413. 71. Benet-Pages A, Orlik P, Strom TM, et al. An FGF23 missense mutation
47. de Seigneux S, Courbebaisse M, Rutkowski JM, et al. Proteinuria causes familial tumoral calcinosis with hyperphosphatemia. Hum Mol
increases plasma phosphate by altering its tubular handling. J Am Soc Genet. 2005;14:385–390.
Nephrol. 2015;26:1608–1618. 72. Larsson T, Yu X, Davis SI, et al. A novel recessive mutation in fibroblast
48. Schafer C, Heiss A, Schwarz A, et al. The serum protein alpha growth factor-23 (FGF23) causes familial tumoral calcinosis. J Clin
2-Heremans-Schmid glycoprotein/fetuin-A is a systemically acting Endocrinol Metab. 2005;90:2424–2427.
inhibitor of ectopic calcification. J Clin Invest. 2003;112:357–366. 73. Araya K, Fukumoto S, Backenroth R, et al. A novel mutation in fibroblast
49. Price PA, Lim JE. The inhibition of calcium phosphate precipitation by growth factor 23 gene as a cause of tumoral calcinosis. J Clin Endocrinol
fetuin is accompanied by the formation of a fetuin-mineral complex. Metab. 2005;90:5523–5527.
J Biol Chem. 2003;278:22144–22152. 74. Ichikawa S, Imel EA, Kreiter ML, et al. A homozygous missense mutation
50. Cai MM, Smith ER, Holt SG. The role of fetuin-A in mineral trafficking in human KLOTHO causes severe tumoral calcinosis. J Clin Invest.
and deposition. Bonekey Rep. 2015;4:672. 2007;117:2684–2691.
51. Herrmann M, Schäfer C, Heiss A, et al. Clearance of fetuin-A-containing 75. Kato K, Jeanneau C, Tarp MA, et al. Polypeptide GalNAc-transferase T3
calciprotein particles is mediated by scavenger receptor-A. Circ Res. and familial tumoral calcinosis. Secretion of fibroblast growth factor 23
2012;111:575–584. requires O-glycosylation. J Biol Chem. 2006;281:18370–18377.
52. Streja E, Lau WL, Goldstein L, et al. Hyperphosphatemia is a combined 76. Frishberg Y, Ito N, Rinat C, et al. Hyperostosis-hyperphosphatemia
function of high serum PTH and high dietary protein intake in dialysis syndrome: a congenital disorder of O-glycosylation associated with
patients. Kidney Int Suppl. 2013;3:462–468. augmented processing of fibroblast growth factor 23. J Bone Miner Res.
53. Koizumi M, Komaba H, Nakanishi S, et al. Cinacalcet treatment and 2007;22:235–242.
serum FGF23 levels in haemodialysis patients with secondary 77. Jono S, McKee MD, Murry CE, et al. Phosphate regulation of vascular
hyperparathyroidism. Nephrol Dial Transplant. 2012;27:784–790. smooth muscle cell calcification. Circ Res. 2000;87:E10–E17.
54. Moe SM, Chertow GM, Parfrey PS, et al. Cinacalcet, fibroblast growth 78. Chen NX, O’Neill KD, Duan D, et al. Phosphorus and uremic serum
factor-23, and cardiovascular disease in hemodialysis: the evaluation of up-regulate osteopontin expression in vascular smooth muscle cells.
cinacalcet HCl therapy to lower cardiovascular events (EVOLVE) trial. Kidney Int. 2002;62:1724–1731.
Circulation. 2015;132:27–39. 79. Moe SM, O’Neill KD, Duan D, et al. Medial artery calcification in ESRD
55. Sato T, Tominaga Y, Ueki T, et al. Total parathyroidectomy reduces patients is associated with deposition of bone matrix proteins. Kidney
elevated circulating fibroblast growth factor 23 in advanced secondary Int. 2002;61:638–647.
hyperparathyroidism. Am J Kidney Dis. 2004;44:481–487. 80. Lin ME, Chen T, Leaf EM, et al. Runx2 expression in smooth muscle cells
56. Takahashi H, Komaba H, Takahashi Y, et al. Impact of parathyroidectomy is required for arterial medial calcification in mice. Am J Pathol.
on serum FGF23 and soluble Klotho in hemodialysis patients with 2015;185:1958–1969.

Kidney International (2016) 90, 753–763 761


review H Komaba and M Fukagawa: Phosphate—a poison for humans?

81. Reynolds JL, Joannides AJ, Skepper JN, et al. Human vascular smooth 104. Schlüter KD, Piper HM. Cardiovascular actions of parathyroid hormone
muscle cells undergo vesicle-mediated calcification in response to and parathyroid hormone-related peptide. Cardiovasc Res. 1998;37:
changes in extracellular calcium and phosphate concentrations: a 34–41.
potential mechanism for accelerated vascular calcification in ESRD. 105. Markowitz GS, Perazella MA. Acute phosphate nephropathy. Kidney Int.
J Am Soc Nephrol. 2004;15:2857–2867. 2009;76:1027–1034.
82. Shroff RC, McNair R, Skepper JN, et al. Chronic mineral dysregulation 106. Haut LL, Alfrey AC, Guggenheim S, et al. Renal toxicity of phosphate in
promotes vascular smooth muscle cell adaptation and extracellular rats. Kidney Int. 1980;17:722–731.
matrix calcification. J Am Soc Nephrol. 2010;21:103–112. 107. Kusano K, Segawa H, Ohnishi R, et al. Role of low protein and low
83. Yamada S, Tokumoto M, Tatsumoto N, et al. Phosphate overload phosphorus diet in the progression of chronic kidney disease in uremic
directly induces systemic inflammation and malnutrition as well as rats. J Nutr Sci Vitaminol (Tokyo). 2008;54:237–243.
vascular calcification in uremia. Am J Physiol Renal Physiol. 2014;306: 108. Koizumi T, Murakami K, Nakayama H, et al. Role of dietary phosphorus
F1418–F1428. in the progression of renal failure. Biochem Biophys Res Commun.
84. Li X, Yang HY, Giachelli CM. Role of the sodium-dependent phosphate 2002;295:917–921.
cotransporter, Pit-1, in vascular smooth muscle cell calcification. Circ 109. Barsotti G, Giannoni A, Morelli E, et al. The decline of renal function
Res. 2006;98:905–912. slowed by very low phosphorus intake in chronic renal patients
85. Sage AP, Lu J, Tintut Y, et al. Hyperphosphatemia-induced nanocrystals following a low nitrogen diet. Clin Nephrol. 1984;21:54–59.
upregulate the expression of bone morphogenetic protein-2 and 110. Schwarz S, Trivedi BK, Kalantar-Zadeh K, et al. Association of disorders
osteopontin genes in mouse smooth muscle cells in vitro. Kidney Int. in mineral metabolism with progression of chronic kidney disease. Clin
2011;79:414–422. J Am Soc Nephrol. 2006;1:825–831.
86. Ewence AE, Bootman M, Roderick HL, et al. Calcium phosphate crystals 111. Fliser D, Kollerits B, Neyer U, et al. Fibroblast growth factor 23
induce cell death in human vascular smooth muscle cells: a potential (FGF23) predicts progression of chronic kidney disease: the Mild to
mechanism in atherosclerotic plaque destabilization. Circ Res. 2008;103: Moderate Kidney Disease (MMKD) Study. J Am Soc Nephrol. 2007;18:
e28–e34. 2600–2608.
87. Hamano T, Matsui I, Mikami S, et al. Fetuin-mineral complex reflects 112. Isakova T, Xie H, Yang W, et al. Fibroblast growth factor 23 and risks of
extraosseous calcification stress in CKD. J Am Soc Nephrol. 2010;21: mortality and end-stage renal disease in patients with chronic kidney
1998–2007. disease. JAMA. 2011;305:2432–2439.
88. Smith ER, Ford ML, Tomlinson LA, et al. Phosphorylated fetuin-A- 113. Nakano C, Hamano T, Fujii N, et al. Combined use of vitamin D status
containing calciprotein particles are associated with aortic stiffness and and FGF23 for risk stratification of renal outcome. Clin J Am Soc Nephrol.
a procalcific milieu in patients with pre-dialysis CKD. Nephrol Dial 2012;7:810–819.
Transplant. 2012;27:1957–1966. 114. Rebholz CM, Grams ME, Coresh J, et al. Serum fibroblast growth factor-23
89. Lim K, Lu TS, Molostvov G, et al. Vascular Klotho deficiency potentiates is associated with incident kidney disease. J Am Soc Nephrol. 2015;26:
the development of human artery calcification and mediates resistance 192–200.
to fibroblast growth factor 23. Circulation. 2012;125:2243–2255. 115. Wolf M. The biomarker niche for fibroblast growth factor 23 testing in
90. Scialla JJ, Lau WL, Reilly MP, et al. Fibroblast growth factor 23 is not CKD. J Am Soc Nephrol. 2015;26:7–9.
associated with and does not induce arterial calcification. Kidney Int. 116. Katsumata K, Kusano K, Hirata M, et al. Sevelamer hydrochloride
2013;83:1159–1168. prevents ectopic calcification and renal osteodystrophy in chronic renal
91. Jimbo R, Kawakami-Mori F, Mu S, et al. Fibroblast growth factor 23 failure rats. Kidney Int. 2003;64:441–450.
accelerates phosphate-induced vascular calcification in the absence of 117. Ferreira A, Frazão JM, Monier-Faugere MC, et al. Effects of sevelamer
Klotho deficiency. Kidney Int. 2014;85:1103–1111. hydrochloride and calcium carbonate on renal osteodystrophy in
92. Di Marco GS, Hausberg M, Hillebrand U, et al. Increased inorganic hemodialysis patients. J Am Soc Nephrol. 2008;19:405–412.
phosphate induces human endothelial cell apoptosis in vitro. Am J 118. Malluche HH, Siami GA, Swanepoel C, et al. Improvements in renal
Physiol Renal Physiol. 2008;294:F1381–F1387. osteodystrophy in patients treated with lanthanum carbonate for two
93. Peng A, Wu T, Zeng C, et al. Adverse effects of simulated hyper- and years. Clin Nephrol. 2008;70:284–295.
hypo-phosphatemia on endothelial cell function and viability. PLoS 119. Wang H, Yoshiko Y, Yamamoto R, et al. Overexpression of fibroblast
One. 2011;6:e23268. growth factor 23 suppresses osteoblast differentiation and matrix
94. Shuto E, Taketani Y, Tanaka R, et al. Dietary phosphorus acutely impairs mineralization in vitro. J Bone Miner Res. 2008;23:939–948.
endothelial function. J Am Soc Nephrol. 2009;20:1504–1512. 120. Murali SK, Roschger P, Zeitz U, et al. FGF23 regulates bone
95. Yilmaz MI, Sonmez A, Saglam M, et al. Comparison of calcium acetate and mineralization in a 1,25(OH)2D3 and Klotho-independent manner.
sevelamer on vascular function and fibroblast growth factor 23 in CKD J Bone Miner Res. 2016;31:129–142.
patients: a randomized clinical trial. Am J Kidney Dis. 2012;59:177–185. 121. Mirza MA, Karlsson MK, Mellström D, et al. Serum fibroblast growth
96. Ayus JC, Mizani MR, Achinger SG, et al. Effects of short daily versus factor-23 (FGF-23) and fracture risk in elderly men. J Bone Miner Res.
conventional hemodialysis on left ventricular hypertrophy and 2011;26:857–864.
inflammatory markers: a prospective, controlled study. J Am Soc 122. Isakova T, Cai X, Lee J, et al. Associations of FGF23 with change in bone
Nephrol. 2005;16:2778–2788. mineral density and fracture risk in older individuals. J Bone Miner Res.
97. Chue CD, Edwards NC, Moody WE, et al. Serum phosphate is associated 2016;31:742–748.
with left ventricular mass in patients with chronic kidney disease: a 123. Morishita K, Shirai A, Kubota M, et al. The progression of aging in klotho
cardiac magnetic resonance study. Heart. 2012;98:219–224. mutant mice can be modified by dietary phosphorus and zinc. J Nutr.
98. Amann K, Tornig J, Kugel B, et al. Hyperphosphatemia aggravates 2001;131:3182–3188.
cardiac fibrosis and microvascular disease in experimental uremia. 124. Ohnishi M, Nakatani T, Lanske B, et al. In vivo genetic evidence for
Kidney Int. 2003;63:1296–1301. suppressing vascular and soft-tissue calcification through the
99. Neves KR, Graciolli FG, dos Reis LM, et al. Adverse effects of reduction of serum phosphate levels, even in the presence of high
hyperphosphatemia on myocardial hypertrophy, renal function, and serum calcium and 1,25-dihydroxyvitamin d levels. Circ Cardiovasc
bone in rats with renal failure. Kidney Int. 2004;66:2237–2244. Genet. 2009;2:583–590.
100. Gutiérrez OM, Januzzi JL, Isakova T, et al. Fibroblast growth factor 23 125. Sitara D, Kim S, Razzaque MS, et al. Genetic evidence of serum
and left ventricular hypertrophy in chronic kidney disease. Circulation. phosphate-independent functions of FGF-23 on bone. PLoS Genet.
2009;119:2545–2552. 2008;4:e1000154.
101. Faul C, Amaral AP, Oskouei B, et al. FGF23 induces left ventricular 126. Ohnishi M, Razzaque MS. Dietary and genetic evidence for phosphate
hypertrophy. J Clin Invest. 2011;121:4393–4408. toxicity accelerating mammalian aging. FASEB J. 2010;24:3562–3571.
102. Grabner A, Amaral AP, Schramm K, et al. Activation of cardiac fibroblast 127. Bergwitz C. Dietary phosphate modifies lifespan in Drosophila. Nephrol
growth factor receptor 4 causes left ventricular hypertrophy. Cell Metab. Dial Transplant. 2012;27:3399–3406.
2015;22:1020–1032. 128. Nguyen TT, Quan X, Hwang KH, et al. Mitochondrial oxidative stress
103. Andrukhova O, Slavic S, Smorodchenko A, et al. FGF23 regulates mediates high-phosphate-induced secretory defects and apoptosis
renal sodium handling and blood pressure. EMBO Mol Med. 2014;6: in insulin-secreting cells. Am J Physiol Endocrinol Metab. 2015;308:
744–759. E933–E941.

762 Kidney International (2016) 90, 753–763


H Komaba and M Fukagawa: Phosphate—a poison for humans? review

129. Jin H, Xu CX, Lim HT, et al. High dietary inorganic phosphate increases Patterns Study (DOPPS): evaluation of possible confounding by
lung tumorigenesis and alters Akt signaling. Am J Respir Crit Care Med. nutritional status. Am J Kidney Dis. 2012;60:90–101.
2009;179:59–68. 139. Cannata-Andía JB, Fernández-Martín JL, Locatelli F, et al. Use of
130. Go AS, Chertow GM, Fan D, et al. Chronic kidney disease and the risks of phosphate-binding agents is associated with a lower risk of mortality.
death, cardiovascular events, and hospitalization. N Engl J Med. Kidney Int. 2013;84:998–1008.
2004;351:1296–1305. 140. Shalhoub V, Shatzen EM, Ward SC, et al. FGF23 neutralization improves
131. Coe LM, Madathil SV, Casu C, et al. FGF-23 is a negative regulator of chronic kidney disease-associated hyperparathyroidism yet increases
prenatal and postnatal erythropoiesis. J Biol Chem. 2014;289:9795–9810. mortality. J Clin Invest. 2012;122:2543–2553.
132. Brancaccio D, Cozzolino M, Gallieni M. Hyperparathyroidism and 141. Lavi-Moshayoff V, Wasserman G, Meir T, et al. PTH increases FGF23
anemia in uremic subjects: a combined therapeutic approach. J Am Soc gene expression and mediates the high-FGF23 levels of experimental
Nephrol. 2004;15(Suppl 1):S21–S24. kidney failure: a bone parathyroid feedback loop. Am J Physiol Renal
133. Rossaint J, Oehmichen J, Van Aken H, et al. FGF23 signaling impairs Physiol. 2010;299:F882–F889.
neutrophil recruitment and host defense during CKD. J Clin Invest.
142. Block GA, Wheeler DC, Persky MS, et al. Effects of phosphate binders in
2016;126:962–974.
moderate CKD. J Am Soc Nephrol. 2012;23:1407–1415.
134. Geara AS, Castellanos MR, Bassil C, et al. Effects of parathyroid hormone
on immune function. Clin Dev Immunol. 2010;2010:418695. 143. Chue CD, Townend JN, Moody WE, et al. Cardiovascular effects of
135. Kir S, Komaba H, Garcia AP, et al. PTH/PTHrP receptor mediates cachexia sevelamer in stage 3 CKD. J Am Soc Nephrol. 2013;24:842–852.
in models of kidney failure and cancer. Cell Metab. 2016;23:315–323. 144. Schiavi SC, Tang W, Bracken C, et al. Npt2b deletion attenuates
136. Kidney Disease: Improving Global Outcomes (KDIGO) CKD-MBD Work hyperphosphatemia associated with CKD. J Am Soc Nephrol. 2012;23:
Group. KDIGO clinical practice guideline for the diagnosis, evaluation, 1691–1700.
prevention, and treatment of chronic kidney disease-mineral and bone 145. Isakova T, Ix JH, Sprague SM, et al. Rationale and approaches to
disorder (CKD-MBD). Kidney Int Suppl. 2009;113:S1–S130. phosphate and fibroblast growth factor 23 reduction in CKD. J Am Soc
137. Isakova T, Gutierrez OM, Chang Y, et al. Phosphorus binders and Nephrol. 2015;26:2328–2339.
survival on hemodialysis. J Am Soc Nephrol. 2009;20:388–396. 146. Bowling CB, Sharma P, Fox CS, et al. Prevalence of reduced estimated
138. Lopes AA, Tong L, Thumma J, et al. Phosphate binder use and mortality glomerular filtration rate among the oldest old from 1988-1994
among hemodialysis patients in the Dialysis Outcomes and Practice through 2005-2010. JAMA. 2013;310:1284–1286.

Kidney International (2016) 90, 753–763 763

You might also like