36
From the Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden RENAL KLOTHO AND MINERAL METABOLISM Risul Amin Stockholm 2014

RENAL KLOTHO AND MINERAL METABOLISM

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: RENAL KLOTHO AND MINERAL METABOLISM

From the Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden

RENAL KLOTHO AND MINERAL METABOLISM

Risul Amin

Stockholm 2014

Page 2: RENAL KLOTHO AND MINERAL METABOLISM

Cover image: Immunofluorescence staining shows Klotho expression (red) in the distal tubules. The sodium-phosphate co-transporter Npt2a (green) is localized at the brush-border membrane of the proximal tubules. DAPI (blue) stains the cell nuclei.

All previously published papers were reproduced with permission from the publisher. Published by Karolinska Institutet. Printed by US-AB. © Risul Amin, 2014 ISBN 978-91-7549-482-1

Page 3: RENAL KLOTHO AND MINERAL METABOLISM

1

I would like to dedicate this thesis to Sweden

Page 4: RENAL KLOTHO AND MINERAL METABOLISM

2

ABSTRACT

Calcium and phosphorous are critical elements in a number of physiological processes,

including maintenance of bone structure, cell signaling and energy metabolism. Their

endocrine regulation is tightly controlled through a number of feedback mechanisms

involving parathyroid hormone (PTH), vitamin D and the recently discovered fibroblast

growth factor 23 (FGF23). The kidney is a key organ in maintaining normal serum

levels of calcium and inorganic phosphorous, and disturbances in mineral metabolism

are commonly observed in patients with chronic kidney disease (CKD). Klotho is a

membrane-bound protein expressed in the renal tubules that acts as a co-receptor for

FGF23. In addition, Klotho can be shedded from the cell surface to extra-cellular

compartments and function as a hormone with effects on mineral metabolism

independent of FGF23. During the progression of CKD the expression of Klotho

rapidly declines, and accumulating evidence point to lack of Klotho as a pathogenic

factor driving clinical complications in CKD. The main focus of this thesis has been to

elucidate the role of renal Klotho in mineral metabolism and on systemic effects.

In Study I we generated distal tubule-specific Klotho knockout mice (Ksp-KL-/-) by

employing cre-lox recombination. Ksp-KL-/- mice were hyperphosphatemic with

elevated serum Fgf23 levels, indicating that distal tubular Klotho affects phosphate

reabsorption in the proximal tubules. The exact mechanism of this proposed distal-to-

proximal tubular signaling remains unknown.

In Study II we generated mice with Klotho deleted throughout the nephron (Six2-KL-/-

). Six2-KL-/- mice were infertile, kyphotic, growth retarded and had a decreased life

span, closely resembling the phenotype seen in systemic Klotho knockout mice. Also

the serum and urine biochemistries, low serum Klotho levels as well as profound

histological abnormalities were indistinguishable from systemic Klotho knockout mice,

unraveling the kidney as the principle contributor to circulating Klotho and mediator of

Klotho anti-ageing traits.

Taken together, the studies presented in this licentiate thesis substantially contribute to

the understanding of renal Klotho function.

Page 5: RENAL KLOTHO AND MINERAL METABOLISM

3

LIST OF PUBLICATIONS

I. Olauson H, Lindberg K, Amin R, Jia T, Wernerson A, Andersson G, Larsson

TE.

Targeted deletion of Klotho in kidney distal tubule disrupts mineral

metabolism.

J Am Soc Nephrol. 2012 Oct; 23 (10): 1641-51.

II. Lindberg K, Amin R, Moe OW, Hu MC, Erben RG, Wernerson A, Lanske B,

Olauson H and Larsson TE.

The kidney is the principal organ mediating Klotho effects.

Accepted for publication in J Am Soc Nephrol (Feb 17, 2014).

Page 6: RENAL KLOTHO AND MINERAL METABOLISM

4

RELATED PUBLICATIONS NOT INCLUDED IN THIS

THESIS

I. Lindberg K, Olauson H, Amin R, Ponnusamy A, Goetz R, Taylor RF,

Mohammadi M, Canfield A, Kublickiene K, Larsson TE.

Arterial Klotho expression and FGF23 effects on vascular calcification and

function.

PLoS One. 2013;8(4):e60658.

II. Olauson H, Lindberg K, Amin R, Sato T, Ting J, Goetz R, Mohammadi M,

Andersson G, Lanske B, Larsson TE.

Parathyroid-specific deletion of the Klotho gene unravels a novel calcineurin

dependent FGF23 signalling pathway that mediates suppression of PTH

secretion.

PLoS Genet. 2013 Dec;9(12):e1003975.

III. Jia T*, Olauson H*, Lindberg K, Amin R, Edvardsson K, Lindholm B,

Andersson G, Wernerson A, Sabbagh Y, Schiavi S, Larsson TE.

A novel model of adenine-induced tubulointerstitial nephropathy in mice.

BMC Nephrol. 2013 May 30;14(1):116.

Page 7: RENAL KLOTHO AND MINERAL METABOLISM

5

CONTENTS

1   Introduction ................................................................................................... 7  1.1   Mineral metabolism ............................................................................ 7  

1.1.1   Calcium homeostasis .............................................................. 7  1.1.2   Phosphate homeostasis ........................................................... 9  1.1.3   Fibroblast growth factor-23 ................................................. 11  1.1.4   Klotho ................................................................................... 13  

1.2   Mineral disturbances in chronic kidney disease ............................... 15  2   Aims ............................................................................................................. 18  3   Material and Methods .................................................................................. 19  

3.1   Ethical approval ................................................................................ 19  3.2   Cre-Lox recombination ..................................................................... 19  3.3   Molecular methods ............................................................................ 20  

3.3.1   Enzyme linked immunosorbent assay (ELISA) .................. 20  3.3.2   Immunohistochemistry and immunofluorescence ............... 20  3.3.3   Immunoblot .......................................................................... 21  3.3.4   Real-time qPCR analysis ..................................................... 21  

3.4   Statistical analysis ............................................................................. 22  4   Results and discussion ................................................................................. 23  

4.1   Distal tubuli-specific Klotho-knockout mice have disrupted mineral metabolism and elevated levels of Fgf23 (Study I) ................................... 23  4.2   The kidney is the main contributor to systemic Klotho and the principal organ mediating Klotho effects (Study II) ................................................. 26  

5   Acknowledgements ..................................................................................... 29  6   References ................................................................................................... 31  

Page 8: RENAL KLOTHO AND MINERAL METABOLISM

6

LIST OF ABBREVIATIONS

1,25(OH)2D 1,25 dihydroxyvitamin D

ADHR Autosomal dominant hypophosphatemic rickets

CaSR Calcium-sensing receptor

CKD Chronic kidney disease

CVD Cardiovascular disease

CYP24A1 1, 25-dihydroxyvitamin D 24-hydroxylase

CYP27B1 25 dihydroxyvitamin D 1-alpha hydroxylase

ESRD End-stage renal disease

FGF Fibroblast growth factor

FGF23 Fibroblast growth factor-23

FGFR Fibroblast growth factor receptor

GFR Glomerular filtration rate

HFTC Hyperphosphatemic familial tumoral calcinosis

IF Immunofluorescence

IHC Immunohistochemistry

mKL Membrane-bound Klotho

cKL Shedded full-length Klotho

sKL Truncated Klotho

Ksp-KL-/- Distal tubule-specific Klotho knockout mice

Npt The type II sodium-dependent phosphate co-transporter

Pi Inorganic phosphorus

PTH Parathyroid hormone

PTH1R Parathyroid hormone receptor 1

qPCR Quantitative real-time polymerase chain reaction

RAAS Renin-angiotensin-aldosterone system

SHPT Secondary hyperparathyroidism

Six2-KL-/- Renal Klotho knockout mice

TIO Tumor-induced osteomalacia

TRPV5 Transient receptor potential cation channel subfamily V, 5

VDR Vitamin D receptor

XLH X-linked hypophosphatemic rickets

Page 9: RENAL KLOTHO AND MINERAL METABOLISM

7

1 INTRODUCTION

1.1 MINERAL METABOLISM Calcium and phosphorus are vital to life and involved in many biological processes,

including maintenance of bone structure, intracellular signaling and energy metabolism.

Mineral homeostasis are preserved through a balanced influx and efflux from the

intestine, bone and kidney, and orchestrated by several hormones in a complex set of

feedback loops. Abnormalities in mineral metabolism are usually a consequence of

hereditary or acquired dysfunctions in any of these organs. The kidney is a pivotal

organ regulating calcium and phosphorus metabolism, and disrupted homeostasis of

these elements are therefore almost universal in patients with chronic kidney disease

(CKD) [1]. Overwhelming epidemiological evidence link these abnormalities to

adverse clinical outcomes, most prominently cardiovascular morbidity and mortality in

patients with CKD as well as in community-based settings [2, 3].

1.1.1 Calcium homeostasis 1.1.1.1 Calcium

Calcium is the most abundant mineral in the human body and has a central role in bone

metabolism, neuromuscular and secretory functions. The daily dietary intake of

calcium in an adult is around 1000 mg. Approximately 50% of the total serum calcium

is free and biologically active, the rest is bound to albumin and to a less degree other

anions. The concentration of calcium within body fluids is tightly controlled by

homeostatic mechanisms involving influx and effluxes of calcium between

extracellular fluids and the bone, kidney and intestine [4]. Two principal hormones

account for this regulation: parathyroid hormone (PTH) and 1,25-dihydroxyvitamin D

(1,25(OH)2D).

Page 10: RENAL KLOTHO AND MINERAL METABOLISM

8

1.1.1.2 Calcium homeostasis and parathyroid hormone

The parathyroid glands are small endocrine glands located adjacent to or embedded in

the thyroid gland. Their main function is to produce and secrete PTH. Low

concentrations of serum calcium inactivate the parathyroid gland resident calcium-

sensing receptor (CaSR), which rapidly results in increased PTH secretion, while high

serum calcium activates the CaSR and suppresses PTH secretion [5]. PTH is an 84

amino acid peptide hormone with a half-life of approximately 4 minutes. PTH acts on

the G protein-coupled parathyroid hormone receptor (PTH1R) and has three main

effects: 1) directly stimulates bone resorption; 2) increases calcium reabsorption in the

renal distal tubules; and 3) stimulates the renal production of 1,25(OH)2D. The

concerted effect of PTH is to rapidly increase serum calcium, and the relationship

between calcium and PTH is characterized by a sigmoidal curve with the steepest slope

at physiological levels of serum calcium [6].

1.1.1.3 Calcium regulation by vitamin D

Vitamin D can be produced either in the skin or ingested with the diet. It is 25-

hydroxylated in the liver, and subsequently 1-hydroxylated in the kidney to its active

form – 1,25(OH)2D. Active vitamin D stimulates calcium absorption in the gut and

reabsorption in the kidney, and resorption in bone, by binding the nuclear vitamin D

receptor (VDR), which acts as a transcription factor promoting genes involved in

calcium ion transport. Calcium and 1,25(OH)2D are reciprocally regulated through a

negative feedback loop. The enzyme 25-dihydroxyvitamin D 1-alpha hydroxylase

(CYP27B1), which fulfils the final activation step of vitamin D, is induced by PTH,

hypocalcemia, and hypophosphatemia, and in contrast, CYP27B1 is repressed by

hypercalcemia and hyperphosphatemia [7, 8].

Page 11: RENAL KLOTHO AND MINERAL METABOLISM

9

1.1.2 Phosphate homeostasis 1.1.2.1 Phosphorus

Phosphorus (P) is the sixth most abundant element in the body. In nature, phosphorus is

predominantly bound to oxygen as a phosphate (PO43-), and is an essential component

of bone, nucleic acids and the phospholipids in the cell membrane. The total body

content of phosphate is about 500-700 g, of which 85% is in the form of hydroxyapatite

in bones and teeth, 15% distributed in the cellular compartment and less than 1% in

extracellular fluids. The physiological range for serum inorganic phosphorous (Pi) is

0.7-1.5 mmol/l. Phosphate balance is regulated by multiple endocrine factors including

PTH, 1,25(OH)2D3 and the more recently discovered hormones fibroblast growth factor

23 (FGF23) and Klotho [9].

1.1.2.2 Phosphate homeostasis

Phosphate homeostasis is maintained by three major mechanisms: intestinal absorption,

bone resorption, and renal reabsorption. The daily intake of phosphate is approximately

1200 mg, of which 70% is absorbed by the small intestine through both active and

passive transport mechanisms. The active transport is mediated through the sodium-

dependent phosphate transporter type 2b (Npt2b). Npt2b is primarily expressed in the

small intestine and in the lungs, and is regulated by two major stimuli; 1,25(OH)2D and

dietary phosphate. The passive diffusion is primarily load-dependent although the exact

regulation is unknown. The relative contribution of these two mechanisms is not

exactly known and may vary across species, but active transport is estimated to account

for at least 50% of total transport based in interventional studies in Npt2b knockout

mice [10]. The kidney is the most important organ for maintaining phosphate

homeostasis. Under basal condition, approximately 70% of the filtered phosphate is

reabsorbed at the brush border membrane of the proximal tubules. Both animal and

human studies suggest that the sodium-dependent phosphate transporters Npt2a and

Page 12: RENAL KLOTHO AND MINERAL METABOLISM

10

Npt2c are responsible for approximately 80% and 20% of the renal phosphate

reabsorption, respectively. Bone is the largest reservoir for phosphate in the body.

Reciprocally, extracellular phosphate concentration is a determinant of bone formation

and resorption. A number of studies suggest that a physiological level of phosphate

inhibits bone resorption whereas dietary deprivation is associated with higher

osteoclastic activity and increased phosphate release [11].

1.1.2.3 Phosphate regulation

PTH was long considered the best-characterized phosphate-regulating factor.

Hyperphosphatemia stimulates the release of PTH, which acts on the proximal tubule to

decrease the brush-border membrane expression of Npt2a and Npt2c, thus causing

increased urinary phosphate excretion [12]. In contrast, 1,25(OH)2D stimulate intestinal

phosphate absorption through increased expression of Npt2b. Bone also plays an

important role to regulate phosphate homeostasis. A decrease in serum Pi leads to

increased phosphate resorption from bone, both directly and through the actions of PTH

and vitamin D. The discovery of the phosphaturic hormones FGF23 and Klotho has

dramatically changed the concepts of phosphate regulation [13].

The regulation of mineral metabolism is summarized in Figure 1.

Page 13: RENAL KLOTHO AND MINERAL METABOLISM

11

Figure 1. Calcium and phosphorus balance is preserved through a balanced influx and efflux

from the intestine, bone and kidney, and orchestrated by several hormones in a complex set of

feedback loops.

1.1.3 Fibroblast growth factor-23 The Fibroblast growth factors (FGFs) are a highly conserved family of genes that can

be divided into seven phylogenetic subfamilies. The human/mouse FGFs gene family

comprises 22 members, from FGF1 to FGF23. These 22 genes encode molecules that

have the ability to bind to one or several of the fibroblast growth factor receptors

(FGFRs) [14]. The FGFs are also divided based on their function into intracellular,

canonical, and hormone-like FGFs. In contrast to the intracellular and

Parathyroid gland

KidneySmall intestine

Calcium ↓

PTH ↑

1,25(OH)2D ↑

Calcium absorption ↑

Bone

1,25(OH)2D ↑

Calcium reabsorption ↑

Phosphate excretion ↑

Bone resorption ↑

CYP27B1 FGF23 ↑

Page 14: RENAL KLOTHO AND MINERAL METABOLISM

12

autocrine/paracrine actions of the canonical FGFs, hormone-like (e.g. FGF19, FGF21

and FGF23) act systemically as endocrine factors [15].

1.1.3.1 Structure and function of FGF23

The human FGF23 gene is located on the chromosome 12p13 and is composed of three

exons separated by two introns. It encodes a 32 kDa glycoprotein containing 251 amino

acids. In the N-terminus there is a 24 amino acid signal peptide; thus the secreted

FGF23 protein consists of 227 amino acids. FGF23 is produced by osteoblasts and

osteocytes in bone. The biologically active full-length FGF23 is cleaved into inactive

N-terminal (20 kDa) and C-terminal (12 kDa) fragments, both within the cell it is being

produced, and in the circulation [16]. The estimated half-life of FGF23 is 20-60

minutes. FGF23 has three main functions in physiology; 1) lower serum Pi through

decreasing the abundance of Npt2a and Npt2c at the brush border membrane of renal

proximal tubules; 2) reduce serum 1,25(OH)2D levels by decreasing the activation and

increasing its catabolism through regulation of CYP27B1 and the 24-hydroxylase

(CYP24A1), respectively; and 3) decrease the synthesis and secretion of PTH from the

parathyroid glands [17, 18]. Hence, the concerted effects of FGF23 is to directly

decrease serum levels Pi, and indirectly decrease serum calcium through effects on

1,25(OH)2D and PTH.

1.1.3.2 Animal models and model diseases

Intravenous administration of Fgf23 or transgenic overexpression of Fgf23 in mice

results in hypophosphatemia due to increased renal phosphate wasting, and decreased

levels of circulating 1,25(OH)2D (and PTH within the short term) [18-20]. A similar

phenotype is seen in patients with increased FGF23 levels, either through abnormal

processing (for example in autosomal dominant hypophosphatemic rickets, ADHR,

OMIM #193100), regulation (X-linked hypophosphatemic rickets, XLH, OMIM

Page 15: RENAL KLOTHO AND MINERAL METABOLISM

13

#307800) or ectopic production (tumor-induced osteomalacia, TIO) [21-23]. In

addition, reduced bone mineral density and growth retardation are observed in mice

over-expressing Fgf23 [24]. In contrast, Fgf23 knockout mice or humans with

homozygous missense mutations in FGF23 (e.g. hyperphosphatemic familial tumoral

calcinosis, HFTC OMIM #21900) suffer from an opposite phenotype with

hyperphosphatemia, hypercalcemia, high serum 1,25(OH)2D levels and soft-tissue

calcifications [25, 26] .

1.1.3.3 Fibroblast growth factor receptors

FGF23 exerts its actions in the kidneys and parathyroid glands through binding to

FGFRs and activate the MAPK/ERK pathway. The binding to a FGFR requires the

presence of Klotho, a type 1 membrane-bound protein that functions as an FGF23 co-

receptor [27]. Klotho enhances the affinity of FGF23 to multiple FGFRs, most

importantly FGFR1, 3 and 4, by forming Klotho-FGFR binary complexes. The

importance of Klotho as a co-receptor is evidenced by the fact that mice lacking Klotho

(Klotho knockout mice) develop an identical phenotype as the Fgf23 knockout mice,

with hyperphosphatemia, high serum 1,25(OH)2D levels, infertility, growth retardation,

shortened lifespan and ectopic calcifications, despite a 1000-fold increase or more in

circulating Fgf23 levels [28] .

1.1.4 Klotho 1.1.4.1 Discovery of Klotho gene

Klotho is considered an ageing-suppressor gene that was first identified by Kuro-o and

colleagues in 1997 when studying mice with random mutations in a sodium proton

exchanger [28]. It was named after the Greek goddess Clotho, who spins the thread of

life and decides over birth and death. Klotho is highly expressed in the distal

convoluted tubules in the kidney, chief cells in the parathyroid glands and the epithelial

Page 16: RENAL KLOTHO AND MINERAL METABOLISM

14

cells of choroid plexus of the brain. Silencing Klotho resulted in a phenotype

resembling human ageing with shortened life span, muscle and skin atrophy,

pulmonary emphysema, osteopenia, infertility, hyperphosphatemia and vascular and

soft tissue calcification. In contrast, overexpression of Klotho extended life span in

mice, speculatively through suppression of insulin/IGF1signaling [29].

1.1.4.2 Structure and isoforms of Klotho

The human KLOTHO gene contains five exons and encodes a type 1 single-pass

transmembrane polypeptide of 1014 amino acids. The Klotho protein has two internal

repeats named KL1 and KL2 that share homology to family 1 beta-glycosidase, and a

short intracellular C-terminus [30]. Membrane bound Klotho (mKL) can be shedded

from the cell surface (cKL) by membrane-anchored proteases (including ADAM10 and

ADAM17) into blood, urine and cerebrospinal fluid [31]. In addition to mKL and cKL,

a secreted isoform (sKL) is also generated through alternative splicing at exon 3 [32]

(Figure 2). The relative quantity of the various isoforms, at least at the protein level, is

currently unknown.

Figure 2. Klotho has three isoforms; a membrane-bound form (mKL), a shedded form (cKL)

and a truncated form generated through alternative splicing (sKL).

KL1

KL2

KL1

KL2

KL1

Proteolytic cleavage

Alternative splicing

mKL cKL sKL

Page 17: RENAL KLOTHO AND MINERAL METABOLISM

15

1.1.4.3 Mode of action of Klotho isoforms

The three Klotho isoforms have distinct functions. In 2006 mKL was identified as a

permissive co-receptor for FGF23, increasing its affinity to FGFRs by multiple folds. In

contrast to mKLs role as a co-receptor, cKL function as an independent endocrine

factor [33, 34]. Recent studies have revealed that cKL is involved in a variety of

biological processes including modulation of ion transport, suppression of the renin-

angiotensin-aldosterone system (RAAS), anti-senescence and anti-oxidation [33, 35,

36]. Furthermore, injection of cKL in mice suppresses insulin/IGF1 signaling leading to

increased insulin resistance. Finally, it was reported that both sKL and cKL are

antagonists of Wnt signaling, and are able to counteract the development of fibrosis in

various mouse models of renal injury [37, 38].

1.1.4.4 Regulation of Klotho

The regulation of Klotho is still incompletely understood. However, the promoter

region of Klotho contains vitamin D responsive elements, and Klotho expression is

upregulated by administration of vitamin D, both in vivo and in vitro [39, 40].

Similarly, Klotho is increased in mice on a low phosphate diet [41]. Conversely, a

number of factors such as phosphate, calcium, inflammation, oxidative stress, uremic

toxins and FGF23 have been shown to reduce Klotho expression [35]. The upstream

regulators of Klotho shedding and alternative splicing are yet to be resolved.

1.2 MINERAL DISTURBANCES IN CHRONIC KIDNEY DISEASE CKD is defined as the presence of kidney damage, or a progressive loss in renal

function measured as decreased glomerular filtration rate (GFR below 60 ml/min/1.73

m2), for a period of three months or more. CKD is divided into five stages (Table 1)

depending on GFR [42]. In CKD stage 5, also known as End-Stage Renal Disease

(ESRD), the five-year survival rate is approximately 50%. To sustain life in ESRD

Page 18: RENAL KLOTHO AND MINERAL METABOLISM

16

patients, renal replacement therapies such as dialysis or kidney transplantation are

needed. People with CKD have a markedly increased risk of developing cardiovascular

disease (CVD) [43]. Other than traditional risk factors such as diabetes and high blood

pressure, there are also CKD-specific factors known to be associated with disease

progression and mortality including albuminuria, hyperphosphatemia, and activation of

RAAS. Current treatment strategies, aimed at slowing progression of disease, and at

reducing CVD risk, are clearly insufficient to prevent development of ESRD and

alleviating the associated cardiovascular high-risk profile.

Stage Description GFR (mL/min/1.73 m2) Albuminuria

1 Normal or high ≥90

<30 mg/g

30-300 mg/g

>300 mg/g

2 Mildly decreased 60-89

3a Mildly to moderately decreased 45-59

3b Moderately to severely decreased 30-44

4 Severely decreased 15-29

5 Kidney failure <15 (or dialysis)

Table 1. Stages of chronic kidney disease. CKD is defined as either kidney damage (usually

quantified by albuminuria) or glomerular filtration rate (GFR) <60 mL/min/1.73 m2 for more

than three months.

As renal function declines, the kidneys fail to maintain normal calcium and Pi levels in

the blood, leading to a disturbed mineral metabolism with increased risk of CVD and

mortality [3]. Biochemical changes usually become evident at CKD stage 3-4, but the

pathogenesis starts already at earlier stages. The dynamics of these changes have been

re-evaluated after the discovery of FGF23, which increases already at a mild reduction

in renal function and precedes other hormonal changes [44, 45]. Initially, elevated

Page 19: RENAL KLOTHO AND MINERAL METABOLISM

17

FGF23 compensate for the reduced number of nephrons and maintain serum Pi within

the normal range. However, this comes at the expense of reduced 1,25(OH)2D levels

and a subsequent development of secondary hyperparathyroidism (sHPT) [46] . At later

stages these hormonal adaptations becomes insufficient and instead progresses into a

maladaptive response, contributing to the increased risk of disease progression and

mortality. A number of studies have convincingly shown that renal and parathyroid

Klotho expression is decreased in animals and humans with acute renal failure or CKD,

regardless of etiology [47-51]. This suppression may partly be mediated by epigenetic

changes since uremic toxins induce hypermethylation of the Klotho gene [52]. The

progressive decrease in Klotho eventually results in end-organ FGF23 resistance, which

is likely to contribute to the extreme elevations in FGF23 observed in ESRD [53]. In

turn, FGF23 was recently identified as a cardiovascular toxin with direct effects on

myocardial growth [54] .

Page 20: RENAL KLOTHO AND MINERAL METABOLISM

18

2 AIMS The overall aim of this thesis was to refine the understanding of renal Klotho function

and role in regulating mineral metabolism.

Specifically, the objectives were to;

• Elucidate the role of distal tubular Klotho in renal phosphate handling and on

regulation of FGF23 (Study I).

• Investigate the role of renal Klotho on systemic effect and its contribution to

circulating Klotho (Study II).

Page 21: RENAL KLOTHO AND MINERAL METABOLISM

19

3 MATERIAL AND METHODS 3.1 ETHICAL APPROVAL The studies in this thesis were conducted in compliance with the guidelines of animal

experiments of Karolinska Institutet and approved by Stockholm South ethical

committee. Ethical approval numbers are S68-10, S116-12 and S118-12.

3.2 CRE-LOX RECOMBINATION Cre-Lox recombination is a widely used technology to generate conditional knockout

mice. The system consists of two components: the Cre enzyme and the LoxP sites. The

LoxP site is a specific 34 base pair sequence derived from bacteriophage P1. When

LoxP sites are inserted into the mouse genome (floxed mice), recombination occurs

between LoxP sites in cells expressing Cre recombinase, resulting in either activation

or repression of a specific DNA sequence or gene (Figure 3).

Figure 3. The whole or parts of the target gene is flanked by LoxP sequences, allowing for

tissue-specific deletion and disruption of gene function.

There are a few potential problems using Cre-Lox recombination. First, lack of

specificity of certain Cre promoters results in recombination in several tissues, which

can make the interpretation of results more difficult. Using a reporter strain is a

Exon 1 Exon 2 Exon 3

Exon 1 Exon 2 Exon 3 Tissue-specific promoter Cre recombinase

Exon 1 Exon 3

X

Wild-type allele

Floxed allele

Floxed allele after cre excision

Page 22: RENAL KLOTHO AND MINERAL METABOLISM

20

common way to confirm that Cre expression is restricted to the intended target tissue.

Second, Cre-Lox recombination is usually less efficient compared to conventional

knockout techniques.

3.3 MOLECULAR METHODS

3.3.1 Enzyme linked immunosorbent assay (ELISA)

Enzyme linked immunosorbent assay (ELISA) is a standard technique used for

detection and quantification of soluble antigens and antibodies in body fluids. Briefly,

an unknown amount of antigen is affixed to the surface of a well coated with a capture

antibody. A detection antibody is then applied over the surface to bind the antigen. The

detection antibody is covalently linked to an enzyme, and in the final step a substrate is

added which the enzyme converts to a detectable signal measured using a

spectrophotometer. In this thesis ELISAs are used to quantify serum levels of FGF23

and PTH.

3.3.2 Immunohistochemistry and immunofluorescence Immunohistochemistry (IHC) and immunofluorescence (IF) are widely used methods

to detect the presence and location of a target protein on a tissue section or in cultured

cells. The technique utilizes antibodies for detection of specific target antigens. Either

monoclonal or polyclonal antibodies may be used. Polyclonal antibodies recognize

different epitopes on the same antigen, whereas monoclonal antibodies bind to a

specific epitope. Usually IHC and IF relies on one primary and one secondary antibody.

The primary antibody is raised against the epitope of interest, and the secondary

antibody is raised against the immunoglobulins of the primary antibody. The secondary

antibody is conjugated with a linker molecule or directly bound to a reporter molecule.

The antibody-antigen interaction is visualized using chromogenic detection (IHC),

Page 23: RENAL KLOTHO AND MINERAL METABOLISM

21

which can be seen in a regular microscope, or fluorescent detection (IF), in which a

fluorophore is conjugated to the antibody, which requires a fluorescence microscope for

detection. Potential drawbacks of IHC and IF are unspecific staining and

autofluorescence. Furthermore, IHC and IF provides only semi-quantitative assessment

of protein expression and are less sensitive than other assays such as immunoblotting or

ELISA. In this thesis, the specificity of all antibodies used for IHC and IF were verified

by immunoblots or other techniques.

3.3.3 Immunoblot Immunoblotting is the golden-standard for specific protein detection and quantification

in cell or tissue lysate. This process is accomplished through three steps: 1) size

separation by gel electrophoresis, 2) transfer to solid support by electrical current

induction, 3) staining with primary and secondary antibodies for visualization. A

reporter enzyme or fluorophore is linked to the secondary antibody that produces a

color that allows detection of the protein of interest. Antibody specificity in

immunoblotting can be confirmed through validation of the protein size, and by using

positive and negative controls.

3.3.4 Real-time qPCR analysis Real-time quantitative PCR (qPCR) analysis is a common and sensitive method to

amplify and quantify specific gene products. The method can use either DNA binding

dyes such as SYBR Green, or sequence specific fluorescent reporter probes. In this

thesis, total RNA was extracted and reverse-transcribed into cDNA, and real-time

qPCR subsequently performed using gene-specific primers in a SYBR Green based

assay. The relative gene expression was calculated with the 2-ΔΔ Cq or Ct method

normalizing the gene of interest to a reference gene in the same sample. GAPDH was

used as a reference gene in study I and β-actin in study II.

Page 24: RENAL KLOTHO AND MINERAL METABOLISM

22

3.4 STATISTICAL ANALYSIS Gaussian distribution was tested using D’Agostino and Pearson omnibus normality test.

Variables fulfilling the criteria for normal distribution were tested with two-tailed t-test.

Non-normally distributed variables were compared using Mann-Whitney test.

Differences were considered statistically significant when p<0.05. Regression analysis

was performed using linear regression. A minimum of five mice of each genotype was

analyzed in all experiments. In study I, Ksp-KL-/- mice with relative Klotho levels >

70% were excluded during comparisons between groups. All mice were included in

correlation tests.

Page 25: RENAL KLOTHO AND MINERAL METABOLISM

23

4 RESULTS AND DISCUSSION

4.1 DISTAL TUBULI-SPECIFIC KLOTHO-KNOCKOUT MICE HAVE DISRUPTED MINERAL METABOLISM AND ELEVATED LEVELS OF FGF23 (STUDY I)

Renal Klotho has a key role in maintaining a normal mineral metabolism, both through

direct regulation of calcium- and phosphate co-transporters, and by facilitating FGF23

signaling. However, there are still a number of unanswered questions regarding Klotho

function and mechanism(s) of action. For example, the exact site by which Klotho

mediates FGF23 effects on urinary phosphate wasting and vitamin D metabolism is

unknown. Klotho is nearly exclusively expressed in the distal tubules whereas

phosphate reabsorption is confined to the proximal tubules. Some studies report that

there is sufficient Klotho expression in the proximal tubules to allow FGF23 signaling,

while other studies have shown activation of downstream signaling events upon FGF23

administration exclusively in the distal tubules [55, 56]. The systemic Klotho knockout

mouse develops a phenotype with severely dysregulated mineral metabolism, and is

therefore unsuitable to dissect Klotho’s role in physiology. To better understand renal

FGF23-Klotho function and regulation, we generated distal tubule specific Klotho

knockout mice (Ksp-KL-/-) employing Cre-Lox combination.

Ksp-KL-/- mice were viable, fertile, had normal longevity and a normal gross

phenotype. The knockout efficiency varied between 0 and 74% as measured with qPCR

and immunofluorescence staining (Figure 4). To determine the impact of distal tubular

Klotho deletion we examined serum and urine biochemistries, renal histology as well as

protein and transcript expression.

Page 26: RENAL KLOTHO AND MINERAL METABOLISM

24

Figure 4. Partial deletion of distal tubular Klotho in Ksp-KL-/- mice was confirmed with

immunofluorescence staining. Klotho co-localized fully with TRPV5 in wild-type mice, and

partially in Ksp-KL-/- mice.

Ksp-KL-/- mice were hyperphosphatemic with elevated Fgf23 levels, corresponding to

the residual Klotho expression. PTH levels were decreased in Ksp-KL-/- mice whereas

calcium and 1,25(OH)2D remained normal. Urinary calcium excretion was increased,

which could speculatively be due to decreased renal levels of TRPV5, as confirmed

with immunoblot. Unexpectedly, urinary phosphate excretion was unaltered in Ksp-KL-

/- mice. In contrast, brush-border membrane expression of Npt2a was markedly

increased, providing a plausible explanation to the observed hyperphosphatemia.

Further, immunoblotting showed increased renal levels of VDR. Transcript levels of

renal Cyp27B1 were increased in Ksp-KL-/- mice and Klotho transcript levels correlated

to those of Cyp27B1, VDR, Npt2a, and FGFR1.

In a subgroup of Ksp-KL-/- mice with normal serum Pi, Fgf23 levels were elevated,

supporting that additional factors than serum Pi contribute to Fgf23 regulation.

20 µm

WT Ksp-KL-/-Kl

otho

TRPV

5M

erge

d

Page 27: RENAL KLOTHO AND MINERAL METABOLISM

25

Speculatively, end-organ resistance or soluble Klotho could be involved. Further

studies are needed to explore this potential relationship. Histological analysis revealed a

normal renal morphology. The lack of renal fibrosis and vascular calcification argues

against direct paracrine effects of Klotho, at least during low cellular stress. These

characteristics might be tested in subsequent studies by induction of renal failure or

with ageing mice.

This is the first study to present genetic and functional evidence that partial deletion of

Klotho in the distal tubules has a major impact on renal phosphate handling in the

proximal tubules. The factor(s) responsible for this proposed distal-to-proximal tubule

signaling are currently unknown, but could speculatively be soluble Klotho (Figure 5).

Although distal tubular Klotho appears essential to renal phosphate handling, our

results indicate a limited effect on vitamin D metabolism. Importantly, our data does

not exclude a role for Klotho in the proximal tubule, and this should be further

examined in future studies.

Figure 5. Deletion of Klotho in the distal tubules has a fundamental impact on renal phosphate

handling in the proximal tubules, suggesting the presence of a distal-to-proximal mechanism.

3 Na+

HPO42-

Proximal tubular cell Distal tubular cell

Luminal side

2 Na+

Npt2a

Npt2c

Klotho

FGFRs

FGF23

Distal-to-proximal mechanism?

Glomerulus

Proximal tubule Distal tubule

Loop of Henle

Page 28: RENAL KLOTHO AND MINERAL METABOLISM

26

4.2 THE KIDNEY IS THE MAIN CONTRIBUTOR TO SYSTEMIC KLOTHO AND THE PRINCIPAL ORGAN MEDIATING KLOTHO EFFECTS (STUDY II)

Klotho has been reported to be expressed in multiple organs including the kidneys,

parathyroid glands, choroid plexus, pancreases, breast, testis, ovaries and urinary

bladder, yet its tissue-specific role outside the kidney remain uncertain. Systemic

Klotho knockout mice develop a phenotype resembling accelerated ageing with organ

abnormalities such as soft tissue calcifications, skin atrophy, pulmonary emphysema

and osteomalacia, and biochemical abnormalities including extremely elevated Fgf23

and 1,25(OH)2D, hypercalcemia and hyperphosphatemia. However, the organ-specific

contribution to the ageing phenotype in Klotho knockout mice, and relevant tissue

source of soluble Klotho are yet to be identified. In order to elucidate these issues we

generated mice with Klotho deleted throughout the nephron (Six2-KL-/-) using Cre-Lox

combination. Successful deletion of renal Klotho was confirmed with

immunofluorescence (Figure 6) and western blotting. Renal Klotho transcript levels

were reduced by approximately 80% compared to wild-type mice.

Figure 6. Immunofluorescence staining reveals efficient deletion of renal Klotho (red) in Six2-

KL-/- mice. Lotus tetragonolobus lectin (LTL, green) is a proximal tubule-specific marker and

DAPI (blue) stains cell nuclei.

Klot

ho/L

TL/D

API

WT Six2-KL-/-

Page 29: RENAL KLOTHO AND MINERAL METABOLISM

27

Six2-KL-/- mice were infertile, kyphotic, growth retarded and had a decreased life span,

closely resembling the phenotype seen in systemic Klotho knockout mice. Their serum

biochemistries were characterized by hyperphosphatemia and remarkably elevated

Fgf23 and 1,25(OH)2D levels. PTH levels were decreased in the face of hypercalcemia.

Circulating levels of Klotho was measured using a combined technique of

immunoprecipitation and immunoblotting, and revealed an 80% reduction in Six2-KL-/-

mice. This strongly supports that the kidney is the principal contributor to circulating

Klotho. To confirm this, renal shedding was examined ex vivo using kidney explants. In

stark contrast to wild type mice, Klotho protein was undetectable in conditioned media

from Six2-KL-/- kidneys. Further, immunostaining showed preserved expression of

parathyroid Klotho in extra-renal organs such as the parathyroid glands and choroid

plexus in Six2-KL-/- mice. Thus, it can be concluded that the kidney is the principle

source of soluble Klotho and that extra renal tissues are not capable of correcting a

systemic deficiency of Klotho.

Next we examined the impact of renal Klotho ablation on kidney histology and

detected a higher proliferation rate with increased cellular density, loss of

differentiation between proximal and distal convoluted tubules as well as loss of

cuboidal Bowman’s epithelium of glomeruli in male Six2-KL-/- mice. Additional

histomorphological changes included mild interstitial fibrosis and widespread

nephrocalcinosis. To evaluate the impact of local Klotho expression on the ageing-like

phenotype we performed an extensive histological comparison between Six2-KL-/- mice

and systemic Klotho knockout mice. Notable histological findings in Six2-KL-/- mice

included pulmonary emphysema, reduced subcutaneous fat and skeletal

Page 30: RENAL KLOTHO AND MINERAL METABOLISM

28

hypomineralization. Importantly, we found no differences in renal or extra renal

abnormalities between Six2-KL-/- and systemic Klotho knockout mice.

In conclusion, our data unequivocally unravels the kidney as the primary source of

circulating Klotho and mediator of Klotho function. This study have several

implications in the field of kidney research and provides support that therapeutic

Klotho delivery in CKD patients, a state of Klotho deficiency, may improve the uremic

complication and help to slow disease progression.

Page 31: RENAL KLOTHO AND MINERAL METABOLISM

29

5 ACKNOWLEDGEMENTS

During the time of these studies, my mother got diagnosed with CKD and my aunt died

from kidney failure, bringing this research to a very personal level for me. Hopefully,

the combined efforts of the scientific community will eventually lead to a greater

understanding of CKD and allow for better treatment(s) for my mother and everyone

else in this world that suffer from this disease.

The work in this licentiate thesis was performed at the Division of Renal Medicine,

Department of Clinical Science, Intervention and Technology at Karolinska Institutet,

Sweden. I would like to express my great appreciation to everyone who has contributed

during the past years. I would especially like to thank the following people:

My supervisor Tobias Larsson Agervald for his excellent guidance that made me go

into research in the first place. Your outstanding knowledge in FGF23 and Klotho

biology and everything surrounding it is very inspiring and indeed I am truly grateful

for your generosity and support during the past years.

Annika Wernerson, my co-supervisor, for your scientific support, and for always

helping out when needed.

Hannes Olauson, my co-supervisor, for guiding me through this journey. You have

always been there, available for scientific discussions and support. You are an

enthusiastic, creative scientist that I learned a lot from. Your encouragement and

friendship never fails to motivate me.

Page 32: RENAL KLOTHO AND MINERAL METABOLISM

30

My former and present colleagues, Karolina Lindberg, and Karin Edvardsson, for

making the endless hours in the lab so much more enjoyable, for all discussions about

work, life and everything else. Christina Hammarstedt, for excellent technical

assistance, always being so helpful and taking care of our last minutes orders. Your

kind attitude towards me made me feel like you were my Swedish mother. Ting Jia, for

your kind friendship and motivation.

Thanks to all the nice people at CLINTEC and Kliniskt Forskningscentrum (KFC).

Peter Stenvinkel, Bengt Lindholm, Tony Qureshi, Karolina Kublickiene, Björn

Anderstam, Monica Eriksson, Ann-Christin Bragfors and Samsul Arefin.

Special thanks to the members of Edvard Smith’s team for their kind help, Leonardo

Vargas, Manuela Gustafsson, Hossain Nawaz, Dara Mohammad, Sylvain Geny,

Burcu Bestas. Also thanks to Göran Andersson, for his kindness and help to allow

me to do imunohistochemistry in his laboratory.

A big thanks you to my all KI friends, including, Philipp, Mei, Brigitte, Sara,

Ibrahim, Abdul, Ferdous, Miraz, Asif Aziz.

Hearty thanks to my Bangladeshi friends, Samsul Boss, Nishu Apu, Jowel Viya and

Vave, for cooking and taking good care of me the last couple of years when I was

spending long hours in the lab.

And finally, my parents Ruhul Amin and Rekha Amin, and my sisters Dr. Ireen

Amin and Safrin Amin, who gave me a happy childhood with love and all mental

support and motivation during these studies.

Page 33: RENAL KLOTHO AND MINERAL METABOLISM

31

6 REFERENCES 1. Levin A, Bakris GL, Molitch M, Smulders M, Tian J, Williams LA, Andress

DL: Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney disease. Kidney Int 2007, 71(1):31-38.

2. Dhingra R, Sullivan LM, Fox CS, Wang TJ, D'Agostino RB, Gaziano JM, Vasan RS: Relations of serum phosphorus and calcium levels to the incidence of cardiovascular disease in the community. Arch Intern Med 2007, 167(9):879-885.

3. Block GA, Klassen PS, Lazarus JM, Ofsthun N, Lowrie EG, Chertow GM: Mineral metabolism, mortality, and morbidity in maintenance hemodialysis. J Am Soc Nephrol 2004, 15(8):2208-2218.

4. Felsenfeld A, Rodriguez M, Levine B: New insights in regulation of calcium homeostasis. Curr Opin Nephrol Hypertens 2013, 22(4):371-376.

5. Chen RA, Goodman WG: Role of the calcium-sensing receptor in parathyroid gland physiology. Am J Physiol Renal Physiol 2004, 286(6):F1005-1011.

6. Brown EM, Gamba G, Riccardi D, Lombardi M, Butters R, Kifor O, Sun A, Hediger MA, Lytton J, Hebert SC: Cloning and characterization of an extracellular Ca(2+)-sensing receptor from bovine parathyroid. Nature 1993, 366(6455):575-580.

7. DeLuca HF: Evolution of our understanding of vitamin D. Nutr Rev 2008, 66(10 Suppl 2):S73-87.

8. Henry HL, Norman AW: Vitamin D: metabolism and biological actions. Annu Rev Nutr 1984, 4:493-520.

9. Farrow EG, White KE: Recent advances in renal phosphate handling. Nat Rev Nephrol 2010, 6(4):207-217.

10. Sabbagh Y, O'Brien SP, Song W, Boulanger JH, Stockmann A, Arbeeny C, Schiavi SC: Intestinal npt2b plays a major role in phosphate absorption and homeostasis. J Am Soc Nephrol 2009, 20(11):2348-2358.

11. Yates AJ, Oreffo RO, Mayor K, Mundy GR: Inhibition of bone resorption by inorganic phosphate is mediated by both reduced osteoclast formation and decreased activity of mature osteoclasts. J Bone Miner Res 1991, 6(5):473-478.

12. Forster IC, Hernando N, Biber J, Murer H: Proximal tubular handling of phosphate: A molecular perspective. Kidney Int 2006, 70(9):1548-1559.

13. Hori M, Shimizu Y, Fukumoto S: Minireview: fibroblast growth factor 23 in phosphate homeostasis and bone metabolism. Endocrinology 2011, 152(1):4-10.

14. Beenken A, Mohammadi M: The structural biology of the FGF19 subfamily. Adv Exp Med Biol 2012, 728:1-24.

15. Angelin B, Larsson TE, Rudling M: Circulating fibroblast growth factors as metabolic regulators--a critical appraisal. Cell Metab 2012, 16(6):693-705.

16. Yamashita T, Yoshioka M, Itoh N: Identification of a novel fibroblast growth factor, FGF-23, preferentially expressed in the ventrolateral thalamic nucleus of the brain. Biochem Biophys Res Commun 2000, 277(2):494-498.

17. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, Goetz R, Kuro-o M, Mohammadi M, Sirkis R, Naveh-Many T, Silver J: The parathyroid is a target organ for FGF23 in rats. J Clin Invest 2007, 117(12):4003-4008.

Page 34: RENAL KLOTHO AND MINERAL METABOLISM

32

18. Liu S, Tang W, Zhou J, Stubbs JR, Luo Q, Pi M, Quarles LD: Fibroblast growth factor 23 is a counter-regulatory phosphaturic hormone for vitamin D. J Am Soc Nephrol 2006, 17(5):1305-1315.

19. Larsson T, Marsell R, Schipani E, Ohlsson C, Ljunggren O, Tenenhouse HS, Jüppner H, Jonsson KB: Transgenic mice expressing fibroblast growth factor 23 under the control of the alpha1(I) collagen promoter exhibit growth retardation, osteomalacia, and disturbed phosphate homeostasis. Endocrinology 2004, 145(7):3087-3094.

20. Shimada T, Urakawa I, Yamazaki Y, Hasegawa H, Hino R, Yoneya T, Takeuchi Y, Fujita T, Fukumoto S, Yamashita T: FGF-23 transgenic mice demonstrate hypophosphatemic rickets with reduced expression of sodium phosphate cotransporter type IIa. Biochem Biophys Res Commun 2004, 314(2):409-414.

21. Consortium A: Autosomal dominant hypophosphataemic rickets is associated with mutations in FGF23. Nat Genet 2000, 26(3):345-348.

22. Jonsson KB, Zahradnik R, Larsson T, White KE, Sugimoto T, Imanishi Y, Yamamoto T, Hampson G, Koshiyama H, Ljunggren O et al: Fibroblast growth factor 23 in oncogenic osteomalacia and X-linked hypophosphatemia. N Engl J Med 2003, 348(17):1656-1663.

23. Shimada T, Mizutani S, Muto T, Yoneya T, Hino R, Takeda S, Takeuchi Y, Fujita T, Fukumoto S, Yamashita T: Cloning and characterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proc Natl Acad Sci U S A 2001, 98(11):6500-6505.

24. Wang H, Yoshiko Y, Yamamoto R, Minamizaki T, Kozai K, Tanne K, Aubin JE, Maeda N: Overexpression of fibroblast growth factor 23 suppresses osteoblast differentiation and matrix mineralization in vitro. J Bone Miner Res 2008, 23(6):939-948.

25. Benet-Pagès A, Orlik P, Strom TM, Lorenz-Depiereux B: An FGF23 missense mutation causes familial tumoral calcinosis with hyperphosphatemia. Hum Mol Genet 2005, 14(3):385-390.

26. Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T, Fukumoto S, Tomizuka K, Yamashita T: Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest 2004, 113(4):561-568.

27. Kurosu H, Ogawa Y, Miyoshi M, Yamamoto M, Nandi A, Rosenblatt KP, Baum MG, Schiavi S, Hu MC, Moe OW et al: Regulation of fibroblast growth factor-23 signaling by klotho. J Biol Chem 2006, 281(10):6120-6123.

28. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, Ohyama Y, Kurabayashi M, Kaname T, Kume E et al: Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 1997, 390(6655):45-51.

29. Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, McGuinness OP, Chikuda H, Yamaguchi M, Kawaguchi H et al: Suppression of aging in mice by the hormone Klotho. Science 2005, 309(5742):1829-1833.

30. Matsumura Y, Aizawa H, Shiraki-Iida T, Nagai R, Kuro-o M, Nabeshima Y: Identification of the human klotho gene and its two transcripts encoding membrane and secreted klotho protein. Biochem Biophys Res Commun 1998, 242(3):626-630.

31. Chen CD, Podvin S, Gillespie E, Leeman SE, Abraham CR: Insulin stimulates the cleavage and release of the extracellular domain of Klotho by ADAM10 and ADAM17. Proc Natl Acad Sci U S A 2007, 104(50):19796-19801.

Page 35: RENAL KLOTHO AND MINERAL METABOLISM

33

32. Shiraki-Iida T, Aizawa H, Matsumura Y, Sekine S, Iida A, Anazawa H, Nagai R, Kuro-o M, Nabeshima Y: Structure of the mouse klotho gene and its two transcripts encoding membrane and secreted protein. FEBS Lett 1998, 424(1-2):6-10.

33. Chang Q, Hoefs S, van der Kemp AW, Topala CN, Bindels RJ, Hoenderop JG: The beta-glucuronidase klotho hydrolyzes and activates the TRPV5 channel. Science 2005, 310(5747):490-493.

34. Hu MC, Shi M, Zhang J, Pastor J, Nakatani T, Lanske B, Razzaque MS, Rosenblatt KP, Baum MG, Kuro-o M et al: Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. FASEB J 2010, 24(9):3438-3450.

35. Hu MC, Kuro-o M, Moe OW: Secreted klotho and chronic kidney disease. Adv Exp Med Biol 2012, 728:126-157.

36. de Borst MH, Vervloet MG, ter Wee PM, Navis G: Cross talk between the renin-angiotensin-aldosterone system and vitamin D-FGF-23-klotho in chronic kidney disease. J Am Soc Nephrol 2011, 22(9):1603-1609.

37. Zhou L, Li Y, Zhou D, Tan RJ, Liu Y: Loss of Klotho contributes to kidney injury by derepression of Wnt/β-catenin signaling. J Am Soc Nephrol 2013, 24(5):771-785.

38. Hu MC, Shi M, Zhang J, Quiñones H, Kuro-o M, Moe OW: Klotho deficiency is an early biomarker of renal ischemia-reperfusion injury and its replacement is protective. Kidney Int 2010, 78(12):1240-1251.

39. Tsujikawa H, Kurotaki Y, Fujimori T, Fukuda K, Nabeshima Y: Klotho, a gene related to a syndrome resembling human premature aging, functions in a negative regulatory circuit of vitamin D endocrine system. Mol Endocrinol 2003, 17(12):2393-2403.

40. Lau WL, Leaf EM, Hu MC, Takeno MM, Kuro-o M, Moe OW, Giachelli CM: Vitamin D receptor agonists increase klotho and osteopontin while decreasing aortic calcification in mice with chronic kidney disease fed a high phosphate diet. Kidney Int 2012, 82(12):1261-1270.

41. Morishita K, Shirai A, Kubota M, Katakura Y, Nabeshima Y, Takeshige K, Kamiya T: The progression of aging in klotho mutant mice can be modified by dietary phosphorus and zinc. J Nutr 2001, 131(12):3182-3188.

42. Levey AS, de Jong PE, Coresh J, El Nahas M, Astor BC, Matsushita K, Gansevoort RT, Kasiske BL, Eckardt KU: The definition, classification, and prognosis of chronic kidney disease: a KDIGO Controversies Conference report. Kidney Int 2011, 80(1):17-28.

43. Herzog CA, Asinger RW, Berger AK, Charytan DM, Díez J, Hart RG, Eckardt KU, Kasiske BL, McCullough PA, Passman RS et al: Cardiovascular disease in chronic kidney disease. A clinical update from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 2011, 80(6):572-586.

44. Larsson T, Nisbeth U, Ljunggren O, Jüppner H, Jonsson KB: Circulating concentration of FGF-23 increases as renal function declines in patients with chronic kidney disease, but does not change in response to variation in phosphate intake in healthy volunteers. Kidney Int 2003, 64(6):2272-2279.

45. Isakova T, Wahl P, Vargas GS, Gutiérrez OM, Scialla J, Xie H, Appleby D, Nessel L, Bellovich K, Chen J et al: Fibroblast growth factor 23 is elevated before parathyroid hormone and phosphate in chronic kidney disease. Kidney Int 2011, 79(12):1370-1378.

46. Shalhoub V, Shatzen EM, Ward SC, Davis J, Stevens J, Bi V, Renshaw L, Hawkins N, Wang W, Chen C et al: FGF23 neutralization improves chronic

Page 36: RENAL KLOTHO AND MINERAL METABOLISM

34

kidney disease-associated hyperparathyroidism yet increases mortality. J Clin Invest 2012, 122(7):2543-2553.

47. Hu MC, Shi M, Zhang J, Quiñones H, Griffith C, Kuro-o M, Moe OW: Klotho deficiency causes vascular calcification in chronic kidney disease. J Am Soc Nephrol 2011, 22(1):124-136.

48. Koh N, Fujimori T, Nishiguchi S, Tamori A, Shiomi S, Nakatani T, Sugimura K, Kishimoto T, Kinoshita S, Kuroki T et al: Severely reduced production of klotho in human chronic renal failure kidney. Biochem Biophys Res Commun 2001, 280(4):1015-1020.

49. Komaba H, Goto S, Fujii H, Hamada Y, Kobayashi A, Shibuya K, Tominaga Y, Otsuki N, Nibu K, Nakagawa K et al: Depressed expression of Klotho and FGF receptor 1 in hyperplastic parathyroid glands from uremic patients. Kidney Int 2010, 77(3):232-238.

50. Krajisnik T, Olauson H, Mirza MA, Hellman P, Akerström G, Westin G, Larsson TE, Björklund P: Parathyroid Klotho and FGF-receptor 1 expression decline with renal function in hyperparathyroid patients with chronic kidney disease and kidney transplant recipients. Kidney Int 2010, 78(10):1024-1032.

51. Galitzer H, Ben-Dov IZ, Silver J, Naveh-Many T: Parathyroid cell resistance to fibroblast growth factor 23 in secondary hyperparathyroidism of chronic kidney disease. Kidney Int 2010, 77(3):211-218.

52. Young GH, Wu VC: KLOTHO methylation is linked to uremic toxins and chronic kidney disease. Kidney Int 2012, 81(7):611-612.

53. Kovesdy CP, Quarles LD: The role of fibroblast growth factor-23 in cardiorenal syndrome. Nephron Clin Pract 2013, 123(3-4):194-201.

54. Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, Gutiérrez OM, Aguillon-Prada R, Lincoln J, Hare JM et al: FGF23 induces left ventricular hypertrophy. J Clin Invest 2011, 121(11):4393-4408.

55. Andrukhova O, Zeitz U, Goetz R, Mohammadi M, Lanske B, Erben RG: FGF23 acts directly on renal proximal tubules to induce phosphaturia through activation of the ERK1/2-SGK1 signaling pathway. Bone 2012, 51(3):621-628.

56. Farrow EG, Davis SI, Summers LJ, White KE: Initial FGF23-mediated signaling occurs in the distal convoluted tubule. J Am Soc Nephrol 2009, 20(5):955-960.