Anda di halaman 1dari 12

Renal and Extrarenal Actions of Klotho

Ming Chang Hu, MD, PhD,*,†,‡ Makoto Kuro-o, MD, PhD,*,§ and Orson W. Moe, MD*,†,储

Summary: Klotho is a single-pass transmembrane protein highly expressed in the kidney. Membrane Klotho
protein acts as a co-receptor for fibroblast growth factor-23. Its extracellular domain is shed from the cell surface
and functions as an endocrine substance that exerts multiple renal and extrarenal functions. An exhaustive review
is beyond the scope and length of this article; thus, only effects with pertinence to mineral metabolism and
renoprotection are highlighted here. Klotho participates in mineral homeostasis via interplay with other calcio-
phosphoregulatory hormones (parathyroid hormone, fibroblast growth factor-23, and 1,25-[OH]2 vitamin D3) in
kidney, bone, intestine, and parathyroid gland. Klotho also may be involved in acute and chronic kidney disease
development and progression. Acute kidney injury is a temporary and reversible state of Klotho deficiency and
chronic kidney disease is a sustained state of systemic Klotho deficiency. Klotho deficiency renders the kidney
more susceptible to acute insults, delays kidney regeneration, and promotes renal fibrosis. In addition to direct
renal effects, Klotho deficiency also triggers and aggravates deranged mineral metabolism, secondary hyperpara-
thyroidism, vascular calcification, and cardiac hypertrophy and fibrosis. Although studies examining the therapeu-
tic effect of Klotho replacement were performed in animal models, it is quite conceivable that supplementation of
exogenous Klotho and/or up-regulation of endogenous Klotho production may be a viable therapeutic strategy for
patients with acute or chronic kidney diseases.
Semin Nephrol 33:118-129 © 2013 Elsevier Inc. All rights reserved.
Keywords: Acute kidney injury, cardiac hypertrophy, chronic kidney disease, hyperparathyroidism, Klotho

T he Klotho gene was identified in 1997 when its


disruption in mice caused a phenotype of prema-
ture multiorgan failure including shortened life
span, growth retardation, accelerated thymic involution,
pulmonary emphysema, cognition impairment, skin atro-
The Klotho gene encodes a single-pass transmembrane
protein and is expressed in multiple tissues, but in particu-
larly high levels in the kidney.1,3 In the mammalian kidney
including mouse, rat, and human, Klotho is expressed prom-
inently in distal convoluted tubules,1,3,4 but also unequivo-
phy, osteopenia, ectopic soft-tissue calcification, hyper- cally is found in the proximal convoluted tubule4 and also in
phosphatemia, and high plasma fibroblast growth factor the inner-medullary collecting duct– derived cell lines.5,6 In
(FGF)-23 levels.1 Most of the features observed in addition to membrane-anchored Klotho, a secreted form of
Klotho hypomorph or knock-out mice could be rescued Klotho protein is generated from the Klotho gene through
by expressing Klotho,1,2 indicating that Klotho is anti- alternative splicing. Secreted Klotho is released di-
aging gene. rectly into the extracellular compartment and is pres-
ent in body fluid. Another important form of soluble
Klotho in body fluid is ectodomain shedding from
*Charles and Jane Pak Center for Mineral Metabolism and Clinical membrane Klotho on cell surface by proteases acro-
Research, University of Texas Southwestern Medical Center, Dallas,
TX. nym for a desintegrin and metalloproteinase 10/17
†Department of Internal Medicine, University of Texas Southwestern (Fig. 1).7 Soluble Klotho protein is present in cerebro-
Medical Center, Dallas, TX. spinal fluid,8 blood,8,9 and urine of mammals.4,9
‡Department of Pediatrics, University of Texas Southwestern Medical Membrane-anchored and soluble Klotho proteins
Center, Dallas, TX. seem to have distinct functions (Table 1). Membrane
§Department of Pathology, University of Texas Southwestern Medical Klotho forms a tetrameric complex with FGF receptors
Center, Dallas, TX.
(FGFRs) and functions as a co-receptor for FGF23,10-12 a
储Department of Physiology, University of Texas Southwestern Medical
Center, Dallas, TX. bone-derived phosphatonin that induces negative phos-
Financial support: supported in part by the National Institutes of Health phate balance through promotion of renal phosphate ex-
(R01-DK091392, R01-DK092461), the George M. O’Brien Kidney Re- cretion (Fig. 1). Soluble Klotho is a pleiotropic protein
search Center at UT Southwestern Medical Center (P30-DK-07938), functioning as an endocrine factor with a multitude of
the American Heart Association (0865235F), the Simmons Family
Foundation, and the Charles and Jane Pak Foundation.
renal and extrarenal effects (Table 1). Recently, several
Conflict of interest statement: none. studies showed that nuclear Klotho13 and cytoplasm
Address reprint requests to Ming Chang Hu, MD, PhD, Charles and Klotho14 are also bioactive molecules to protect cells
Jane Pak Center of Mineral Metabolism and Clinical Research, from senescence and apoptosis (Table 1).
University of Texas Southwestern Medical Center at Dallas, 5323
Harry Hines Blvd, Dallas, TX 75390-8885. E-mail:
ming.chang.hu@utsouthwestern.edu PHYSIOLOGICAL ROLE OF KLOTHO IN THE KIDNEY
0270-9295/ - see front matter
© 2013 Elsevier Inc. All rights reserved. The kidney is not a mere excretory organ but also a hor-
http://dx.doi.org/10.1016/j.semnephrol.2012.12.013 monal source producing several active molecules such as

118 Seminars in Nephrology, Vol 33, No 2, March 2013, pp 118-129


Renal and extrarenal klotho 119

Figure 1. Physiologic roles of Klotho on solute channels and transporters and vitamin D metabolism in the kidney. Klotho is expressed prominently
in distal convoluted tubules, and less in proximal convoluted tubules.4 In proximal convoluted tubules, membrane Klotho at the basolateral side4
functions as a co-receptor of FGFRs and drives FGF23 signal transduction to inhibit NaPi cotransporters (NaPi: 2a/c and Pit2) and to suppress
cyp27␤1 encoding for 1-hydroxylase, and to stimulate cyp24␣1 encoding for 24-hydroxylase. The role of Klotho in the cytoplasm of renal tubules is
unclear. Whether membrane Klotho at the luminal side4 inhibits NaPi cotransporters (2a/c and Pit2) in an autocrine mode is not known (dashed line).
In distal convoluted tubules, membrane Klotho at the basolateral side4 functions as a co-receptor of FGFRs to induce FGF23 signal transduction.
What intermediate(s) are released from distal convoluted tubules and how intermediate(s) affect proximal convoluted tubules in a paracrine mode is
not known. One possible candidate is Klotho release from the distal convoluted tubules to act on the PCT. Whether membrane Klotho at the luminal
side directly regulates TRPV5 in an autocrine mode is unknown. Soluble Klotho in luminal urine derived from either blood or urine exerts regulatory
action on NaPi cotransporters in proximal convoluted tubules, and on TRPV5 in DCT. Dashed line, suspected action.

1,25-(OH)2-vitamin D3 (1,25 VD3), renin, erythropoietin, Interaction With Renal 1,25-(OH)2-Vitamin D3 Production
and Klotho. Klotho exerts multiple actions on the kidney but Klotho and the vitamin D system reciprocally regulate
only selected functions are highlighted in this article. This each other. In homozygous Klotho-deficient (Kl⫺/⫺)
includes regulation of 1,25 VD3 production and modulation mice, extremely high plasma 1,25 VD3 levels were noted
of urinary phosphate (Pi), Ca, and K excretion. as well as up-regulation of 1␣-hydroxylase and down-

Table 1. Biological Functions of Klotho Protein


Form of Klotho Nuclear Intracellular Membrane Extracellular

Locale Nucleus Cytoplasm Cell surface Blood, urine, cerebrospinal fluid


Domain N/A Kl1 or full length Full length Ectodomain containing Kl1 and Kl2
Biologic function Anti-age 2 cytokine production (1) FGF23-dependent: (FGF23 (1) FGF23-dependent: same as
Anti-senescence co-receptor for FGF23 transmembrane but in
Anti-age signal) endocrine or paracrine mode
Modulation of Anti-age (2) FGF23-independent:
Na/K-ATPase Anti-IGF Anti-oxidation
PTH release Calciophospho-regulatory Modulation of renal ion channels
Ca homeostasis hormone Anti-Wnt signal
Suppress PTH, 1,25 VD3 Anti-apoptosis
(2) FGF23-independent: Antisenescence
release soluble Klotho Anti-RAA
CSF, cerebrospinal fluid; IGF, insulin-like growth factor; N/A, no information; RAA, rennin-angiotensin-aldosterone.
120 M.C. Hu, M. Kuro-o, and O.W. Moe

regulation of 24-hydrolase,15 which provides in vivo ge- pear when hyperphosphatemia is induced in mice with
netic, but indirect, evidence that the high circulating 1,25 double deletion of NaPi-2a and Klotho by feeding with a
VD3 levels are the result of overproduction and low high-phosphate diet.18
degradation of 1,25 VD3 (Fig. 1).15 In addition, Yoshida 1,25 VD3 administration up-regulates renal Klotho
et al15 found that normal genetic responses to vitamin D expression in normal animals.16 An in vitro study per-
supplementation, including down-regulation of 1␣-hy- formed by Forster et al5 showed that there are vitamin
droxylase transcripts and up-regulation of 24-hydroxy- D–responsive elements in the vicinity of the Klotho gene
lase and vitamin D receptor (VDR) transcripts, were promoter, and that 1,25 VD3 induces Klotho transcripts
impaired in Kl⫺/⫺ mice, suggesting that normal expres- in cultured mouse and human kidney cell lines. Thus 1,25
sion of renal Klotho is required for normal vitamin D VD3 and Klotho forms a negative feedback control loop
homeostasis. On the other hand, the dysregulation of the similar to that of parathyroid hormone (PTH) and vitamin
vitamin D system may be associated with high mortality D. A primary increase in 1,25 VD3 up-regulates Klotho
in both Klotho-deficient and FGF23-deficient mice be- expression, which in turn suppresses 1,25 VD3 produc-
cause diminution of vitamin D activity by a vitamin tion and likewise an increase in Klotho will suppress 1,25
D– deficient diet16 or genetically deleting the 1␣-hydrox- VD3 to remove a major stimulator of Klotho production
ylase gene17 successfully normalizes plasma 1,25 VD3 (Fig. 2, left panel). Novel insights into Klotho–vitamin D
levels and rescues a significant fraction of the phenotypes interaction will be valuable in understanding 1,25 VD3
including renal Pi retention, vascular calcification, and therapy in chronic kidney disease (CKD) patients.
short life span in Kl⫺/⫺ mice. However, the mechanism
whereby inactivation of vitamin D activity rescues most
Modulation of Renal Ion Channel and Transporters
phenotypes in Kl⫺/⫺ mice is unknown. Recently, Ohnishi
and Razzaque18 proposed the role of phosphate toxicity Klotho protein was considered one of the novel phospha-
in mammalian aging because reducing blood Pi levels by tonins soon after its discovery because the Klotho-deficient
genetic deletion of Na-Pi cotransporter-2a (NaPi-2a) res- mouse has severe hyperphosphatemia and the transgenic
cues most phenotypes in Kl⫺/⫺ mice. These beneficial mouse overexpressing Klotho has hypophosphatemia. Sub-
effects disappear and premature aging-like features reap- sequent animal and cell culture studies revealed that Klotho

Figure 2. Proposed physiological role of Klotho in mineral metabolism and pathophysiological consequences of Klotho deficiency in CKD. Left
panel: in the setting of normal kidney function with normal Klotho levels, Klotho may suppress FGF23 production and release from the bone.
But there are no data to date to prove a direct effect of Klotho on FGF23 production in the bone. Klotho functions as a co-receptor of FGFR
to allow FGF23 to suppress PTH production and release from the parathyroid. PTH stimulates and increases plasma levels of FGF23 and 1,25
VD3. Increased 1,25 VD3 further stimulates FGF23, and directly and indirectly suppresses PTH levels. Increased 1,25 VD3 also stimulates
Klotho production in the kidney. Taken together, through several negative or positive feedback loops, Klotho functions as both a phosphate and
calcium regulatory hormone to directly or indirectly suppress PTH, 1,25 VD3, and FGF23 production and release. The final outcome of Klotho’s
action on the kidney is to prevent renal Pi retention and to prevent renal Ca loss. Right panel: in CDK and end-stage renal disease, the network
is deranged (red arrows). Renal Klotho is decreased followed by a decrease in plasma Klotho. The down-regulation of Klotho increases FGF23
production via unknown mechanisms, which in turn suppresses 1,25 VD3 production in the kidney. Whether low plasma Klotho renders the
parathyroid gland resistant to the suppressive effect of FGF23 on PTH production is not proven. However, decreased FGFR1/3 and Klotho
expression in the uremic parathyroid gland could make the gland resistant to FGF23, and triggers and/or promotes secondary hyperparathy-
roidism. Low plasma Ca levels also participate in SHPT development. Hyperphosphatemia amplifies the high FGF23 and PTH, and low Klotho
in the blood. The high plasma PTH, Pi, and FGF23, and low plasma 1,25 VD3 and Klotho, in concert, contribute to the development of
complications such as metabolic bone disease, secondary hyperparathyroidism, cardiomyopathy, and vascular calcification. Dashed line:
unproven putative roles of Klotho.
Renal and extrarenal klotho 121

not only controls phosphate homeostasis through modulat- ceptible to proteases residing in brush-border membrane.
ing Na-dependent phosphate cotransporters (NaPi-2a and Although protease inhibitors abolish the proteolysis, they
2c),4,19,20 but also regulates calcium homeostasis by modu- do not reverse the Klotho-induced inhibition of trans-
lating the renal calcium channel, transient receptor potential port,4 supporting the theory that Klotho-induced degly-
ion channel (TRPV5),21,22 and potassium homeostasis by cosylation is sufficient to suppress NaPi cotransport ac-
regulation of the renal outer medullary K channel 1 tivity and that subsequent proteolysis is not required to
(ROMK1).23 One unique aspect is that as a regulator of ion suppress NaPi transport during the acute phase (Fig. 1).
transport, Klotho functions as an enzyme. The two Kl1 and
Kl2 repeats in the extracellular domain of membrane Klotho Regulation of TRPV5
share 20% to 40% amino acid sequence homology to family In addition to being a potent phosphaturic hormone,
1 glycosidase.1,24 But two important and highly preserved Klotho also maintains calcium homeostasis.15 Through
glutamate residues critical for the glycosidase activity are its suppression of PTH28 and 1,25 VD3,15 Klotho indi-
replaced by an asparagine within the Kl1 domain and an rectly decreases intestinal calcium absorption. Suppres-
alanine or a serine within the Kl2 domain, rendering the sion of PTH also is expected to promote calciuria, but the
glycosidase activity null.1,24 However, in vitro studies potent action of Klotho on distal calcium reabsorption
showed that Klotho possesses ␤-glucuronidase4,22,24 or siali- partially counteracts the reduction in gut absorption. The
dase activity21,23 through which Klotho exerts regulation of net effect at the whole organism level is maintenance of
several renal ion channels and transporters. Therefore, balance but with lower turnover. This is supported by the
Klotho is emerging as a principal calciophosphoregulatory fact that Klotho gene deletion or overexpression has
hormone. rather mild effects on plasma calcium as opposed to the
severe hyperphosphatemia and hypophosphatemia in
Inhibition of NaPi-2a activity Klotho-deficient and Klotho-excess animals, respec-
Three NaPi cotransporters, NaPi-2a, NaPi-2c, and Pit-2, tively.1,4 We focus on the most novel aspect, which is the
with different expression in the kidney proximal tubules, direct effect of Klotho on renal calcium reabsorption
participate in renal regulation of phosphate homeostasis (Fig. 1).
with different isoforms possessing different substrate Chang et al22 showed that soluble Klotho stimulates
specificities, pH sensitivities, and kinetics of response in TRPV5, one of the key regulators for urinary calcium
regulation.25 excretion, by stabilizing TRPV5 on the cell surface and
Two lines of independent studies showed that hyper- proposed that Klotho functions as a glucuronidase.
phosphatemic Kl⫺/⫺ mice display an increase in activity Glucuronic acid is a very uncommon moiety of N-glycan
of NaPi cotransport in the kidney,4 and in NaPi-2a and chains on a membrane channel such as TRPV5. A second
NaPi-2c protein expression compared with wild-type study by Cha et al21 supported an alternative mode of
(WT) mice,4,19 suggesting that hyperphosphatemia at least Klotho effect on TRPV5. Klotho functions as a sialidase,
in part is of renal origin. Most importantly, phosphate which removes ␣2,6-sialic acids from the N-glycan
restriction26,27 or induction of renal phosphate leak by chains on TRPV5 and exposes underlying N-acetyl-D-
deletion of NaPi-2a,18 successfully rescues the multior- lactosamine for binding to galectin-1.21 This association
gan failure in the Klotho-deficient mice, indicating that prevents TRPV5 from internalization via a dynamin-
phosphotoxicity is a principal pathogenic factor in these dependent process, leading to stabilization of more
animals. Transgenic Klotho-overexpressing mice have TRPV5 on the cell surface. These results clearly support
lower plasma Pi, whereas renal fractional excretion of Pi the concept that Klotho is a calciotropic protein that
is increased,4 indicating a renal leak of Pi. Soluble Klotho prevents renal calcium loss.
increases renal fractional excretion of Pi and decreases
plasma Pi in the normal rat and in FGF23 knock-out Regulation of ROMK1
mice,4 indicating that the Klotho-induced phosphaturia is ROMK1 is one of the major mediators of urinary K
FGF23-dependent as well as FGF23-independent. reabsorption. Cha et al23 found that acute infusion of
The direct suppression of NaPi-2a by soluble Klotho Klotho leads to antikaliuresis mediated by increased api-
protein in a FGF23-independent fashion is mediated by cal membrane abundance of ROMK1. They proposed a
direct inhibition of NaPi cotransport activity without mechanism of Klotho action similar to that of TRPV5.23
change in protein abundance on the cell surface, which is Klotho removes terminal sialic acids from N-glycan of
a novel mode of regulation for this class of transporters.4 ROMK1, and exposes underlying disaccharide galactose-
This can be mimicked by recombinant ␤-glucuronidase N-acetylglucosamine, a ligand for a ubiquitous galec-
and blocked by glucuronidase inhibitor, but not affected tin-1. Binding to galectin-1 at the cell surface prevents
by sialidase.4 The model of a direct effect of Klotho on clathrin-mediated endocytosis and causes accumulation
NaPi-2a protein is proposed as follows: glucuronate on of functional ROMK1 on the plasma membrane.23 Pres-
an as yet unknown substrate is removed by Klotho, ently, the role of Klotho as a potassium homeostatic
leading to suppression of NaPi cotransport activity, hormone is unclear because there does not appear to be
which subsequently renders NaPi-2a protein more sus- disturbances in plasma potassium concentrations in either
122 M.C. Hu, M. Kuro-o, and O.W. Moe

the Klotho-deficient or Klotho-overexpressing mice. If tively facilitate the action of FGF23 on suppression of
Klotho has significant antikaliuretic effects, one should PTH production. The expression of Pit-1, one of the
not anticipate undesirable effects in Klotho-deficient NaPi-3 isoforms, was found in the parathyroid gland
states in acute kidney injury (AKI) and CKD but theo- and was up-regulated by a 1,25 VD3 and low Pi diet,
retically should be more concerned with therapeutic ad- and down-regulated by vitamin D– deficient diet.31 Hu
ministration of Klotho leading to hyperkalemia. These et al9 showed that Klotho suppresses the activity and
theoretical effects remain to be explored. expression of Pit-1 in the rat vascular smooth muscle
cell line in vitro, but to date, there is no evidence for
PHYSIOLOGICAL ROLE OF a Klotho effect on Pit-1 in parathyroid.
KLOTHO IN EXTRARENAL ORGANS Ionized calcium activity is a key modulator of PTH
synthesis and release from parathyroid. An intriguing
Membrane Klotho and soluble Klotho protein exert dis- model was proposed as an alternative mode of Klotho
tinct but possibly overlapping actions. FGFRs are ex-
action on the parathyroid cells.32 In this model, intracel-
pressed ubiquitously, but FGF23 signal transduction is
lular Klotho binds to Na/K-adenosine triphosphatase
controlled by co-expression with membrane Klotho.12
(ATPase) to form a complex in response to low intracel-
The restricted expression of Klotho protein in very few
organs (kidney, heart, brain, and parathyroid gland) in lular [Ca2⫹] and bring Na/K-ATPase to the cell surface.32
concert with multiple functions in multiple tissues (Table The resultant change in electrochemical gradient was
1) suggests that soluble Klotho may function indepen- proposed to trigger the release of PTH through an un-
dently of FGFRs as an endocrine hormone and an en- identified signal pathway.32 Thus, Klotho expression in
zyme to directly modulate target proteins. the parathyroid may suppress PTH production by the
FGF23 signal pathway and increase PTH production by
the Na/K-ATPase signal pathway stimulated by low
Modulation of PTH Production
blood Ca levels. However, how the complex of Na/K-
Klotho modulates PTH directly and indirectly. Klotho ATPase with Klotho is formed in response to low Ca2⫹
indirectly regulates PTH production through modulation levels, how Na/K-ATPase activity is stimulated, and
of plasma levels of 1,25 VD3, Pi, and FGF23. In addition, what intracellular signal is required to couple Na/K-
Klotho may exert a direct effect on PTH production and ATPase to PTH release remain to be illustrated.
release (Fig. 2, left panel).
In genetically manipulated mice whose Klotho gene is
replaced by a LacZ reporter, ␤-X-gal staining is clearly Inhibition of Intestinal Phosphate Absorption
positive in the kidney, the sinoatrial node region of the In addition to increased renal reabsorption of Pi, in-
heart, choroid plexus of brain, and the parathyroid creased absorption of dietary Pi may exacerbate hyper-
gland,29 which is compatible with earlier findings by phosphatemia in Kl⫺/⫺ mice19 because expression of the
reverse-transcription polymerase chain reaction.1 Immu- intestinal phosphate transporter NaPi-2b is significantly
nohistochemistry further confirmed the localization of higher in Kl⫺/⫺ mice than in WT mice.19 In vitro studies
Klotho in the parathyroid and but not the surrounding revealed that Klotho directly decreases Pi-induced cur-
thyroid tissue,28 thus excluding the possibility that the rent in NaPi-2b– expressing oocytes,20 which supports the
positive reverse-transcription polymerase chain reaction model that Klotho inhibits Pi absorption in the intestine.
results are caused by contamination. Ben-Dov et al28 In addition, a recent study showed that another
found FGFR1 and FGFR3 in the parathyroid tissue, sug- Na-coupled phosphate transporter Pit-1 protein also is
gesting that FGFRs may form a complex with Klotho and
present in the apical membrane of enterocytes of rat
FGF23 in the parathyroid gland. FGF23 binds to Klotho/
duodenum and jejunum, but not in the ileum.33 Unlike
FGFR and activates the mitogen-activated protein kinase
NaPi-2b, PiT-1 protein in the brush-border membrane
cascade signal pathway in cultured cells transfected with
and Pit-1 messenger RNA (mRNA) expression are not
Klotho,12 parathyroid cells,28 as well as in the kidney,4,10
indicating that the parathyroid gland is a likely target changed in the duodenum or jejunum as a function of
organ of FGF23, and Klotho is a prerequisite for FGF23 dietary Pi.33 Therefore, the relative contribution of
to modulate PTH production. PiT-1 to intestinal Pi absorption remains to be deter-
In vivo animal and in vitro cell culture experiments mined, and Klotho’s effect on intestinal Pit-1 remains
performed in independent laboratories revealed that to be addressed.
FGF23 decreases PTH production, increases expression Taken together, Klotho is a calciophosphoregulatory
of both the parathyroid calcium-sensing receptor and the protein. The direct effect of Klotho on the kidney is to
vitamin D receptor (both of which contributes to sup- promote phosphaturia and to prevent renal calcium loss.
pression of PTH), and decreases cell proliferation28,30 Klotho regulates Pi and Ca homeostasis directly and by
when normal Klotho and FGFR are expressed in para- interplay with other calcium and phosphate regulatory
thyroid glands. Interestingly, FGF23 seems to increase hormones: FGF23, PTH, and 1,25 VD3 (Figs. 1 and 2,
Klotho in the parathyroid gland,28 which may posi- left panel).
Renal and extrarenal klotho 123

PATHOPHYSIOLOGICAL ROLE OF Transforming growth factor (TGF)-␤ is considered to


KLOTHO DEFICIENCY IN KIDNEY DISEASE contribute to renal fibrosis.45 Kl⫺/⫺ mice have more glo-
merular and tubulointerstitial fibrin deposition and higher
Klotho Deficiency Renders the active plasminogen activator inhibitor-1 antigen in
Kidney More Susceptible to Injury plasma and plasminogen activator inhibitor-1 mRNA ex-
Liu et al34 showed increased senescence in progenitor pression in the kidney,46 and Kl-/⫹ mice have more fibro-
cells in many tissues of Kl⫺/⫺ mice. Knock-down of sis than WT mice do at baseline and after unilateral
endogenous Klotho promotes augmentation of senes- ureteral ligation to induce unilateral ureteral obstruction
cence in cultured cells.34 Administration of exogenous (UUO).47 Kl-/⫹ mice with UUO also have higher TGF-␤
Klotho significantly decreases senescence in endothelial levels, and lower Klotho mRNA and protein than WT
cells35 and fibroblasts.36 Furthermore, Klotho depletion- mice.47 The obstructed kidneys from Kl-/⫹ mice express
induced cell senescence may be associated with up-reg- significantly higher levels of fibrosis markers such as
ulation of Wnt signaling activity because administration ␣–smooth muscle actin, fibronectin, and TGF-␤ than
of exogenous Wnt accelerates cell senescence in vivo and those from WT mice.47 In cultured rat kidney cell lines,
in vitro, and Wnt signaling is increased significantly in Klotho alleviates TGF-␤–induced epithelial-mesenchy-
Kl⫺/⫺ mice, and suppressed by genetic Klotho overex- mal transition or phenotype, suppresses TGF-␤–induced
pression.34 Soluble Klotho can bind to various Wnt fam- target genes activation, and reduces TGF-␤1–induced
ily members and inhibit their biological activity.34 Re- Smad2 phosphorylation,48 suggesting that Klotho protein
cently, intracellular Klotho was shown to be able to inhibits renal fibrosis primarily through inhibiting
suppress cell senescence by inhibiting retinoic-acid–in- TGF-␤1 signaling. More importantly and interestingly,
ducible gene-I–induced expression of interleukin-6 and Doi et al48 recently showed that intraperitoneal injection
interleukin-8 both in vitro and in vivo.14 Klotho defi- of soluble Klotho protein suppresses renal fibrosis in-
ciency from kidney diseases enhances cell senescence duced by UUO, suggesting that soluble Klotho protein
induced by oxidative stress.37-39 Excessive senescence or may be a novel therapeutic agent for renal fibrosis. In
resultant apoptosis and stem cell deletion may decrease addition to the effect of Klotho on TGF-␤1, suppressive
the kidney’s ability to defend against renal insults and effects of Klotho on the insulin-like growth factor path-
impair regeneration. way2 and on the Wnt signal pathway34 also may be
Klotho is an anti-apoptotic protein. Klotho deficiency associated with Klotho’s inhibitory action on renal fibro-
significantly increases apoptosis in cultured kidney and sis, but to date there is no direct evidence to confirm this
endothelial cells,35,40 and in the kidney.39 Oxidative stress concept.
down-regulates Klotho expression and in turn increases We propose that Klotho deficiency renders the kidney
cell damage when exposed to H2O2.6 Increase of Klotho more susceptible to injury, accelerates renal fibrogenesis,
by genetic manipulation or viral delivery decreases the retards renal tissue regeneration, and eventually promotes
number of apoptotic cells and improves kidney function chronic progression. Supplementation of Klotho may
and renal morphology after acute40 and chronic kidney provide beneficial impact in preventing and slowing
damage.39 These results form the basis for potential clin- down CKD progression.
ical application of Klotho in the treatment of AKI and
CKD. PATHOPHYSIOLOGICAL ROLE OF
Klotho Deficiency Delays Kidney Recovery KLOTHO IN THE METABOLIC SYNDROME
Two independent studies showed that Kl⫺/⫺ mice have The metabolic syndrome (MS) is characterized by obe-
stem cell depletion in several organs34 and progenitor cell sity, serum lipid profile alterations, hypertension, and
senescence in the kidney,39 which may delay tissue re- fasting hyperglycemia, and is a risk factor for the devel-
covery after kidney injury. In addition, Kl⫺/⫺ mice have opment of diabetes and cardiovascular disease.49,50 Re-
severe abnormal endothelial function41 and integrity,42 cent studies have indicated that the MS also is associated
and impairment of angiogenesis and vasculogenesis after independently with an increased risk for incident CKD in
ischemic limbs.43 If those results were translated into nondiabetic adults51 because MS patients are at signifi-
ischemic kidney damage, kidney regeneration may be cantly higher risk for microalbuminuria and/or CKD, and
delayed after kidney injury. Increased Klotho by genetic the level of risk is related to the number of components
manipulation was shown to accelerate angiogenesis and of the syndrome. Any component of MS may indepen-
vasculogenesis, decrease limb loss, and promote limb dently favor the development of renal abnormalities and
recovery after ischemic injury in Kl⫺/⫺ mice,44 which potentially is considered a modifiable risk factor for
suggest that Klotho replacement may promote kidney CKD. Thus, correction of any component should be a
recovery. rationale for intervention of MS and effectively can pre-
vent the development and progression of renal damage.
Klotho Deficiency Promotes Renal Fibrosis One cross-sectional study examining genetic variants
Renal fibrosis is not only a histologic characteristic in of the Klotho gene polymorphism with MS showed as-
CKD, but also is pathogenic for chronic progression. sociation of the KL-VS variant (F352V and/or C370S
124 M.C. Hu, M. Kuro-o, and O.W. Moe

within exon 2, simply termed VS alleles) to high blood PATHOPHYSIOLOGICAL ROLE OF KLOTHO DEFICIENCY
glucose level, high blood pressure, insulin resistance, and IN COMPLICATIONS OF CHRONIC KIDNEY DISEASE
a trend toward its association with hypertriglyceridemia
in Asian Indians.52 Several animal models such as dia- Secondary Hyperparathyroidism
betes induced by streptozotocin,53 diabetes from leptin Secondary hyperparathyroidism (SHPT) is a common
deficiency,54 spontaneous non–insulin-dependent diabe- and severe complication in CKD,62 contributing to renal
tes (Otsuka Long-Evans Tokushima Fatty),55,56 and hy- metabolic bone disease, cardiac disease, and anemia. In
pertension (spontaneous hypertension and volume-de- addition, hyperphosphatemia, hypovitaminosis D, and
pendent hypertension)55,57 have disclosed a dramatic low expression of VDR are proposed to contribute to
reduction of renal Klotho mRNA and/or protein expres- maintaining increased PTH levels in CKD patients. But if
sion, suggesting that a decrease in Klotho may be part of one analyzes the profile of changes of those parameters,
the MS. At present, we do not know whether Klotho is a it is more likely that these abnormalities are involved in
cause or a result of MS, or a parallel phenomenon. the acceleration of SHPT in later stages rather than
The administration of thiazolidinedione, an agonist triggering them early.62 Epidemiologic observation63 and
of peroxisome proliferator-activated receptor-␥ in- animal studies64 show that a plasma FGF23 increase is a
creases renal Klotho mRNA expression, attenuates ab- very early event. Plasma Klotho decreases may even be
normal lipid and glucose metabolism, and reduces an earlier event,65 which in turn can stimulate FGF23
systolic blood pressure in Otsuka Long-Evans overproduction. Thus, the conceptual mode of develop-
Tokushima Fatty rats.56 More interestingly, adenovi- ment of SHPT is revised to include aberrant FGF23/
rus-mediated Klotho gene delivery can repeat thiazo- Klotho activity to play a more fundamental role in SHPT
lidinedione’s action,58 indicating the therapeutic po- development (Fig. 2, right panel).
tential of Klotho gene delivery in MS. CKD in both human beings and animals is a state of
In rats with streptozotocin-induced diabetes,59 high plasma levels of FGF2366 and low FGFR1 and
Klotho protein in the kidney is notably decreased Klotho expression in the parathyroid gland,67-69 which
along with kidney destruction.59 Both insulin and phlo- renders FGF23 to lose its inhibitory actions on PTH
ridzin corrects hyperglycemia, reverses the reduced production, and to fail to increase the calcium-sensing
renal Klotho expression, and improves kidney function receptor and VDR.30 Indeed, Galitzer et al70 confirmed
and histology of diabetic rats. Klotho protein in Ma- that FGF23 fails to decrease plasma PTH, suppress cell
din–Darby canine kidney cells is reduced by incuba- proliferation, and activate the mitogen-activated protein
kinase pathway in parathyroid glands of rats with ad-
tion in high glucose medium.7 Insulin has been shown
vanced CKD, indicating that the gland is resistant to
to stimulate shedding of the extracellular domain of
FGF23, possibly from low FGFR1 and Klotho (Fig. 2,
membrane Klotho protein,7 which may increase the
right panel). In addition, the aberrant Klotho-Na/K-
blood Klotho concentrations.
ATPase axis, an FGF23-independent mode, also may be
Similarly, in rats with spontaneous hypertension,
involved in SHPT development in CKD. Hofman-Bang
Klotho gene delivery via adeno-associated virus carrying
et al71 reported an unexpected finding of an increase in
mouse Klotho full-length complementary DNA reverses Klotho, FGFR, and Na/K ATPase in the parathyroid
reduced Klotho expression in the kidney, controls blood glands of early CKD rats, which is seemingly opposite of
pressure, improves kidney function, and attenuates renal findings by other laboratories,30,68-70 but may be owing to
fibrosis.57 different stages of the CKD model and different levels of
Taken together, the MS appears to be a state of Klotho plasma calcium concentrations. These investigators fur-
deficiency before development of kidney injury. Klotho ther reasoned that when plasma calcium levels are in the
deficiency may render the kidneys more susceptible to normal range, the increased expression of Klotho in
acute and chronic renal insults, and kidney damage fur- parathyroid glands will be reduced,71 because Imura
ther exacerbates Klotho deficiency. Improvement of glu- et al32 proposed that low plasma calcium increases intra-
cose metabolism or control of blood pressure could in- cellular Klotho. Thus far, a model can be constructed in
crease Klotho expression in the kidney considerably. On which PTH production in CKD may be controlled by two
the other hand, Klotho possesses anti-insulin activity and Klotho-dependent signal pathways. In early CKD,
induces insulin resistance.2 Unger60 in 2006 proposed Klotho-Na/K-ATPase signaling is overactive owing to
that insulin resistance induced by Klotho may decrease higher expression of Klotho and FGFR in the parathyroid
insulin-stimulated intracellular glucose availability and glands and possibly regulated by plasma calcium con-
may prevent intracellular caloric and lipid overload and centration. Hypocalcemia promotes formation of the
toxicity. A frequently used medication, peroxisome pro- Klotho/Na/K-ATPase complex and induces PTH synthe-
liferator-activated receptor-␥ agonist for type II diabetes sis in the parathyroid. The increase of plasma FGF23
mellitus, has been confirmed to increase Klotho.61 Thus, with a high expression of Klotho and FGFR in the
modulation of Klotho expression in the kidney is a po- parathyroid glands should decrease PTH secretion to
tential future treatment option for the MS. maintain normal mineral metabolism. However, in ad-
Renal and extrarenal klotho 125

vanced CKD, a low expression of Klotho and FGFR in


the parathyroid glands blunts suppressive activity of Table 2. Comparison of Phenotypes Klotho Deficiency and
FGF23/Klotho signaling. The nonunanimous blood lev- CKD
els of PTH in CKD patients would be interpreted by the Chronic
Klotho Deficiency Kidney Disease
complicated mechanisms of PTH modulation. Thus,
Klotho restoration in the parathyroid with normalization Blood chemistry
of plasma Ca levels may be a novel approach to block Phosphate 1111 1 or 111*
Calcium 1 ↔ or 22
SHPT development.
Creatinine 1 111
1,25 VD3 111 222
Cardiovascular Calcification PTH ↔ or 2 11
FGF23 111 11
Cardiovascular disease is the leading cause of mortality Klotho 222 or disappear 22 at ESRD†
of CKD and cardiovascular calcification is prevalent in Gross phenotypes
CKD. In addition to the classic traditional factors, FGF23 Body weight 222 22
and Klotho are novel contributors to ectopic calcification Growth retardation 2222 22 in children
in soft tissues including the aorta (Fig. 2B).1,9,72,73 Thus Physical activity 222 2
far, there are no effective targeted therapies for cardio- Fertility 2222 22
Life span 2222 22
vascular disease in CKD other than manipulation of the
Cardiovascular
classic risk factors. disease
Tangri et al74 examined the association of the func- Cardiac 11 111
tional Kl-VS variant of Klotho with valvular and vascular hypertrophy
calcifications on 1389 cases and 2139 controls from the Cardiac fibrosis 11 111
Framingham Heart Study Offspring Cohort and did not Vascular 1111 11
calcification
observe any association of the Kl-VS variant of Klotho
Arthrosclerosis 11 111
valvular or vascular calcifications. However, experimen- Blood pressure 1 1111
tal animal studies documented that CKD is a state of Hematocrit levels 2 2222
systemic Klotho deficiency (Table 2). Kl⫺/⫺ mice have Bone disease 222 222
extensive ectopic calcification in soft tissues akin to that Abbreviation: ESRD, end-stage renal disease.
observed in CKD subjects, suggesting a pathogenetic *During early CKD the blood Pi is in the normal range.
association between Klotho deficiency and calcification. †Blood Klotho may be increased in early CKD.89
An increase in Klotho by genetic manipulation inhibits
vascular calcification in CKD animals.9 The suppressive
influence of Klotho on vascular calcification in CKD is
multifactorial, including the following: (1) decreasing human arterial organ cultures from CKD patients.77 Further-
plasma Pi by promoting a negative Pi balance4,72,73; (2) more, VDR activators exert their anticalcific effects by
inhibiting Pi-induced Pit-1 and Pit-2 activation in the restoration of Klotho and regaining FGF23 responsive-
vasculature9; (3) suppressing cell senescence, apoptosis, ness.77 This study provides some novel insights into vascu-
and death in vascular endothelial cells and smooth mus- lar calcification in uremia: both chronic metabolic and
cle cells induced by a variety of insults including mechanistic stress could induce vascular calcification by
Pi36,38,75; and (4) serving as an anti-inflammatory modu- dysregulation of Klotho/FGF23 signaling. Administration
lator.14,54 of VDR activators can restore Klotho expression and un-
Data from independent laboratories showed that Klotho mask the FGF23 anticalcific effect.77 This concept is similar
and FGFR1/3 protein and/or mRNA, but not FGF23 and to the mode of the effect of defect in FGF23/Klotho signal-
FGFR4, are expressed in human vasculature,76,77 and human ing in SHPT in CKD (Fig. 2, right panel).
aorta– derived smooth muscle cells.77 Interestingly, both Recently, one study examined the role of stanniocalcin
Klotho and FGFR1/3 expression are down-regulated in hu- (STC) 1 and 2, which are up-regulated in the kidney of
man arteries from CKD patients who have vascular calcifi- Kl⫺/⫺ mice,78 in ectopic calcification. STC2 protein is
cation.77 When human aorta-derived smooth muscle cells localized focally with the calcified lesions of renal arte-
are incubated with pooled uremic blood, high Pi, and Ca, rioles, renal tubular cells, heart, and aorta in Kl⫺/⫺ mice.
both FGFR1/3 and Klotho expressions are down-regulated High Pi medium increases STC2 mRNA levels as well as
and vascular smooth muscle cells undergoing “osteogenic- that of osteocalcin, osteopontin, and PiT-1 in rat aortic
chondrogenic” transdifferentiation are triggered. Vascular vascular smooth muscle. Interestingly, knockdown with a
Klotho deficiency by Klotho knockdown potentiates the small interfering RNA or the overexpression of STC2
development of calcification and decreases the ability of showed acceleration and inhibition of Pi-induced calci-
vascular tissue to respond to FGF23.77 Lim et al also fication, respectively, in this cell line.79 These results
showed that vascular Klotho deficiency driven by pro-cal- suggest that STC2 might represent a novel target for
cific stressors could be restored by VDR activators in an in ectopic calcification in the condition of Klotho defi-
vitro cell culture model and an ex vivo model by using ciency.
126 M.C. Hu, M. Kuro-o, and O.W. Moe

The prospect of Klotho restoration affecting the course Administration of Exogenous Klotho
of CKD in terms of prevention, forestalling, slowing, or Animal studies have shown that delivery of Klotho gene
reversal of vascular calcification is a high priority ques- via viral carrier efficiently protects the kidney from acute
tion in terms of therapeutics and requires investigation. injury induced by ischemia reperfusion,83 and also affects
the course of disease progression of CKD.57,58,84 Com-
Cardiac Remodeling pared with Klotho gene delivery to animal subjects, ad-
ministration of recombinant Klotho protein might be a
Cardiac remodeling, which often is used as a general
safer and easier approach to correct endocrine Klotho
term of changes in cardiac structure and function with
deficiency.
implied negative connotations, is used here to describe
In vitro studies have shown that soluble Klotho, a full
the cardiac hypertrophy and fibrosis in CKD; sometimes length of the extracellular domain, is active in inhibition
referred to as uremic cardiomyopathy, which distin- of insulin-like growth factor signal transduction,2 sup-
guishes it pathogenically from highly prevalent hyperten- pression of Wnt signal,34 modulation of several ion chan-
sive and ischemic cardiomyopathy. In addition to tradi- nels or transporters,4,21,23 or control of FGF23 signal
tional cardiovascular risk factors, novel risk factors such transduction.12 In vivo studies further showed its thera-
as vitamin D deficiency,80 high plasma FGF23,81,82 and peutic potential in several kidney disease models. Klotho
low plasma Klotho81 all can contribute to cardiac hyper- administration has been proven successful in the protec-
trophy (Fig. 2, right panel). tion of kidney function in AKI animals induced by isch-
In the heart, Klotho is expressed solely at the sinoatrial emia-reperfusion injury (IRI).85 More recently, the same
node.29 Kl⫺/⫺ mice have sinoatrial conduction defects,29 Klotho preparation also was documented to be efficient
which may be a cause of sudden death under restraint in the inhibition of renal fibrosis and in better preserva-
stress. The absence of degenerative structural change in tion of renal function in the UUO model.48 Thus, exog-
the sinoatrial node of Kl⫺/⫺ mice29 suggests that haplo- enous Klotho protein supplementation may be a poten-
sufficient levels of Klotho might be sufficient to maintain tially feasible way of replacement therapy in Klotho-
the function of sinoatrial node as a pacemaker. deficient states.
One recent study showed that Klotho deficiency is
associated with cardiac hypertrophy.81 Klotho-deficient Up-Regulation of Endogenous Klotho Protein
mice have left ventricular hypertrophy, which is not Transgenic overexpression of Klotho to plasma levels
hypertension-dependent, but may be FGF23-dependent about twice that of normal2 is effective in preventing
because either intramyocardial or intravenous injection kidney injury acutely from ischemia-reperfusion,85 im-
of FGF23 results in cardiac hypertrophy in WT mice, or mune-mediated glomerulonephritis,39 and in a renal ab-
an FGFR blocker attenuates cardiac hypertrophy in 5/6 lation model of CKD.9 This provides the basis for devel-
nephrectomized CKD, but does not change blood pres- oping ways to increase endogenous Klotho expression
sure.81 One interpretation is the importance of FGF23, via stimulation or removal of suppression of Klotho
but the effect of low Klotho is equally likely owing to the when residual kidney function is somewhat preserved
possible interplay between Klotho and FGF23 in the and endogenous Klotho-producing cells in the kidney are
pathogenesis of cardiac remodeling in CKD. It is clear not destroyed but simply suppressed. Furthermore, strat-
that uremic cardiomyopathy is a complex metabolic dis- egies to increase Klotho production in extrarenal tissues
ease that is way beyond the classic cardiac risk factors. In might be of particular importance for end-stage renal
addition to FGF23 and Klotho, aberrant vitamin D and disease patients whose functional kidney tissue is totally
phosphate metabolism and cardiac renin-angiotension- lost.
aldosterone activation also may participate in cardiac It has been shown that the peroxisome proliferator-
hypertrophy in CKD (Fig. 2, right panel). activated receptor-␥ agonist,86 and anti-oxidants6 in-
crease renal Klotho expression in AKI animals. Some
animal experiments and cell culture studies showed that
KLOTHO AS A POTENTIAL THERAPEUTIC AGENT restricting Pi intake,26 active vitamin D,16 inhibition of
Although there are no published clinical studies showing 3-hydroxy-3-methylglutaryl CoA reductase,87 and inhibi-
the therapeutic efficacy of Klotho administration in either tion of Ang II88 increase Klotho expression in the kidney
acute or chronic kidney disease, animal experiments thus and/or in cultured cell lines. Therefore, these conven-
far have shown unequivocal therapeutic effects of Klotho tional maneuvers potentially offer additional therapeutic
mechanisms for treating Klotho deficiency and their clin-
gene delivery or direct administration of Klotho protein
ical utility needs to be explored.
on several models. Restoration of endogenous Klotho or
administration of exogenous Klotho potentially provides
novel treatment strategies for CKD patients. We high-
CONCLUSIONS AND PERSPECTIVES
light the recent advances of Klotho administration in In concert, animal data and the limited clinical observa-
animal models of kidney disease. tions to date are overwhelmingly strong to suggest that
Renal and extrarenal klotho 127

Klotho is a pleiotropic protein and plays multiple phys- 11. Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H,
iological roles in modulation of kidney function and Okawa K, et al. Klotho converts canonical FGF receptor into a
specific receptor for FGF23. Nature. 2006;444:770-4.
pathophysiological roles in acute kidney damage, pro- 12. Kurosu H, Ogawa Y, Miyoshi M, Yamamoto M, Nandi A, Rosen-
gression of CKD, and extrarenal complications in CKD. blatt KP, et al. Regulation of fibroblast growth factor-23 signaling
Klotho is not merely an early biomarker for kidney by klotho. J Biol Chem. 2006;281:6120-3.
damage, but also has a pathogenic role for kidney dis- 13. German DC, Khobahy I, Pastor J, Kuro OM, Liu X. Nuclear
ease. The understanding of the renal and extrarenal ac- localization of Klotho in brain: an anti-aging protein. Neurobiol
Aging. 2012;33:1483.e25-30.
tions of Klotho will advance novel strategies for both 14. Liu F, Wu S, Ren H,Gu J. Klotho suppresses RIG-I-mediated
diagnosis and treatment of AKI and/or CKD. senescence-associated inflammation. Nat Cell Biol. 2011;13:
The potential utility of Klotho in clinical practice is 254-62.
anticipated to be at least two-fold. First, Klotho may 15. Yoshida T, Fujimori T, Nabeshima Y. Mediation of unusually
serve as an early and sensitive biomarker to kidney high concentrations of 1,25-dihydroxyvitamin D in homozygous
klotho mutant mice by increased expression of renal 1alpha-
diseases. But its specificity and its prognostic value and
hydroxylase gene. Endocrinology. 2002;143:683-9.
differential diagnostic value in human beings remain to 16. Tsujikawa H, Kurotaki Y, Fujimori T, Fukuda K, Nabeshima Y.
be examined. Second, Klotho exogenous supplementa- Klotho, a gene related to a syndrome resembling human prema-
tion and/or up-regulation of endogenous Klotho produc- ture aging, functions in a negative regulatory circuit of vitamin D
tion may provide novel therapy for AKI patients to retard endocrine system. Mol Endocrinol. 2003;17:2393-403.
17. Ohnishi M, Nakatani T, Lanske B, Razzaque MS. Reversal of
or block its progression to CKD and for CKD by arrest-
mineral ion homeostasis and soft-tissue calcification of klotho
ing or slowing progression as well as preventing and knockout mice by deletion of vitamin D 1alpha-hydroxylase.
reversing complications. Because we are moving to use Kidney Int. 2009;75:1166-72.
Klotho as a novel diagnostic, prognostic, and therapeutic 18. Ohnishi M, Razzaque MS. Dietary and genetic evidence for
strategy for CKD patients, identification of proper indi- phosphate toxicity accelerating mammalian aging. FASEB J.
2010;24:3562-71.
cations including CKD stage, composition of abnormal
19. Segawa H, Yamanaka S, Ohno Y, Onitsuka A, Shiozawa K,
mineral metabolism, and state of complications is of the Aranami F, et al. Correlation between hyperphosphatemia and
highest priority. type II Na-Pi cotransporter activity in klotho mice. Am J Physiol
Renal Physiol. 2007;292:F769-79.
20. Dermaku-Sopjani M, Sopjani M, Saxena A, Shojaiefard M, Bo-
REFERENCES gatikov E, Alesutan I, et al. Downregulation of NaPi-IIa and
1. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, NaPi-IIb Na-coupled phosphate transporters by coexpression of
Utsugi T, et al. Mutation of the mouse klotho gene leads to a Klotho. Cell Physiol Biochem. 2011;28:251-8.
syndrome resembling ageing. Nature. 1997;390:45-51. 21. Cha SK, Ortega B, Kurosu H, Rosenblatt KP, Kuro OM, Huang
2. Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani CL. Removal of sialic acid involving Klotho causes cell-surface
P, et al. Suppression of aging in mice by the hormone Klotho. retention of TRPV5 channel via binding to galectin-1. Proc Natl
Science. 2005;309:1829-33. Acad Sci U S A. 2008;105:9805-10.
3. Kato Y, Arakawa E, Kinoshita S, Shirai A, Furuya A, Yamano K, 22. Chang Q, Hoefs S, van der Kemp AW, Topala CN, Bindels RJ,
et al. Establishment of the anti-Klotho monoclonal antibodies and Hoenderop JG. The beta-glucuronidase klotho hydrolyzes and
detection of Klotho protein in kidneys. Biochem Biophys Res activates the TRPV5 channel. Science. 2005;310:490-3.
Commun. 2000;267:597-602. 23. Cha SK, Hu MC, Kurosu H, Kuro-o M, Moe O, Huang CL.
4. Hu MC, Shi M, Zhang J, Pastor J, Nakatani T, Lanske B, et al. Regulation of renal outer medullary potassium channel and renal
Klotho: a novel phosphaturic substance acting as an autocrine K(⫹) excretion by Klotho. Mol Pharmacol. 2009;76:38-46.
enzyme in the renal proximal tubule. FASEB J. 2010;24: 24. Tohyama O, Imura A, Iwano A, Freund JN, Henrissat B, Fujimori T,
3438-50. et al. Klotho is a novel beta-glucuronidase capable of hydrolyzing
5. Forster RE, Jurutka PW, Hsieh JC, Haussler CA, Lowmiller CL, steroid beta-glucuronides. J Biol Chem. 2004;279:9777-84.
Kaneko I, et al. Vitamin D receptor controls expression of the 25. Moe OW. PiT-2 coming out of the pits. Am J Physiol Renal
anti-aging klotho gene in mouse and human renal cells. Biochem Physiol. 2009;296:F689-90.
Biophys Res Commun. 2011;414:557-62. 26. Morishita K, Shirai A, Kubota M, Katakura Y, Nabeshima Y,
6. Mitobe M, Yoshida T, Sugiura H, Shirota S, Tsuchiya K, Nihei H. Takeshige K, et al. The progression of aging in klotho mutant
Oxidative stress decreases klotho expression in a mouse kidney mice can be modified by dietary phosphorus and zinc. J Nutr.
cell line. Nephron Exp Nephrol. 2005;101:e67-74. 2001;131:3182-8.
7. Chen CD, Podvin S, Gillespie E, Leeman SE, Abraham CR. 27. Takeda E, Yamamoto H, Nashiki K, Sato T, Arai H, Taketani Y.
Insulin stimulates the cleavage and release of the extracellular Inorganic phosphate homeostasis and the role of dietary phospho-
domain of Klotho by ADAM10 and ADAM17. Proc Natl Acad rus. J Cell Mol Med. 2004;8:191-200.
Sci U S A. 2007;104:19796-801. 28. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, Goetz R, Kuro-o M,
8. Imura A, Iwano A, Tohyama O, Tsuji Y, Nozaki K, Hashimoto N, Mohammadi M, et al. The parathyroid is a target organ for FGF23
et al. Secreted Klotho protein in sera and CSF: implication for in rats. J Clin Invest. 2007;117:4003-8.
post-translational cleavage in release of Klotho protein from cell 29. Takeshita K, Fujimori T, Kurotaki Y, Honjo H, Tsujikawa H,
membrane. FEBS Lett. 2004;565:143-7. Yasui K, et al. Sinoatrial node dysfunction and early unexpected
9. Hu MC, Shi M, Zhang J, Quinones H, Griffith C, Kuro-o M, et al. death of mice with a defect of klotho gene expression. Circula-
Klotho deficiency causes vascular calcification in chronic kidney tion. 2004;109:1776-82.
disease. J Am Soc Nephrol. 2011;22:124-36. 30. Canalejo R, Canalejo A, Martinez-Moreno JM, Rodriguez-Ortiz
10. Goetz R, Nakada Y, Hu MC, Kurosu H, Wang L, Nakatani T, et ME, Estepa JC, Mendoza FJ, et al. FGF23 fails to inhibit uremic
al. Isolated C-terminal tail of FGF23 alleviates hypophosphatemia parathyroid glands. J Am Soc Nephrol. 2010;21:1125-35.
by inhibiting FGF23-FGFR-Klotho complex formation. Proc Natl 31. Tatsumi S, Segawa H, Morita K, Haga H, Kouda T, Yamamoto H,
Acad Sci U S A. 2010;107:407-12. et al. Molecular cloning and hormonal regulation of PiT-1, a
128 M.C. Hu, M. Kuro-o, and O.W. Moe

sodium-dependent phosphate cotransporter from rat parathyroid 51. Kurella M, Lo JC,Chertow GM. Metabolic syndrome and the risk
glands. Endocrinology. 1998;139:1692-9. for chronic kidney disease among nondiabetic adults. J Am Soc
32. Imura A, Tsuji Y, Murata M, Maeda R, Kubota K, Iwano A, et al. Nephrol. 2005;16:2134-40.
alpha-Klotho as a regulator of calcium homeostasis. Science. 52. Majumdar V,Christopher R. Association of exonic variants of
2007;316:1615-8. Klotho with metabolic syndrome in Asian Indians. Clin Chim
33. Giral H, Caldas Y, Sutherland E, Wilson P, Breusegem S, Barry Acta. 2011;412:1116-21.
N, et al. Regulation of rat intestinal Na-dependent phosphate 53. Asai O, Nakatani K, Tanaka T, Sakan H, Imura A, Yoshimoto S,
transporters by dietary phosphate. Am J Physiol Renal Physiol. et al. Decreased renal alpha-Klotho expression in early diabetic
2009;297:F1466-75. nephropathy in humans and mice and its possible role in urinary
34. Liu H, Fergusson MM, Castilho RM, Liu J, Cao L, Chen J, et al. calcium excretion. Kidney Int. 2012;81:539-47.
Augmented Wnt signaling in a mammalian model of accelerated 54. Zhao Y, Banerjee S, Dey N, LeJeune WS, Sarkar PS, Brobey R,
aging. Science. 2007;317:803-6. et al. Klotho depletion contributes to increased inflammation in
35. Maekawa Y, Ohishi M, Ikushima M, Yamamoto K, Yasuda O, kidney of the db/db mouse model of diabetes via RelA (ser-
Oguro R, et al. Klotho protein diminishes endothelial apoptosis ine)536 phosphorylation. Diabetes. 2011;60:1907-16.
and senescence via a mitogen-activated kinase pathway. Geriatr 55. Aizawa H, Saito Y, Nakamura T, Inoue M, Imanari T, Ohyama Y,
Gerontol Int. 2011;11:510-6. et al. Downregulation of the Klotho gene in the kidney under
36. de Oliveira RM. Klotho RNAi induces premature senescence of sustained circulatory stress in rats. Biochem Biophys Res Com-
human cells via a p53/p21 dependent pathway. FEBS Lett. 2006; mun. 1998;249:865-71.
580:5753-8. 56. Yamagishi T, Saito Y, Nakamura T, Takeda S, Kanai H, Sumino
37. Dmitrieva NI, Burg MB. High NaCl promotes cellular senes- H, et al. Troglitazone improves endothelial function and augments
cence. Cell Cycle. 2007;6:3108-13. renal klotho mRNA expression in Otsuka Long-Evans Tokushima
38. Nakano-Kurimoto R, Ikeda K, Uraoka M, Nakagawa Y, Yutaka Fatty (OLETF) rats with multiple atherogenic risk factors. Hy-
K, Koide M, et al. Replicative senescence of vascular smooth pertens Res. 2001;24:705-9.
muscle cells enhances the calcification through initiating the 57. Wang Y,Sun Z. Klotho gene delivery prevents the progression of
osteoblastic transition. Am J Physiol Heart Circ Physiol. 2009; spontaneous hypertension and renal damage. Hypertension. 2009;
297:H1673-84. 54:810-7.
39. Haruna Y, Kashihara N, Satoh M, Tomita N, Namikoshi T, Sasaki 58. Saito Y, Nakamura T, Ohyama Y, Suzuki T, Iida A, Shiraki-Iida
T, et al. Amelioration of progressive renal injury by genetic T, et al. In vivo klotho gene delivery protects against endothelial
manipulation of Klotho gene. Proc Natl Acad Sci U S A. 2007; dysfunction in multiple risk factor syndrome. Biochem Biophys
104:2331-6. Res Commun. 2000;276:767-72.
40. Sugiura H, Yoshida T, Mitobe M, Yoshida S, Shiohira S, Nitta K, 59. Cheng MF, Chen LJ,Cheng JT. Decrease of Klotho in the kidney
et al. Klotho reduces apoptosis in experimental ischaemic acute of streptozotocin-induced diabetic rats. J Biomed Biotechnol.
kidney injury via HSP-70. Nephrol Dial Transplant. 2010;25: 2010;2010:513853.
60-8. 60. Unger RH. Klotho-induced insulin resistance: a blessing in dis-
41. Saito Y, Yamagishi T, Nakamura T, Ohyama Y, Aizawa H, Suga guise? Nat Med. 2006;12:56-7.
T, et al. Klotho protein protects against endothelial dysfunction. 61. Wang X, Liu X, Zhan Y, Lavallie ER, Diblasio-Smith L, Collins-
Biochem Biophys Res Commun. 1998;248:324-9. Racie L, et al. Pharmacogenomic, physiological, and biochemical
42. Nagai R, Saito Y, Ohyama Y, Aizawa H, Suga T, Nakamura T, et investigations on safety and efficacy biomarkers associated with
al. Endothelial dysfunction in the klotho mouse and downregula- the peroxisome proliferator-activated receptor-gamma activator
tion of klotho gene expression in various animal models of rosiglitazone in rodents: a translational medicine investigation.
vascular and metabolic diseases. Cell Mol Life Sci. 2000;57: J Pharmacol Exp Ther. 2010;334:820-9.
738-46. 62. Schumock GT, Andress DL, Marx SE, Sterz R, Joyce AT,
43. Shimada T, Takeshita Y, Murohara T, Sasaki K, Egami K, Shin- Kalantar-Zadeh K. Association of secondary hyperparathyroid-
tani S, et al. Angiogenesis and vasculogenesis are impaired in the ism with CKD progression, health care costs and survival in
precocious-aging klotho mouse. Circulation. 2004;110:1148-55. diabetic predialysis CKD patients. Nephron Clin Pract. 2009;
44. Fukino K, Suzuki T, Saito Y, Shindo T, Amaki T, Kurabayashi 113:c54-61.
M, et al. Regulation of angiogenesis by the aging suppressor gene 63. Isakova T, Wahl P, Vargas GS, Gutierrez OM, Scialla J, Xie H,
klotho. Biochem Biophys Res Commun. 2002;293:332-7. et al. Fibroblast growth factor 23 is elevated before parathyroid
45. Sato M, Muragaki Y, Saika S, Roberts AB, Ooshima A. Targeted hormone and phosphate in chronic kidney disease. Kidney Int.
disruption of TGF-beta1/Smad3 signaling protects against renal 2011;79:1370-8.
tubulointerstitial fibrosis induced by unilateral ureteral obstruc- 64. Hasegawa H, Nagano N, Urakawa I, Yamazaki Y, Iijima K, Fujita
tion. J Clin Invest. 2003;112:1486-94. T, et al. Direct evidence for a causative role of FGF23 in the
46. Takeshita K, Yamamoto K, Ito M, Kondo T, Matsushita T, Hirai abnormal renal phosphate handling and vitamin D metabolism in
M, et al. Increased expression of plasminogen activator inhibi- rats with early-stage chronic kidney disease. Kidney Int. 2010;
tor-1 with fibrin deposition in a murine model of aging, “Klotho” 78:975-80.
mouse. Semin Thromb Hemost. 2002;28:545-54. 65. Shimamura Y, Hamada K, Inoue K, Ogata K, Ishihara M, Kagawa
47. Sugiura H, Yoshida T, Shiohira S, Kohei J, Mitobe M, Kurosu T, et al. Serum levels of soluble secreted alpha-Klotho are de-
H, et al. Reduced klotho expression level in kidney aggravates creased in the early stages of chronic kidney disease, making it a
renal interstitial fibrosis. Am J Physiol Renal Physiol. 2012; probable novel biomarker for early diagnosis. Clin Exp Nephrol.
302:F1252-64. 2012;16:722-9.
48. Doi S, Zou Y, Togao O, Pastor JV, John GB, Wang L, et al. 66. Shimada T, Urakawa I, Isakova T, Yamazaki Y, Epstein M,
Klotho inhibits transforming growth factor-beta1 (TGF-beta1) Wesseling-Perry K, et al. Circulating fibroblast growth factor 23
signaling and suppresses renal fibrosis and cancer metastasis in in patients with end-stage renal disease treated by peritoneal
mice. J Biol Chem. 2011;286:8655-65. dialysis is intact and biologically active. J Clin Endocrinol Metab.
49. Cleland SJ. Cardiovascular risk in double diabetes mellitus-when 2010;95:578-85.
two worlds collide. Nat Rev Endocrinol. 2012;8:476-85. 67. Kumata C, Mizobuchi M, Ogata H, Koiwa F, Nakazawa A,
50. Saito I. Epidemiological evidence of type 2 diabetes mellitus, Kondo F, et al. Involvement of alpha-klotho and fibroblast growth
metabolic syndrome, and cardiovascular disease in Japan. Circ J. factor receptor in the development of secondary hyperparathy-
2012;76:1066-73. roidism. Am J Nephrol. 2010;31:230-8.
Renal and extrarenal klotho 129

68. Komaba H, Goto S, Fujii H, Hamada Y, Kobayashi A, Shibuya K, 78. Yahata K, Mori K, Mukoyama M, Sugawara A, Suganami T,
et al. Depressed expression of Klotho and FGF receptor 1 in Makino H, et al. Regulation of stanniocalcin 1 and 2 expression in
hyperplastic parathyroid glands from uremic patients. Kidney Int. the kidney by klotho gene. Biochem Biophys Res Commun.
2010;77:232-8. 2003;310:128-34.
69. Krajisnik T, Olauson H, Mirza MA, Hellman P, Akerstrom G, 79. Takei Y, Yamamoto H, Sato T, Otani A, Kozai M, Masuda M, et
Westin G, et al. Parathyroid Klotho and FGF-receptor 1 expres- al. Stanniocalcin 2 is associated with ectopic calcification in
sion decline with renal function in hyperparathyroid patients with alpha-klotho mutant mice and inhibits hyperphosphatemia-in-
chronic kidney disease and kidney transplant recipients. Kidney duced calcification in aortic vascular smooth muscle cells. Bone.
Int. 2010;78:1024-32. 2012;50:998-1005.
70. Galitzer H, Ben-Dov IZ, Silver J,Naveh-Many T. Parathyroid 80. Achinger SG, Ayus JC: The role of vitamin D in left ventricular hyper-
cell resistance to fibroblast growth factor 23 in secondary trophy and cardiac function. Kidney Int Suppl. 2005;95:S37-42.
hyperparathyroidism of chronic kidney disease. Kidney Int. 81. Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, et
2010;77:211-8. al. FGF23 induces left ventricular hypertrophy. J Clin Invest.
71. Hofman-Bang J, Martuseviciene G, Santini MA, Olgaard K, 2011;121:4393-408.
Lewin E. Increased parathyroid expression of klotho in uremic 82. Mirza MA, Larsson A, Melhus H, Lind L, Larsson TE. Serum intact
rats. Kidney Int. 2010;78:1119-27. FGF23 associate with left ventricular mass, hypertrophy and geometry in
72. Ichikawa S, Imel EA, Kreiter ML, Yu X, Mackenzie DS, Soren- an elderly population. Atherosclerosis. 2009;207:546-51.
son AH, et al. A homozygous missense mutation in human 83. Sugiura H, Yoshida T, Tsuchiya K, Mitobe M, Nishimura S, Shirota
KLOTHO causes severe tumoral calcinosis. J Clin Invest. 2007; S, et al. Klotho reduces apoptosis in experimental ischaemic acute
117:2684-91. renal failure. Nephrol Dial Transplant. 2005;20:2636-45.
73. Nakatani T, Sarraj B, Ohnishi M, Densmore MJ, Taguchi T, 84. Mitani H, Ishizaka N, Aizawa T, Ohno M, Usui S, Suzuki T, et al.
Goetz R, et al. In vivo genetic evidence for klotho-dependent, In vivo klotho gene transfer ameliorates angiotensin II-induced
fibroblast growth factor 23 (Fgf23) -mediated regulation of sys- renal damage. Hypertension. 2002;39:838-43.
temic phosphate homeostasis. FASEB J. 2009;23:433-41. 85. Hu MC, Shi M, Zhang J, Quinones H, Kuro-o M, Moe OW.
74. Tangri N, Alam A, Wooten EC, Huggins GS. Lack of association Klotho deficiency is an early biomarker of renal ischemia-reper-
of Klotho gene variants with valvular and vascular calcification in fusion injury and its replacement is protective. Kidney Int. 2010;
Caucasians: a candidate gene study of the Framingham Offspring 78:1240-51.
Cohort. Nephrol Dial Transplant. 2011;26:3998-4002. 86. Zhang H, Li Y, Fan Y, Wu J, Zhao B, Guan Y, et al. Klotho is a
75. Ikushima M, Rakugi H, Ishikawa K, Maekawa Y, Yamamoto K, target gene of PPAR-gamma. Kidney Int. 2008;74:732-9.
Ohta J, et al. Anti-apoptotic and anti-senescence effects of Klotho 87. Narumiya H, Sasaki S, Kuwahara N, Irie H, Kusaba T, Ka-
on vascular endothelial cells. Biochem Biophys Res Commun. meyama H, et al. HMG-CoA reductase inhibitors up-regulate
2006;339:827-32. anti-aging klotho mRNA via RhoA inactivation in IMCD3 cells.
76. Donate-Correa J, Mora-Fernandez C, Martinez-Sanz R, Muros- Cardiovasc Res. 2004;64:331-6.
de-Fuentes M, Perez H, Meneses-Perez B, et al. Expression of 88. Yoon HE, Ghee JY, Piao S, Song JH, Han DH, Kim S, et al.
FGF23/KLOTHO system in human vascular tissue. Int J Cardiol. Angiotensin II blockade upregulates the expression of Klotho, the
2011. Epub ahead of print. anti-ageing gene, in an experimental model of chronic cyclospor-
77. Lim K, Lu TS, Molostvov G, Lee C, Lam F, Zehnder D, et al. ine nephropathy. Nephrol Dial Transplant. 2011;26:800-13.
Vascular klotho deficiency potentiates the development of human 89. Sugiura H, Tsuchiya K, Nitta K. Circulating levels of soluble
artery calcification and mediates resistance to FGF23. Circulation. alpha-Klotho in patients with chronic kidney disease. Clin Exp
2012;125:2243-55. Nephrol. 2011;15:795-6.

Anda mungkin juga menyukai