Você está na página 1de 15

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/257765277

Pathophysiology of CKD-MBD

Article  in  Clinical Reviews in Bone and Mineral Metabolism · September 2011


DOI: 10.1007/s12018-011-9120-8

CITATION READS

1 967

1 author:

Grahame J Elder
Westmead Hospital
83 PUBLICATIONS   2,224 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Chronic kidney disease mineral and bone disorders; controversies and directions View project

All content following this page was uploaded by Grahame J Elder on 13 March 2016.

The user has requested enhancement of the downloaded file.


Clinic Rev Bone Miner Metab
DOI 10.1007/s12018-011-9120-8

ORIGINAL PAPER

Pathophysiology of CKD-MBD
Grahame J. Elder

Ó Springer Science+Business Media, LLC 2011

Abstract To maintain calcium and phosphate balance and mortality to the biochemical and hormonal conse-
despite declining kidney function, adaptations must begin quences of kidney damage (Fig. 1). Defining a new dis-
in the earliest stages of chronic kidney disease (CKD). order by consensus does not happen frequently in
These are generally successful in maintaining calcium and medicine, so it is intriguing to reflect how greatly a new
phosphate levels within the normal range through CKD appreciation of these connections has impacted on the
stages 1–4. Although routine biochemistry is not informa- clinical management of CKD and integrated areas of lab-
tive about net gain of these minerals, newer markers such oratory research that previously seemed more distantly
as Klotho and fibroblast growth factor 23 may provide related.
clues to these early adjustments and be of prognostic value. Maintaining the body’s mineral balance is a matter of
Bone cells play a central role in modulating responses to matching input to output, which becomes more challenging
CKD. This review describes that role and highlights the as kidney function declines. At the renal level, the ‘‘intact
dynamic communication between bone and the kidneys. nephron hypothesis’’ [1] describes how maintaining min-
eral homeostasis will demand greater work from fewer
Keywords CKD-MBD  Bone  Osteocyte  Osteoblast  nephrons. As this occurs, signal pathways that control
Osteoclast  Fibroblast growth factor 23  Klotho  mineral homeostasis need to reset, and bone is compelled
Parathyroid hormone  Renal adaptation  Pathophysiology to assume functions of mineral homeostasis that were
previously shared with the kidneys. These interconnections
are fascinating, but some understanding of bone cells and
Introduction how they work is necessary to savor their complexity—and
potential for failure. So, I will start by reviewing the rel-
The laboratory, bone and vascular abnormalities that evant bone biology and signals that affect it and then
develop with CKD have been recognized for many years, proceed to changes that occur with progressive CKD.
so why create a new condition called CKD-mineral and
bone disorder (MBD)? The reason is connections. Normal Bone; Basic Bone Biology
Although the associations are not new, the defining feature
of CKD-MBD is the way that pathophysiology so inti- Normal bone turnover, mineralization and the maintenance
mately links fracture risk, vascular calcification, morbidity of bone volume are essential for a healthy skeleton that is
stable enough for locomotion, strong enough to resist
fractures and provides for the homeostasis and storage of
G. J. Elder (&)
Department of Renal Medicine, Westmead Hospital, Westmead, calcium and phosphate. To maintain strength and stability,
Sydney, NSW 2145, Australia the skeleton must detect mechanical strain that indicates
e-mail: g.elder@garvan.unsw.edu.au where extra bone is required, repair microdamage that
occurs with daily activities and renew aging bone. Cortical
G. J. Elder
Osteoporosis and Bone Biology Program, Garvan Institute bone (as found in the shafts of long bones) is commonly
of Medical Research, Sydney, NSW, Australia described as ‘‘compact’’; a misnomer if the extensive

123
Clinic Rev Bone Miner Metab

Initiation of Remodeling

Bone regions share common (or stochastic) remodeling rates


that may vary with factors that include age and menopausal
status, and conditions such as thyroid disease or glucocorti-
coid use. Remodeling can also be pathological (or redundant)
when new bone is preferentially remodeled or targeted in
response to mechanical strain [4, 5]. Targeted remodeling
originates when loading leads to strain, or fatigue and micro
cracks that disrupt osteocyte canalicular networks [6]
(Fig. 2). When this is sensed by osteocytes, they initiate
remodeling by the generation of new BMUs (Fig. 2).
Affected osteocytes recruit haematopoietic stem cells from
nearby ‘stem cell niches’ [7] or circulating pre-osteoclasts,
and these differentiate into osteoclasts under the influence of
the receptor activator of nuclear factor jB-ligand (RANKL)
expressed by cells of the osteoblast lineage [8]. Next, the
osteoclasts resorb damaged bone over a period of 2–3 weeks;
during which time some will undergo apoptosis, but new
osteoclasts are recruited to continue the job. Osteoblast
teams then lay down a matrix of osteoid which they then
Fig. 1 The term ‘‘chronic kidney disease mineral and bone disorder’’
unifies the important elements of this clinical cluster
mineralize and finally the bone becomes quiescent while the
mineral crystals mature and the bone strengthens. In cortical
bone, the BMU forms a tunnel that is re-mineralized to form
a new osteon. In cancellous bone, the BMU excavates a
cortical porosity of ageing or hyperparathyroidism is trench across the bone surface that is filled to form a hemi-
present. Cortical bone contains yellow marrow and has osteon. ‘‘Activation frequency’’ is a measure of the overall
relatively slow turnover, particularly at peripheral sites [2]. intensity of bone turnover—although not necessarily the
By comparison, cancellous bone within vertebrae and at the birth of new BMUs.
ends of long bones consists of bony plates or trabeculae, Within the BMU, the distance between the cutting cone
which provides a large surface area. Cancellous and that releases mineral and the osteoblast team that deposits
endocortical bone surfaces near red marrow have higher osteoid and mineral can be varied in a ‘‘concertina-like’’
turnover because they are more active in haematopoiesis fashion [9]. Consequently, the release of calcium and
and mineral homeostasis [2]. phosphate can vary with BMU activity. In young animals,
this osteoclastic bone resorption appears to play a signifi-
Bone Remodeling cant role in calcium homeostasis, but in mature animals
calcium ion release or uptake from the osteocyte lacunar
Bone remodeling is the term used for the process of bone and canalicular network seems most important for rapid
turnover and renewal. The teams of cells involved are calcium adjustments [10–12] [Table 1].
termed bone multicellular units (BMUs) and consist of a
cutting cone of osteoclasts derived from hematopoietic
stem cells, followed by osteoblasts that are derived from
mesenchymal stem cells (MSC) that lay down matrix and
mineral [3]. Not surprisingly this is a tightly coupled pro-
cess, because as the BMU advances at 10–20 lm/day, any
imbalance between bone resorption and formation could
lead to unwanted changes in bone volume, microarchitec-
ture, strength or mineral homeostasis [4]. To avoid these
consequences, the cells of the BMU must be in close
communication with each other and with osteocytes; ter- Fig. 2 Microcrack disruption of the osteocyte canalicular system
minally differentiated osteoblasts that are the most abun- initiates a remodeling cycle by the Bone Multicellular Unit:
osteoclasts remove damaged bone followed by osteoblast teams,
dant cells in bone and principal conductors of the ‘‘bone which produce matrix and re-mineralized bone, after which there is a
orchestra.’’ quiescent period

123
Clinic Rev Bone Miner Metab

Table 1 Principal cells found within bone


Osteoclasts are derived from hematopoietic stem cells that fuse to form large multinucleated bone resorbing cells. Osteoclast precursors
commit to the osteoclast lineage when receptor activator of nuclear factor jB (RANK) cell surface receptors bind RANK ligand (RANKL),
which is produced by osteoblasts, osteocytes and bone marrow stromal cells
Osteoblasts are bone forming cells derived from mesenchymal stem cells (MSC), which have potential to undergo differentiation to
chondrocytes, adipocytes and myocytes. MSCs commit to the osteoblast lineage and develop to mature osteoblasts under the influence of
numerous downstream factors that include Runx2 (also known as Cbfa1), Osterix and members of the Wnt signaling pathway [13]. They
express alkaline phosphatase (bone-specific alkaline phosphatase; BALP) and osteocalcin, secrete collagen and non-collagenous matrix
proteins and mineralize bone. Osteoblasts may flatten to become lining cells, be buried in the bone matrix aligned in the direction of bone
formation to become osteocytes, or undergo apoptosis
Osteocytes are terminally differentiated osteoblasts that have become embedded in the bone matrix. They maintain intimate connections with
each other and with mineralizing osteoblasts on the bone surface through networks of cell processes that traverse the bone canaliculi [14]
(Fig. 3). Osteocytes act as mechanosensors and regulate osteoid matrix formation and mineralization, responding to parathyroid hormone
(PTH) and c-terminal PTH fragments [15], estrogens, prostaglandins and 1,25-dihydroxyvitamin D [1,25(OH)2D]. Osteocytes act as ion
sensors and are likely to control calcium homeostasis by altering its concentration in the fluid bathing the lacunae and canaliculi. Osteocytes
have recently been reported to produce RANKL and they may in fact be the major cells controlling osteoclastic bone resorption in adults
[16, 17]

Fig. 4 An example of direct signaling to osteoclasts is by the


production of cell surface and soluble receptor activator of nuclear
factor jB-ligand (RANKL) and osteoprotegerin (OPG). Indirect
osteoclast to osteoblast signaling can occur by the release of cytokines
from the bone matrix

osteoclast activity and potentiate bone anabolic activity


(Fig. 4). An example of indirect osteoclast to osteoblast
signaling is the release of cytokines such as insulin-like
growth factor (IGF) and transforming growth factor (TGF)-
b from the bone matrix during osteoclastic resorption.
Fig. 3 Fluorescence image of calvarial fragment demonstrating Upon release, these factors activate the osteoblast teams
osteocytes and their dense network of cell processes overlying and can also recruit MSCs to the site [19–21]. Direct,
osteoblasts on the bone surface. Reproduced with permission from
[14]
bidirectional signaling can also occur between osteoblasts
and osteoclasts. An example is the proposed role for for-
ward signaling by Eph receptor-expressing osteoblasts to
Coupling Bone Cell Activity stimulate bone formation and reverse signaling through
ephrin-expressed by osteoclasts to inhibit bone resorption
The coupling of bone resorption and formation depends [22]. Additional osteoclast factors influencing bone turn-
upon direct and indirect signals between bone cells, as well over have recently been reported including the migration
as ‘‘whole-body’’ controls such as metabolic or sympa- and mineralization stimulating agents BMP-6 and sphin-
thetic nervous system signals [18]. An example of direct gosine 1-phosphate (S1P) [23], interferon regulatory fac-
osteoblast to osteoclast signaling is the production by tor-8 (IRF-8) [24], semaphorin 4d (Sema4D) [125] and
osteoblasts of RANKL to facilitate osteoclast differentia- osteoclast-derived sclerostin [25], which may reduce
tion and activity, or production of the RANKL decoy osteoblast anabolic activity with aging. Since direct
receptor osteoprotegerin (OPG) to bind RANKL, reduce osteoclast to osteoblast contact does not occur in the

123
Clinic Rev Bone Miner Metab

human BMU, details of these mechanisms need further


development.

Bone Turnover in Response to Mechanotransduction

The concept of mechanotransduction was 1st introduced by


Julius Wolff in 1892, when he published ‘‘The Law of
Transformation of Bone.’’. This work stated that ‘‘As a
consequence of primary shape variations and continuous
loading, or even due to loading alone, bone changes its inner
architecture according to mathematical rules and, as a sec-
ondary effect and governed by the same mathematical rules,
also changes its shape.’’ Now known as ‘‘Wolff’s Law’’ he
regarded this finding as a ‘‘brick to complete the building of
Fig. 5 The Wnt signaling pathway is anabolic for bone. Wnts are
Charles Darwin’’ (http://jwi.charite.de/en/ accessed 25th
produced by stromal cells/osteoblast progenitors and bind to osteo-
September 2011); a prescient statement, given the now blast LRP 5/6 and LRP 4 receptors. Sclerostin inhibits this binding.
familiar concept of gene-environment interactions. SOST gene expression is itself inhibited by mechanical strain
Osteocytes are the most numerous and long-lived bone
cells. The osteocyte networks are highly integrated and pathway [32, 33] (Fig. 5). This pathway is anabolic for
ideally suited to being mechanical strain receptors and for bone and is described below (Fig. 5). SOST gene expres-
the conversion of strain into biochemical signals, or sion is influenced by a number of factors that influence
‘‘mechanotransduction’’ [26]). Although the process is not bone strength, particularly mechanical stimulation of bone,
fully understood, mechanical stimulation of bone causes which inhibits sclerostin production and increases osteo-
motion and shear stress within the fluid or gel-filled can- blast activity [34]. Illustrating this, osteocytes prone to
alicular network. The osteocyte processes in these tube-like mechanical stimulation nearer the bone surface are more
structures possess a central filament bundle that is tethered often sclerostin negative than osteocytes further from the
to the canalicular wall by transverse tethering elements surface and areas of remodeling [31]. One of the most
[27]. When stimulated, these elements transmit a signal to dramatic human examples of sclerostin suppression is
the actin cytoskeleton of the osteocyte process, causing a illustrated by the condition Van Buchem disease charac-
biochemical response. Recent investigations have provided terized by a symmetrical high bone mass phenotype, gen-
direct evidence for this load-induced fluid flow [28], which erally by increased bone formation markers [35] and
is also essential for maintaining bone elasticity and osteo- caused by deletions downstream of the SOST gene [36].
cyte hydration and survival. The estrogen receptor (ER)-a As opposed to this pathological inhibition of SOST expres-
appears to participate in this signaling cascade, as dem- sion, sclerostin suppression occurs every day when PTH is
onstrated by reduced bone formation following mechanical released and binds to osteocyte PTH/PTHrP receptors
loading in ER-a knockout mice compared to the wild type (PPR). This circadian PTH pulse is imitated by the daily
[29]. Osteoblasts also respond to shear, stretch and injections of PTH [1–34] or PTH [1–84] that are used to
hydrostatic pressure by activation of mechanosensitive treat osteoporosis.
channels, with downstream signaling possibly amplified by Dickkopf-1 (Dkk-1) is another potent, soluble inhibitor
the osteocyte network. It appears likely that ER-a receptors of canonical Wnt signaling and bone formation, which is
interact with membrane IGF-receptors to sensitize osteo- highly expressed in osteocytes. Dkk-1 binds LRP5/6 and a
blasts to the anabolic effects of IGFs [30]. These findings transmembrane protein, Kremen [37] (Fig. 5). The tertiary
help to explain the action of selective ER modulators complex that is formed is internalized, removing LRP5
(SERMS) such as raloxifene, which are ER-a agonists. from the cell surface and therefore preventing Wnt sig-
naling [38].
Humoral Bone Pathways

Sclerostin is a soluble protein encoded by the SOST gene Wnt Signaling


and produced predominantly by mature osteocytes. After
release, sclerostin is believed to reach osteoblasts via the The Wnts are a family of secreted glycoprotein growth
canalicular system [31] where it binds to LDL-receptor- factors named after the WNT gene of Drosophila melano-
related protein (LRP) 5/6 and LRP4 and inhibits signaling gaster that results in a wingless mutation, and the
through the canonical (or stereotypical) Wnt signaling integration 1 (INT) gene, which is activated in some mouse

123
Clinic Rev Bone Miner Metab

mesenchymal precursors commit to differentiation along


the osteoblast lineage. Wnts inhibit mesenchymal precur-
sors from developing into adipocytes by blocking the
master adipogenic transcription factors [46] and the dif-
ferentiation of MSCs along the chondrocyte lineage [47].

Adaptations to CKD and Development of the Mineral


and Bone Disorder

The Intact Nephron Hypothesis

With this background in bone biology, we can now focus


on renal adaptations to CKD, described by Bricker et al. [1]
in 1960 (as the intact nephron hypothesis) and further
developed in 1972 as the trade-off hypothesis [48]. Both
hypotheses sought to explain the changes that occur as
Fig. 6 The Wnt signaling pathway: Frizzled receptor protein (FRP), glomerular filtration rate (GFR) falls and adaptive mecha-
low density lipoprotein-receptor-related protein (LRP), dishevelled nisms are triggered to maintain mineral homeostasis. The
(Dvl), the ‘‘destruction complex’’ comprising Axin, adenomatous authors saw ‘‘a common denominator’’ in the response of
polyposis coli (APC), glycogen synthase kinase (GSK), lymphoid
disparate forms of renal disease to reductions in GFR. This
enhancer binding factor (LEF). Dickkopf-1 (Dkk-1) removes LRP
from the cell surface resulted in an increased demand on fewer residual neph-
rons to excrete increased quantities of solute; a concept that
relies upon most remaining nephrons being functionally
tumors. Wnts bind to cell surface Frizzled receptor proteins intact. To maintain homeostasis as GFR declines, sub-
(FRP) and members of the transmembrane low density stances such as creatinine that are filtered with little tubular
lipoprotein-receptor-related protein (LRP) family, LRP5/6 handling remain in balance because rising plasma levels
[39] and LRP4 [40] (Fig. 6). Binding activates Dishevelled increase their filtration (and excretion) by each remaining
(Dvl), with the presence of b arrestin required for full nephron. However, potassium, calcium and phosphate
activation [41]. Dvl activation inhibits a b-catenin undergo tubular handling (and would kill the patient if their
‘‘destruction complex’’ that would otherwise promote the levels rose to maintain balance like creatinine does) and for
phosphorylation and proteosomal destruction of cytoplas- these solutes, balance is maintained by each intact nephron
mic b-catenin [42]. This destruction complex consists of excreting a greater fraction of its filtered load. For example,
Axin and the proteins adenomatous polyposis coli (APC) while 10% of phosphate is excreted at normal GFR, 90%
and glycogen synthase kinase-3 (GSK-3). When the com- needs to be excreted at lower levels of GFR to maintain
plex is inactivated, b-catenin is released, its levels rise, and homeostasis. On the other hand, this adaptation is unnec-
it is translocated to the nucleus. There it binds to T-cell essary if a reduction in GFR is accompanied by a simul-
factor/lymphoid enhancer binding factor 1 (TCF/LEF1), a taneous restriction of dietary phosphate [49].
transcriptional effector that increases OPG production and
promotes the expression of other target genes [43]. It now Osteocytes Control Mineral Homeostasis and Adaptations
appears that other potent regulators of osteoblast differen- to CKD-MBD in Bone and Kidney
tiation such as BMP-2, Wnt3a and Runx2 may also influ-
ence osteoblast differentiation by affecting the balance As mentioned earlier, by controlling BMU recruitment and
between LEF1 and alternative LEF1 isoforms that have remodeling cycles, osteocytes and osteoblasts have an
lower affinity for binding of b-catenin [44]. As osteoblasts ability to modulate homeostasis of calcium and phosphate.
differentiate due to activation of the Wnt signaling path- But the enormous lacunar-canalicular network, which is
way, they produce progressively less RANKL and estimated to have a surface area more than 100-times that
increasingly more OPG [45]. Hence, in addition to stimu- of the trabecular bone surface [50], also provides osteo-
lating osteoblast activity, activation of the Wnt signaling cytes an opportunity to influence ion levels by direct
cascade alters the balance of OPG to RANKL, resulting in interaction with their adjacent bony matrix [51]. This
a reduction of osteoclastic bone resorption. process is termed ‘‘osteocytic osteolysis’’ and involves
Clearly many molecules influence Wnt/b-catenin sig- osteocytic production of RANKL, downregulation of
naling, so that the activity of this pathway can be thought OPG and promotion of osteoclast formation and activity
of as a series of molecular switches that determine whether [16, 17].

123
Clinic Rev Bone Miner Metab

Osteocytes also influence systemic control of calcium, FGFs by having a low affinity for heparan sulfate (HS),
phosphate and 1,25(OH)2D levels, through production of which allows them to escape from HS-rich extracellular
fibroblast growth factor-23 (FGF23), which is a member of matrices and to circulate systemically. However, it also
the FGF ligand superfamily. FGF23 is a 251 amino acid reduces their ability to bind to FGF receptors, because
glycoprotein that undergoes post-translational modification efficient binding relies upon the affinity of FGFs for HS.
before being secreted as FGF23 (25-251). FGF23 gene FGF23 therefore requires a separate cofactor in order to
expression is upregulated by serum phosphate [52], bind to the FGF23 receptor 1 (FGFR1) involved with
although whether this is a direct effect in healthy individ- phosphate control or FGFR3/FGFR4 involved with vitamin
uals or those with CKD is still unclear, because a number D activation [63]. This binding factor was identified as
of studies have reported that FGF23 levels do not rise Klotho, a 130-kDa single-pass transmembrane protein
acutely in response to phosphate loading [53–55]. As named after the 1st of the Moirae, who were the three
opposed to the evidence for phosphate, there is good evi- Greek goddesses controlling man’s fate. Klotho spun the
dence that FGF23 gene expression and FGF23 levels are thread of life, so it was highly appropriate to assign her
directly upregulated by 1,25(OJH)2D acting through oste- name to a transgenic mouse gene that conferred longevity
ocyte/osteoblast vitamin D receptors (VDR) [56, 57] and above the wild type when over-expressed, but upon dele-
that PTH also increases FGF23 directly, in addition to tion resulted in premature aging and a shortened life span
indirectly by conversion of 25(OH)D to 1,25(OH)2D [58, [64]. When it was realized that Klotho and FGF knockout
59] (Fig. 7). Increased serum phosphate levels also stim- mice shared identical phenotypes, Klotho was discovered
ulate the post-translational O-glycosylation of FGF23, to be the co-receptor for FGF23, compensating for
which protects it from cleavage and increases its stability FGF23’s low HS binding affinity [65]–and emphasizing the
[60]; the enzyme involved being coded by the polypeptide importance of phosphate homeostasis to survival!
N-acetylgalactosaminyltransferase 3 (GALNT3) gene. In response to an increase in serum phosphate levels,
Expression of FGF23 is suppressed by both the phosphate- FGF23 is secreted and binds to the FGFR-Klotho complex
regulating gene with homologies to endopeptidases on the at a number of sites to restore phosphate homeostasis. In
X chromosome (PHEX) and dentin matrix protein 1 the proximal tubule, FGF23 acts as a ‘‘phosphatonin’’ by
(DMP1), most likely by indirect mechanisms [61]. reducing the expression of proximal tubular sodium-
FGF23 together with FGF15 and FGF21 are designated dependent phosphate co-transporter (NPT) 2a and increases
‘‘hormone-like’’ FGFs [62]. They differ from nonhormonal the fractional excretion of phosphate [66]. Increased
FGF23 also downregulates the activity of 1-alpha
hydroxylase (cytochrome P450 (CYP) 27B1) in the prox-
imal tubule, reducing the renal conversion of 25(OH)D to
1,25(OH) [67]. Additionally, FGF23 increases the activity
of the catabolic 24-hydroxylase enzyme (CYP 24A1),
which enhances the conversion of 1,25(OH)2D to 1,24,25
(OH)3D and reduces the availability of substrate to the
1-alpha hydroxylase enzyme by converting 25(OH)D to
24,25(OH)2D [67] (Fig. 7).

Klotho FGF23 Interactions

Because Klotho is required for hormonal FGF23 binding,


FGF23 activity is targeted to tissues where Klotho
expression occurs. These are predominantly the distal
convoluted tubule, to a lesser extent the proximal tubule,
the parathyroid and the choroid plexus and to minor
degrees other endocrine organs [68]. So, it remains a
Fig. 7 Serum phosphate, 1,25(OH)2D and PTH upregulate osteocyte
conundrum that Klotho expression is principally located in
FGF23 gene expression. In turn, FGF23 reduces levels of phosphate
and 1,25(OH)2D by downregulating renal sodium-dependent phos- distal tubular cells, while the phosphaturic target of FGF23
phate co-transporter 2a (NPT 2a) expression and 1-alpha hydroxylase appears to be in the proximal tubule. One explanation is
activity, while upregulating 24-hydroxylase activity. FGF23 reduces that despite lower levels of Klotho in the proximal tubule,
PTH by binding to the FGF receptor (FGFR)-Klotho complex.
FGF23 may still be able to signal through the proximal
Additionally, FGF23 increases parathyroid 1-alpha hydroxylase
activity and thereby local 1,25(OH)2D levels. Renal—parathyroid FGFR-Klotho complex to regulate phosphate absorption.
pathways are indicated by the outer arrows Another possibility, made more feasible by the close

123
Clinic Rev Bone Miner Metab

anatomical proximity of distal and proximal tubules, is that FGF23, Klotho and PTH in Health
when FGF23 binds to receptors in the distal tubule a sec-
ond signal is produced that influences proximal tubular An interesting recent finding highlights the interdepen-
phosphate transport [69, 70]. In addition to targeting the dence of FGF23, Klotho and PTH. In a rat model, renal
action of FGF23, Klotho is independently active, being Klotho expression was reduced in the absence of PTH,
shed from cellular membranes in the distal tubule and making the proximal tubule refractory to even high levels
cleaved to circulate in soluble, lower molecular weight of FGF23 [126]. However, when the animals received PTH
forms with enzymatic activity [71, 72]. This soluble Klotho supplementation, renal Klotho levels were restored and
has recently been reported to enter the proximal tubule and FGF23 could induce phosphaturia and normalize serum
inhibit phosphate transport, independent of FGF23 activity phosphate. These experiments demonstrate ‘‘two-way reg-
[73]. Secreted Klotho also co-localize with calcium chan- ulation’’ between FGF23 and PTH when renal function is
nels (transient receptor potential ion channel 5; TRPV5) on normal, with PTH stimulating renal Klotho and osteocyte
distal tubular plasma membranes, which are responsible for FGF23 expression. Parathyroid glands express both FGFR1
approximately 15% of overall calcium reabsorption [74]. and Klotho and as demonstrated in experimental animal
Klotho stabilizes these channels, or prevents their degra- studies, the feedback loop is completed by the rise in
dation, and in this way appears responsible for controlling FGF23 binding to the FGFR-Klotho complex and sup-
levels of urinary calcium [75]. pressing PTH [80]. In addition to this direct feedback,
FGF23 may also suppress PTH release by stimulation of
Hormonal Adaptations to CKD; FGF, Klotho parathyroid 1-a hydroxylase activity, increasing the con-
and Phosphate Excretion version of 25(OH)D to 1,25(OH)2D to in turn suppress
PTH [81] (Fig. 7).
FGF23 levels rise early in the course of CKD, probably in
response to incremental rises of serum phosphate and Changes with CKD
possibly in response to even earlier reductions in Klotho
expression [76]. Initially, the increased plasma FGF23 is an Despite these elegant feedback loops, as renal function
appropriate, compensatory response and the resultant declines FGF23 fails to inhibit PTH (possibly due to
increase in fractional excretion of phosphate can maintain reduced parathyroid Klotho-FGFR1) or to maintain renal
serum phosphate levels within the normal range through to phosphate excretion [82], a circumstance not dissimilar to
CKD stage 3 or 4 in up to 90% of patients [77]. However, FGF or Klotho knockout mouse models, except that these
even extreme FGF23 levels will not prevent hyperphos- animals retain the capacity to produce calcitriol. Their
phatemia if nephron loss continues. At this point, FGF23 phenotypes bear a striking resemblance to that of severe
levels may equal or exceed those seen in conditions of CKD, comprising hyperphosphatemia, growth retardation,
severe FGF-23 excess such as hypophosphatemic rickets skin and muscle atrophy, hypogonadism, vascular and soft
and tumor-induced osteomalacia (TIO) [78] (Fig. 8) and tissue calcification, reduced bone density, premature aging
have been associated with increased mortality and cardio- and a reduced lifespan [83].
vascular disease, although a causative relationship remains
to be established [79]. This rise in FGF23 also downreg- PTH Responses to Altered Mineral Metabolism
ulates production while increasing catabolism of
1,25(OH)2D, and initially this might also be regarded as In 1849 and 1850, Sir Richard Owen dissected a Great
appropriate. Lower 1,25(OH)2D levels reduce dietary Indian Rhinoceros, which till its death had been the prop-
phosphate absorption and de-repress PTH production, erty of the London Zoological Society. He discovered the
increasing phosphaturia, but the risk for this adaptation is Glands of Owen, later named the parathyroids and by 1925
that over time severe hyperparathyroidism may ensue. the role of PTH in calcium regulation had been recognized

Fig. 8 Levels of FGF23 in


CKD greatly exceed those seen
in most other conditions of
FGF23 excess. Reproduced with
permission from [78]

123
Clinic Rev Bone Miner Metab

[84]. However, the intricate mechanisms by which calcium, or vitamin D levels and the underlying mechanisms have
phosphate, 1,25(OH)2D, FGF23 and Klotho influence PTH recently been clarified. Adenine uridine-rich binding factor
transcription and/or post-transcriptional regulation con- 1 (AUF1) is the protein that stabilizes PTH mRNA while
tinue to be illuminated. the destabilizing protein K-homology-type splicing regu-
Fluctuations in ionized calcium rapidly alter PTH latory protein (KSRP) recruits degradation machinery that
secretion, keeping ionized calcium levels within a narrow reduces PTH mRNA stability [89]. These parathyroid
range. Reduction of extracellular calcium levels below the proteins bind to the 3’ untranslated region (UTR) of PTH
‘‘calcium set point’’; the calcium level corresponding to the mRNA. The KSRP destabilizing protein is in turn con-
midpoint of the steep, reverse sigmoidal PTH response trolled by the peptidyl-prolyl cis/trans isomerase Pin1. In
curve, activates calcium sensing receptor (CaR) signaling hypocalcaemia and experimental CKD, Pin1 activity is
via an inhibitory G-coupled protein to release preformed reduced, resulting in KSRP phosphorylation and inactiva-
PTH from stored secretory granules. On the other hand, tion. This allows increased binding of the stabilizing pro-
PTH secretion is suppressed when elevated levels of tein AUF1 to PTH mRNA. On the other hand, in phosphate
extracellular calcium acting through an alternative G-cou- depletion experiments KSRP activity is increased, leading
pled protein cause the release of calcium from endoplasmic to a reduction in AUF1 binding and to reduced PTH
reticulum. mRNA stability [90].
Even when calcium levels are strikingly elevated, PTH
secretion cannot be completely suppressed. However, the Parathyroid Hyperplasia
parathyroid can respond to elevation of calcium levels by
increasing the degradation of preformed PTH to PTH As secondary hyperparathyroidism develops, parathyroid
fragments and constituent amino acids within parathyroid cells undergo mitosis causing parathyroid enlargement.
cells. This results in the secretion of a higher proportion of This hyperplasia is a consequence of hypocalcaemia,
PTH carboxyl-terminal fragments, which may lack the hyperphosphatemia and reduced levels of 1,25-di-
calcaemic activity of PTH 1-84. The best characterized of hydroxyvitamin D [91] and can be prevented by phosphate
these is PTH [7–84], which appears to act as an antagonist restriction or therapy with 1,25(OH)2D [87, 92].
of the PPR and to exert biological effects through a novel 1,25(OH)2D exerts its anti-proliferative effects through
c-terminal receptor expressed solely by osteocytes [15, 85]. induction of p21 gene expression [93]. Similarly, reducing
This receptor is possibly part of a larger receptor complex dietary phosphate induces p21, independent of changes in
and may require levels of PTH above the normal upper 1,25(OH)2D [94]. On the other hand, increased cellular
range for activity. proliferation cannot be ascribed solely to downregulation
of p21 expression.
Transcriptional Regulation of PTH A further mechanism has recently been identified.
Reduced levels of 1,25(OH)2D or high levels of dietary
In contrast to FGF23, PTH increases renal Cyp27B1 phosphate induce translocation to the cell surface of a
activity and the conversion of 25(OH)D to 1,25(OH)2D tumor necrosis factor-a-converting enzyme (TACE), which
(Fig. 7). This provides direct negative feedback by down- results in release of the growth promoter TGF-a [95, 96]. In
regulation of PTH mRNA transcription, as well as indirect turn, TGF-a binds to and activates the epidermal growth
feedback by activation of osteocyte FGF23 release. factor receptor (EGFR). Activated EGFR then translocates
Increased levels of 1,25(OH)2D also increase CaR to the nucleus, where it functions as an autocrine signal for
expression, sensitizing parathyroid cells to inhibition of cell growth. With phosphate restriction parathyroid TGF-a
PTH secretion by calcium [86]. Compared to the rapid returns to normal levels rapidly, which suggests that low-
effects of calcium on PTH secretion, these transcriptional ering dietary phosphate may counteract uremia-induced
pathways influence PTH over hours, with much longer parathyroid hyperplasia by preventing parathyroid TGF-a
periods required for changes to occur in cellular enhancement in addition to inducing expression of p21.
proliferation. In order to impede the development of secondary
hyperparathyroidism and parathyroid hyperplasia, treat-
Post-Transcriptional PTH Regulation ment with calcitriol or its analogs is often initiated in the in
the early stages of CKD. Given that osteocytes respond to
Phosphate levels directly influence PTH (Fig. 7) by alter- 1,25(OH)2D by increasing the secretion of FGF23, this
ing the stability of PTH mRNA [87], with a rise in the approach has been questioned [56]. Nevertheless, some
serum phosphate increasing PTH mRNA stability and a studies suggest that the additional effect of calcitriol ther-
reduction in dietary phosphate reducing PTH mRNA sta- apy on already elevated FGF23 levels may be relatively
bility [88]. These changes occur independently of calcium modest [97]. Moreover, treatment with calcitriol or its

123
Clinic Rev Bone Miner Metab

analogs has been associated with reduced or stable mor- [110]. BMP-7 has been used successfully to treat the
tality [98, 99] and in animal studies, with increased renal adynamic bone disorder that developed in adult mice after
expression of Klotho [100]. Optimal treatment should in the induction of renal impairment [111]. Because BMP-7
fact be low dose or ‘physiological’ replacement, aimed to upregulates the expression of osteoblast PTH receptors,
avoid hypercalcemia, hyperphosphatemia and the pro- higher levels of PTH may be required for regulation of
gression of vascular calcification that can be aggravated by stem cell niches when levels of BMP-7 are reduced by
aggressive therapy. We should also avoid overzealous renal damage [108]. Alterations to Wnt signaling and levels
suppression of PTH levels toward the ‘‘normal’’ range in of Wnt inhibitory proteins have also been recently descri-
patients with CKD stages 5 or 5D, which can result in bed in animal models of CKD [112, 113] and in patients
exposure of a low bone turnover phenotype [101]. undergoing dialysis [114]. In studies using the Jck mouse
model of polycystic kidney disease, an early rise in FGF23
PTH and Stem Cell Niches was reported, with a later rise in levels of PTH, phosphate
and the expression of genes associated with osteoblast
Mesenchymal and hematopoietic stem cells survive and function [112]. The authors proposed that an early increase
regenerate in a complex and protected bone micro-envi- in SOST expression reduced Wnt signaling and conse-
ronment, influenced by surrounding cells including osteo- quently the OPG to RANKL ratio. In turn, this would
blasts, osteocytes and sympathetic neurones [102, 103]. increase osteoclast activity and predispose to osteitis fib-
PTH, prostaglandins and beta-adrenergic stimuli all appear rosa, which was the pattern described on bone biopsy.
to play important roles by signaling through osteoblast A later rise in PTH levels sustained high bone turnover and
lineage cells to maintain stem cell survival and activity reduced SOST gene expression, but elevated levels of
[104], osteogenesis and erythropoiesis. PTH has been secreted frizzled related protein 4 (sFRP4) were considered
shown to increase the proliferation rate of mesenchymal sufficient to maintain suppression of Wnt and osteoblast
stem cells (MSC) with a diminution of senescence and gene expression. In a rat model of renal failure, PTH and
apoptosis [105], a putative protective action thought to phosphate levels were elevated after 20 weeks and the Wnt
reduce MSC stress and preserve genome integrity. This inhibiting proteins sFRP and Dkk-1 were over-expressed
ability was highlighted in mice following lethal irradiation [113]. In a recent report, which included 630 bone biopsies
and marrow transplantation; animals treated with PTH from patients undergoing dialysis, 62% of white and 32%
survived, while those treated with mock injection had less of black subjects were diagnosed to have low bone turn-
than 50% survival at 10 days [106]. over [107]. In 60 patients, sclerostin levels were assessed
Low bone turnover is a common form of renal osteo- and found to be approximately 5 times greater than pre-
dystrophy estimated to be present in over 60% of white menopausal women and twice those of post menopausal
patients on dialysis [107]. It has been suggested that when women [114]. After adjustment for age, gender, dialysis
Wnt signaling and osteoblast activity is inhibited early in duration, diabetes and treatment with vitamin D, sclerostin
CKD (for which low turnover, ‘‘hypodynamic’’ or ady- levels correlated negatively with activation frequency
namic bone might be the histomorphometric phenotype), (p \ 0.003) and bone formation rate/bone surface
the development of secondary hyperparathyroidism may be (p = 0.007). However, there were no associations between
a compensatory mechanism to maintain or ‘‘rescue’’ stem levels of Dkk-1 and these bone parameters.
cell niches [108]. Of course, with progressive CKD such Whether low (or absent) bone remodeling increases
‘‘compensatory’’ PTH responses becomes inappropriate bone micro damage and fracture risk remains the subject of
and sustained elevation of PTH can result in bone changes a longstanding debate, as does the use of the term ‘‘ady-
of hyperparathyroidism including the proliferation of namic bone disease’’ to describe this condition [115]. At
fibroblast-like cells in the bone marrow, termed osteitis the other extreme, poorly mineralized and/or high turnover
fibrosa. These cells are now considered to be pre-osteo- ‘‘woven’’ bone undoubtedly reduces bone’s structural
blasts that have proliferated in the presence of elevated integrity. However, the extremes of turnover have impli-
PTH levels and will fail to mature into osteoblasts unless cations for more than bone health. Bone’s exchangeable
normal PTH levels are restored [109]. calcium pool is located at least in part in the hydrated
Understanding changes that may lead to reduced osteoid covering the bone surfaces [9]. With low turnover,
osteoblast activity and bone turnover in early CKD is still quiescent, mineralized bone surfaces increase at the
in the developmental stage. However, abnormalities of expense of osteoid, so that the surface available for calcium
bone morphogenic protein (BMP) levels and of Wnt sig- exchange diminishes. In the context of CKD and a reduced
naling are likely contributors. BMP-7 is an important renal excretory capacity for calcium, loss of this bone
promoter of osteoblast differentiation that is produced ‘‘sink’’ for calcium sequestration may increase the risk of
primarily by the kidneys, so that levels are reduced in CKD vascular calcification, particularly when calcium balance is

123
Clinic Rev Bone Miner Metab

positive. High bone turnover with a mismatch of formation others, such as associations between low turnover and
to resorption may also increase the net efflux of calcium diabetes that may be mediated by advanced glycation end-
and phosphate from bone and similarly predispose to vas- products, neurological or circulatory effects. Together with
cular calcification. other articles in this edition, I hope this ‘‘nephrologists
guide to bone biology’’ might highlight the beautiful
Markers of Early CKD complexity of this fascinating area for clinical and labo-
ratory research. Certainly, it is an area that seems to
Because rises in FGF23 occur as early as CKD stage 1 and acquire momentum every day.
2 when the estimated GFR is still 60–90 ml/min per
1.73 m2 [116, 117] (Fig. 8), FGF23 measurement may
provide a sensitive marker of early disturbances in phos-
phate metabolism, progressive kidney failure and risk of References
mortality. This could be used as a guide to commencing
dietary phosphate restriction or other interventions to 1. Bricker NS, Morrin PA, Kime SW Jr. The pathologic physiology
of chronic Bright’s disease. An exposition of the ‘‘intact nephron
reduce dietary phosphate absorption, and to commencing hypothesis’’. Am J Med. 1960;28:77–98.
other measures aimed to reduce progression of kidney 2. Parfitt AM. Misconceptions (2): turnover is always higher in
damage [118]. As well as being a prognostic marker, the cancellous than in cortical bone. Bone. 2002;30(6):807–9.
rise in levels of FGF23 that occurs with progressive CKD 3. Parfitt AM. Osteonal and hemi-osteonal remodeling: the spatial
and temporal framework for signal traffic in adult human bone.
may be of independent clinical significance, having been J Cell Biochem. 1994;55(3):273–86.
associated with disorders of myocardial mass and perfor- 4. Parfitt AM. Targeted and nontargeted bone remodeling: rela-
mance, increased risk of vascular calcification, CKD pro- tionship to basic multicellular unit origination and progression.
gression and mortality [119–121]. Urinary levels of Bone. 2002;30(1):5–7.
5. Polig E, Jee WS. Bone age and remodeling: a mathematical
secreted Klotho might also be utilized for the detection of treatise. Calcif Tissue Int. 1987;41(3):130–6.
nascent renal damage, because declines have been reported 6. Verborgt O, Gibson GJ, Schaffler MB. Loss of osteocyte
as early as CKD stage 1 [122, 123]. Intervention studies are integrity in association with microdamage and bone remodeling
currently underway, which will help to define the utility of after fatigue in vivo. J Bone Miner Res. 2000;15(1):60–7.
7. Porter RL, Calvi LM. Communications between bone cells and
estimating and treating these early markers by dietary and hematopoietic stem cells. Arch Biochem Biophys. 2008;473(2):
other means [124]. It is hoped these strategies might reduce 193–200.
CKD progression and complications. 8. Yasuda H, Shima N, Nakagawa N, Yamaguchi K, Kinosaki M,
Mochizuki S, et al. Osteoclast differentiation factor is a ligand
for osteoprotegerin/osteoclastogenesis-inhibitory factor and is
identical to TRANCE/RANKL. Proc Natl Acad Sci USA.
Summary 1998;95(7):3597–602.
9. Parfitt AM. Calcium homeostasis. In: Mundy GR, Martin TJ,
The development of CKD produces compensatory altera- editors. Handbook of experimental pharmacology. Heidelberg:
Springer; 1993. p. 1–65.
tions to bone and mineral metabolism that progress as renal 10. Parfitt AM. The actions of parathyroid hormone on bone: rela-
function declines, often resulting in end-organ damage. tion to bone remodeling and turnover, calcium homeostasis, and
The complexity of these interactions is only now being metabolic bone diseases. II. PTH and bone cells: bone turnover
appreciated, with intricate feedback pathways demon- and plasma calcium regulation. Metabolism. 1976;25(8):
909–55.
strated between bone and the kidneys, parathyroids, the 11. Parfitt AM. The actions of parathyroid hormone on bone: rela-
brain and sympathetic nervous system, the pancreas, gas- tion to bone remodeling and turnover, calcium homeostasis, and
trointestinal tract, adipose tissues and of course the vas- metabolic bone disease. Part I of IV parts: mechanisms of cal-
cular tree. Many factors in addition to those highlighted cium transfer between blood and bone and their cellular basis:
morphological and kinetic approaches to bone turnover.
here influence the balance between these systems, ranging Metabolism. 1976;25(7):809–44.
from reactive oxygen species to bone marrow temperature 12. Powell WF Jr, Barry KJ, Tulum I, Kobayashi T, Harris SE,
regulation by brown fat! All of these are likely to be Bringhurst FR, et al. Targeted ablation of the PTH/PTHrP
influenced by the development of CKD. receptor in osteocytes impairs bone structure and homeostatic
calcemic responses. J Endocrinol. 2011;209(1):21–32.
The response of bone turnover to these physiological 13. Harada S, Rodan GA. Control of osteoblast function and regu-
changes forms a continuum, ranging from adynamic or lation of bone mass. Nature. 2003;423(6937):349–55.
hypodynamic, with implications for stem cell niches, 14. Kamioka H, Honjo T, Takano-Yamamoto T. A three-dimen-
hematopoiesis and osteogenesis, to severe hyperparathy- sional distribution of osteocyte processes revealed by the com-
bination of confocal laser scanning microscopy and differential
roidism and osteitis fibrosa. I have described some of the interference contrast microscopy. Bone. 2001;28(2):145–9.
contributing mechanisms, such as altered Wnt signaling 15. Divieti P, Geller AI, Suliman G, Juppner H, Bringhurst FR.
and reduction in levels of BMP-7, but have not addressed Receptors specific for the carboxyl-terminal region of

123
Clinic Rev Bone Miner Metab

parathyroid hormone on bone-derived cells: determinants of 34. Robling AG, Niziolek PJ, Baldridge LA, Condon KW, Allen
ligand binding and bioactivity. Endocrinology. 2005;146(4): MR, Alam I, et al. Mechanical stimulation of bone in vivo
1863–70. reduces osteocyte expression of Sost/sclerostin. J Biol Chem.
16. Xiong J, Onal M, Jilka RL, Weinstein RS, Manolagas SC, 2008;283(9):5866–75.
O’Brien CA. Matrix-embedded cells control osteoclast forma- 35. Wergedal JE, Veskovic K, Hellan M, Nyght C, Balemans W,
tion. Nat Med 2011;17(10):1235–1241. Libanati C, et al. Patients with Van Buchem disease, an osteo-
17. Nakashima T, Hayashi M, Fukunaga T, Kurata K, Oh-Hora M, sclerotic genetic disease, have elevated bone formation markers,
Feng JQ, et al. Evidence for osteocyte regulation of bone higher bone density, and greater derived polar moment of inertia
homeostasis through RANKL expression. Nat Med 2011;17(10): than normal. J Clin Endocrinol Metab. 2003;88(12):5778–83.
1231–1234. 36. Balemans W, Patel N, Ebeling M, Van Hul E, Wuyts W, Lacza
18. Weiss AJ, Iqbal J, Zaidi N, Mechanick JI. The skeletal sub- C, et al. Identification of a 52 kb deletion downstream of the
system as an integrative physiology paradigm. Curr Osteoporos SOST gene in patients with van Buchem disease. J Med Genet.
Rep. 2010;8(4):168–77. 2002;39(2):91–7.
19. Hayden JM, Mohan S, Baylink DJ. The insulin-like growth 37. Mao B, Wu W, Davidson G, Marhold J, Li M, Mechler BM,
factor system and the coupling of formation to resorption. Bone. et al. Kremen proteins are Dickkopf receptors that regulate Wnt/
1995;17(Suppl 2):93S–8S. beta-catenin signalling. Nature. 2002;417(6889):664–7.
20. Centrella M, McCarthy TL, Canalis E. Transforming growth 38. Bonewald LF, Johnson ML. Osteocytes, mechanosensing and
factor-beta and remodeling of bone. J Bone Joint Surg Am. Wnt signaling. Bone. 2008;42(4):606–15.
1991;73(9):1418–28. 39. Mikels AJ, Nusse R. Purified Wnt5a protein activates or inhibits
21. Tang Y, Wu X, Lei W, Pang L, Wan C, Shi Z, et al. TGF-beta1- beta-catenin-TCF signaling depending on receptor context.
induced migration of bone mesenchymal stem cells couples PLoS Biol. 2006;4(4):e115.
bone resorption with formation. Nat Med. 2009;15(7):757–65. 40. Choi HY, Dieckmann M, Herz J, Niemeier A. Lrp4, a novel
22. Edwards CM, Mundy GR. Eph receptors and ephrin signaling receptor for Dickkopf 1 and sclerostin, is expressed by osteo-
pathways: a role in bone homeostasis. Int J Med Sci. 2008;5(5): blasts and regulates bone growth and turnover in vivo. PLoS
263–72. One. 2009;4(11):e7930.
23. Pederson L, Ruan M, Westendorf JJ, Khosla S, Oursler MJ. 41. Chen W, ten Berge D, Brown J, Ahn S, Hu LA, Miller WE, et al.
Regulation of bone formation by osteoclasts involves Wnt/BMP Dishevelled 2 recruits beta-arrestin 2 to mediate Wnt5A-stim-
signaling and the chemokine sphingosine-1-phosphate. Proc ulated endocytosis of Frizzled 4. Science. 2003;301(5638):
Natl Acad Sci USA. 2008;105(52):20764–9. 1391–4.
24. Zhao B, Takami M, Yamada A, Wang X, Koga T, Hu X, et al. 42. Fiedler M, Mendoza-Topaz C, Rutherford TJ, Mieszczanek J,
Interferon regulatory factor-8 regulates bone metabolism by Bienz M. Dishevelled interacts with the DIX domain polymer-
suppressing osteoclastogenesis. Nat Med. 2009;15(9):1066–71. ization interface of Axin to interfere with its function in down-
25. Kusu N, Laurikkala J, Imanishi M, Usui H, Konishi M, Miyake regulating beta-catenin. Proc Natl Acad Sci USA. 2011;108(5):
A, et al. Sclerostin is a novel secreted osteoclast-derived bone 1937–42.
morphogenetic protein antagonist with unique ligand specificity. 43. Tutter AV, Fryer CJ, Jones KA. Chromatin-specific regulation
J Biol Chem. 2003;278(26):24113–7. of LEF-1-beta-catenin transcription activation and inhibition in
26. Klein-Nulend J, van der Plas A, Semeins CM, Ajubi NE, vitro. Genes Dev. 2001;15(24):3342–54.
Frangos JA, Nijweide PJ, et al. Sensitivity of osteocytes to 44. Hoeppner LH, Secreto F, Jensen ED, Li X, Kahler RA,
biomechanical stress in vitro. FASEB J. 1995;9(5):441–5. Westendorf JJ. Runx2 and bone morphogenic protein 2 regulate
27. Han Y, Cowin SC, Schaffler MB, Weinbaum S. Mechano- the expression of an alternative Lef1 transcript during osteoblast
transduction and strain amplification in osteocyte cell processes. maturation. J Cell Physiol. 2009;221(2):480–9.
Proc Natl Acad Sci USA. 2004;101(47):16689–94. 45. Glass DA 2nd, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers
28. Price C, Zhou X, Li W, Wang L. Real-time measurement of H, et al. Canonical Wnt signaling in differentiated osteo-
solute transport within the lacunar-canalicular system of blasts controls osteoclast differentiation. Dev Cell. 2005;8(5):
mechanically loaded bone: direct evidence for load-induced 751–64.
fluid flow. J Bone Miner Res. 2011;26(2):277–85. 46. Ross SE, Hemati N, Longo KA, Bennett CN, Lucas PC,
29. Aguirre JI, Plotkin LI, Gortazar AR, Millan MM, O’Brien CA, Erickson RL, et al. Inhibition of adipogenesis by Wnt signaling.
Manolagas SC, et al. A novel ligand-independent function of the Science. 2000;289(5481):950–3.
estrogen receptor is essential for osteocyte and osteoblast 47. Hill TP, Spater D, Taketo MM, Birchmeier W, Hartmann C.
mechanotransduction. J Biol Chem. 2007;282(35):25501–8. Canonical Wnt/beta-catenin signaling prevents osteoblasts from
30. Jessop HL, Suswillo RF, Rawlinson SC, Zaman G, Lee K, differentiating into chondrocytes. Dev Cell. 2005;8(5):727–38.
Das-Gupta V, et al. Osteoblast-like cells from estrogen receptor 48. Bricker NS. On the pathogenesis of the uremic state. An exposi-
alpha knockout mice have deficient responses to mechanical tion of the ‘‘trade-off hypothesis’’. N Engl J Med. 1972;286(20):
strain. J Bone Miner Res. 2004;19(6):938–46. 1093–9.
31. Poole KE, van Bezooijen RL, Loveridge N, Hamersma H, 49. Slatopolsky E, Caglar S, Gradowska L, Canterbury J, Reiss E,
Papapoulos SE, Lowik CW, et al. Sclerostin is a delayed Bricker NS. On the prevention of secondary hyperparathyroid-
secreted product of osteocytes that inhibits bone formation. ism in experimental chronic renal disease using ‘‘proportional
FASEB J. 2005;19(13):1842–4. reduction’’ of dietary phosphorus intake. Kidney Int. 1972;2(3):
32. Li X, Zhang Y, Kang H, Liu W, Liu P, Zhang J, et al. Sclerostin 147–51.
binds to LRP5/6 and antagonizes canonical Wnt signaling. 50. Marotti G, Ferretti M, Remaggi F, Palumbo C. Quantitative
J Biol Chem. 2005;280(20):19883–7. evaluation on osteocyte canalicular density in human secondary
33. Veverka V, Henry AJ, Slocombe PM, Ventom A, Mulloy B, osteons. Bone. 1995;16(1):125–8.
Muskett FW, et al. Characterization of the structural features 51. Aarden EM, Burger EH, Nijweide PJ. Function of osteocytes in
and interactions of sclerostin: molecular insight into a key reg- bone. J Cell Biochem. 1994;55(3):287–99.
ulator of Wnt-mediated bone formation. J Biol Chem. 2009; 52. Burnett SM, Gunawardene SC, Bringhurst FR, Juppner H, Lee
284(16):10890–900. H, Finkelstein JS. Regulation of C-terminal and intact FGF-23

123
Clinic Rev Bone Miner Metab

by dietary phosphate in men and women. J Bone Miner Res. 71. Matsumura Y, Aizawa H, Shiraki-Iida T, Nagai R, Kuro-o M,
2006;21(8):1187–96. Nabeshima Y. Identification of the human klotho gene and its
53. Nishida Y, Taketani Y, Yamanaka-Okumura H, Imamura F, two transcripts encoding membrane and secreted klotho protein.
Taniguchi A, Sato T, et al. Acute effect of oral phosphate Biochem Biophys Res Commun. 1998;242(3):626–30.
loading on serum fibroblast growth factor 23 levels in healthy 72. Imura A, Iwano A, Tohyama O, Tsuji Y, Nozaki K, Hashimoto
men. Kidney Int. 2006;70(12):2141–7. N, et al. Secreted Klotho protein in sera and CSF: implication
54. Ito N, Fukumoto S, Takeuchi Y, Takeda S, Suzuki H, Yamashita for post-translational cleavage in release of Klotho protein from
T, et al. Effect of acute changes of serum phosphate on fibroblast cell membrane. FEBS Lett. 2004;565(1–3):143–7.
growth factor (FGF)23 levels in humans. J Bone Miner Metab. 73. Hu MC, Shi M, Zhang J, Pastor J, Nakatani T, Lanske B, et al. Klotho:
2007;25(6):419–22. a novel phosphaturic substance acting as an autocrine enzyme in the
55. Isakova T, Gutierrez O, Shah A, Castaldo L, Holmes J, Lee H, renal proximal tubule. FASEB J. 2010;24(9):3438–50.
et al. Postprandial mineral metabolism and secondary hyper- 74. Chang Q, Hoefs S, van der Kemp AW, Topala CN, Bindels RJ,
parathyroidism in early CKD. J Am Soc Nephrol. 2008;19(3): Hoenderop JG. The beta-glucuronidase klotho hydrolyzes and
615–23. activates the TRPV5 channel. Science. 2005;310(5747):490–3.
56. Nishi H, Nii-Kono T, Nakanishi S, Yamazaki Y, Yamashita T, 75. Cha SK, Ortega B, Kurosu H, Rosenblatt KP, Kuro OM, Huang
Fukumoto S, et al. Intravenous calcitriol therapy increases CL. Removal of sialic acid involving Klotho causes cell-surface
serum concentrations of fibroblast growth factor-23 in dialysis retention of TRPV5 channel via binding to galectin-1. Proc Natl
patients with secondary hyperparathyroidism. Nephron Clin Acad Sci USA. 2008;105(28):9805–10.
Pract. 2005;101(2):c94–9. 76. Bernheim J, Benchetrit S. The potential roles of FGF23 and
57. Liu S, Tang W, Zhou J, Stubbs JR, Luo Q, Pi M, et al. Fibroblast Klotho in the prognosis of renal and cardiovascular diseases.
growth factor 23 is a counter-regulatory phosphaturic hormone Nephrol Dial Transplant. 2011;26(8):2433–8.
for vitamin D. J Am Soc Nephrol. 2006;17(5):1305–15. 77. Gutierrez O, Isakova T, Rhee E, Shah A, Holmes J, Collerone G,
58. Lavi-Moshayoff V, Wasserman G, Meir T, Silver J, Naveh- et al. Fibroblast growth factor-23 mitigates hyperphosphatemia
Many T. PTH increases FGF23 gene expression and mediates but accentuates calcitriol deficiency in chronic kidney disease.
the high-FGF23 levels of experimental kidney failure: a bone J Am Soc Nephrol. 2005;16(7):2205–15.
parathyroid feedback loop. Am J Physiol Renal Physiol. 2010; 78. Isakova T, Gutierrez OM, Wolf M. A blueprint for randomized
299(4):F882–9. trials targeting phosphorus metabolism in chronic kidney dis-
59. Lopez I, Rodriguez-Ortiz ME, Almaden Y, Guerrero F, Oca ease. Kidney Int. 2009;76(7):705–16.
AM, Pineda C, et al. Direct and indirect effects of parathyroid 79. Fukumoto S, Shimizu Y. Fibroblast growth factor 23 as a phos-
hormone on circulating levels of fibroblast growth factor 23 in photropic hormone and beyond. J Bone Miner Metab. 2011;29(5):
vivo. Kidney Int. 2011;80(5):475–82. 507–14.
60. Chefetz I, Kohno K, Izumi H, Uitto J, Richard G, Sprecher E. 80. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, Goetz R, Kuro-o
GALNT3, a gene associated with hyperphosphatemic familial M, Mohammadi M, et al. The parathyroid is a target organ for
tumoral calcinosis, is transcriptionally regulated by extracellular FGF23 in rats. J Clin Invest. 2007;117(12):4003–8.
phosphate and modulates matrix metalloproteinase activity. 81. Krajisnik T, Bjorklund P, Marsell R, Ljunggren O, Akerstrom G,
Biochim Biophys Acta. 2009;1792(1):61–7. Jonsson KB, et al. Fibroblast growth factor-23 regulates para-
61. Quarles LD. Endocrine functions of bone in mineral metabolism thyroid hormone and 1alpha-hydroxylase expression in cultured
regulation. J Clin Invest. 2008;118(12):3820–8. bovine parathyroid cells. J Endocrinol. 2007;195(1):125–31.
62. Itoh N, Ornitz DM. Functional evolutionary history of the mouse 82. Canalejo R, Canalejo A, Martinez-Moreno JM, Rodriguez-Ortiz
Fgf gene family. Dev Dyn. 2008;237(1):18–27. ME, Estepa JC, Mendoza FJ, et al. FGF23 fails to inhibit uremic
63. Gattineni J, Twombley K, Goetz R, Mohammadi M, Baum M. parathyroid glands. J Am Soc Nephrol. 2010;21(7):1125–35.
Regulation of serum 1, 25(OH)2 vitamin D3 levels by fibroblast 83. Memon F, El-Abbadi M, Nakatani T, Taguchi T, Lanske B,
growth factor 23 is mediated by FGF receptors 3 and 4. Am J Razzaque MS. Does Fgf23-klotho activity influence vascular
Physiol Renal Physiol. 2011;301(2):F371–7. and soft tissue calcification through regulating mineral ion
64. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, metabolism? Kidney Int. 2008;74(5):566–70.
Utsugi T, et al. Mutation of the mouse klotho gene leads to a 84. Collip JB, Leitch DB. A case of tetany treated with Parathyrin.
syndrome resembling ageing. Nature. 1997;390(6655):45–51. Can Med Assoc J. 1925;15(1):59–60.
65. Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, 85. Divieti P, Inomata N, Chapin K, Singh R, Juppner H, Bringhurst
Okawa K, et al. Klotho converts canonical FGF receptor into a FR. Receptors for the carboxyl-terminal region of pth(1–84) are
specific receptor for FGF23. Nature. 2006;444(7120):770–4. highly expressed in osteocytic cells. Endocrinology. 2001;142(2):
66. Yan X, Yokote H, Jing X, Yao L, Sawada T, Zhang Y, et al. 916–25.
Fibroblast growth factor 23 reduces expression of type IIa Na?/ 86. Canaff L, Hendy GN. Human calcium-sensing receptor gene.
Pi co-transporter by signaling through a receptor functionally Vitamin D response elements in promoters P1 and P2 confer
distinct from the known FGFRs in opossum kidney cells. Genes transcriptional responsiveness to 1, 25-dihydroxyvitamin D.
Cells. 2005;10(5):489–502. J Biol Chem. 2002;277(33):30337–50.
67. Inoue Y, Segawa H, Kaneko I, Yamanaka S, Kusano K, Ka- 87. Slatopolsky E, Finch J, Denda M, Ritter C, Zhong M, Dusso A,
wakami E, et al. Role of the vitamin D receptor in FGF23 action et al. Phosphorus restriction prevents parathyroid gland growth.
on phosphate metabolism. Biochem J. 2005;390(Pt 1):325–31. High phosphorus directly stimulates PTH secretion in vitro.
68. Nabeshima Y. Klotho: a fundamental regulator of aging. Ageing J Clin Invest. 1996;97(11):2534–40.
Res Rev. 2002;1(4):627–38. 88. Moallem E, Kilav R, Silver J, Naveh-Many T. RNA-Protein
69. Liu S, Vierthaler L, Tang W, Zhou J, Quarles LD. FGFR3 and binding and post-transcriptional regulation of parathyroid hor-
FGFR4 do not mediate renal effects of FGF23. J Am Soc mone gene expression by calcium and phosphate. J Biol Chem.
Nephrol. 2008;19(12):2342–50. 1998;273(9):5253–9.
70. Farrow EG, Davis SI, Summers LJ, White KE. Initial FGF23- 89. Naveh-Many T. Minireview: the play of proteins on the para-
mediated signaling occurs in the distal convoluted tubule. J Am thyroid hormone messenger ribonucleic Acid regulates its
Soc Nephrol. 2009;20(5):955–60. expression. Endocrinology. 2010;151(4):1398–402.

123
Clinic Rev Bone Miner Metab

90. Nechama M, Ben-Dov IZ, Briata P, Gherzi R, Naveh-Many T. 108. Hruska KA, Saab G, Mathew S, Lund R. Renal osteodystrophy,
The mRNA decay promoting factor K-homology splicing reg- phosphate homeostasis, and vascular calcification. Semin Dial.
ulator protein post-transcriptionally determines parathyroid 2007;20(4):309–15.
hormone mRNA levels. FASEB J. 2008;22(10):3458–68. 109. Lotinun S, Sibonga JD, Turner RT. Evidence that the cells
91. Parfitt AM. The hyperparathyroidism of chronic renal failure: a responsible for marrow fibrosis in a rat model for hyperpara-
disorder of growth. Kidney Int. 1997;52(1):3–9. thyroidism are preosteoblasts. Endocrinology. 2005;146(9):
92. Szabo A, Merke J, Beier E, Mall G, Ritz E. 1, 25(OH)2 vitamin 4074–81.
D3 inhibits parathyroid cell proliferation in experimental uremia. 110. Chaudhary LR, Hofmeister AM, Hruska KA. Differential
Kidney Int. 1989;35(4):1049–56. growth factor control of bone formation through osteoprogenitor
93. Liu M, Lee MH, Cohen M, Bommakanti M, Freedman LP. differentiation. Bone. 2004;34(3):402–11.
Transcriptional activation of the Cdk inhibitor p21 by vitamin 111. Lund RJ, Davies MR, Brown AJ, Hruska KA. Successful
D3 leads to the induced differentiation of the myelomonocytic treatment of an adynamic bone disorder with bone morphoge-
cell line U937. Genes Dev. 1996;10(2):142–53. netic protein-7 in a renal ablation model. J Am Soc Nephrol.
94. Dusso AS, Pavlopoulos T, Naumovich L, Lu Y, Finch J, Brown 2004;15(2):359–69.
AJ, et al. p21(WAF1) and transforming growth factor-alpha 112. Sabbagh Y, Tan W, Bracken C, O’Brien S, Phillips L, Ryan S,
mediate dietary phosphate regulation of parathyroid cell growth. et al. High turnover renal osteodystrophy is associated with beta-
Kidney Int. 2001;59(3):855–65. catenin repression in osteocytes (Abstract).. J Bone Miner Res
95. Cozzolino M, Lu Y, Finch J, Slatopolsky E, Dusso AS. 2011;ASBMR Annual Meeting:SA0177.
p21WAF1 and TGF-alpha mediate parathyroid growth arrest by 113. Carrillo-Lopez N, Martin-Gutierez L, Roman-Garcia P, Panizo
vitamin D and high calcium. Kidney Int. 2001;60(6):2109–17. S, Naves-Diaz M, Cannata Andia J. Evidence that PTH can
96. Dusso A, Arcidiacono MV, Yang J, Tokumoto M. Vitamin D be directly involved in the regulation of the secreted frizzled
inhibition of TACE and prevention of renal osteodystrophy and related proteins (sFRPs) and the Wnt signaling pathway
cardiovascular mortality. J Steroid Biochem Mol Biol. 2010; (Abstract). J Bone Miner Res 2011; ASBMR Annual Meeting:
121(1–2):193–8. SU0176.
97. Imel EA, DiMeglio LA, Hui SL, Carpenter TO, Econs MJ. 114. Cejka D, Herberth J, Branscum AJ, Fardo DW, Monier-
Treatment of X-linked hypophosphatemia with calcitriol and Faugere MC, Diarra D, et al. Sclerostin and Dickkopf-1 in
phosphate increases circulating fibroblast growth factor 23 renal osteodystrophy. Clin J Am Soc Nephrol. 2011;6(4):
concentrations. J Clin Endocrinol Metab. 2010;95(4):1846–50. 877–82.
98. Andress DL. Vitamin D in chronic kidney disease: a systemic 115. Parfitt AM. Renal bone disease: a new conceptual framework for
role for selective vitamin D receptor activation. Kidney Int. the interpretation of bone histomorphometry. Curr Opin Nephrol
2006;69(1):33–43. Hypertens. 2003;12(4):387–403.
99. Palmer SC, McGregor DO, Macaskill P, Craig JC, Elder GJ, 116. Ix JH, Shlipak MG, Wassel CL, Whooley MA. Fibroblast
Strippoli GF. Meta-analysis: vitamin D compounds in chronic growth factor-23 and early decrements in kidney function: the
kidney disease. Ann Intern Med. 2007;147(12):840–53. Heart and Soul Study. Nephrol Dial Transplant. 2010;25(3):
100. Tsujikawa H, Kurotaki Y, Fujimori T, Fukuda K, Nabeshima Y. 993–7.
Klotho, a gene related to a syndrome resembling human pre- 117. Isakova T, Xie H, Yang W, Xie D, Anderson AH, Scialla J, et al.
mature aging, functions in a negative regulatory circuit of Fibroblast growth factor 23 and risks of mortality and end-stage
vitamin D endocrine system. Mol Endocrinol. 2003;17(12): renal disease in patients with chronic kidney disease. JAMA.
2393–403. 2011;305(23):2432–9.
101. Gal-Moscovici A, Popovtzer MM. New worldwide trends in 118. Oliveira RB, Cancela AL, Graciolli FG, Dos Reis LM, Draibe
presentation of renal osteodystrophy and its relationship to SA, Cuppari L, et al. Early control of PTH and FGF23 in nor-
parathyroid hormone levels. Clin Nephrol. 2005;63(4):284–9. mophosphatemic CKD patients: a new target in CKD-MBD
102. Zhu J, Garrett R, Jung Y, Zhang Y, Kim N, Wang J, et al. therapy? Clin J Am Soc Nephrol. 2010;5(2):286–91.
Osteoblasts support B-lymphocyte commitment and differenti- 119. Gutierrez OM, Januzzi JL, Isakova T, Laliberte K, Smith K,
ation from hematopoietic stem cells. Blood. 2007;109(9): Collerone G, et al. Fibroblast growth factor 23 and left ven-
3706–12. tricular hypertrophy in chronic kidney disease. Circulation.
103. Chow A, Lucas D, Hidalgo A, Mendez-Ferrer S, Hashimoto D, 2009;119(19):2545–52.
Scheiermann C, et al. Bone marrow CD169? macrophages 120. Juppner H, Wolf M, Salusky IB. FGF-23: more than a regulator
promote the retention of hematopoietic stem and progenitor cells of renal phosphate handling? J Bone Miner Res. 2010;25(10):
in the mesenchymal stem cell niche. J Exp Med. 2011;208(2): 2091–7.
261–71. 121. Kirkpantur A, Balci M, Gurbuz OA, Afsar B, Canbakan B,
104. Wu JY, Purton LE, Rodda SJ, Chen M, Weinstein LS, McMa- Akdemir R, et al. Serum fibroblast growth factor-23 (FGF-23)
hon AP, et al. Osteoblastic regulation of B lymphopoiesis is levels are independently associated with left ventricular mass
mediated by Gs{alpha}-dependent signaling pathways. Proc and myocardial performance index in maintenance haemodi-
Natl Acad Sci USA. 2008;105(44):16976–81. alysis patients. Nephrol Dial Transplant. 2011;26(4):
105. Di Bernardo G, Galderisi U, Fiorito C, Squillaro T, Cito L, 1346–54.
Cipollaro M, et al. Dual role of parathyroid hormone in endo- 122. Hu MC, Shi M, Zhang J, Quinones H, Kuro-o M, Moe OW.
thelial progenitor cells and marrow stromal mesenchymal stem Klotho deficiency is an early biomarker of renal ischemia-
cells. J Cell Physiol. 2010;222(2):474–80. reperfusion injury and its replacement is protective. Kidney Int.
106. Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, 2010;78(12):1240–51.
Knight MC, et al. Osteoblastic cells regulate the haematopoietic 123. Hu MC, Shi M, Zhang J, Quinones H, Griffith C, Kuro-o M,
stem cell niche. Nature. 2003;425(6960):841–6. et al. Klotho deficiency causes vascular calcification in chronic
107. Malluche HH, Mawad HW, Monier-Faugere MC. Renal osteo- kidney disease. J Am Soc Nephrol. 2011;22(1):124–36.
dystrophy in the first decade of the new millennium: analysis of 124. Block GA, Persky MS, Ketteler M, Kestenbaum B, Thadhani R,
630 bone biopsies in black and white patients. J Bone Miner Kooienga L, et al. A randomized double-blind pilot study of
Res. 2011;26(6):1368–76. serum phosphorus normalization in chronic kidney disease: a

123
Clinic Rev Bone Miner Metab

new paradigm for clinical outcomes studies in nephrology. 126. López I, Rodrı́guez-Ortiz ME, Almadén Y, Guerrero F, de Oca
Hemodial Int. 2009;13(3):360–2. AM, Pineda C, Shalhoub V, Rodrı́guez M, Aguilera-Tejero E
125. Negishi-Koga T, Shinohara M, Komatsu N, Bito H, Kodama T, (2011) Direct and indirect effects of parathyroid hormone on
Friedel RH, Takayanagi H (2011) Suppression of bone forma- circulating levels of fibroblast growth factor 23 in vivo. Kidney
tion by osteoclastic expression of semaphorin 4D. Nat Med. Int 80(5):475–82. doi:10.1038/ki.2011.107
doi:10.1038/nm.2489

123

View publication stats

Você também pode gostar