• No results found

University of Groningen Klotho in vascular biology Mencke, Rik

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Klotho in vascular biology Mencke, Rik"

Copied!
27
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Klotho in vascular biology

Mencke, Rik

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2018

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Mencke, R. (2018). Klotho in vascular biology. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

165

Chapter 5

Assessment of vascular Klotho expression

and functionality

R. Mencke Z. Zhuge M. Burmakin G. Harms M.G. Vervloet M. Carlström H. Olauson J.L. Hillebrands on behalf of the NIGRAM Consortium

(3)

166

Abstract

The renal anti-ageing protein Klotho is being increasingly viewed as a possible link between chronic kidney disease (CKD) and its cardiovascular complications. Klotho is primarily expressed in the kidney, where it is proteolytically cleaved and circulates as soluble Klotho, having beneficial effects on the vasculature. While renal Klotho and soluble Klotho are potentially useful in targeting the vasculature, endogenous Klotho expression in the vasculature is a controversial topic. Various studies conclude that Klotho is not expressed while a multitude of other studies reports high smooth muscle Klotho expression.

In furtherance of resolving this issue, we investigated vascular Klotho expression in mouse and human using Western blotting, immunohistochemistry, PCR, and RNA in situ hybridization. We performed various controls using Klotho-/- mice, Six2-Cre/Klothoflox/flox mice, and

SM22α-Cre/Klothoflox/flox mice and further investigated whether SM22α-Cre/Klothoflox/flox mice have a

vascular phenotype as reflected by impaired vascular function.

We detected a 116 kDa band on Western blot in cultured vascular smooth muscle cells, as well as a vascular staining pattern in tissues, using anti-Klotho antibody Ab69208. This staining pattern did not overlap with the known renal Klotho expression pattern. The Ab69208 staining pattern was present in both Klotho-/- mice and SM22α-Cre/Klothoflox/flox mice, indicating that

it is not specific for Klotho. We corroborate, however, that Klotho is expressed at an extremely low level in the vasculature as detected by RT-PCR, which is further underlined by the finding that SM22α-Cre/Klothoflox/flox mice have a phenotype including endothelial dysfunction and

impaired contractility.

In conclusion, our data indicate that detection of Klotho as a 116 kDa band or as a vascular staining pattern is not related to Klotho, but there appears to be a role for the very low Klotho expression level in regulating endothelial function and smooth muscle cell contractility.

(4)

167

Introduction

There has been a growing interest in the relationship between the pathophysiology of chronic kidney disease (CKD), the occurrence of cardiovascular disease (CVD) in CKD patients, and the possibility that the anti-ageing protein Klotho could be a factor relevant to both. Klotho is a renal transmembrane protein expressed primarily in the distal convoluted tubule (1), where it functions as an obligate co-factor to fibroblast growth factor (FGF) receptor 1c facilitating FGF23 signaling (2, 3) and where it is cleaved off and released as soluble Klotho in blood and urine (4, 5). Deficiency of Klotho in mice causes a syndrome resembling human ageing (1), while Klotho overexpression extends lifespan (6) and protects against various ageing-related diseases (7-16). The combination of findings that CKD is an acquired state of Klotho deficiency (9, 17, 18), Klotho deficiency itself induces a mild form of CKD (9, 19), deficiency of Klotho especially affects the heart and the vasculature (1, 7, 9, 20), and Klotho treatment is beneficial to both experimental CKD and its cardiovascular complications (7, 9, 21), strengthens the hypothesis that Klotho constitutes an important link between CKD and CVD. One promising area of research therefore explores the targeting of Klotho as a treatment for the vasculature, which is afflicted with vascular calcification (1), endothelial dysfunction (22, 23), arterial stiffening (24-26), and hypertension (19, 27) in both murine Klotho deficiency and CKD. Which form of Klotho to target, however, is an open-ended question. Soluble Klotho has been shown to be able to ameliorate Klotho deficiency-induced vascular calcification (28). The alternatively spliced transcript long thought to encode a putative “secreted Klotho”, however, is not actively translated to a protein (29), but therapeutic strategies focused on the KL1 internal repeat that comprises most of the putative “secreted Klotho” are likely viable (30-32). Finally, membrane-bound vascular Klotho expression is a matter of great debate. Stimulating Klotho expression systemically is expected to affect the vasculature by a potentially higher level of soluble Klotho that is derived from the kidney. Whether Klotho is expressed in the vasculature itself and whether stimulating endogenous vascular Klotho expression would therefore be an option to be explored, is currently highly controversial. We and others have previously shown that membrane-bound Klotho protein could not be detected in human or mouse arteries and that, lacking Klotho as an obligate co-receptor, the vasculature does not partake in down-stream FGF23 signaling like the kidney does (33, 34). Our data also indicate, though, that Klotho mRNA is expressed at an extremely low level in the vasculature. This finding is widely echoed in recent literature, but interestingly, many authors also report the presence of high levels of Klotho protein in arteries and smooth muscle cells (35-37). Therefore, in this study, we investigated arterial Klotho expression in furtherance of resolving this issue.

(5)

168

Materials and Methods

Human tissue

Healthy renal artery pieces from kidney donor (leftover from surgery; N = 6) and kidney graft recipient iliac arteries (leftover during creation of the end-to-side anastomosis; N = 6) were collected at the VU University Medical Center, after approval by the Institutional Review Board and after informed consent was given. Renal cell carcinoma-adjacent healthy renal tissue removed during nephrectomy was used as control tissue. Use of human material was conform the Declaration of Helsinki.

(Immuno)histochemistry and immunofluorescence

For immunohistochemistry and immunofluorescence, we used 4 µm thick cryosections, which were dried, fixed in acetone at RT for 10 minutes, followed by incubation with anti-Klotho antibodies Ab69208 (1:100; Abcam, UK) or KM2076 (1:20; TransGenic Inc., Japan), or with E29 (for Epithelial Membrane Antigen, Dako, Denmark) or 1A4 (for α-Smooth Muscle Actin, Dako) for 1 hour. Endogenous peroxidase was inactivated in 0.075% H2O2 in PBS for 30 minutes.

Polyclonal secondary and tertiary antibodies were applied as appropriate (rabbit anti-rat-HRP (P0450, Dako), goat anti-rabbit-HRP (P0448, Dako), rabbit anti-goat-HRP (P0449, Dako), donkey anti-rabbit-Alexa Fluor 488 (Invitrogen), goat anti-mouse IgG2a-Alexa Fluor 555 (Invitrogen)). As a chromogen, 3-amino-9-ethyl carbazole (AEC) was used (with 0.03% H2O2 in

0.02% AEC/50 mM acetate) for 15 minutes. Nuclei were counterstained with hematoxylin or DAPI. Hematoxylin-eosin stainings, periodic acid-Schiff stainings, and Von Kossa stainings were performed according to routine protocols on FFPE sections.

Cell culture

Human aortic smooth muscle cells (HASMCs, ScienCell, USA) were cultured in Smooth Muscle Cell Medium with 2% Fetal Calf Serum, 1% penicillin/streptomycin, and 1% Smooth Muscle Cell Growth Supplement (all from ScienCell), at 37 °C and 5% CO2. Human kidney epithelial cell line HK-2 was maintained in DMEM with 10% FCS, 1% L-glutamine, and 1% penicillin/streptomycin (all from Lonza, USA). For cytofluorescence, HASMCs were cultured on glass coverslips, fixed in 2% paraformaldehyde, and stained according to the protocols described above, but with Tyramide-TRITC Signal Amplification (Perkin Elmer, USA).

(6)

169

Western blotting

Cells were lysed in radioimmunoprecipitation assay (RIPA) buffer and equal amounts of protein were enzymatically de-glycosylated in loading buffer using Endoglycosidase F (a kind gift from Prof. Joost Hoenderop, Randboud University Nijmegen, The Netherlands) for 1 hour at 37 °C. Proteins were separated on 8% polyacrylamide SDS-PAGE gels and blotted onto nitrocellulose membranes. After blocking with 5% non-fat dried milk/TBST, blots were incubated with Ab69208 or KM2076 1:1000 overnight, followed by appropriate secondary antibodies, and detection with Super Signal West Femto Chemiluminescence (Thermy Scientific, USA) on a ChemiDoc MP Imaging System (Bio-Rad, USA).

PCR

RNA was extracted using the TRIzol (Thermo Fisher Scientific) method with chloroform and 2-propanol. Synthesis of cDNA was performed using SuperScript II (Thermo Fisher Scientific) and random hexamer primers. For PCR 20 ng of cDNA input was used, for 40 cycles of 30 s at 94 °C, 30 s at 59 °C and 30 s at 72 °C. Primer sequences are as follows: exon 1 forward (5’-ACTACCGCTTCTCCATCTCG-3’) with exon 2 reverse (5’-TCAAGGTCAATCCAGGAAAG-3’), exon 2 forward CCACAGCATCAAAGAATGTC-3’) with exon 3 reverse (5’-CCACAGCATCAAAGAATGTC-3’), exon 3 forward (5’-CTAAGCCAGGACAAGATG-3’) with exon 4 reverse (5’-TCAGGTCGGTAAACTGAG-3’), and exon 4 forward (5’-CTCCAGGAAATGCACGTTAC-3’) with exon 5 reverse (5’-AAAGCCAGTAAAGACTTTCG-(5’-CTCCAGGAAATGCACGTTAC-3’), followed by resolving on a 2% agarose gel.

RNA in situ hybridization

Formalin-fixed, paraffin-embedded kidney sections from WT and SM22α-Cre/Klothoflox/flox

mice were baked at 60 °C for 1 hour, followed by antigen retrieval and RNA-FISH or RNA-BRISH according to manufacturer instructions (ACDBio, Italy). The probes we used were directed against the mouse Klotho gene (422081) and the human Klotho gene (422041) with the RNAscope 2.5 HD Reagent kit or the RNAscope Multiplex Fluorescent kit v2.

Animal experiments

Klotho+/- mice were provided by Prof. Joost Hoenderop (Radboud University Medical Center,

Nijmegen, The Netherlands). Breeding at the University Medical Center Groningen Central Animal Facility produced WT, Klotho+/-, and Klotho-/- mice. Tissue from Six2-Cre/Klothoflox/flox

mice was used (38) and SM22α-Cre/Klothoflox/flox mice were generated as described previously

(7)

170

the Klotho gene. All animal experiments were conform the NIH Guide for the Use and Care of Laboratory Animals. For vascular function measurements, male and female SM22α-Cre/Klothoflox/flox and WT mice (N = 4 per group) 8-12 weeks of age with an average weight of

25g mice were kept under standard conditions with free access to standard pellet chow and tap water. The mice were sacrificed by cervical dislocation. The whole intestine with attached mesenterical tissues were harvested and mesenteric arteries were dissected (215+/-25 um in diameter, 2 mm in length) and mounted in a myograph chamber (Model 620 M, Danish Myo Technology, Denmark), with physiological salt solution (PSS; composition in mM: 119 NaCl, 4.7 KCl, 1.6 CaCl2, 1.2 KH2PO4, 1.2 MgSO4, 25.1 NaHCO3, 5.5 glucose, 0.026 EDTA). Isometric tension was recorded with a Powerlab system (Powerlab 4/30, AD Instruments, Australia). After being mounted, the vessels were equilibrated for 30 min in PSS bubbling with carbogen (95% O2; 5% CO2) at 37 degree, pH 7.4. Resting tension of the arteries was set as described previously, which is in accordance with Mulvany’s normalization procedure. The vascular viability was verified by a contractile response to potassium chloride (KCl, 100 mM). After washing, the mesenteric arteries were pre-contracted with increasing concentrations of Phenylephrine (PE, 0.0001–10 μM) to reach approximately 80% of KCl-induced contraction. After reaching a stable plateau phase, the endothelium-dependent relaxation was induced by a cumulative concentration of acetylcholine (ACh, 0.001–100 μM). For the endothelium-independent relaxation, a cumulative concentration-dependent response for sodium nitroprusside (SNP, 0.001–100μM) was induced. Between each concentration-response curve, the arteries were washed three times with PSS solution and left to stabilize for 30 minutes.

Statistical analysis

Data are presented as mean ± SEM. Differences between groups are compared using Student’s

t test or the Mann-Whitney U test, depending on the normality of the distribution, which was

tested using the Kruskal-Wallis test. A p value < 0.05 was considered significant and statistical analyses were performed using GraphPad Prism version 5 (Graphpad Inc, USA).

Results

Ab69208 detects a 116 kDa band and displays a vascular staining pattern

We first attempted to find an anti-Klotho antibody that detects a 116 kDa protein, since this is the size of the protein detected by anti-Klotho antibodies in arteries and smooth muscle cells (SMCs). Ab69208 indeed displayed immunoreactivity in human SMCs at 116 kDa (Figure 1A). Additionally, established anti-Klotho antibody KM2076 detected Klotho in renal HK-2 cells at 130 kDa (HK2 –endoF), which exhibited a mobility shift to around 116 kDa by enzymatic

(8)

de-171

glycosylation (HK2 +endoF). No such mobility shift occurred with the SMC band, allowing for the possibility that the 116 kDa band constitutes an non-glycosylated Klotho protein. Assessing the staining pattern of Ab69208, we found that Ab69208 is highly reactive with arterial SMCs in both healthy kidney donor renal arteries and in CKD kidney graft recipient iliac arteries (Figure 1B, C). Additionally, cultured human aortic SMCs stain positive with Ab69208 (Figure 1D). Of note, while KM2076 detects Klotho protein in distal tubules and not in arteries (Figure 1E, F), Ab69208 binds to arteries and not to distal tubules (Figure 1G, H).

Figure 1. Ab69208 is immunoreactive with a vascular epitope. (A) Western blot using anti-Klotho antibodies

Ab69208 on human aortic smooth muscle cells (with and without endoglycosidase F treatment) and KM2076 on human kidney (HK-)2 cells (with and without endoglycosidase F treatment). Note that the vascular band detected with Ab69208 is about 116 kDa in size and does not display a mobility shift after endoglycosidase F treatment and that the renal Klotho as detected with KM2076 is 130 kDa in size and exhibits a mobility shift after enzymatic de-glycosylation. (B) Ab69208 displays a smooth muscle cell staining pattern in healthy donor renal arteries.

Original magnification: 400×. (C) Ab69208 displays a smooth muscle cell staining pattern in kidney graft recipient

iliac arteries. Original magnification: 400×. (D) Cultured smooth muscle cells stain positive with Ab69208. Original

magnification: 400× (inset 630×). (E) KM2076 detects Klotho in distal tubules in human kidney but not in

intrarenal arteries. Original magnification: 400×. (F) Conjugate control for (E). Original magnification: 400×. (G)

Ab69208 detects smooth muscle cells in human kidney, but no distal tubules. Original magnification: 400×. (I) Conjugate control for (G). Original magnifications: 400×.

(9)

172

Ab69208 is not immunoreactive with tubules

To further determine the Ab69208 staining pattern, we performed double stainings for Ab69208 and epithelial membrane antigen (EMA), a distal tubular marker, or α-smooth muscle actin (α-SMA), a smooth muscle cell marker. We found that the Ab69208 epitope was mutually exclusive with EMA (Figure 2A-F), whereas it showed full co-localization with α-SMA (Figure 2G-L). If Ab69208 detects a smaller Klotho protein, its expression pattern would be specifically regulated in different cell types.

Figure 2. The Ab69208 staining pattern is a smooth muscle cell staining pattern without overlap with a distal tubule staining pattern. (A) Epithelial membrane antigen (EMA) staining of human kidney. (B) Ab69208 staining

of human kidney. (C) Overlay, indicating no co-localization of EMA and Ab69208, with neither antibody staining

the glomerulus. (D) Epithelial membrane antigen (EMA) staining of human kidney. (E) Ab69208 staining of human

kidney including a larger artery. (F) Overlay, indicating no co-localization of EMA and Ab69208. (G) α-Smooth

muscle actin staining of human kidney. (H) Ab69208 staining of human kidney. (I) Overlay, indicating full

co-localization of α-SMA and Ab69208 in arterioles. (J) α-Smooth muscle actin staining of human kidney, including a

larger artery. (K) Ab69208 staining of human kidney, including a larger artery. (L) Overlay, indicating full

co-localization of α-SMA and Ab69208 in arteries. Glomeruli are indicated with g. Arterioles are indicated with open arrows. The media of arteries is indicated with m. Original magnifications are 400×.

(10)

173

Ab69208 is immunoreactive with Klotho knockout tissue

To assess whether the protein detected by Ab69208 could be one originating from the Klotho gene, we used various Klotho knockout strains. In WT mouse kidneys, KM2076 detected Klotho in distal tubules (Figure 3A) and not in Klotho-/- mouse distal tubules (Figure 3B), as

expected. However, the vascular staining pattern detected by Ab69208 in WT mice (Figure 3C) was still present in Klotho-/- mice, indicating that it does not detect a Klotho protein. To further

substantiate this finding, we used kidney-specific (Six2-Cre) and artery-specific (SM22α-Cre) Klotho knockout mice (34, 38). Six2-Cre/Klothoflox/flox mice did not display tubular Klotho

staining with KM2076 (Figure 3F), which was present in their WT littermates (Figure 3E), and Ab69208 vascular staining was similar in mice from both genotypes (Figure G, H). SM22α-Cre/Klothoflox/flox mice also displayed arterial staining with Ab69208 (Figure 3L) like their WT

littermates (Figure 3K) while renal Klotho expression as

Figure 3. Staining patterns of anti-Klotho antibodies in Klotho knockout mice. (A) KM2076 detects tubules in

WT mice. (B) KM2076 does not detect tubules in Klotho-/- mice. (C) Ab69208 detects arteries in WT mice. (D) Ab69208 detects arteries in Klotho-/- mice. (E) KM2076 detects tubules in WT mice. (F) KM2076 does not detect tubules in Six2-Cre/Klothoflox/flox mice with a kidney-specific Klotho deletion. (G) Ab69208 detects arteries in WT mice. (H) Ab69208 detects arteries in Six2-Cre/Klothoflox/flox mice with a kidney-specific Klotho deletion. (I) KM2076 detects tubules in WT mice. (J) KM2076 detects tubules in SM22α-Cre/Klothoflox/flox mice with an artery-specific Klotho deletion. (K) Ab69208 detects arteries in WT mice. (L) Ab69208 detects arteries in

(11)

174

detected with KM2076 was also similar (Figure 3I, J). Together, these data from different and independent strains of Klotho knockout mice indicate that vascular staining detected by Ab69208 does not concern a Klotho protein (be it glycosylated or non-glycosylated).

Klotho mRNA is expressed at an extremely low level in arteries

Many studies detail that Klotho is expressed at a very low level in arteries and in cultured SMCs. We also previously (33) and in this study detect Klotho mRNA expression in SMCs (Figure 4A). However, to reliably detect Klotho mRNA expression, a higher input of cDNA (20 ng) was required for a PCR run of 40 cycles and even then detection depends on the part of the transcript and primers. Note the lack of a band after using primers spanning the exon 1-2 boundary, illustrating that the level of Klotho mRNA is low to the extent that it is often rendered undetectable. Given the sensitivity of PCR as a detection method, we also wanted to use RNA in situ hybridization as a method of providing information about Klotho mRNA

expression in a morphological context. Klotho mRNA was readily and similarly detected in distal (and to a lesser extent, proximal) tubules in kidneys from both WT and SM22α- Cre/Klothoflox/flox mice (Figure 4B-E). In arteries, however, Klotho mRNA could not be detected

regardless of genotype (Figure 4D-I). In human kidney, Klotho mRNA was highly expressed in distal tubules, expressed at a very low level in proximal tubules, and not detectable in arteries (Supplemental Figure 1).

Phenotyping of SM22α-Cre/Klothoflox/flox mice

Recent studies have found distinct and functional effects for Klotho endogenously expressed at a very low level in bone cells (in Dmp1-Cre/Klothoflox/flox (39) and Prx1-Cre/Klothoflox/flox mice

(40)) and in proximal tubular epithelium (in Scl34a1-Cre/Klothoflox/flox, PEPCK-Cre/Klothoflox/flox,

and Kap-Cre/Klothoflox/flox mice (41)). To assess whether this could also be the case for the

vasculature, we investigated the aortas of SM22α-Cre/Klothoflox/flox mice. As reported before

(34), compared to WT littermates, artery-specific Klotho knockout aortas displayed normal morphology (Figure 5A, B, D, E) and did not display vascular calcification (Figure 5C, F), a prominent feature in systemic Klotho deficiency.

(12)

175

Figure 4. Investigation of Klotho mRNA expression. (A) RT-PCR for Klotho in human kidney and human smooth

muscle cells, for 40 cycles and with 20 ng of input, yielding a Klotho band that is usually, but not always detected, which is illustrative of the low expression pattern. (B) RNA in situ hybridization for Klotho mRNA in WT mouse

kidney. (C) RNA in situ hybridization for Klotho mRNA in WT mouse kidney. (D) RNA in situ hybridization for Klotho

mRNA in SM22α-Cre/Klothoflox/flox mouse kidney. (E) RNA in situ hybridization for Klotho mRNA in SM22α-Cre/Klothoflox/flox mouse kidney. Note the artery and that Klotho mRNA expression is similar when comparing both genotypes. (F) RNA in situ hybridization for Klotho mRNA in WT mouse artery. (G) RNA in situ hybridization for

Klotho mRNA in WT mouse artery. (H) RNA in situ hybridization for Klotho mRNA in SM22α-Cre/Klothoflox/flox mouse artery. (I) RNA in situ hybridization for Klotho mRNA in SM22α-Cre/Klothoflox/flox mouse artery. Note that Klotho mRNA is not detected in arteries from either genotype. Original magnif×ication: 630×.

(13)

176

Artery-specific Klotho knockout mice have endothelial dysfunction

To assess whether artery-specific Klotho knockout mice display a local vascular phenotype, we performed vascular function measurements. In SM22α-Cre/Klothoflox/flox mesenteric artery

rings, endothelium-dependent relaxation was significantly impaired (Figure 6A), whereas endothelium-independent relaxation was similar (Figure 6B). These results indicate that Klotho-deficient SMCs are capable of normal relaxation, but not when dependent on endothelium-derived NO, suggesting that cross-talk that at least includes communication from smooth muscle to the endothelium is impaired in the absence of a low level of endogenous SMC Klotho.

Figure 5. Phenotyping of the SM22α-Cre/Klothoflox/flox mouse aorta. (A) Hematoxylin-eosin staining of WT mouse

aorta. (B) Periodic acid-Schiff staining of WT mouse aorta. (C) Von Kossa staining of WT mouse aorta. (D)

Hematoxylin-eosin staining of SM22α-Cre/Klothoflox/flox mouse aorta. (E) Periodic acid-Schiff staining of SM22α-Cre/Klothoflox/flox mouse aorta. (F) Von Kossa staining of SM22α-Cre/Klothoflox/flox mouse aorta. No morphological abnormalities or calcifications were noted. Original magnification: 400×.

(14)

177

Artery-specific Klotho knockout mice display impaired arterial contractility

In addition to the relaxation phenotype dependent on thendothelium, we found that artery-specific Klotho knockout mouse arterial rings exhibit impaired contractility in response to phenylepinephrine (Figure 7A), concerning an absolute difference in exerted force, rather than a relative difference compared to KPSS-induced contraction (Figure 7B). Induction of contraction by angiotensin II revealed a similar trend, which did not reach significance (Figure 7C, D).

Figure 6. Artery-specific Klotho deletion results in endothelial dysfunction. (A) Endothelium-dependent vascular

relaxation in response to acetylcholine is significantly reduced in mesenteric arteries from SM22α-Cre/Klothoflox/flox mice. (B) Endothelium-independent vascular relaxation in response to sodium nitroprusside is similar when comparing WT mice and SM22α-Cre/Klothoflox/flox mice. Plotted are means ± SEM (N = 16 rings from 4 mice, for each group). * p < 0.05.

(15)

178

Discussion

The most important findings of this study are the determination that the vascular Klotho immunoreactivity of Ab69208 is non-specific and that, on the other hand, the very low expression level of Klotho does have local functions as its deficiency leads to a phenotype of endothelial dysfunction and impaired contractility.

Figure 7. Artery-specific Klotho deletion results in impaired vascular contractility. (A) Force exerted by

mesenteric artery rings from SM22α-Cre/Klothoflox/flox mice in response to phenylephrine is significantly lower compared to WT littermates. (B) Phenylephrine-induced constriction relative to KPSS-induced constriction is

similar across genotypes. (C) Force exerted by mesenteric artery rings from SM22α-Cre/Klothoflox/flox mice in response to angiotensin II tends to be lower compared to WT littermates, but the difference does not reach statistical significance. (D) Angiotensin II-induced constriction relative to KPSS-induced constriction is similar

(16)

179

In recent years, many authors have addressed the question of endogenous vascular Klotho expression. With regard to Klotho protein, very discrepant findings have been reported. The first extensive study of the Klotho expression pattern was performed using a LacZ reporter system, which did not reveal appreciable vascular expression (42). Klotho expression was not detected in rat aorta using KM2076 (33, 34, 43, 44), nor with KM2119 (34) or AF1819 (25, 45-47), all antibodies known to be mostly specific for renal Klotho and to detect Klotho at 130 kDa in protein lysates using Western blotting. Other studies, however, included data on a vascular 116 kDa Klotho protein and/or staining pattern, using Ab75023, Ab69208, Ab1813373 and Ab203576 (35, 36, 48-51), unspecified anti-Klotho antibodies (37, 52-56), as well as ABIN502138 (57), and AF1819 (in mouse) (58, 59). In particular, Zhu et al. show that the alleged vascular Klotho protein is smaller than recombinant Klotho (52) and Jimbo et al. detect Klotho in rat aorta (with rat monoclonal KM2076), like Hortells et al. (60), but not in SMCs unless transfected with a Klotho construct (61). This summary suggests that the reason for the discrepant findings is likely technical in nature, since the major difference between these studies is which anti-Klotho antibody was used. Our data indicate that the vascular staining pattern and 116 kDa band are likely non-specific, because they are readily detected in both systemic and artery-specific Klotho knockout mice. Our data also render unlikely the possibilities that a Klotho protein that has undergone differential post-translational modification or that a differentially spliced variant is the underlying explanation.

Although our experiments with Ab69208 can not necessarily be extrapolated to other anti-Klotho antibodies that exhibit a vascular staining pattern, the determination that Ab69208 antibody preferentially binds to an SMC epitope with a protein mass of 116 kDa instead of to the renal 130 kDa protein does raise the bar for validation of other anti-Klotho antibodies that exhibit a similar staining pattern. The mutual exclusivity of renal Klotho and non-specific vascular staining patterns (Figures 1, 2) is a convenient characteristic of Ab69208, because other anti-Klotho antibodies (even established antibodies like KM2076 and AF1819) may generally or under certain antigen retrieval conditions detect both the normal Klotho protein and the vascular epitope, while our data indicate that the latter would still be non-specific. Additionally, the mutual exclusivity shows that the detection of a vascular Klotho staining pattern is not a matter of a greater sensitivity, rendering the low vascular protein level detectable with more suitable antibodies, because in the presence of a strong vascular staining pattern with Ab69208, the distal tubules in the kidney remained negative. We recommend that novel Klotho expression patterns be validated using knockout tissue and RNA

in situ hybridization (62).

While the data in the literature are highly discrepant on the protein level, as discussed, the data on the mRNA level have always been much more in agreement. Using PCR, Klotho is generally detected in the vasculature around the detection limit, resulting either in low expression levels (1, 33, 34, 52, 53, 55, 58, 59, 61, 63-70) or in negative data (25, 33, 44-47, 71-75), the detection limit-adjacent nature of which the RT-PCR data in Figure 4A suitably exemplify. Note that we have, on occasion, also detected the exon 1-2 junction in SMCs, as

(17)

180

well as every other exon-exon junction (as seen in Figure 4A), precluding the possibility of a shorter splice variant. A better assessment of Klotho mRNA expression levels is provided by RNA in situ hybridization, for morphological and cellular context (41, 62), or by RNA-seq, for comparison to other genes (53, 74).

Combining our Klotho protein and mRNA data, we conclude that the strong vascular staining pattern and 116 kDa band are not specific for Klotho, while a very low level of Klotho mRNA is constitutively expressed in smooth muscle cells. This results presumably in a very low level of Klotho protein expression, which is too low to be detected. Its deficiency, however, results in a phenotype. Recent studies on endogenous Klotho expression have more frequently produced similar results, especially with regard to bone-specific Klotho knockout mice (39, 40) and in experiments with endothelial cells from WT mice and Klotho-deficient mice suggestive of an extremely low constitutive expression level in WT cells (76). Although the expression in proximal tubule epithelium is comparatively higher than in bone or in the vasculature, there was also for a long time a debate on whether Klotho is expressed in the proximal tubule, which has recently been detailed using RNA-ISH and the demonstration of a phenotype (41). Similarly, there is apparently a very low level of SMC Klotho expression, which affects the endothelium (causing endothelium-dependent relaxation impairment when deficient) and the smooth muscle cells themselves (causing impaired contractility). The finding that loss of SMC Klotho impairs the functionality of the endothelium, in the presence of presumably normal systemic Klotho levels (given the normal renal expression levels) on the luminal side, is highly interesting. The effect is likely either autocrine, with a loss of SMC Klotho affecting SMC-to-EC signaling mediated by other molecules, or it is a paracrine function that targets a highly polarized basolateral EC pathway, otherwise it is unlikely that the comparatively miniscule loss of SMC Klotho could not be offset by the normal Klotho levels in the circulation. We hypothesize that the impaired contractility after artery-specific Klotho deletion is a reflection of partial de-differentiation of SMCs. Relaxation, however, was not affected, except for the endothelial component.

In conclusion, we found that the vascular Klotho staining pattern corresponding to a 116 kDa protein, as is often reported, is non-specific, but that the extremely low constitutive Klotho expression level does have relevant effects, as its SMC-specific deletion leads to endothelial dysfunction and impaired SMC contractility. It is yet to be determined how this miniscule amount of Klotho affects the vasculature and by which mechanisms, but it is becoming increasingly clear that low levels of endogenous Klotho expression have important local functions in various cell types in addition to its functions as a protein present in the blood, urine, and cerebrospinal fluid.

(18)

181

Acknowledgments

This study was funded by a Dutch Kidney Foundation Consortium grant [CP10.11]. NIGRAM PIs: Joost Hoenderop and René Bindels, Radboud University Medical Center, Nijmegen, The Netherlands; Marc Vervloet and Piet ter Wee, VU University Medical Center, Amsterdam, The Netherlands; Gerjan Navis, Martin de Borst, and Jan-Luuk Hillebrands, University Medical Center Groningen, Groningen, The Netherlands.

(19)

182

References

1. M. Kuro-o, Y. Matsumura, H. Aizawa, H. Kawaguchi, T. Suga, T. Utsugi, Y. Ohyama, M. Kurabayashi, T. Kaname, E. Kume, H. Iwasaki, A. Iida, T. Shiraki-Iida, S. Nishikawa, R. Nagai, Y. I. Nabeshima, Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 390, 45-51 (1997).

2. H. Kurosu, Y. Ogawa, M. Miyoshi, M. Yamamoto, A. Nandi, K. P. Rosenblatt, M. G. Baum, S. Schiavi, M. C. Hu, O. W. Moe, M. Kuro-o, Regulation of fibroblast growth factor-23 signaling by klotho. J. Biol. Chem. 281,

6120-6123 (2006).

3. I. Urakawa, Y. Yamazaki, T. Shimada, K. Iijima, H. Hasegawa, K. Okawa, T. Fujita, S. Fukumoto, T. Yamashita, Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature. 444, 770-774 (2006).

4. A. Imura, A. Iwano, O. Tohyama, Y. Tsuji, K. Nozaki, N. Hashimoto, T. Fujimori, Y. Nabeshima, Secreted Klotho protein in sera and CSF: implication for post-translational cleavage in release of Klotho protein from cell membrane. FEBS Lett. 565, 143-147 (2004).

5. C. D. Chen, S. Podvin, E. Gillespie, S. E. Leeman, C. R. Abraham, Insulin stimulates the cleavage and release of the extracellular domain of Klotho by ADAM10 and ADAM17. Proc. Natl. Acad. Sci. U. S. A. 104, 19796-19801

(2007).

6. H. Kurosu, M. Yamamoto, J. D. Clark, J. V. Pastor, A. Nandi, P. Gurnani, O. P. McGuinness, H. Chikuda, M. Yamaguchi, H. Kawaguchi, I. Shimomura, Y. Takayama, J. Herz, C. R. Kahn, K. P. Rosenblatt, M. Kuro-o, Suppression of aging in mice by the hormone Klotho. Science. 309, 1829-1833 (2005).

7. M. C. Hu, M. Shi, N. Gillings, B. Flores, M. Takahashi, M. Kuro-O, O. W. Moe, Recombinant alpha-Klotho may be prophylactic and therapeutic for acute to chronic kidney disease progression and uremic cardiomyopathy.

Kidney Int. 91, 1104-1114 (2017).

8. M. C. Hu, M. Shi, H. J. Cho, B. Adams-Huet, J. Paek, K. Hill, J. Shelton, A. P. Amaral, C. Faul, M. Taniguchi, M. Wolf, M. Brand, M. Takahashi, M. Kuro-O, J. A. Hill, O. W. Moe, Klotho and Phosphate Are Modulators of Pathologic Uremic Cardiac Remodeling. J. Am. Soc. Nephrol. 26, 1290-1302 (2015).

9. M. C. Hu, M. Shi, J. Zhang, H. Quinones, C. Griffith, M. Kuro-o, O. W. Moe, Klotho deficiency causes vascular calcification in chronic kidney disease. J. Am. Soc. Nephrol. 22, 124-136 (2011).

10. M. C. Hu, M. Shi, J. Zhang, H. Quinones, M. Kuro-o, O. W. Moe, Klotho deficiency is an early biomarker of renal ischemia-reperfusion injury and its replacement is protective. Kidney Int. 78, 1240-1251 (2010).

11. D. B. Dubal, L. Zhu, P. E. Sanchez, K. Worden, L. Broestl, E. Johnson, K. Ho, G. Q. Yu, D. Kim, A. Betourne, M. Kuro-O, E. Masliah, C. R. Abraham, L. Mucke, Life extension factor klotho prevents mortality and enhances cognition in hAPP transgenic mice. J. Neurosci. 35, 2358-2371 (2015).

12. D. B. Dubal, J. S. Yokoyama, L. Zhu, L. Broestl, K. Worden, D. Wang, V. E. Sturm, D. Kim, E. Klein, G. Q. Yu, K. Ho, K. E. Eilertson, L. Yu, M. Kuro-o, P. L. De Jager, G. Coppola, G. W. Small, D. A. Bennett, J. H. Kramer, C. R. Abraham, B. L. Miller, L. Mucke, Life extension factor klotho enhances cognition. Cell. Rep. 7, 1065-1076 (2014).

(20)

183

13. P. Ravikumar, J. Ye, J. Zhang, S. N. Pinch, M. C. Hu, M. Kuro-o, C. C. Hsia, O. W. Moe, alpha-Klotho protects against oxidative damage in pulmonary epithelia. Am. J. Physiol. Lung Cell. Mol. Physiol. 307, L566-75 (2014).

14. P. Ravikumar, L. Li, J. Ye, M. Shi, M. Taniguchi, J. Zhang, M. Kuro-O, M. C. Hu, O. W. Moe, C. C. Hsia, Alpha-Klotho deficiency in Acute Kidney Injury Contributes to Lung Damage. J. Appl. Physiol. (1985). 120, 723-732

(2016).

15. J. Xie, J. Yoon, S. W. An, M. Kuro-O, C. L. Huang, Soluble Klotho Protects against Uremic Cardiomyopathy Independently of Fibroblast Growth Factor 23 and Phosphate. J. Am. Soc. Nephrol. 26, 1150-1160 (2015).

16. J. Xie, S. K. Cha, S. W. An, M. Kuro-O, L. Birnbaumer, C. L. Huang, Cardioprotection by Klotho through downregulation of TRPC6 channels in the mouse heart. Nat. Commun. 3, 1238 (2012).

17. N. Koh, T. Fujimori, S. Nishiguchi, A. Tamori, S. Shiomi, T. Nakatani, K. Sugimura, T. Kishimoto, S. Kinoshita, T. Kuroki, Y. Nabeshima, Severely reduced production of klotho in human chronic renal failure kidney. Biochem.

Biophys. Res. Commun. 280, 1015-1020 (2001).

18. S. L. Barker, J. Pastor, D. Carranza, H. Quinones, C. Griffith, R. Goetz, M. Mohammadi, J. Ye, J. Zhang, M. C. Hu, M. Kuro-o, O. W. Moe, S. S. Sidhu, The demonstration of alphaKlotho deficiency in human chronic kidney disease with a novel synthetic antibody. Nephrol. Dial. Transplant. 30, 223-233 (2015).

19. X. Zhou, K. Chen, H. Lei, Z. Sun, Klotho gene deficiency causes salt-sensitive hypertension via monocyte chemotactic protein-1/CC chemokine receptor 2-mediated inflammation. J. Am. Soc. Nephrol. 26, 121-132

(2015).

20. K. Yang, C. Wang, L. Nie, X. Zhao, J. Gu, X. Guan, S. Wang, T. Xiao, X. Xu, T. He, X. Xia, J. Wang, J. Zhao, Klotho Protects Against Indoxyl Sulphate-Induced Myocardial Hypertrophy. J. Am. Soc. Nephrol. 26, 2434-2446 (2015).

21. M. Shi, B. Flores, N. Gillings, A. Bian, H. J. Cho, S. Yan, Y. Liu, B. Levine, O. W. Moe, M. C. Hu, alphaKlotho Mitigates Progression of AKI to CKD through Activation of Autophagy. J. Am. Soc. Nephrol. 27, 2331-2345 (2016).

22. T. Nakamura, Y. Saito, Y. Ohyama, H. Masuda, H. Sumino, M. Kuro-o, Y. Nabeshima, R. Nagai, M. Kurabayashi, Production of nitric oxide, but not prostacyclin, is reduced in klotho mice. Jpn. J. Pharmacol. 89, 149-156 (2002).

23. Y. Saito, T. Yamagishi, T. Nakamura, Y. Ohyama, H. Aizawa, T. Suga, Y. Matsumura, H. Masuda, M. Kurabayashi, M. Kuro-o, Y. Nabeshima, R. Nagai, Klotho protein protects against endothelial dysfunction. Biochem. Biophys.

Res. Commun. 248, 324-329 (1998).

24. K. Chen, X. Zhou, Z. Sun, Haplodeficiency of Klotho Gene Causes Arterial Stiffening via Upregulation of Scleraxis Expression and Induction of Autophagy. Hypertension. 66, 1006-1013 (2015).

25. Y. Lin, J. Chen, Z. Sun, Antiaging Gene Klotho Deficiency Promoted High-Fat Diet-Induced Arterial Stiffening via Inactivation of AMP-Activated Protein Kinase. Hypertension. 67, 564-573 (2016).

26. D. Gao, Z. Zuo, J. Tian, Q. Ali, Y. Lin, H. Lei, Z. Sun, Activation of SIRT1 Attenuates Klotho Deficiency-Induced Arterial Stiffness and Hypertension by Enhancing AMP-Activated Protein Kinase Activity. Hypertension. 68,

1191-1199 (2016).

27. X. Zhou, K. Chen, Y. Wang, M. Schuman, H. Lei, Z. Sun, Antiaging Gene Klotho Regulates Adrenal CYP11B2 Expression and Aldosterone Synthesis. J. Am. Soc. Nephrol. 27, 1765-1776 (2016).

(21)

184

28. J. M. Hum, L. M. O'Bryan, A. K. Tatiparthi, T. A. Cass, E. L. Clinkenbeard, M. S. Cramer, M. Bhaskaran, R. L. Johnson, J. M. Wilson, R. C. Smith, K. E. White, Chronic Hyperphosphatemia and Vascular Calcification Are Reduced by Stable Delivery of Soluble Klotho. J. Am. Soc. Nephrol. 28, 1162-1174 (2017).

29. R. Mencke, G. Harms, J. Moser, M. van Meurs, A. Diepstra, H. G. Leuvenink, J. L. Hillebrands, Human alternative Klotho mRNA is a nonsense-mediated mRNA decay target inefficiently spliced in renal disease. JCI

Insight. 2, 10.1172/jci.insight.94375 (2017).

30. L. Abramovitz, T. Rubinek, H. Ligumsky, S. Bose, I. Barshack, C. Avivi, B. Kaufman, I. Wolf, KL1 internal repeat mediates klotho tumor suppressor activities and inhibits bFGF and IGF-I signaling in pancreatic cancer. Clin.

Cancer Res. 17, 4254-4266 (2011).

31. G. Dalton, S. W. An, S. I. Al-Juboori, N. Nischan, J. Yoon, E. Dobrinskikh, D. W. Hilgemann, J. Xie, K. Luby-Phelps, J. J. Kohler, L. Birnbaumer, C. L. Huang, Soluble klotho binds monosialoganglioside to regulate membrane microdomains and growth factor signaling. Proc. Natl. Acad. Sci. U. S. A. 114, 752-757 (2017).

32. J. D. Wright, S. W. An, J. Xie, J. Yoon, N. Nischan, J. J. Kohler, N. Oliver, C. Lim, C. L. Huang, Modeled structural basis for the recognition of alpha2-3-sialyllactose by soluble Klotho. FASEB J. 31, 3574-3586 (2017).

33. R. Mencke, G. Harms, K. Mirkovic, J. Struik, J. Van Ark, E. Van Loon, M. Verkaik, M. H. De Borst, C. J. Zeebregts, J. G. Hoenderop, M. G. Vervloet, J. L. Hillebrands, NIGRAM Consortium, NIGRAM consortium, Membrane-bound Klotho is not expressed endogenously in healthy or uraemic human vascular tissue. Cardiovasc. Res. 108,

220-231 (2015).

34. K. Lindberg, H. Olauson, R. Amin, A. Ponnusamy, R. Goetz, R. F. Taylor, M. Mohammadi, A. Canfield, K. Kublickiene, T. E. Larsson, Arterial klotho expression and FGF23 effects on vascular calcification and function.

PLoS One. 8, e60658 (2013).

35. K. Lim, T. S. Lu, G. Molostvov, C. Lee, F. Lam, D. Zehnder, L. L. Hsiao, Vascular Klotho Deficiency Potentiates the Development of Human Artery Calcification and Mediates Resistance to FGF-23. Circulation. 125, 2243-2255

(2012).

36. K. Lim, A. Groen, G. Molostvov, T. Lu, K. S. Lilley, D. Snead, S. James, I. B. Wilkinson, S. Ting, L. L. Hsiao, T. F. Hiemstra, D. Zehnder, alpha-Klotho expression in human tissues. J. Clin. Endocrinol. Metab. 100, 1308-1318

(2015).

37. Y. Zhao, M. M. Zhao, Y. Cai, M. F. Zheng, W. L. Sun, S. Y. Zhang, W. Kong, J. Gu, X. Wang, M. J. Xu, Mammalian target of rapamycin signaling inhibition ameliorates vascular calcification via Klotho upregulation. Kidney Int. 88,

711-721 (2015).

38. K. Lindberg, R. Amin, O. W. Moe, M. C. Hu, R. G. Erben, A. Ostman Wernerson, B. Lanske, H. Olauson, T. E. Larsson, The kidney is the principal organ mediating klotho effects. J. Am. Soc. Nephrol. 25, 2169-2175 (2014).

39. H. Komaba, J. Kaludjerovic, D. Z. Hu, K. Nagano, K. Amano, N. Ide, T. Sato, M. J. Densmore, J. I. Hanai, H. Olauson, T. Bellido, T. E. Larsson, R. Baron, B. Lanske, Klotho expression in osteocytes regulates bone metabolism and controls bone formation. Kidney Int. 92, 599-611 (2017).

40. J. Kaludjerovic, H. Komaba, T. Sato, R. G. Erben, R. Baron, H. Olauson, T. E. Larsson, B. Lanske, Klotho expression in long bones regulates FGF23 production during renal failure. FASEB J. 31, 2050-2064 (2017).

(22)

185

41. N. Ide, H. Olauson, T. Sato, M. J. Densmore, H. Wang, J. I. Hanai, T. E. Larsson, B. Lanske, In vivo evidence for a limited role of proximal tubular Klotho in renal phosphate handling. Kidney Int. 90, 348-362 (2016).

42. K. Takeshita, T. Fujimori, Y. Kurotaki, H. Honjo, H. Tsujikawa, K. Yasui, J. K. Lee, K. Kamiya, K. Kitaichi, K. Yamamoto, M. Ito, T. Kondo, S. Iino, Y. Inden, M. Hirai, T. Murohara, I. Kodama, Y. Nabeshima, Sinoatrial node dysfunction and early unexpected death of mice with a defect of klotho gene expression. Circulation. 109,

1776-1782 (2004).

43. H. Mitani, N. Ishizaka, T. Aizawa, M. Ohno, S. Usui, T. Suzuki, T. Amaki, I. Mori, Y. Nakamura, M. Sato, M. Nangaku, Y. Hirata, R. Nagai, In vivo klotho gene transfer ameliorates angiotensin II-induced renal damage.

Hypertension. 39, 838-843 (2002).

44. W. L. Lau, E. M. Leaf, M. C. Hu, M. M. Takeno, M. Kuro-O, O. W. Moe, C. M. Giachelli, 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. 82, 1261-1270 (2012).

45. Y. Wang, Z. Sun, Klotho gene delivery prevents the progression of spontaneous hypertension and renal damage. Hypertension. 54, 810-817 (2009).

46. X. Wang, Z. Sun, RNAi silencing of brain klotho potentiates cold-induced elevation of blood pressure via the endothelin pathway. Physiol. Genomics. 41, 120-126 (2010).

47. Y. Wang, M. Kuro-o, Z. Sun, Klotho gene delivery suppresses Nox2 expression and attenuates oxidative stress in rat aortic smooth muscle cells via the cAMP-PKA pathway. Aging Cell. 11, 410-417 (2012).

48. J. Chen, X. Zhang, H. Zhang, T. Liu, H. Zhang, J. Teng, J. Ji, X. Ding, Indoxyl Sulfate Enhance the Hypermethylation of Klotho and Promote the Process of Vascular Calcification in Chronic Kidney Disease. Int. J.

Biol. Sci. 12, 1236-1246 (2016).

49. B. Richter, J. Haller, D. Haffner, M. Leifheit-Nestler, Klotho modulates FGF23-mediated NO synthesis and oxidative stress in human coronary artery endothelial cells. Pflugers Arch. 468, 1621-1635 (2016).

50. L. Cheng, L. Zhang, J. Yang, L. Hao, Activation of peroxisome proliferator-activated receptor gamma inhibits vascular calcification by upregulating Klotho. Exp. Ther. Med. 13, 467-474 (2017).

51. C. P. Chung, Y. C. Chang, Y. Ding, K. Lim, Q. Liu, L. Zhu, W. Zhang, T. S. Lu, G. Molostvov, D. Zehnder, L. L. Hsiao, alpha-Klotho expression determines nitric oxide synthesis in response to FGF-23 in human aortic endothelial cells. PLoS One. 12, e0176817 (2017).

52. D. Zhu, N. C. Mackenzie, J. L. Millan, C. Farquharson, V. E. MacRae, A protective role for FGF-23 in local defence against disrupted arterial wall integrity? Mol. Cell. Endocrinol. 372, 1-11 (2013).

53. J. R. Chang, J. Guo, Y. Wang, Y. L. Hou, W. W. Lu, J. S. Zhang, Y. R. Yu, M. J. Xu, X. Y. Liu, X. J. Wang, Y. F. Guan, Y. Zhu, J. Du, C. S. Tang, Y. F. Qi, Intermedin1-53 attenuates vascular calcification in rats with chronic kidney disease by upregulation of alpha-Klotho. Kidney Int. 89, 586-600 (2016).

54. J. Zhang, J. R. Chang, X. H. Duan, Y. R. Yu, B. H. Zhang, Thyroid hormone attenuates vascular calcification induced by vitamin D3 plus nicotine in rats. Calcif. Tissue Int. 96, 80-87 (2015).

(23)

186

55. N. A. van Venrooij, R. C. Pereira, Y. Tintut, M. C. Fishbein, N. Tumber, L. L. Demer, I. B. Salusky, K. Wesseling-Perry, FGF23 protein expression in coronary arteries is associated with impaired kidney function. Nephrol. Dial.

Transplant. 29, 1525-1532 (2014).

56. S. Zheng, S. Zhang, Y. Song, W. Guo, W. Zhai, X. Qiu, J. Li, MicroRNA-297a regulates vascular calcification by targeting fibroblast growth factor 23. Iran. J. Basic Med. Sci. 19, 1331-1336 (2016).

57. C. S. Ritter, S. Zhang, J. Delmez, J. L. Finch, E. Slatopolsky, Differential expression and regulation of Klotho by paricalcitol in the kidney, parathyroid, and aorta of uremic rats. Kidney Int. 87, 1141-1152 (2015).

58. Y. Fang, C. Ginsberg, T. Sugatani, M. C. Monier-Faugere, H. Malluche, K. A. Hruska, Early chronic kidney disease-mineral bone disorder stimulates vascular calcification. Kidney Int. 85, 142-150 (2014).

59. Y. Fang, C. Ginsberg, M. Seifert, O. Agapova, T. Sugatani, T. C. Register, B. I. Freedman, M. C. Monier-Faugere, H. Malluche, K. A. Hruska, CKD-induced wingless/integration1 inhibitors and phosphorus cause the CKD-mineral and bone disorder. J. Am. Soc. Nephrol. 25, 1760-1773 (2014).

60. L. Hortells, C. Sosa, N. Guillen, S. Lucea, A. Millan, V. Sorribas, Identifying early pathogenic events during vascular calcification in uremic rats. Kidney Int. 92, 1384-1394 (2017).

61. R. Jimbo, F. Kawakami-Mori, S. Mu, D. Hirohama, B. Majtan, Y. Shimizu, Y. Yatomi, S. Fukumoto, T. Fujita, T. Shimosawa, Fibroblast growth factor 23 accelerates phosphate-induced vascular calcification in the absence of Klotho deficiency. Kidney Int. 85, 1103-1111 (2014).

62. H. Olauson, R. Mencke, J. L. Hillebrands, T. E. Larsson, Tissue expression and source of circulating alphaKlotho.

Bone. 100, 19-35 (2017).

63. J. Donate-Correa, C. Mora-Fernandez, R. Martinez-Sanz, M. Muros-de-Fuentes, H. Perez, B. Meneses-Perez, V. Cazana-Perez, J. F. Navarro-Gonzalez, Expression of FGF23/KLOTHO system in human vascular tissue. Int. J.

Cardiol. 165, 179-183 (2013).

64. J. F. Navarro-Gonzalez, J. Donate-Correa, M. Muros de Fuentes, H. Perez-Hernandez, R. Martinez-Sanz, C. Mora-Fernandez, Reduced Klotho is associated with the presence and severity of coronary artery disease. Heart.

100, 34-40 (2014).

65. N. Silswal, C. D. Touchberry, D. R. Daniel, D. L. McCarthy, S. Zhang, J. Andresen, J. R. Stubbs, M. J. Wacker, FGF23 directly impairs endothelium-dependent vasorelaxation by increasing superoxide levels and reducing nitric oxide bioavailability. Am. J. Physiol. Endocrinol. Metab. 307, E426-36 (2014).

66. J. Donate-Correa, E. Martin-Nunez, R. Martinez-Sanz, M. Muros-de-Fuentes, C. Mora-Fernandez, N. Perez-Delgado, J. F. Navarro-Gonzalez, Influence of Klotho gene polymorphisms on vascular gene expression and its relationship to cardiovascular disease. J. Cell. Mol. Med. 20, 128-133 (2016).

67. R. Nakano-Kurimoto, K. Ikeda, M. Uraoka, Y. Nakagawa, K. Yutaka, M. Koide, T. Takahashi, S. Matoba, H. Yamada, M. Okigaki, H. Matsubara, Replicative senescence of vascular smooth muscle cells enhances the calcification through initiating the osteoblastic transition. Am. J. Physiol. Heart Circ. Physiol. 297, H1673-84

(24)

187

68. J. Voelkl, I. Alesutan, C. B. Leibrock, L. Quintanilla-Martinez, V. Kuhn, M. Feger, S. Mia, M. S. Ahmed, K. P. Rosenblatt, M. Kuro-O, F. Lang, Spironolactone ameliorates PIT1-dependent vascular osteoinduction in klotho-hypomorphic mice. J. Clin. Invest. 123, 812-822 (2013).

69. P. Zhang, Y. Li, Y. Du, G. Li, L. Wang, F. Zhou, Resveratrol Ameliorated Vascular Calcification by Regulating Sirt-1 and Nrf2. Transplant. Proc. 48, 3378-3386 (2016).

70. T. Nakahara, K. Kawai-Kowase, H. Matsui, H. Sunaga, T. Utsugi, T. Iso, M. Arai, S. Tomono, M. Kurabayashi, Fibroblast growth factor 23 inhibits osteoblastic gene expression and induces osteoprotegerin in vascular smooth muscle cells. Atherosclerosis. 253, 102-110 (2016).

71. T. Shiraki-Iida, H. Aizawa, Y. Matsumura, S. Sekine, A. Iida, H. Anazawa, R. Nagai, M. Kuro-o, Y. Nabeshima, Structure of the mouse klotho gene and its two transcripts encoding membrane and secreted protein. FEBS Lett.

424, 6-10 (1998).

72. K. Fon Tacer, A. L. Bookout, X. Ding, H. Kurosu, G. B. John, L. Wang, R. Goetz, M. Mohammadi, M. Kuro-o, D. J. Mangelsdorf, S. A. Kliewer, Research resource: Comprehensive expression atlas of the fibroblast growth factor system in adult mouse. Mol. Endocrinol. 24, 2050-2064 (2010).

73. J. J. Scialla, W. L. Lau, M. P. Reilly, T. Isakova, H. Y. Yang, M. H. Crouthamel, N. W. Chavkin, M. Rahman, P. Wahl, A. P. Amaral, T. Hamano, S. R. Master, L. Nessel, B. Chai, D. Xie, R. R. Kallem, J. Chen, J. P. Lash, J. W. Kusek, M. J. Budoff, C. M. Giachelli, M. Wolf, Chronic Renal Insufficiency Cohort Study Investigators, Fibroblast growth factor 23 is not associated with and does not induce arterial calcification. Kidney Int. 83, 1159-1168 (2013).

74. J. L. Rukov, E. Gravesen, M. L. Mace, J. Hofman-Bang, J. Vinther, C. B. Andersen, E. Lewin, K. Olgaard, Effect of chronic uremia on the transcriptional profile of the calcified aorta analyzed by RNA sequencing. Am. J. Physiol.

Renal Physiol. 310, F477-91 (2016).

75. D. Jung, Y. Xu, Z. Sun, Induction of anti-aging gene klotho with a small chemical compound that demethylates CpG islands. Oncotarget. 8, 46745-46755 (2017).

76. T. Kusaba, M. Okigaki, A. Matui, M. Murakami, K. Ishikawa, T. Kimura, K. Sonomura, Y. Adachi, M. Shibuya, T. Shirayama, S. Tanda, T. Hatta, S. Sasaki, Y. Mori, H. Matsubara, Klotho is associated with VEGF receptor-2 and the transient receptor potential canonical-1 Ca2+ channel to maintain endothelial integrity. Proc. Natl. Acad. Sci.

(25)

188

Supplementary Figure 1. Bright-field RNA in situ hybridization for Klotho mRNA in human kidney. (A) Klotho

mRNA is expressed at a very low level in proximal tubules but expression in arteries cannot be discerned. (B)

Klotho mRNA is expressed in distal tubules, but not in arterioles. (C) Klotho mRNA is expressed in distal tubules

and at a low level in proximal tubules. Black arrows indicate very low Klotho expression. Green arrows indicate arterioles. Original magnification: 400×.

(26)
(27)

Referenties

GERELATEERDE DOCUMENTEN

(C) Autoradiography on aortas from Klotho -/- mice at 7 weeks of age reveals that the pattern develops in the aortic arch and in the abdominal aorta. Patchy involvement of the

Given the profound protective effects of Klotho on endothelial function, smooth muscle cell differentiation, and the kidney, we explored whether a relationship exists

To address whether there is a relationship between Klotho and intimal hyperplasia, we investigated various Klotho-deficient mouse strains and we studied the development of

To assess whether Klotho deficiency induces endothelial dysfunction, we used aortic rings from Klotho -/- , Klotho +/- , and WT mice for ex vivo vascular function measurements..

GBM cell lines (A172, U251, U87, and GSC23) were stimulated with recombinant human Klotho (aa 34-981; R&amp;D Systems, USA) at a concentration of 40 pM, equivalent to 5.2 ng/ml) for

To summarise, reports indicate that soluble Klotho, either directly supplemented as recombinant protein or derived from induced or constitutive overexpression, is capable of

We have assessed studies on Klotho gene variants, Klotho promoter methylation, Klotho mRNA expression, Klotho protein expression, and soluble Klotho levels, with regard to

In short, we found that certain Klotho SNPs are more frequent in ESRD patients and that rs577912 and rs553791 in recipients are associated with an increased risk of graft loss, which