ArticlePDF AvailableLiterature Review

Hyperphosphatemia of Chronic Kidney Disease

Authors:

Abstract and Figures

Observational studies have determined hyperphosphatemia to be a cardiovascular risk factor in chronic kidney disease. Mechanistic studies have elucidated that hyperphosphatemia is a direct stimulus to vascular calcification, which is one cause of morbid cardiovascular events contributing to the excess mortality of chronic kidney disease. This review describes the pathobiology of hyperphosphatemia that develops as a consequence of positive phosphate balance in chronic kidney disease and the mechanisms by which hyperphosphatemia acts on neointimal vascular cells that are stimulated to mineralize in chronic kidney disease. The characterization of hyperphosphatemia of chronic kidney disease as a distinct syndrome in clinical medicine with unique disordered skeletal remodeling, heterotopic mineralization and cardiovascular morbidity is presented.
Content may be subject to copyright.
Hyperphosphatemia of Chronic Kidney Disease
Keith A Hruska, M.D.
1,2
, Suresh Mathew, M.D.
1
, Richard Lund, M.D.
3
, Ping Qiu
4
, and
Raymond Pratt
4
1
Renal Division, Department of Pediatrics, Washington University, St Louis, Missouri
2
Renal Division, Department of Medicine, Washington University, St Louis, Missouri
3
Creighton University, Omaha, Nebraska
4
Shire Pharmaceuticals, Wayne, PA
Abstract
Observational studies have determined hyperphosphatemia to be a cardiovascular risk factor in
chronic kidney disease. Mechanistic studies have elucidated that hyperphosphatemia is a direct
stimulus to vascular calcification, which is one cause of morbid cardiovascular events contributing
to the excess mortality of chronic kidney disease. This review describes the pathobiology of
hyperphosphatemia that develops as a consequence of positive phosphate balance in chronic kidney
disease and the mechanisms by which hyperphosphatemia acts on neointimal vascular cells that are
stimulated to mineralize in chronic kidney disease. The characterization of hyperphosphatemia of
chronic kidney disease as a distinct syndrome in clinical medicine with unique disordered skeletal
remodeling, heterotopic mineralization and cardiovascular morbidity is presented.
Introduction
Hyperphosphatemia is associated with significant pathophysiology in chronic kidney disease
(CKD). This pathophysiology contributes to the high rates of mortality observed in CKD (1).
Approximately 11–15% of Americans have CKD (2–4), and their risk of death due to a
cardiovascular event related cause is higher than their risk of surviving and needing renal
replacement therapy for end stage kidney disease (ESKD) (1,2,4). The mortality rates of
patients surviving CKD and receiving hemodialysis are extremely high such that a 30 year old
patient with ESKD has a life expectancy similar to a ninety year old with normal renal function
(2). The mechanisms of this excess risk of cardiovascular disease are not completely
understood. The well characterized risks of cardiovascular disease in the general population
do not explain the increased risk in CKD (1,3). Observational studies suggest that the well
known propensity of ESKD patients to develop heterotopic mineralization of soft tissues
including the vasculature is an important component of the cardiovascular risks of ESKD (5,
6). Furthermore, several observational studies demonstrate that hyperphosphatemia is an
independent cardiovascular risk factor in CKD (7–9). Hyperphosphatemia has been linked to
vascular calcification (5,10)(11).
Pathogenesis
To consider the pathogenesis of hyperphosphatemia in CKD, it is useful to review the
mechanisms of phosphate homeostasis (Figure 1). We ingest approximately 1000–1200 mg of
Corresponding Author: Keith A Hruska, M.D., Division of Pediatric Nephrology, Department of Pediatrics, Washington University,
McDonnell Pediatric Research Bldg, Room 5109, Campus Box 8208, 660 S. Euclid Avenue, St. Louis, Mo 63110, Ph 314-286-2772,
Fax 314-286-2894, Hruska_k@kids.wustl.edu.
NIH Public Access
Author Manuscript
Kidney Int. Author manuscript; available in PMC 2009 August 31.
Published in final edited form as:
Kidney Int. 2008 July ; 74(2): 148–157. doi:10.1038/ki.2008.130.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
phosphorus in the average American diet of 2007. Of this, a net of about 800 mg is absorbed
into the exchangeable phosphorus pool. This pool consists of intracellular phosphorus (70%),
the skeletal mineralization front (29%) and the serum phosphorus (<1%). Exit from the
exchangeable pool is through skeletal deposition, renal excretion, and intestinal secretion
(Figure 1). Exit from the exchangeable pool into the skeleton is matched by entry from the
skeleton into the pool in adulthood, and therefore, we do not normally think of the skeleton as
a key contributor to the level of phosphorus concentration in the pool, but in chronic kidney
disease, as we will show below, it is very important. Regulation of phosphorus excretion by
the kidney is the key mechanism of maintaining phosphate balancein normal day to day life.
Kidney injury impairs the ability of mammals to maintain phosphorus balance, and in human
chronic kidney disease, phosphorus homeostasis is lost and positive phosphate balance occurs
in the later stages (4 and 5) of kidney diseases (12,13). Loss of phosphorus homeostasis due
to excretion failure in chronic kidney disease results in hyperphosphatemia (14) due to positive
balance increasing the concentration in the exchangeable phosphorus pool, often when the pool
size is reduced as in the adynamic bone disorder (Figure 2). Surprisingly and not generally
adequately considered, the skeleton contributes to hyperphosphatemia in CKD and ESKD
through the effects of disordered bone remodeling. There are multiple skeletal remodeling
disorders discussed below in CKD, but all of them are associated with excess bone resorption
compared to bone formation. Thereby, they contribute to hyperphosphatemia and effectively
block the skeleton from exerting its normal reservoir function when positive phosphate balance
occurs.
The normal function of the skeleton as a reservoir when phosphate balance is positive is seen
in several syndromes of hyperphosphatemia in mammalian pathophysiology (Table 1). All of
these except immobilization and chronic kidney disease are associated with increased skeletal
mass and mineralization due to phosphorus deposition into the skeletal storage reservoir (see
section on “Other Hyperphosphatemic Syndromes”). In chronic kidney disease, there is a
complex set of losses and adaptations in skeletal function that produce bone disorders that
complicate the state (see section on “Renal Osteodystrophy”). Recent discoveries characterize
all forms of skeletal function disorder in chronic kidney disease as having excess bone
resorption rates compared to bone formation rates (see section on “Osteoporosis in chronic
kidney disease”). Therefore, the skeleton is contributing to hyperphosphatemia in chronic
kidney disease, and the reservoir function of the skeleton that is supposed to act in the presence
of positive phosphorus balance is blocked. The outcome of this change in physiology to a new
pathophysiology requires that a new phosphate reservoir for the positive balance is established.
This new reservoir is soft tissue organs including the vasculature (Figure 2). The problem with
establishing the new reservoirs of phosphate storage is that they produce disease.
Regulation of phosphate balance during CKD before loss of balance occurs is complex. Loss
of calcitriol production capacity is an important factor leading to a decrease in Ca absorption,
hypocalcemia and stimulation of parathyroid hormone (PTH) secretion (Figure 3). The increase
in PTH levels reduces the fraction of the filtered phosphate load and maintains phosphate
excretion at normal levels despite the reduction in the filtered load of phosphorus due to the
decrease in glomerular filtration. The decrease in calcitriol production also represents a
decrease in the signal to the osteocytes and osteoblasts for the production of fibroblast growth
factor 23 (FGF23) (15). However, the tendency for decreased production is overcome by the
fact that FGF23 is normally catabolized by glomerular filtration and proximal tubular
degradation. Thus, the protein levels increase in CKD as the GFR decreases (16,17). This is a
second potent stimulus to phosphorus excretion as kidney function is diminished. The relative
contributions PTH and FGF23 make to phosphate homeostasis during CKD have not been
determined, but in the absence of either homeostasis is lost. A third stimulus for increased
phosphate excretion is phosphate itself which also potently inhibits the activity of the proximal
Hruska et al. Page 2
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
tubular sodium dependent Pi transport proteins (Na/Pi co-transporters, NaPi2a and NaPi2c
(18)(19)).
The sodium dependent phosphate transport protein, NaPi2a (approximately 70% of proximal
tubular phosphate transport), is regulated mainly by endocytic vesicle cycling in an out of the
apical membrane of the epithelial cell (20,21)(19). It is directly regulated by Pi concentrations
(20,21)(18). Low Pi increases decreases vesicular retrieval from the plasma membrane while
high Pi concentrations stimulate it (20). A late effect of Pi concentration is a slight change in
gene transcription of NaPi2a with greater effects on protein levels, and stimulation of
transcription of NaPi2a associated proteins (such as (diphor-1) by low and inhibition by high
Pi (20,22). Similar experiments have not been performed for the minor, but very important
NaPi2c of the proximal tubule. NaPi2b of the duodenal enterocyte is a transcriptional target of
the vitamin D receptor (VDR) and dietary phosphorus (23), but calcitriol deficiency or
hyperphosphatemia in CKD do not result in decreased intestinal phosphate absorption, because
the decrease in active duodenal transport is compensated by passive transport through
enterocyte paracellular pathways in the rest of the intestine.
One of the new reservoirs for phosphate deposition established when excretion is no longer
sufficient to maintain balance, the vasculature (Figure 2), is especially disease causing.
Vascular calcification in CKD is not well tolerated as it produces blood vessel stiffness (Figure
4). There are two forms of vascular calcification prominent in CKD, calcification of
atherosclerotic neointimal plaques and arterial medial calcification. CKD markedly stimulates
both forms. The atherosclerotic calcification is especially appreciated in the coronary arteries
as it is measured by the increasingly popular imaging techniques for determining coronary
artery calcification (24,25). However, arterial medial calcification is as clinically important,
as it is the likely the most important factor in vascular stiffness and increased pulse pressure
in CKD.
In order to study the mechanisms of CKD stimulated vascular calcification, we developed a
translational animal model. We started with a model of atherosclerosis that develops cardiac
valvular and atherosclerotic neointimal plaque calcification, the low density lipoprotein
receptor deficient mouse (LDLR/) fed a high fat diet. To the model we added ablation
induced kidney failure, and demonstrated marked stimulation of aortic atherosclerotic
calcification (26). We discovered that bone morphogenetic protein-7 (BMP-7) prevented the
development of CKD stimulated vascular calcification (26). Furthermore, the high fat fed CKD
mice exhibited hyperphosphatemia that was corrected by BMP-7 (27). In investigating the
mechanism of hyperphosphatemia correction by BMP-7 we found that bone formation was
stimulated correcting the adynamic bone disorder that complicates the kidney failure in these
mice (27). Renal phosphate excretion was not increased, and we questioned how much of the
BMP-7 effect on vascular calcification was due to bone formation induced correction of
hyperphosphatemia. To examine this, we added phosphate binders to the high fat diet in an
attempt to isolate hyperphosphatemia correction as a single entity separate from the other
actions of BMP-7. To our surprise, phosphate binders were very effective in preventing
vascular calcification. We first used CaCO3 (27), but subsequently have studied sevelamer
carbonate (Figure 5) (28) and LaCO3 with similar results.
The LDLR/ high fat fed mouse is characterized by obesity, hypertension and insulin
resistance that progresses to diabetes and severe hypercholesterolemia. Thus, the mouse model
is relevant to the human metabolic syndrome, and the development of kidney disease in obese
diabetics. Even the renal osteodystrophy complicating CKD, the adynamic bone disorder, is
the same as observed in patients with diabetic nephropathy. The vascular calcification of the
model was discovered by Towler and Semenkovich (29). They found that osteoblastic
transcriptional activity was present in the aorta, and their model is the starting point to which
Hruska et al. Page 3
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
we add CKD. At the beginning, in other words our high fat fed sham operated animals,
expressed BMP-2, BMP-4, Runx2, Msx2, osteocalcin and osteopontin in the vasculature,
especially the aorta. This is relevant because several investigators have demonstrated that the
vasculature of patients with CKD/ESKD expresses osteoblastic markers (30–33). However,
our high fat fed sham operated animals had a low level of vascular calcification that was
stimulated two to four fold by the induction of CKD (26).
To further investigate mechanisms of atherosclerotic calcification, we developed an in vitro
model to study in cell culture with the strategy of confirming discoveries made in vitro using
our animal model. We began by obtaining primary cultures of human vascular smooth muscle
cells (hVSMC) from areas of aortic atherosclerotic plaques, which were expanded and then
exposed to an increase in media phosphorus concentrations (Figure 6). We demonstrated that
the starting tissue and the primary cultures in regular media expressed BMP-2, BMP-4, Runx2,
Msx2, osteocalcin and osteopontin. Thus, it appeared that BMP-2 and 4 as bone morphogens
were stimulating an osteoblastic differentiation program in vascular cells directed by the
osteoblast specific transcription factors. This data is in agreement with other investigators
(34), including those who reported increased levels of BMP-2 and 4 in atherosclerotic lesions
(35,36). However, the primary hVSMC cultures did not mineralize the extracellular matrix as
osteoblast cultures do (Figure 6) (37). When media phosphorus was increased by 1 or 2 mM
to 2 or 3 mM (equal to a serum phosphorus of 6 to 9 mg/dl), heavy matrix mineralization
ensued. Analysis of the osteoblastic transcription program revealed that the very low levels of
osterix expression in the starting cultures were increased several fold by the increase in media
phosphorus. Blocking the increase in osterix expression in high phosphate media prevented
matrix mineralization (38). Both our translational model in vivo and our cell culture model in
vivo represent mineralization due to the atherosclerotic process. We observe mainly neointimal
calcification in vivo, and the medial calcification we observe is in proximity to atherosclerotic
plaques.
When we examined our high fat fed LDLR/ aortas from the various groups of animals, we
found that the sham operated high fat fed animals had low or undetectable levels of osterix
expression that were increased several fold when CKD was induced (38). Most importantly,
treatment of CKD high fat fed hyperphosphatemic animals with phosphate binders inhibited
osterix expression (Figure 7) (38). As expected without a critical osteoblastic transcription
factor, matrix mineralization (neointimal calcification) was severely compromised similar to
results shown in Figure 5.
The mechanism of phosphorus stimulation of matrix mineralization in vitro has been studied
by Jono et al (39) and Li et al (40) in VSMC and by Beck et al in osteoblastic cells (41)(42).
These authors have demonstrated that the effect of media phosphorus was through activation
of ERK1/2, and it was blocked by an inhibitor of sodium dependent phosphate transport
proteins, phosphonoformic acid (PFA). However the effects of PFA are relatively specific to
the type 2 Na/Pi cotransporters (43). A sodium dependent phosphate transport protein of the
VSMC is Pit-1, and an RNAi to Pit-1 also inhibited the actions of high phosphorus media
(40,43). The effects of PFA in VSMC mineralization may have been due to the role of
phosphonates to inhibit phosphate crystal formation, although NaPi2a has recently been found
in osteoblast-like cells (43,44). Other recent studies of Pi transport in VSMC indicate that Pit-1
and Pit-2 account for Na-dependent transport, which is only modestly inhibited by high
concentrations of PFA(45). In subsequent studies reported in abstract form at this writing, the
authors demonstrate that these concentrations of PFA induce cytotoxicity (46). Despite the
recent clarification regarding effects of PFA on VSMC, the studies with RNAi to Pit-1 (45)
(40) indicate that blocking the actions of high phosphorus culture media in vitro are similar to
the effects of lowering the serum phosphorus in vivo, that of inhibiting osteoblastic stimulation
of matrix mineralization.
Hruska et al. Page 4
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Thus, phosphorus is more than a stimulator of vascular calcification acting through an elevated
calcium-phosphorus product in CKD and ESKD. It is a signaling molecule serving to complete
osteoblastic differentiation in the aorta, and an important component of the action of CKD to
stimulate atherosclerotic calcification. The results of the translational animal studies and the
studies in vitro just discussed are in agreement with a new clinical consensus that has lead to
renaming of renal osteodystrophy by the KDIGO Foundation as the chronic kidney disease
mineral bone disorder (CKD-MBD) in recognition of the roles of the skeleton in
hyperphosphatemia and vascular calcification (47).
Other Hyperphosphatemic Syndromes
Hyperphosphatemic syndromes occur due to a variety of causes (Table 1). Hyperphosphatemia
due to intravenous hyperalimentation (TPN) in an immobilized patient is the most common
cause of hyperphosphatemia in clinical medicine. Here the phosphate content of the TPN
prescription equivalent to normal dietary intake is associated with hyperphosphatemia leading
to removal of phosphate from the parenteral alimentation. Sometimes removal of phosphate
intake does not correct the hyperphosphatemia and patients require inhibition of bone
resorption to correct the hyperphosphatemia. The immobilization syndrome has at least two
key features in common with the hyperphosphatemia of CKD. The first is the severe inhibition
of bone formation associated with immobilization that resembles the inhibition of bone
formation in the adynamic bone disorder of CKD/ESKD. The second is the excess bone
resorption contributing to the hyperphosphatemia. The major difference from the
hyperphosphatemia of CKD may be duration of the syndrome which in the case of
immobilization is insufficient to calcify the vasculature and produce cardiac events due to the
hyperphosphatemia.
Transcellular shifts of phosphate associated with catabolism, tumor lysis or rhabdomyolysis
are also relatively common causes of hyperphosphatemia (Table 1). Hyperphosphatemic
syndromes due to decreased renal excretion besides chronic kidney disease are uncommon in
clinical medicine. Of these, FGF23 deficiency genetically produced in the mouse is especially
instructive. FGF23 null mice exhibit hyperphosphatemia, elevated calcitriol levels and
increased skeletal mineralization including chondrosseous junctions, the primary spongiosa,
and heterotopic sites such as various organs and the vasculature (48). The mice die at about
thirteen weeks due to cardiovascular complications. The phenotype of the FGF23 null mice is
rescued by the production of a double genetic deficiency for FGF23 and 25
hydroxycholecalciferol 1 alpha hydroxylase (49). This demonstrates the role of excess
calcitriol in the FGF23 null phenotype. Interestingly, the vascular calcification of the FGF23
null phenotype is also rescued by low phosphate diets (50).
FGF23 is a recently discovered phosphaturic hormone, responsible for autosomal dominant
hypophosphatemic rickets through an activating mutation (51). The recent discovery that
inactivating mutations cause the rare human disease of familial tumoral calcinosis completes
the story told by the FGF23 null mice due to similarities in the phenotypes. The premature
cardiac death in FGF23 deficient mice is reminiscent of the early coronary artery disease in
Klotho mice (52,53). Klotho has recently been discovered to function as a co-receptor for
FGF23 in the proximal tubule (54), and Klotho inactivation has also been found to cause
familial tumoral calcinosis (55). Thus, the role of hyperphosphatemia in stimulating heterotopic
mineralization has clearly been demonstrated in mice and men. The unique aspect of
hyperphosphatemia in CKD is that the skeletal response to hyperphosphatemia, increased
deposition of phosphate, is blocked.
Hruska et al. Page 5
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Renal Osteodystrophy
There are several disorders of bone remodeling that complicate CKD and ESKD. These have
been characterized histomorphometrically and correlated to PTH levels to estimate bone
turnover rates. This process has to be replaced in order to monitor the skeleton more closely
as is necessary to optimize cardiovascular therapy in CKD. Our current practice of using PTH
levels to correlate with bone turnover is insufficient in terms of sensitivity and fails to detect
excess bone resorption. Presently, high PTH levels (>500 pg/ml with an intact hormone assay)
are thought to indicate increased skeletal remodeling due to secondary hyperparathyroidism.
While this is generally correct, it does not measure the impact of elevated remodeling in CKD
on bone mass or strength. The elevated remodeling associated with secondary
hyperparathyroidism, produces an osteoblast phenotype that has reduced secretion of type 1
collagen and increased production of RANK ligand (RANKL), the critical osteoclast
differentiation factor. This results in bone resorption outpacing bone formation. In addition,
the high remodeling rates are characterized by insufficient replacement of newly formed
atypical “woven” bone with bone formed on collagen lamellae. Thus, even with normal bone
mass, skeletal frailty may be problematic in high turnover osteodystrophy in CKD/ESKD.
A secondary effect of hyperphosphatemia in CKD is stimulation of nodular hyperplasia of the
parathyroid glands (56–59). Hyperplastic chief cells from the nodular areas are of clonal origin
demonstrating less of cell cycle control (60). Clinically, this phenomenon accounts for the loss
of control of the adaptive function of secondary hyperparathyroidism in CKD, and results in
very elevated PTH levels that are difficult to treat and clinical manifestations of severe
secondary hyperparathyroidism. This often leads to parathyroidectomy in order to treat the
clinical complications of the uncontrolled PTH levels.
With prevalent administration of high doses of vitamin D analogs in CKD/ESKD, a newer
(discovered in the 1980’s) low turnover form of renal osteodystrophy has become increasingly
common, termed the adynamic bone disorder (ABD) (61–63). The ABD was originally thought
to be due to suppression of osteoblast function with high doses of vitamin D analogs (61,63).
The finding that vitamin D analogs stimulate, not inhibit, bone formation and osteoblast
function has put this contention to rest (64–66). What is likely the case is that the negative
effects of CKD on skeletal anabolism are uncovered by suppression of PTH. This demonstrates
that higher than normal PTH levels are required to maintain bone remodeling in CKD (67).
Many different mechanisms of resistance to the actions of PTH in CKD have been proposed
including desensitization of the PTH receptor by persistent high PTH levels (68). While this
is likely, another mechanism is probably central and critical. The endosteal osteoblast forms
the niche of the hematopoietic stem cell (HSC) (69–71), and loss of osteoblast surface and
number as a result of loss of skeletal anabolism due to kidney injury threatens hematopoiesis.
There are three principles that regulate the HSC niche, the bone morphogenetic proteins
(BMPs), the Wnts, and PTH (69,71). Loss of BMP or Wnt influence as a result of kidney injury
would lead to a need for higher levels of PTH to protect niche function and skeletal remodeling.
The point that is well established is that in ESKD, higher than normal PTH levels are required
to maintain normal rates of skeletal remodeling, i.e. PTH levels recommended in the KDOQI
guidelines.
The loss of skeletal remodeling after renal injury produces a mechanism of increasing PTH
secretion due to the decrease in the exchangeable phosphorus pool size (Figure 2). Now boluses
of phosphorus, as with meals, distribute in a smaller pool and produce intermittent short-lived
stimuli to increased PTH secretion either through secondary effects on serum Ca or due to
direct effects of transient increased in serum phosphorus that may be within the normal range.
This all occurs in the face of normal fasting levels of Ca and PO4. Renal osteodystrophy begins
early in CKD. It is often first detected by elevated PTH levels in the face of normal Ca, PO4
Hruska et al. Page 6
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
and calcitriol levels (14). In this setting, the cause of the increase in PTH levels is a decrease
in the exchangeable PO4 pool size due to a loss of the skeletal mineralization front that occurs
when the normal stimulus to bone formation is lost due to kidney injury. The role of FGF23
which is increased before changes in Ca, Pi or calcitriol on PTH secretion are unclear despite
recent studies (72). The underpinning of renal osteodystrophy by a loss of skeletal anabolic
potential due to kidney injury is poorly appreciated, but it is often uncovered when PTH levels
are suppressed to near normal levels, producing the adynamic bone disorder (62,63,73).
Osteoporosis in chronic kidney disease
The balance between bone formation and resorption may be either negative or positive in CKD.
When positive, osteosclerosis results, but this is rare in modern medicine. In the case of negative
bone balance, bone loss occurs in cortical and cancellous bone and is more rapid when bone
turnover is high. In those cases, bone densitometry will detect osteopenia or osteoporosis. The
prevalence of osteoporosis in the population with CKD exceeds the prevalence in the general
population (74–76). Osteoporosis is observed in CKD before dialysis is required for end stage
kidney failure (77). When bone turnover is high, as in secondary hyperparathyroidism with
osteitis fibrosa, bone resorption rates are in excess of bone formation and osteopenia
progressing to osteoporosis may result. When bone turnover is low, although both bone
formation rates and bone resorption may be reduced, resorption is in excess and loss of bone
mass occurs. Thus, osteoporosis may be observed with either high turnover (77–80) or low
turnover (81) forms of osteodystrophy. When bone resorption exceeds bone formation rates in
CKD, phosphorus and calcium release contribute to hyperphosphatemia and hypercalcemia.
The increase in skeletal mineral deposition that should result from hyperphosphatemia or
hypercalcemia is blocked, and heterotopic mineralization is stimulated, especially in the
vasculature. The failure of the skeleton to absorb positive phosphate balance in CKD is an
important stimulus to heterotopic mineralization, and links the skeleton and osteoporosis in
CKD to cardiovascular events and mortality. This link between osteoporosis and vascular
calcification, now partly defined in CKD, is not specific to CKD. Type 2 osteoporosis is
strongly associated with vascular calcification, and perhaps, the mechanisms defined in CKD
related to the serum phosphorus as a signal and positive phosphate balance also apply.
The discussion of osteoporosis in CKD above is focused on osteoporosis caused by CKD itself.
However, many patients with CKD have osteoporosis independent of CKD. These patients
may be elderly or may have post-menopausal osteoporosis. In addition, it is clear that gonadal
hormone deficiency, as in post-menopausal osteoporosis, is also caused by CKD and is another
factor in the pathogenesis of osteoporosis in CKD. Thus, osteoporosis in CKD presents a
difficult differential diagnosis between the type 2 osteoporosis of aging, gonadal hormone
deficiency, and excess bone resorption associated with the CDK-BMD.
Conclusion
In conclusion, hyperphosphatemia in CKD is a distinct syndrome. It represents one component
of the increased risk of cardiovascular disease in CKD that has been successfully analyzed.
Hyperphosphatemia in CKD represents a signal that heterotopic sites of mineralization are
being used to compensate for the failure of reservoir function of the skeleton in positive
phosphate balance. In fact, hyperphosphatemia itself is one of the signals activating heterotopic
deposition sites, and functions as a signaling molecule in stimulating atherosclerotic neointimal
mineralization that is markedly increased in CKD. Unique features of hyperphosphatemia in
CKD, especially the failure of the skeletal reservoir function, qualify it as a distinct syndrome
characterized by phosphate excretion failure, contribution of the skeleton to
hyperphosphatemia, heterotopic mineralization including the vasculature, and severe
cardiovascular disease leading to morbid cardiac events and often to demise.
Hruska et al. Page 7
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Acknowledgments
We would like to thank Helen Odle and Frank Strebeck for administrative and technical support, Celina Mount of
CDRAC for artistic support, and Jose Menoyo (Genzyme) for valuable discussion.
Disclosure:
The manuscript and the studies reported here were supported by NIH grants DK070790 and AR41677, and by grants-
in-aid from Genzyme and Shire. KAH received consultation fees from Genzyme and Shire Pharmaceutical. PQ and
RP are employees of Shire Pharmaceuticals. SM received consultation fees from Genzyme, and RL received
consultation fees from Abbott.
References
1. Go AS, Chertow GM, Fan D, et al. Chronic kidney disease and the risks of death, cardiovascular events,
and hospitalization. New Engl J Med 2004;351:1296–1305. [PubMed: 15385656]
2. Foley RN, Parfrey PS, Sarnak MJ. Clinical epidemiology of cardiovascular disease in chronic renal
disease. Am JKid Dis 1998;32:S112–S119. [PubMed: 9820470]
3. Sarnak MJ, Levey AS, Schoolwerth AC, et al. Kidney Disease as a Risk Factor for Development of
Cardiovascular Disease: A Statement From the American Heart Association Councils on Kidney in
Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and
Prevention. Hypertension 2003;42:1050–1065. [PubMed: 14604997]
4. Coresh J, Selvin E, Stevens LA, et al. Prevalence of Chronic Kidney Disease in the United States. J
Am Med Assoc 2007;298:2038–2047.
5. London GM, Guerin AP, Marchais SJ, et al. Arterial media calcification in end-stage renal diseases:
impact on all-cause and cardiovascular mortality. Nephrol Dial Transplant 2003;18:1731–1740.
[PubMed: 12937218]
6. Raggi P, Boulay A, Chasan-Taber S, et al. Cardiac calcification in adult hemodialysis patients. A link
between end-stage renal disease and cardiovascular disease? J Am Coll Cardiol 2002;39:695–701.
[PubMed: 11849871]
7. Block GA, Hulbert-Shearon TE, Levin NW, et al. Association of serum phosphorus and calcium X
phosphate product with mortality risk in chronic hemodialysis patients: a national study. Am J Kidney
Dis 1998;31:607–617. [PubMed: 9531176]
8. Kestenbaum B, Sampson JN, Rudser KD, et al. Serum phosphate levels and mortality risk among
people with chronic kidney disease. J Am Soc Nephrol 2005;16:520–528. [PubMed: 15615819]
9. Slinin Y, Foley RN, Collins AJ. Calcium, Phosphorus, Parathyroid Hormone, and Cardiovascular
Disease in Hemodialysis Patients: The USRDS Waves 1, 3, and 4 Study. J Am Soc Nephrol
2005;16:1788–1793. [PubMed: 15814832]
10. Marchais SJ, Metivier F, Guerin AP, et al. Association of hyperphosphataemia with haemodynamic
disturbances in end-stage renal disease. Nephrol Dial Transplant 1999;14:2178–2183. [PubMed:
10489228]
11. Block GA, Raggi P, Bellasi A, et al. Mortality effect of coronary calcification and phosphate binder
choice in incident hemodialysis patients. Kidney Int 2007;71:438–441. [PubMed: 17200680]
12. Slatopolsky E, Robson AM, Elkan I, et al. Control of phosphate excretion in uremic man. J Clin Invest
1968;47:1865–1874. [PubMed: 5666116]
13. Slatopolsky E, Gradowska L, Kashemsant C. The control of phosphate excretion in uremia. J Clin
Invest 1966;45:672–677. [PubMed: 5935357]
14. Craver L, Marco MP, Martinez I, et al. Mineral metabolism parameters throughout chronic kidney
disease stages 1–5--achievement of K/DOQI target ranges. Nephrol Dial Transplant 2007;22:1171–
1176. [PubMed: 17205962]
15. Liu S, Tang W, Zhou J, et al. Fibroblast Growth Factor 23 Is a Counter-Regulatory Phosphaturic
Hormone for Vitamin D. J Am Soc Nephrol 2006;17:1305–1315. [PubMed: 16597685]
16. Mayan H, Vered I, Mouallem M, et al. Pseudohypoaldosteronism Type II: Marked Sensitivity to
Thiazides, Hypercalciuria, Normomagnesemia, and Low Bone Mineral Density. JClin Endo Metab
2002;87:3248–3254.
Hruska et al. Page 8
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
17. Pande S, Ritter CS, Rothstein M, et al. FGF-23 and sFRP-4 in chronic kidney disease and post-renal
transplantation. Nephron Physiol 2006;104:23–32.
18. Takahashi F, Morita K, Katai K, et al. Effects of dietary Pi on the renal Na+-dependent Pi transporter
NaPi-2 in thyroparathyroidectomized rats. Biochem J 1998;333:175–181. [PubMed: 9639577]
19. Miyamoto K-I, Segawa H, Ito M, et al. Physiological regulation of renal sodium-dependent phosphate
cotransporters. Japanese J of Physiology 2004;54:93–102.
20. Murer H, Hernando N, Forster I, et al. Regulation of Na/Pi transporter in the proximal tubule. Annu
Rev Physiol 2003;65:531–542. [PubMed: 12517995]
21. Levi M, Kempson SA, Lotscher M, et al. Molecular regulation of renal phosphate transport. J
Membrane Biol 1996;154:1–9. [PubMed: 8881022]
22. Custer M, Spindler B, Verrey F, et al. Identification of a new gene product (diphor-1) regulated by
dietary phosphate. Am J Physiol 1997;273:F801–F806. [PubMed: 9374845]
23. Hattenhauer O, Traebert M, Murer H, et al. Regulation of small intestinal Na-P(i) type IIb
cotransporter by dietary phosphate intake. Am J Physiol 1999;277:756–62.
24. Wang L, Jerosch-Herold M, Jacobs J, et al. Coronary Artery Calcification and Myocardial Perfusion
in Asymptomatic Adults: The MESA (Multi-Ethnic Study of Atherosclerosis). J Am Coll Cardiol
2006;48:1018–1026. [PubMed: 16949496]
25. Pletcher MJ, Tice JA, Pignone M, et al. Using the Coronary Artery Calcium Score to Predict Coronary
Heart Disease Events: A Systematic Review and Meta-analysis. Arch Int Med 2004;164:1285–1292.
[PubMed: 15226161]
26. Davies MR, Lund RJ, Hruska KA. BMP-7 is an efficacious treatment of vascular calcification in a
murine model of atherosclerosis and chronic renal failure. J Am Soc Nephrol 2003;14:1559–1567.
[PubMed: 12761256]
27. Davies MR, Lund RJ, Mathew S, et al. Low turnover osteodystrophy and vascular calcification are
amenable to skeletal anabolism in an animal model of chronic kidney disease and the metabolic
syndrome. J Am Soc Nephrol 2005;16:917–928. [PubMed: 15743994]
28. Mathew S, Lund R, Strebeck F, et al. Reversal of the adynamic bone disorder and decreased vascular
calcification in chronic kidney disease by sevelamer carbonate therapy. J Am Soc Nephrol
2007;18:122–130. [PubMed: 17182886]
29. Towler DA, Bidder M, Latifi T, et al. Diet-induced diabetes activates an osteogenic gene regulatory
program in the aortas of low density lipoprotein receptor-deficient mice. J Biol Chem
1998;273:30427–30434. [PubMed: 9804809]
30. Demer LL. A skeleton in the atherosclerosis closet. Circulation 1995;92:2029–2032. [PubMed:
7554176]
31. Moe SM, Duan D, Doehle BP, et al. Uremia induces the osteoblast differentiation factor Cbfa1 in
human blood vessels. Kidney Int 2003;63:1003–1011. [PubMed: 12631081]
32. Fischer JW, Steitz SA, Johnson PY, et al. Decorin Promotes Aortic Smooth Muscle Cell Calcification
and Colocalizes to Calcified Regions in Human Atherosclerotic Lesions. Arterioscler Thromb Vasc
Biol 2004;24:2391–2396. [PubMed: 15472131]
33. Shanahan CM, Cary NRB, Metcalfe JC, et al. High expression of genes for calcification-regulating
proteins in human atherosclerotic placques. J Clin Invest 1994;93:2393–2402. [PubMed: 8200973]
34. Li X, Yang HY, Giachelli CM. BMP-2 promotes phosphate uptake, phenotypic modulation, and
calcification of human vascular smooth muscle cells. Atherosclerosis. In Press, Corrected Proof:
35. Boström K, Watson KE, Horn S, et al. Bone morphogenetic protein expression in human
atherosclerotic lesions. J Clin Invest 1993;91:1800–1809. [PubMed: 8473518]
36. Dhore CR, Cleutjens J, Lutgens E, et al. Differential expression of bone matrix regulatory proteins
in human atherosclerotic plaques. Arterioscler Thromb Vasc Biol 2001;21:1998–2003. [PubMed:
11742876]
37. Chaudhary LR, Hofmeister AM, Hruska KA. Differential growth factor control of bone formation
through osteoprogenitor differentiation. Bone 2004;34:402–411. [PubMed: 15003788]
38. Mathew S, Tustison K, Sugatani T, et al. The mechanism of phosphorus as a cardiovascular risk factor
in chronic kidney disease. J Am Soc Nephrol. 2007
Hruska et al. Page 9
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
39. Jono S, McKee MD, Murry CE, et al. Phosphate regulation of vascular smooth muscle cell
calcification. Circ Res 2000;87:e10–e17. [PubMed: 11009570]
40. Li X, Yang HY, Giachelli CM. Role of the Sodium-Dependent Phosphate Cotransporter, Pit-1, in
Vascular Smooth Muscle Cell Calcification. Circ Res 2006;98:905–912. [PubMed: 16527991]
41. Beck GR Jr, Zerler B, Moran E. Phosphate is a specific signal for induction of osteopontin gene
expression. Proc Natl Acad Sci 2000;97:8352–8357. [PubMed: 10890885]
42. Beck GR Jr. Inorganic phosphate as a signaling molecule in osteoblast differentiation. J Cell Biochem
2003;90:234–243. [PubMed: 14505340]
43. Virkki LV, Biber J, Murer H, et al. Phosphate transporters: a tale of two solute carrier families. AJP
-Renal Physiology 2007;293:F643–F654. [PubMed: 17581921]
44. Gray RW, Caldas AE, Wilz DR, et al. Metabolism and excretion of 3H-1,25(OH)2 vitamin D3 in
healthy adults. J Clin Endocrinol Metab 1978;46:756–750. [PubMed: 263717]
45. Villa-Bellosta R, Bogaert YE, Levi M, et al. Characterization of Phosphate Transport in Rat Vascular
Smooth Muscle Cells: Implications for Vascular Calcification. Arterioscler Thromb Vasc Biol
2007;27:1030–1036. [PubMed: 17322102]
46. Villa-Bellosta R, Bogaert Y, Levi M, et al. Toxicity of phosphonoformic acid in vascular smooth
muscle cells: relationship to vascular calcification. FASEB J 2007;21:A1244–A124a.
47. Moe S, Drueke T, Cunningham J, et al. Definition, evaluation, and classification of renal
osteodystrophy: A position statement from Kidney Disease: Improving Global Outcomes (KDIGO).
Kidney Int 2006;69:1945–1953. [PubMed: 16641930]
48. Sitara D, Razzaque MS, Hesse M, et al. Homozygous ablation of fibroblast growth factor-23 results
in hyperphosphatemia and impaired skeletogenesis, and reverses hypophosphatemia in Phex-
deficient mice. Matrix Biol 2004;23:421–32. [PubMed: 15579309]
49. Razzaque MS, Sitara D, Taguchi T, et al. Premature aging-like phenotype in fibroblast growth factor
23 null mice is a vitamin D-mediated process. FASEB J 2006;05-5432fje
50. Stubbs JR, Liu S, Tang W, et al. Role of Hyperphosphatemia and 1,25 Dihydroxyvitamin D in
Vascular Calcification and Mortality in Fibroblastic Growth Factor 23 Null Mice. J Am Soc Nephrol
2007;18:2116–2124. [PubMed: 17554146]
51. White KE, Lorenz B, Evans WE, et al. Autosomal dominant hypophosphatemic Rickets is caused by
mutations in a novel gene, FGF23, that shares homology with the fibroblast growth factor family. J
Bone Miner Res 2000;15:S153.
52. Arking DE, Becker DM, Yanek LR, et al. KLOTHO allele status and the risk of early-onset occult
coronary artery disease. Am J Hum Genet 2003;72:1154–1161. [PubMed: 12669274]
53. Arking DE, Krebsova A, Macek M Sr, et al. Association of human aging with a functional variant of
klotho. Proc Natl Acad Sci 2002;99:856–861. [PubMed: 11792841]
54. Kurosu H, Ogawa Y, Miyoshi M, et al. Regulation of fibroblast growth factor-23 signaling by Klotho.
J Biol Chem 2006;281:6120–6123. [PubMed: 16436388]
55. Ichikawa S, Imel EA, Kreiter ML, et al. A homozygous missense mutation in human KLOTHO causes
severe tumoral calcinosis. J Clin Invest 2007;117:2684–2691. [PubMed: 17710231]
56. Dusso AS, Lu Y, Pavlopoulos T, et al. A role of enhanced expression of transforming growth factor
alpha (TGF-alpha) in the mitogenic effect of high dietary phosphorus on parathyroid cell growth in
uremia. J Am Soc Nephrol 1999;10:617.
57. Fukuda N, Tanaka H, Tominaga Y. Decreased 1,25 dihydroxyvitamin D3 receptor density is
associated with a more severe form of parathyroid hyperplasia in chronic uremic patients. J Clin
Invest 1993;92:1436–1443. [PubMed: 8397225]
58. Naveh-Many T, Rahamimov R, Livni N, et al. Parathyroid cell proliferation in normal and chronic
renal failure rats. The effects of calcium, phosphate, and vitamin D. J Clin Invest 1995;4:1786–1793.
[PubMed: 7560070]
59. Denda M, Finch J, Slatopolsky E. Phosphorus accelerates the development of parathyroid hyperplasia
and secondary hyperparathyroidism in rats with renal failure. Am J Kid Dis 1996;28:596–602.
[PubMed: 8840952]
60. Tominaga Y, Kohara S, Namii Y, et al. Clonal analysis of nodular parathyroid hyperplasia in renal
hyperparathyroidism. World J Surg 1996;20:744–752. [PubMed: 8678945]
Hruska et al. Page 10
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
61. Salusky IB, Ramirez JA, Oppenheim WL, et al. Biochemical markers of renal osteodystrophy in
pediatric patients undergoing CAPD/CCPD. Kidney Int 1994;45:253–258. [PubMed: 8127016]
62. Hercz G, Pei Y, Greenwood C, et al. Aplastic osteodystrophy without aluminum: The role of
“suppressed” parathyroid function. Kidney Int 1993;44:860–866. [PubMed: 8258962]
63. Salusky IB, Goodman WG, Kuizon BD. Implications of intermittent calcitriol therapy on growth and
secondary hyperparthyroidism. Pediatr Nephrol 2000;14:641–645. [PubMed: 10912534]
64. Mathew S, Lund RJ, Strebeck F, et al. Effects of paricalcitol therapy in the adynamic bone disorder.
J Am Soc Nephrol 2005;16:32A.
65. Hendy GN, Hruska KA, Mathew S, et al. New insights into mineral and skeletal regulation by active
forms of vitamin D. Kidney Int 2006;69:218–223. [PubMed: 16408109]
66. Panda DK, Miao D, Bolivar I, et al. Inactivation of the 25-hydroxyvitamin D 1α hydroxylase and
vitamin D receptor demonstrates independent and interdependent effects of calcium and vitamin D
on skeletal and mineral homeostasis. J Biol Chem 2004;279:16754–16766. [PubMed: 14739296]
67. Wang M, Hercz G, Sherrard DJ, et al. Relationship between intact 1–84 parathyroid hormone and
levels for bone turnover in patients on chronic maintenance dialysis. Am J Kidney Dis 1995;26:836–
844. [PubMed: 7485142]
68. Slatopolsky E, Finch J, Clay P, et al. A novel mechanism for skeletal resistance in uremia. Kidney
Int 2000;58:753–761. [PubMed: 10916099]
69. Calvi LM, Adams GB, Weibrecht KW, et al. Osteoblastic cells regulate the haematopoietic stem cell
niche. Nature 2003;425:841–846. [PubMed: 14574413]
70. Kuznetsov SA, Riminucci M, Ziran N, et al. The interplay of osteogenesis and hematopoiesis:
expression of a constitutively active PTH/PTHrP receptor in osteogenic cells perturbs the
establishment of hematopoiesis in bone and of skeletal stem cells in the bone marrow. J Cell Biol
2004;167:1113–1122. [PubMed: 15611335]
71. Zhang J, Niu C, Ye L, et al. Identification of the haematopoietic stem cell niche and control of the
niche size. Nature 2003;425:836–841. [PubMed: 14574412]
72. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, et al. The parathyroid is a target organ for FGF23 in
rats. J Clin Invest 2007;JCI32409
73. Goodman WG, Ramirez JA, Belin TR, et al. Development of adynamic bone in patients with
secondary hyperparathyroidism after intermittent calcitriol therapy. Kidney Int 1994;46:1160–1166.
[PubMed: 7861712]
74. Alem AM, Sherrard DJ, Gillen DL, et al. Increased risk of hip fracture among patients with end-stage
renal disease. Kidney Int 2000;58:396–399. [PubMed: 10886587]
75. Cunningham J, Sprague S, Cannata-Andia J, et al. Osteoporosis in chronic kidney disease. Am JKid
Dis 2004;43:566–571. [PubMed: 14981616]
76. Stehman-Breen C. Osteoporosis and chronic kidney disease. Seminars in Nephrology 2004;24:78–
81. [PubMed: 14730513]
77. Rix M, Andreassen H, Eskildsen P, et al. Bone mineral density and biochemical markers of bone
turnover in patients with predialysis chronic renal failure. Kidney Int 1999;56:1084–1093. [PubMed:
10469378]
78. Bonyadi M, Waldman SD, Liu D, et al. Mesenchymal progenitor self-renewal deficiency leads to
age-dependent osteoporosis in Sca-1/Ly-6A null mice. Proc Natl Acad Sci 2003;100:5840–5845.
[PubMed: 12732718]
79. Stehman-Breen C. Bone Mineral Density Measurements in Dialysis Patients. Seminars in Dialysis
2001;14:228–229. [PubMed: 11422932]
80. Stehman-Breen C, Sherrard D, Walker A, et al. Racial differences in bone mineral density and bone
loss among end-stage renal disease patients. Am JKid Dis 1999;33:941–946. [PubMed: 10213653]
81. Coco M, Rush H. Increased incidence of hip fractures in dialysis patients with low serum parathyroid
hormone. Am JKid Dis 2000;36:1115–1121. [PubMed: 11096034]
Hruska et al. Page 11
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 1.
Phosphorus balance in normal physiology. The kidney is the main regulator of human
phosphate homeostasis. In adulthood, exit from the exchangeable phosphorus pool into the
skeleton (bone formation) is roughly equal to entry into the exchangeable pool due to bone
resorption. The skeleton is a storage depot for Pi and contains 85% of the total body phosphorus.
Hruska et al. Page 12
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 2.
Phosphorus homeostasis is lost in chronic kidney disease due to failure of excretion. Despite
reductions in the fraction of filtered phosphorus that is reabsorbed, eventually the filtered load
becomes insufficient to maintain homeostasis, and positive phosphorus balance ensues. Kidney
disease decreases the exchangeable phosphorus pool size by inhibiting bone formation. The
skeletal mineralization fronts at the sites of new bone formation are significant components of
the exchangeable phosphorus pool. Positive phosphate balance is associated with establishment
of heterotopic mineralization sites in soft tissue organs and the vasculature. Exit from the
exchangeable phosphorus pool into the vasculature is portrayed as a bidirectional process
because we have been able to demonstrate that stopping the exit into the vasculature results in
diminishment of established vascular calcification levels.
Hruska et al. Page 13
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 3.
Regulation of phosphorus balance in chronic kidney disease. Regulation of phosphorus
homeostasis is complex. In chronic kidney disease, a decrease in calcitriol production leads to
a decrease in calcium absorption, hypocalcemia, and hyperparathyroidism.
Hyperparathyroidism is one factor contributing to the decrease in the fraction of filtered
phosphorus reabsorbed (decrease in the TRP). Additionally, high levels of FGF23 and
hyperphosphatemia itself, contribute to reducing the TRP. However, when kidney failure
becomes too severe, hyperphosphatemia ensues.
Hruska et al. Page 14
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 4.
Vascular calcification causes cardiac morbidity and mortality in chronic kidney disease.
Vascular calcification increases arterial stiffness leading to an increase in pulse wave velocity
and pulse pressure both of which contribute to development of cardiac ischemia, and left
ventricular hypertrophy and cardiac failure.
Hruska et al. Page 15
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 5.
Control of hyperphosphatemia in translational studies results in reduction of established
vascular calcium levels. LDLR/ mice on high-fat diets with chronic kidney disease have
established vascular calcification at 22 wks (baseline) which increases in vehicle treated
animals sacrificed at 28 wks (veh). However, in animals treated with sevelamer carbonate (1%
sev and 3% sev), aortic calcium levels were significantly reduced compared to baseline, as the
hyperphosphatemia was controlled (28).
Hruska et al. Page 16
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 6.
Schematic of the experimental plan for in vitro studies demonstrating that high phosphorus
causes vascular calcification and osteoblastic gene expression. Human vascular smooth muscle
cells (hVSMC) derived from atherosclerotic aortas expressed increased levels of morphogens,
specific transcription factors and biomarkers of the osteoblast and decreased levels of those
corresponding to contractile hVSMC. Yet the cells did not mineralize until media Pi was
increased from 1 to 2mM. High media Pi stimulated osterix expression, and when osterix
expression was diminished in the presence of high media Pi there was no mineralization (38).
Hruska et al. Page 17
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 7.
Expression of osterix in the aortas of LDLR/ high-fat fed mice. High fat feeding had a small
effect to increase aortic osterix expression in sham operated animals (sham fat) compared to
wild type mice. Induction of chronic kidney disease (CKD high fat) produced a several-fold
increase in osterix expression which was eliminated by treatment with phosphate binders, in
this case lanthanum carbonate 1% or 3% added to the diet (38).
Hruska et al. Page 18
Kidney Int. Author manuscript; available in PMC 2009 August 31.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Hruska et al. Page 19
Table 1
Hyperphosphatemic Syndromes
Increased intake
Transcellular shifts from intracellular to extracellular spaces
Excess bone resorption
Decreased renal excretion
Idiopathic hyperparathyroidism
Pseudohypoparathyroidism
FGF23 deficiency
Tumoral calcinosis
Chronic kidney disease
Acromegaly
Artifactual
Kidney Int. Author manuscript; available in PMC 2009 August 31.
... 21 Furthermore, our study results are consistent with the study by Singh et al which found that virtually all patients develop hyperphosphatemia, and those with serum phosphorus (>6.5 mg/dL) are at higher risk of death. 23 The increase in PTH levels reduces the filtered phosphate load and maintains phosphate elimination at a normal level, although the decrease in the filtered phosphorus load is due to the reduction in the glomerular filtration. 19 For dialysis CKD serum phosphate with high phosphorus levels, 49 (69.0%) ...
... Singh et al found that almost all patients with end-stage renal disease develop hyperphosphatemia and those with serum phosphate levels >6.5 mg/dl have a significantly higher risk of mortality, mainly due to calcification of the heart, lungs and Soft tissue. 23,29 In conclusion, treatment of hyperphosphatemia is critical factor in preventing death and disability in CKD patients and in CKD stage III and IV patients between >2.7 and <4.6 and in CKD or dialysis patients in stage V should be between 3.5 and 5.5. ...
Article
Full-text available
Background: Chronic kidney disease is a major public health problem worldwide. As kidney function declines, it leads to several metabolic abnormalities including dysregulation of mineral metabolism. It is also reported that hyperphosphatemia in patients with advanced kidney disease is associated with an increased risk of mortality and cardiovascular events, and is higher in dialysis-dependent chronic kidney disease (CKD) patients compared to non-dialysis CKD. However, data in the Indian context is limited. Objectives were to evaluate and compare serum phosphorus levels and associated factors in non-dialysis and dialysis CKD patients. Also, the impact of dietary phosphate restriction and the use of phosphate binders on serum phosphorus is analysed. Methods: A cross-sectional study was conducted at a tertiary care hospital in Kolkata, India, with 100 CKD patients: 50 non-dialysis CKD patients and 50 dialysis-dependent CKD patients. Relevant demographic, clinical and laboratory parameters including serum phosphorus, calcium, parathyroid hormone (PTH), alkaline phosphatase, albumin and estimated glomerular filtration rate (eGFR) were collected. Data was analyzed using appropriate statistical tests. Results: Mean serum phosphorus was significantly higher in the dialysis CKD group (6.12±0.34 mg/dl) compared to the non-dialysis CKD group (4.56±0.80 mg/dl). Serum calcium and PTH were also higher while eGFR and albumin were lower in the dialysis CKD group. Serum phosphorus levels increased with advancing CKD stages in the non-dialysis group. Phosphate binder helped phosphorus control in dialysis CKD patients. Conclusions: Our study is in confluence with other reports and dietary phosphate restriction and the use of phosphate binders help optimize phosphorus levels in CKD patients.
... Effects of nutrients dissolved in water have been studied, and there is evidence that nitrate, phosphate and ammonia produce certain diseases in high levels; for example, phosphates are associated with urine damage and osteoporosis [34]. Additionally, there is scientific evidence that ammonium can affect the developing central nervous system when it enters the human body [35]. ...
Article
Full-text available
This review critically analyzed the use of biosorbents derived from rice husks and corn residues for nutrient removal from aqueous solutions. Additionally, this review highlighted the use of such biosorbents in wastewater treatment. Furthermore, novel approaches for sustainable nutrient removal from aqueous solutions were identified. A comprehensive understanding of the implementation of biosorption processes using agro-industrial residues based on corn and rice crops is critical for the sustainable management of residues and water bodies in the world to protect and conserve natural resources. Specifically, the review focuses on the exploration, preparation and application of innovative biosorbents to remove various forms of nutrients such as total nitrogen, total phosphorus, nitrates, ammonium and phosphates from aqueous solution, analyzing the sustainability of treatments applied to biomass, such as thermal transformation or chemical modification to reduce environmental impacts. It was found that 95 to 99% of nitrogen and phosphorus can be removed with biosorbents made from rice husks and corn residues, analyzing approximately 50 scientific articles related to these plant materials. Research opportunities were identified, such as the recovery of removed nutrients for soil improvement, life cycle analysis to assess the concept of zero waste, among other aspects. Finally, a scheme is proposed for the selection and application of sustainable biosorbents for the removal of nutrients from aqueous solutions.
Article
Full-text available
Food quality assessment is becoming a global priority due to population growth and the rise of ionic pollutants derived from anthropogenic sources. However, the current methods used to quantify toxic ions are expensive and their operation is complex. Consequently, there is a need for affordable and accessible methods for the accurate determination of ion concentrations in food. Electrochemical sensors based on potentiometry represent a promising approach in this field, with the potential to overcome limitations of the currently available systems. This review summarizes the current advances in the electrochemical quantification of heavy metals and toxic anions in the food industry using potentiometric sensors. The healthcare impact of common heavy metal contaminants (Cd2+, Hg2+, Pb2+, As3+) and anions (ClO4−, F−, HPO4−, SO42−, NO3−, NO2−) is discussed, alongside current regulations, and gold standard methods for analysis. Sensor performances are compared to current benchmarks in terms of selectivity and the limit of detection. Given the complexity of food samples, the percentage recovery values (%) and the methodologies employed for ion extraction are also described. Finally, a summary of the challenges and future directions of the field is provided. An overview of technologies that can overcome the limitations of current electrochemical sensors is shown, including new extraction methods for ions in food.
Article
Introduction Case reports have suggested a causative role between sevelamer use and subsequent gastrointestinal bleeding (GIB), but no large observational studies have evaluated this association. Methods Using the United States Renal Data System database from 2015 to 2019, we examined the association between initiation of sevelamer (versus non-sevelamer containing phosphate binders) and GIB hospitalization as well as all-cause mortality among individuals on hemodialysis. We emulated a target trial using Cox regression models and inverse probability of treatment weights to estimate the adjusted hazard ratios (HR) across outcomes and subgroups. Results Among 21,354 new users of phosphate binders (11,276 sevelamer and 10,078 non-sevelamer) with baseline lab data (calcium, phosphorus, hemoglobin, and albumin), there were 2,811 GIB hospitalizations and 5,920 deaths after a median follow-up of 1.3 years. Compared with the initiation of non-sevelamer binders, sevelamer was not associated with an increased risk of GIB hospitalization (89 vs. 90 events per 1000 person-years; IPTW-HR 0.98, 95% CI 0.91 – 1.06) or all-cause mortality (220 vs. 224 events per 1000 person-years; IPTW-HR 0.98 95% CI 0.93 – 1.03). Subgroup analyses (such as diabetes and anti-coagulation use) were generally consistent, and there was no association between sevelamer dose and GIB hospitalization. Conclusion Among patients requiring hemodialysis, sevelamer (vs non-sevelamer) containing phosphate binders was not associated with increased risk of GIB hospitalization.
Article
Background: Aging increases fracture risk through bone loss and microarchitecture deterioration due to an age-related imbalance in bone resorption and formation during bone remodelling. We examined the associations between levels of phosphate, calcium, and alkaline phosphatase, and fracture risk in initially-healthy older individuals. Methods: A post-hoc analysis of the Aspirin in Reducing Events in the Elderly (ASPREE) trial recruited 16,703 Australian participants aged ≥70 years and 2,411 US participants aged ≥65 years. Analyses were conducted on ASPREE-Fracture substudy participants from Australia with serum calcium, phosphate, and alkaline phosphatase measurement. Fracture data were collected post-randomization. Cox regression was used to calculate hazard ratios (HR) and 95% confidence intervals (CIs). Phosphate, calcium, and alkaline phosphatase were analysed in deciles (D1-D10), with deciles 4-7 (31-70%) as the reference category. Restricted cubic spline curves were used to identify nonlinear associations. Results: Of the 9915 participants, 907 (9·2%) persons had incident fractures recorded over 3·9 (SD 1·4) years. In the fully adjusted model, males in the top decile (D10) of phosphate had 78% higher risk of incident fracture (HR 1·78, 95% CI 1·25-2·54). No such association was observed for females (HR 1·09, 95% CI 0·83-1·44). The population attributable fraction in men within the D10 phosphate category is 6·9%. Conclusion: This result confirms that, high-normal serum phosphate levels are associated with increased fracture risk in older men.
Article
Full-text available
Background: Chronic Kidney Disease (CKD) is a disease in which the kidney’s functionality declines gradually. The aim of this study was to identify significant laboratory prognostic factors on death due to CKD in a clinical complex. Materials and Methods: A retrospective study including 109 patients with the end-stage renal disease treated at Iran Helal pharmaceutical and the clinical complex was conducted between 2014-2018. The survival time was set as the time interval between starting dialysis until death due to CKD. Also, the transplantation was considered as competing risk, which was occurred for a few patients. A three-parameter Gompertz model was used that considers both the event of interest and the competing event simultaneously. Results: Death due to CKD occurred in 29 (26.6%) of the patients and 19(17.4%) with transplantation. Serum uric acid was a significant prognostic factor that decreased the hazard of mortality by 21%. Serum phosphorus and age by increasing the risk of death, were poor prognoses for the event of interest. Serum uric acid and phosphorus 6.9-9.9 (mg/dl) were associated with 72% and 4.05-fold increased hazard of transplant, respectively. The 4-year cumulative incidence of death and transplant was 48.4% and 29.2%, respectively. Conclusion: We have deduced that high serum phosphorus levels and increased levels of age were associated with worse outcomes. High serum uric acid level was related to better survival, which could be explained by having a better protein-rich diet alongside the high albumin level. [GMJ.2020;9:e1798]
Article
Objectives We sought to determine clinical and laboratory correlates of calcification of the coronary arteries (CAs), aorta and mitral and aortic valves in adult subjects with end-stage renal disease (ESRD) receiving hemodialysis.
Article
Objectives We sought to determine clinical and laboratory correlates of calcification of the coronary arteries (CAs), aorta and mitral and aortic valves in adult subjects with end-stage renal disease (ESRD) receiving hemodialysis.
Article
Although it is well known that chronic renal failure induces parathyroid hyperplasia, the pathogenesis and development of this parathyroid lesion in this disease are poorly understood. Histopathologically, there is progression from diffuse to nodular hyperplasia, and each nodule consists of a single cell type with aggressive proliferative potential. Pathophysiologic and clinical investigations have suggested that neoplastic tumors may emerge from nodular hyperplasia. In this study the clonality of parathyroid tissue in nodular and diffuse hyperplasia in renal hyperparathyroidism was analyzed by a method based on restriction fragment length polymorphism of the X chromosome-linked phosphoglycerokinase gene and on random inactivation of the gene by methylation. DNA of peripheral lymphocytes was screened in 43 women undergoing parathyroidectomy for advanced renal hyperparathyroidism, and 10 of these patients appeared to be heterozygous. Fourteen specimens from these patients were available for clonal analysis. The analysis showed that all four specimens of diffuse hyperplasia were polyclonal, whereas all seven specimens from nodules in nodular hyperplasia and all three samples representing parathyroid tissue removed from forearm because of graft-dependent recurrence were revealed to be monoclonal. It is likely that the clonal origin of each nodule is independent. These results suggest that in renal hyperparathyroidism parathyroid glands initially grow diffusely and polyclonally, and then the cells in the nodules are later transformed monoclonally and proliferate aggressively. From the present study it can be concluded that nodular hyperplasia represents monoclonal parathyroid neoplasia, which might explain why patients with nodular hyperplasia in renal hyperparathyroidism are refractory to medical treatment, requiring parathyroidectomy. To prevent recurrences, nodular hyperplastic tissue should not be left at surgery.
Article
Elevated serum phosphorus is a predictable accompaniment of end-stage renal disease (ESRD) in the absence of dietary phosphate restriction or supplemental phosphate binders. The consequences of hyperphosphatemia include the development and progression of secondary hyperparathyroidism and a predisposition to metastatic calcification when the product of serum calcium and phosphorus (Ca x PO4) is elevated. Both of these conditions may contribute to the substantial morbidity and mortality seen in patients with ESRD. We have analyzed the distribution of serum phosphorus in two large national, random, cross-sectional samples of hemodialysis patients who have been receiving dialysis for at least 1 year. Data were obtained from two special studies of the United States Renal Data System, the Case Mix Adequacy Study (1990) and the Dialysis Morbidity and Mortality Study Wave 1 (1993). The relative risk of death by serum phosphorus quintiles is described after adjusting for age at onset of ESRD, race, sex, smoking status, and the presence of diabetes, the acquired immunodeficiency syndrome, and/or neoplasm. Logistic regression analysis is then used to describe the demographic, comorbid, and laboratory parameters associated with high serum phosphorus. Serum phosphorus was similar in these two study populations and averaged 6.2 mg/dL. Ten percent of patients had levels greater than 9 mg/dL and at least 30% of each group had serum phosphorus levels greater than 7 mg/dL. The adjusted relative risk of death by serum phosphorus level was not uniform across all quintiles, being constant below a level of 6.5 mg/dL and increasing significantly above this level. The relative risk of death for those with a serum phosphorus greater than 6.5 mg/dL was 1.27 relative to those with a serum phosphorus of 2.4 to 6.5 mg/dL. This increased risk was not diminished by statistical adjustment for coexisting medical conditions, delivered dose of dialysis, nutritional parameters, or markers of noncompliance. Evaluation of predictors of serum phosphorus greater than 6.5 mg/dL revealed in multivariate analysis that younger age at onset of ESRD, female sex, white race, diabetes, active smoking, and higher serum creatinine levels were all significant predictors. Analysis of serum calcium revealed no correlation with relative risk of death. The Ca x PO4 product, however, showed a mortality risk trend similar to that seen with serum phosphorus alone. Those in the highest quintile of the Ca x PO4 product (>72 mg2/dL2) had a relative mortality risk of 1.34 relative to those with products of 42 to 52 mg2/dL2. The relative mortality risk by log parathyroid hormone (PTH) level was elevated for patients with higher levels, but the mortality risk associated with hyperphosphatemia was independent of PTH. For hemodialysis patients who have been receiving dialysis for at least 1 year, we conclude that a large percentage have a serum phosphorus level above 6.5 mg/dL and that this places them at increased risk of death. This increased risk is independent of PTH. The mechanism(s) responsible for death is unknown, but may be related to an abnormally high Ca x PO4 product. Although mechanisms are not clearly established, this study supports the need for vigorous control of hyperphosphatemia to improve patient survival.
Article
The synthesis of very high specific activity 25-OH-vitamin D3 (78 Ci/mmol) has made possible the study of the metabolism and plasma disappearance of 3H after a single dose of 3H-1,25-(OH)2-D3 in quantities that are only 10-20% of the endogenous plasma pool. We studied seven healthy adults who were given doses of 1,25-(OH)2-D3 ranging from 30-2300 pmol. Plasma disappearance was rapid with only 14 +/- 2% of administered 3H remaining in the plasma pool 4 h after labeling. Plasma metabolite profiles during the first 4 h showed only 1,25-(OH)2-D3. Thereafter, significant amounts of other metabolites were detected. The 6-day cumulative excretion of 3H in urine and feces (virtually all associated with metabolites of 1,25-(OH)2-D3) averaged 16 +/- 3% and 49 +/- 11% of the dose, respectively. Compartmental analysis of the isotope data for two subjects who received the smallest doses of 1,25-(OH)2-D3 indicated that endogenous renal 1,25-(OH)2-D3 synthesis rates approximate 0.8-2.4 nmol/day (0.3-1.0 microgram/day).