• 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)

259

Chapter 8

Klotho deficiency induces arteriolar

hyalinosis in a trade-off with vascular

calcification

R. Mencke J. Voelkl A.T. Umbach H. Olauson G. Harms M. Bulthuis L. Quintanilla-Martinez H. van Goor F. Lang J.L. Hillebrands Submitted

(3)

260

Abstract

Hyalinosis is a vascular lesion that affects the renal vasculature and contributes to renal function decline in ageing. We wanted to assess whether arteriolar hyalinosis is caused by Klotho deficiency – a state known to induce both renal and vascular phenotypes associated with ageing.

We used histochemistry to assess the presence of hyalinosis in Klotho-/- kidneys, as compared

to Klotho+/- and wild-type littermates. We also used histochemistry and

immunohistochemistry to investigate the composition of the vascular lesions and the different layers of the vascular wall. Finally, we used spironolactone to inhibit calcification in kl/kl mice and characterized the vascular lesions in the kidney.

We detected arteriolar hyalinosis in Klotho-/- mice, which was found to be present up to the

afferent arterioles. Hyalinosis was accompanied by loss of α-smooth muscle actin expression, while the endothelial lining was mostly intact. Hyalinous lesions were positive for IgM and iC3b/c/d, indicating subendothelial leakage of plasma proteins. The presence of extracellular matrix proteins suggested increased production by smooth muscle cells. Finally, in Klotho

-/-mice that developed marked vascular calcification, treatment with spironolactone allowed for replacement of calcification by hyalinosis.

Klotho deficiency potentiates both endothelial hyperpermeability and smooth muscle cell de-differentiation. In the absence of a stimulus capable of inducing calcification, smooth muscle cells will assume a synthetic phenotype in response to subendothelial leakage of plasma proteins. In the kidney, this results in hyalinosis of the afferent arterioles, which contributes to the decline in renal function. Klotho may play a role in preventing ageing-related or calcineurin inhibitor-induced arteriolar hyalinosis.

(4)

261

Introduction

Arteriolar hyalinosis, or hyaline arteriolosclerosis, is a vascular lesion often found in the kidney in ageing and hypertension (1), or caused by calcineurin inhibitor use after transplantation (2, 3). It consists of accumulated hyaline material, which is thought to be the result of subendothelial leakage of plasma proteins (4, 5) and of subsequently increased extracellular matrix production by smooth muscle cells (SMCs) (6). In the kidney, the afferent arterioles are the most commonly affected vessels, likely owing to their role of major resistance arterioles, placing great local stress on the endothelium. It is thought that hyalinosis of the afferent arterioles leads to loss of autoregulation, which in turn leads to glomerular damage and the decline of renal function (7, 8).

We sought to address whether murine Klotho deficiency, as a model for premature ageing that also leads to a spontaneous decrease in renal function, is affected by hyalinosis as well. Klotho is a renal anti-ageing protein that is predominantly expressed in the distal convoluted tubule (9). Its marked protective effects on the kidney (10-21) and on the vasculature (22-26)

are intricately interrelated, involving both direct effects on renal and vascular cells and indirect effects via the regulation of phosphate (27-29) and calcium (30-32) homeostasis. In mice, Klotho deficiency leads to a renal phenotype of albuminuria, elevated serum creatinine and urea nitrogen levels (33), both interstitial and glomerular fibrosis (33, 34), and mesangial matrix expansion (35). Furthermore, Klotho-deficient kidneys are more prone to the development of renal disease in various damage models (10, 14, 17, 20, 34, 35). With regard to the vasculature, full Klotho deficiency leads to a phenotype of medial calcification (9) and endothelial hyperpermeability (36), while heterozygous mice display endothelial dysfunction (37-39), arterial stiffening (40-42), and hypertension (33, 43). Given the profound protective effects of Klotho on endothelial function, smooth muscle cell differentiation, and the kidney, we explored whether a relationship exists between hyalinosis as an ageing-associated vascular lesion and Klotho deficiency as a state of accelerated ageing.

Results

Klotho deficiency induces arteriolar hyalinosis

In 7-week-old Klotho-/- mouse kidneys, we detected vascular lesions that were strongly

PAS-positive (Figure 1, G-H) and were typically eosinophilic and “glassy” on HE staining (Figure 1, I), indicative of hyalinosis. These lesions were not observed in Klotho+/- or wild-type littermates

(5)

262

lesions did not represent calcifications (Supplemental Figure 1, A-C). Oil Red O stainings were also negative, indicating that these lesions did not contain a fatty component (Supplemental Figure 1, D-E).

Hyalinosis is mostly found in terminal interlobular arteries and proximal afferent arterioles

The relatively large segmental arteries generally contained affected regions (Figure 2A) and luminal measurements of all α-SMA-positive arteries indicate that this pattern remained patchy with some or some parts of interlobar and arcuate arteries being affected. Most affected arteries were interlobular arteries and proximal afferent arterioles (Figure 2B), with most having a lumen with a diameter of 6-42 µm (Figure 2D). However, the vast majority of

Figure 1. Arteriolar hyalinosis develops in Klotho deficiency. (A) Periodic acid-Schiff (PAS) staining on WT kidney (original magnification 100×), (B) close-up of a normal arteriole (original magnification 400×). (C) Hematoxylin-eosin (HE) staining on WT kidney, with a normal arteriole (original magnification 400×). (D) PAS staining on Klotho+/- kidney (original magnification 100×), (E) close-up of a normal arteriole (original magnification 400×). (F)

HE staining on Klotho+/- kidney, with a normal arteriole (original magnification 400×). (G) PAS staining on Klotho -/- kidney with PAS-positive hyaline lesions throughout the cortex (original magnification 100×), (H) close-up of

arteriole affected by hyalinosis (original magnification 400×). (I) HE staining on Klotho-/- kidney, with arterioles

affected by hyalinosis, displaying a typically eosinophilic and “glassy” aspect (original magnification 400×). Arrows indicate arterioles.

(6)

263

arterioles smaller than 15 µm (including terminal afferent arterioles and efferent arterioles) was generally unaffected (Figure 2D) and renin/PAS double stainings did not reveal co-localization of renin expression and hyalinosis in terminal afferent arterioles (Figure 2C).

Figure 2. Determination of affected segments of the renal vasculature. (A) α-smooth muscle actin (α-SMA)/PAS double staining on Klotho-/- kidney with a large segmental artery, showing severe hyalinosis, with loss of α-SMA

expression in the lesion, spanning the vascular wall. (B) α-SMA/PAS double staining on Klotho-/- kidney with a

terminal interlobular artery and afferent arteriole, showing hyalinosis up until halfway the afferent arteriole. (C) Renin/PAS double staining on Klotho-/- kidney, showing that the terminal afferent arteriole is not affected by

hyalinosis. Original magnifications are 400×. Closed arrows indicate hyalinosis and open arrows indicate unaffected arteries. (D) Histogram of lumen diameters of all renal α-SMA-positive arteries, showing that hyalinous arteries are generally 6-42 µm in diameter, with the smallest arterioles generally being unaffected.

(7)

264

Arteriolar morphology

PAS/CD31 double stainings indicate that the endothelial lining of affected arterioles is generally still intact (Figure 3A, B). Verhoeff stainings indicate that the elastic lamin is intact in unaffected arterioles, but cannot be discerned in affected areas (Figure 3C, D). PAS/α-SMA double stainings indicate that arteries and arterioles affected by hyalinosis lose their smooth muscle cell expression of α-SMA (Figure 3E, F).

Figure 3. Evaluation of morphology of the vascular wall in Klotho deficiency-induced arteriolar hyalinosis. (A) CD31/PAS double staining on Klotho-/- kidney (original magnification 100×), (B) close-up of CD31/PAS staining

showing that in arteries/arterioles both unaffected and affected by hyalinosis, the endothelial lining is intact (original magnification 100x). (C) Verhoeff staining on Klotho-/- kidney (original magnification 100×), (D) close-up

of Verhoeff staining showing that the elastic lamin is intact in unaffected arteries, which cannot be ascertained in arteries affected by hyalinosis (original magnification 100x). (E) α-SMA/PAS double staining on Klotho-/- kidney

(original magnification 100×), (F) close-up of α-SMA/PAS staining showing a normal media in unaffected arteries, whereas the media in arteries affected by hyalinosis mostly consists of accumulated hyaline material, with smooth muscle cells having lost their α-SMA expression (original magnification 100x). Closed arrows indicate hyalinosis and open arrows indicate unaffected arteries.

(8)

265

Hyaline depositions contain plasma proteins

Using immunohistochemistry, we found that hyalinous lesions in Klotho-/- kidneys are positive

for IgM (Figure 4, A-C), and for activated C3 proteins iC3b/c/d (Figure 4, D-F), indicating that the integrity of the endothelial barrier function is locally compromised and that leaked plasma proteins accumulate in the subendothelial space. Vascular positivity for IgM and iC3b/c/d was not found in WT kidneys (Figure 4, A, D).

De-differentiation of smooth muscle cells to a synthetic phenotype

In Klotho-/- mouse arteries, lesional smooth muscle cells were found to have lost expression

of α-SMA, a contractile apparatus protein that is also a marker for differentiated SMCs (Figures 5H, 2A). In turn, lesional SMCs gained expression of S100A4 (Figure 5J), which is considered a marker of a synthetic phenotype in SMCs. Indeed, in Klotho-/- kidneys, we also detected

collagen I, collagen III, and Masson Trichrome positivity, most prominently in

Figure 4. Accumulation of plasma proteins in hyalinous lesions. (A) Immunohistochemistry for IgM on WT kidney, showing interstitial staining, but no IgM in the vascular wall. (B) Immunohistochemistry for IgM on Klotho -/- kidney, showing an artery with immunoreactivity for IgM in the vascular wall. (C) Immunohistochemistry for

IgM on WT mouse spleen, as a positive control. (D) Immunohistochemistry for iC3b/c/d on WT kidney, showing no vascular expression. (E) Immunohistochemistry for iC3b/c/d on Klotho-/- kidney, depicting immunoreactivity

in the vascular wall of an arteriole. (F) Immunohistochemistry for iC3b/c/d on brain-dead mouse kidney, as a positive control. Arrows indicate arteries/arterioles. Original magnifications are 400×.

(9)

266

hyalinised parts of segmental arteries (Figure 5B, D, F), indicating increased deposition of extracellular matrix proteins. WT renal arteries of similar size did not have medial/subendothelial collagen expression (Figure 5A, C, E), loss of α-SMA expression (Figure 5G), or SMC expression of S100A4 (Figure 5I).

Vascular lesions in different Klotho knockout mouse strains

Because not only the development of hyalinosis but also the lack of vascular calcification in our Klotho-/- mice is an unconventional finding, we wanted to assess whether hyalinosis is a

trait also found in other strains of Klotho knockout mice. Therefore, we performed PAS and Von Kossa stainings on kidney sections from kl/kl mice (which have a disrupted promoter), β-actin-Cre/Klothoflox/flox (or: β-actin-KL-/-) mice (which have a deletion of exon 2 of the Klotho

gene in cells that express β-actin, which is ubiquitous), to compare to Klotho-/- mice (which

have a deletion of exon 2 in all cells). We found that kl/kl mouse kidneys almost exclusively display vascular calcification (Figure 6A), whereas β-actin-KL-/- mice exhibit a mix of vascular

calcification and some hyalinosis (Figure 6B), while Klotho-/- mice only have prominent

hyalinosis (Figure 6C). Quantative assessment is depicted in Figure 6G-I. These findings indicate that at least β-actin-KL-/- mice can also develop arteriolar hyalinosis, even though their

phenotype is quite variable, possibly due to slight variations in recombination.

Spironolactone inhibits vascular calcification and allows for its replacement by hyalinosis

Finally, to further examine whether the development of arteriolar hyalinosis is a consequence of Klotho deficiency in general, we wanted to see whether inhibition of vascular calcification in kl/kl would also potentiate the development of arteriolar hyalinosis. We treated kl/kl mice with 80 mg/L spironolactone for 8 weeks, which was previously found to inhibit the development of vascular calcification (44). Kl/kl mice spontaneously displayed severe nephrocalcinosis (86 ± 8% calcified arteries) (Figure 7A, B, F) and very little hyalinosis (4 ± 2%) (Figure 7G). However, spironolactone treatment allowed for the replacement of calcified arteries (51 ± 27%) with hyalinosis (32 ± 23%) (Figure 7C-E, G).

(10)

267

Figure 5. De-differentiation of smooth muscle cells into a synthetic phenotype results in the deposition of extracellular matrix in hyalinous lesions. (A) Masson Trichrome staining on WT mouse kidney, showing part of a normal segmental artery. (B) Masson Trichrome staining on Klotho-/- kidney, showing

(11)

268

(Figure 4. Cont’d.) Klotho-/- kidney, showing an artery with immunoreactivity for IgM in the vascular wall. (C)

Immunohistochemistry for IgM on WT mouse spleen, as a positive control. (D) Immunohistochemistry for iC3b/c/d on WT kidney, showing no vascular expression. (E) Immunohistochemistry for iC3b/c/d on Klotho

-/-kidney, depicting immunoreactivity in the vascular wall of an arteriole. (F) Immunohistochemistry for iC3b/c/d on brain-dead mouse kidney, as a positive control. Arrows indicate arteries/arterioles. Original magnifications are 400×.

(Figure 5. Cont’d.) part of a segmental artery affected by hyalinosis, with increased collagen deposition (blue) at the site of the lesion. (C) Collagen I/PAS double staining on WT mouse kidney, showing part of a normal segmental artery. (D) Collagen I/PAS double staining on Klotho-/- mouse kidney, showing part of a segmental artery affected

by hyalinosis, with locally increased collagen I expression in the affected vessel wall. (E) Collagen III/PAS double staining on WT mouse kidney, showing part of a normal segmental artery. (F) Collagen III/PAS double staining on Klotho-/- mouse kidney, showing part of a segmental artery affected by hyalinosis, with locally increased collagen

III expression in the affected vessel wall. (G) α-SMA/PAS double staining on WT mouse kidney, showing part of a normal segmental artery. (H) α-SMA/PAS double staining on Klotho-/- mouse kidney, showing part of a segmental

artery affected by hyalinosis, with loss of α-SMA positivity in lesional smooth muscle cells. (I) S100A4/PAS double staining on WT mouse kidney, showing part of a normal segmental artery. (J) S100A4/PAS double staining on Klotho-/- mouse kidney, showing part of a segmental artery affected by hyalinosis, with S100A4-positive smooth

muscle cells in the affected area. Original magnifications are 400×.

Figure 6. Assessment of vascular pathologies in different Klotho knockout mouse strains. (A) PAS staining shows prominent vascular calcification in kl/kl mouse kidney. (B) PAS staining shows mild arteriolar hyalinosis in β-actin-KL-/- mouse kidney. (C) PAS staining shows marked arteriolar hyalinosis in Klotho-/- mouse kidney. (D) Von Kossa

staining shows prominent vascular calcification in kl/kl mouse kidney. (E) Von Kossa staining shows no medial calcification in β-actin-KL-/- mouse kidney. (F) Von Kossa staining shows no vascular calcification in Klotho-/- mouse

kidney. (G) Quantification of arteries assessed as calcified in different Klotho-deficient mouse strains, showing marked vascular calcification in kl/kl mice, variable vascular calcification in β- actin-KL-/- mice, and no vascular

calcification in Klotho-/- mice. (H) Quantification of arteries assessed as affected by hyalinosis in different

Klotho-deficient mouse strains, showing virtually no hyalinosis in kl/kl mice, some hyalinosis in β-actin-KL-/- mice, and

prominent presence of hyalinosis in Klotho-/- mice. (I) Quantification of arteries assessed as normal in different

Klotho-deficient strains. Original magnifications are 400×. Arrows indicate arterioles affected by hyalinosis. * p < 0.05, ** p < 0.01, *** p < 0.001.

(12)

269

Figure 7. Inhibition of vascular calcification by spironolactone potentiates the replacement of vascular calcification with hyalinosis. (A) PAS staining on untreated kl/kl mouse kidney, displaying severe nephrocalcinosis throughout the cortex. (B) Von Kossa staining on untreated kl/kl mouse kidney from (A), displaying severe nephrocalcinosis (black). (C) PAS staining on spironolactone-treated kl/kl mouse kidney, exhibiting no vascular (or tubular) calcification, but marked hyalinosis. (D) Von Kossa staining on spironolactone-treated kl/kl mouse kidney from (C), displaying no vascular (or tubular) calcification. Original magnifications 80×; inserts 400×. (E) Correlation between the percentages of arteries assessed as calcified and as hyalinous, showing a linear, negative correlation (p < 0.001, Spearman’s ρ). (F) Quantification of the arteries assessed as calcified on both PAS and Von Kossa staining in untreated and spironolactone-treated kl/kl mouse kidneys. (G) Quantification of the arteries assessed as affected by hyalinosis on both PAS and Von Kossa staining in untreated and spironolactone-treated kl/kl mouse kidneys. *** p < 0.001.

(13)

270

Figure 8. Paradigm of the effect of Klotho on smooth muscle cell phenotypic transitions. Klotho inhibits smooth muscle cell (SMC) de-differentiation from a contractile phenotype to either a calcifying or a synthetic phenotype. Depending on the stimuli that SMCs are exposed to, de-differentiation can go in either direction. High phosphate and calcium levels, and cellular senescence promote the development of vascular calcification, whereas subendothelial leakage of plasma proteins will foster the development of hyalinosis. In the absence of sufficiently strong pro-calcific stimuli (which can be either before these stimuli properly develop, in the context of greater resistance against these stimuli, or upon inhibition of calcification, like by spironolactone), SMCs respond to endothelial hyperpermeability in contributing to the development of arteriolar hyalinosis.

(14)

271

Discussion

The key finding of this study is the observation that Klotho deficiency induces arteriolar hyalinosis. The emergence of an ageing-related vascular lesion as part of the phenotype of Klotho deficiency compounds the hypothesis that Klotho exerts anti-ageing effects. Given the prevalent view that arteriolar hyalinosis, particularly in afferent arterioles, leads to the loss of renal autoregulation and contributes to the ageing-related loss of renal function (7, 8), the potential involvement of Klotho is of particular interest to the ageing population. The ageing-related decrease in Klotho expression may be causally involved in this process, and considering the protective effects Klotho has been shown to have experimentally on renal function (10, 14, 17, 22), increasing Klotho levels may prove to be a promising approach to the preservation of renal function during ageing.

We have shown that Klotho deficiency allows for plasma proteins to accumulate in the subendothelial space in hyaline lesions, attesting to the poor barrier function of the endothelium. Indeed, endothelial hyperpermeability (36) and endothelial dysfunction (37-39)

have been shown before to develop in the absence of Klotho. It is therefore clear that adequate Klotho levels are paramount to endothelial integrity. However, more interesting in our study is the observed plasticity of the smooth muscle cell phenotype in Klotho deficiency, considering the previously reported direct in vitro effects of Klotho on SMC differentiation

(22). The Klotho-/- mouse is well-studied with respect to its development of severe vascular

calcification (45-49), in particular of the media, as occurs in chronic kidney disease (CKD) or in ageing. However, we found that, likely due to differences in genetic background, diet, and other environmental factors, Klotho-deficient mice display a certain phenotypic variability. Klotho-/- mice had not developed vascular calcification in the kidney at the age of 7 weeks. We

hypothesize that the accumulated pro-calcific triggers were not potent enough to induce vascular calcification, which in turn allowed for the development of arteriolar hyalinosis instead. The Klotho-/- mice have a mixed genetic background, but one which is mostly C57BL/6,

which is generally more resistant to the development of vascular calcification. To address this phenomenon, we also studied kl/kl mice (on a mixed background which was mostly 129/Sv) that displayed severe vascular calcification at 7-8 weeks of age, and β-actin-KL-/- mice (also on

a mixed background predominantly C57BL/6 (50)). The observation that β-actin-KL-/- mice

spontaneously display some arteriolar hyalinosis and that kl/kl mice do so after treatment with spironolactone, which inhibited the development of calcification by blocking VSMC-derived aldosterone inducing calcification via a Pit1-dependent mechanism (44, 51), allows us to speculate that the development of hyalinosis is a function of the degree to which vascular calcification develops, which can be influenced by the genetic background, as well as by dietary factors. The development of arteriolar hyalinosis in lieu of vascular calcification in spironolactone-treated mice lets us conclude that in the absence of sufficiently strong pro-calcific stimulus smooth muscle cells de-differentiate to a more synthetic phenotype. We

(15)

272

hypothesize that Klotho inhibits pathological de-differentiation of smooth muscle cells and the consequently assumed phenotype is the net result of the stimuli to which the SMCs are exposed. In the case of hyalinosis, it is thought that the subendothelial presence of accumulated plasma proteins triggers the SMC response of de-differentiation and synthesis of ECM proteins (52, 53). This concept is schematically depicted in Figure 8.

To expand on the topic of phenotypic variability in Klotho deficiency, it has long been known that different Klotho levels induce different vascular phenotypes. Klotho-/- mice display severe

vascular calcification (9), whereas Klotho+/- mice do not do so spontaneously, although they

are more prone to the development of vascular calcification upon induction of CKD (22). While Klotho+/- mice develop endothelial dysfunction, vascular function in full knockouts has proven

difficult to evaluate due to severe calcification (37-39). Klotho+/- mice develop arterial

stiffening after 14 weeks of age with increased aortic collagen deposition and elastin degradation (40-42), which probably does not develop in Klotho-/- mice due to the dominance

of the calcification phenotype. Of note, arteriolar hyalinosis and arterial stiffening have in common the aberrant SMC behaviour of excessive ECM deposition. Naturally, the short lifespan of full knockout mice also limits the window for the development of other pathologies. The same probably holds true for the development of hypertension from 15-16 weeks of age onward in Klotho+/- mice (33, 40, 43), while Klotho-/- mice are generally

hypotensive due to hypovolemia (44, 54, 55). Our study indicates that a similar example of phenotypic divergence is the uncovering of arteriolar hyalinosis if the development of calcification is mildly delayed and/or inhibited. The comparison of these phenotypes between different Klotho knockout genotypes under different conditions may shed light on the development of vascular disease in patients. The rapid and severe development of vascular calcification in kl/kl mice may make them a suitable model for vascular pathologies in CKD patients, who are also more severely Klotho-deficient than the general ageing population. Heterozygote mice may constitute a suitable model for general ageing or other patients with intermediate Klotho levels, displaying milder pathologies like arterial stiffening, endothelial dysfunction, and hypertension. Our study suggests that even in ageing patients in which vascular calcification in the kidney plays a limited role, Klotho deficiency may still contribute to the development of arteriolar hyalinosis and the subsequent decline in renal function. The development of hyalinosis has not been widely studied in animal models, but calcineurin appears to be a key modulator. Calcineurin inhibitors (CNI) like tacrolimus and cyclosporin A, genetic deletion of calcineurin α (56), and endothelial deletion of FK-506-binding protein 12 (FKBP12, to which tacrolimus binds) (57) readily induce hyalinosis. This effect is thought to be mostly mediated by TGFβ1 signaling, considering the evidence that anti-TGFβ1 antibodies mitigated hyalinization (58) and deletion of FKPB12 constitutively activates TGFβRI and downstream Smad2/3 signaling (57). Given that Klotho inhibits TGFβ1 signaling by binding to TGFβRII and decreasing its affinity for TGFβ1 (15), leading to less activation of TGFβRI, it appears that Klotho may act upstream of CNI-induced hyalinosis. This leads to the hypothesis that Klotho may be able to mitigate the TGFβ1-induced side effects of CNI use. Of note are

(16)

273

also the down-regulation of Klotho in CsA nephropathy (59-63), producing functional Klotho deficiency, and the general renoprotective and anti-inflammatory effects of Klotho overexpression in CsA nephropathy (63, 64). Investigation of Klotho as a therapeutic target in inhibiting development of chronic graft dysfunction in renal transplantation patients may therefore also be warranted.

In short, arteriolar hyalinosis appears to be an important feature of the Klotho deficiency phenotype, which is likely generally masked by the development of vascular calcification and is uncovered upon inhibition of or resistance against vascular calcification. The finding that Klotho deficiency induces arteriolar hyalinosis raises new questions and hypotheses on the effects of Klotho on endothelial integrity and smooth muscle cell de-differentiation, on the role of Klotho in ageing and in ageing-related renal function decline, and on the potential of Klotho in ageing and in CNI nephrotoxicity.

Materials and Methods

Experimental animals

All animal experiments were conducted in accordance with the NIH guideline on care and use of laboratory animals. Klotho-/- mice were housed at the central animal facility of the

University Medical Center Groningen. Klotho-/- mice were housed in individually ventilated

cages with abundant nesting material and WT or Klotho+/- buddies to prevent hypothermia.

Klotho-/- mice were provided with wetted or soaked food and drinking water (with long

drinking nipples to ensure easy accessibility) ad libitum. They were monitored daily. At the age of 7 weeks, mice were sacrificed under deep isoflurane anaesthesia by cardiac puncture. Kidneys were collected and snap-frozen in liquid nitrogen or fixed in formalin and embedded in paraffin. Klotho-/- (N=3), Klotho+/- (N=4), and WT (N=3) mice were analysed for the presence

of arteriolar hyalinosis. Kl/kl mice were housed at the Institute for Physiology at the University of Tübingen and were not treated (N=13) or treated with spironolactone (80 mg/L; N=10) in drinking water for 8 weeks. Kidney tissue was used from β-actin-Cre/Klothoflox/flox mice bred

as previously described (50).

Histochemistry

Paraffin sections (4 µm) were cut, de-paraffinized in xylene and re-hydrated in a graded ethanol series. Periodic acid-Schiff staining was performed by incubating sections in 1% periodic acid for 10 minutes and in Schiff’s reagent for 15 minutes (or for 5 minutes as a mild counterstaining after immunohistochemistry with DAB), followed by hematoxylin

(17)

274

counterstaining. Von Kossa stainings were performed by incubating sections in 1% silver nitrate for 1 hour under sunlight exposure, followed by incubation in 3% sodium thiosulfate for 5 minutes and counterstaining with nuclear fast red. Masson Trichrome stainings were performed by incubating sections for 5 minutes in celestine blue, 5 minutes in hematoxylin, 5 minutes in a 1:2 mixture of 1% acid fuchsin and 1% xylidine ponceau, 5 minutes in 1% phosphomolybdic acid, and 1 minute in 1% aniline blue. Verhoeff stainings were performed by incubating sections for 15 minutes in Verhoeff staining solution (5:2:2 mixture of 5% alcoholic hematoxylin, 10% ferric chloride, and 2% potassium iodide/1% iodine), followed by destaining 2% ferric chloride and staining with Van Gieson’s solution. Oil red O staining was performed on cryosections, which were fixed for 10 minutes in 8% formalin, dipped twice in 60% 2-propanol, followed by incubation for 10 minutes in Oil red O solution, de-staining in 60% 2-propanol, and counterstaining in hematoxylin.

Immunohistochemistry

Paraffin sections (4 µm) were cut, de-paraffinzed in xylene and re-hydrated in a graded ethanol series. Antigen retrieval was performed by heating sections at 500 W for 15 minutes in 10 mM citric acid (pH 6) for α-SMA, CD31, and collagen I, in 170 mM Tris/1 mM EDTA (pH 9) for renin and S100A4, and in 1 mM EDTA (pH 8) for collagen III. After blocking endogenous peroxidase (0.3% H2O2/PBS) for 30 minutes, sections were incubated with primary antibodies

(α-SMA: 1:300, mouse monoclonal 1A4, Dako, Denmark; CD31: 1:50, SZ31, Dianova, Germany; collagen I: 1:100, 1310-01, Southern Biotech, USA; collagen III: 1:75, 1330-01, Southern Biotech; renin: 1:2500, polyclonal antibody kindly provided by Dr. T Inagami (Vanderbilt University School of Medicine, Nashville, USA);S100A4: 1:2000, A5114, Dako) for 1 hour in 1% BSA/PBS. If necessary, avidin and biotin blocking solutions were applied (Vector Labs, USA). Sections were incubated with the appropriate secondary and tertiary antibodies for 30 minutes (goat anti-mouse-biotin, rabbit anti-rat-HRP, goat anti-rabbit-HRP, rabbit anti-goat-HRP, Dako) and the chromogenic reaction was performed using DAB in 0.03% H2O2/PBS,

followed by hematoxylin counterstaining. IgM and iC3b/c/d stainings were performed on 4 µm frozen sections, which were dried, fixed in acetone for 10 minutes at RT, followed by endogenous peroxidase blocking in 0.075% H2O2/PBS for 30 minutes and incubation with

primary antibodies for 1 hour (iC3b/c/d: 1:20, HM1065, Hycult Biotech, USA; IgM-HRP: 1:100, 1020-05, Southern Biotech). Then, the appropriate secondary and tertiary antibodies were applied for 30 minutes (goat anti-mouse-HRP, rabbit anti-goat-HRP, goat anti-rabbit-HRP, Dako) and the chromogenic reaction was performed with DAB or AEC. Nuclei were counterstained with hematoxylin. All slides were scanned using a Hamamatsu Nanozoomer 2.0HT (Hamamatsu Photonics, Hamamatsu, Japan) and scans were analysed using Aperio ImageScope software (Leica Biosystems, Germany).

(18)

275

Statistical analysis

Normally distributed data are presented as mean ± SD and normally distributed data are presented as median [interquartile range]. Differences between groups were tested with ANOVA followed by Bonferroni’s post-hoc correction, or Student’s t test, or by a Kruskal-Wallis test followed by Dunn’s post-hoc correction, or Mann-Whitney U test, after prior Kolmogorov-Smirnov testing for normality. Spearman’s ρ was used for correlation analysis. A p value < 0.05 was considered statistically significant. All data analysis was performed using SPSS version 23 (IBM, USA) and GraphPad Prism version 5 (GraphPad, USA).

(19)

276

Acknowledgments

We would like to thank Wierd Kooistra and Marian Reinders for technical support. 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), Piet ter Wee and Marc Vervloet (VU University Medical Center, Amsterdam), and Gerjan Navis, Martin de Borst, and Jan-Luuk Hillebrands (University Medical Center Groningen). This study was also funded by the University Medical Center Groningen MD/PhD program.

(20)

277

References

1. A. R. Moritz, M. R. Oldt, Arteriolar Sclerosis in Hypertensive and Non-Hypertensive Individuals. Am. J. Pathol. 13, 679-728.7 (1937).

2. A. C. Matos, N. O. Camara, L. R. REQUIaO-Moura, E. J. Tonato, T. C. Filiponi, M. Souza-DURaO Jr, D. M. Malheiros, M. Fregonesi, M. Borrelli, A. Pacheco-Silva, Presence of arteriolar hyalinosis in post-reperfusion biopsies represents an additional risk to ischaemic injury in renal transplant. Nephrology (Carlton). 21, 923-929 (2016).

3. N. Kambham, S. Nagarajan, S. Shah, L. Li, O. Salvatierra, M. M. Sarwal, A novel, semiquantitative, clinically correlated calcineurin inhibitor toxicity score for renal allograft biopsies. Clin. J. Am. Soc. Nephrol. 2, 135-142 (2007).

4. S. Sund, A. V. Reisaeter, H. Scott, P. Fauchald, O. Bentdal, G. Sodal, T. Hovig, Morphological studies of baseline needle biopsies from living donor kidneys: light microscopic, immunohistochemical and ultrastructural findings. APMIS. 106, 1017-1034 (1998).

5. C. N. Gamble, The pathogenesis of hyaline arteriolosclerosis. Am. J. Pathol. 122, 410-420 (1986).

6. J. J. Morrissey, Direct or indirect endothelial cell transforming growth factor-beta receptor activation initiates arteriolar hyalinosis. Kidney Int. 82, 838-839 (2012).

7. G. S. Hill, D. Heudes, J. Bariety, Morphometric study of arterioles and glomeruli in the aging kidney suggests focal loss of autoregulation. Kidney Int. 63, 1027-1036 (2003).

8. B. F. Palmer, Renal dysfunction complicating the treatment of hypertension. N. Engl. J. Med. 347, 1256-1261 (2002).

9. 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).

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. L. Zhou, H. Mo, J. Miao, D. Zhou, R. J. Tan, F. F. Hou, Y. Liu, Klotho Ameliorates Kidney Injury and Fibrosis and Normalizes Blood Pressure by Targeting the Renin-Angiotensin System. Am. J. Pathol. 185, 3211-3223 (2015). 12. L. Zhou, Y. Li, D. Zhou, R. J. Tan, Y. Liu, Loss of Klotho contributes to kidney injury by derepression of Wnt/beta-catenin signaling. J. Am. Soc. Nephrol. 24, 771-785 (2013).

13. L. Zhou, Y. Li, W. He, D. Zhou, R. J. Tan, J. Nie, F. F. Hou, Y. Liu, Mutual antagonism of Wilms' tumor 1 and beta-catenin dictates podocyte health and disease. J. Am. Soc. Nephrol. 26, 677-691 (2015).

14. 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).

(21)

278

15. S. Doi, Y. Zou, O. Togao, J. V. Pastor, G. B. John, L. Wang, K. Shiizaki, R. Gotschall, S. Schiavi, N. Yorioka, M. Takahashi, D. A. Boothman, M. Kuro-o, Klotho inhibits transforming growth factor-beta1 (TGF-beta1) signaling and suppresses renal fibrosis and cancer metastasis in mice. J. Biol. Chem. 286, 8655-8665 (2011).

16. Y. Haruna, N. Kashihara, M. Satoh, N. Tomita, T. Namikoshi, T. Sasaki, T. Fujimori, P. Xie, Y. S. Kanwar, Amelioration of progressive renal injury by genetic manipulation of Klotho gene. Proc. Natl. Acad. Sci. U. S. A. 104, 2331-2336 (2007).

17. M. C. Panesso, M. Shi, H. J. Cho, J. Paek, J. Ye, O. W. Moe, M. C. Hu, Klotho has dual protective effects on cisplatin-induced acute kidney injury. Kidney Int. 85, 855-870 (2014).

18. 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).

19. X. Guan, L. Nie, T. He, K. Yang, T. Xiao, S. Wang, Y. Huang, J. Zhang, J. Wang, K. Sharma, Y. Liu, J. Zhao, Klotho suppresses renal tubulo-interstitial fibrosis by controlling basic fibroblast growth factor-2 signalling. J. Pathol. 234, 560-572 (2014).

20. J. H. Kim, J. Xie, K. H. Hwang, Y. L. Wu, N. Oliver, M. Eom, K. S. Park, N. Barrezueta, I. D. Kong, R. P. Fracasso, C. L. Huang, S. K. Cha, Klotho May Ameliorate Proteinuria by Targeting TRPC6 Channels in Podocytes. J. Am. Soc. Nephrol. 28, 140-151 (2017).

21. Y. L. Wu, J. Xie, S. W. An, N. Oliver, N. X. Barrezueta, M. H. Lin, L. Birnbaumer, C. L. Huang, Inhibition of TRPC6 channels ameliorates renal fibrosis and contributes to renal protection by soluble klotho. Kidney Int. 91, 830-841 (2017).

22. 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).

23. Y. Saito, T. Nakamura, Y. Ohyama, T. Suzuki, A. Iida, T. Shiraki-Iida, M. Kuro-o, Y. Nabeshima, M. Kurabayashi, R. Nagai, In vivo klotho gene delivery protects against endothelial dysfunction in multiple risk factor syndrome. Biochem. Biophys. Res. Commun. 276, 767-772 (2000).

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

25. K. Yang, C. Du, X. Wang, F. Li, Y. Xu, S. Wang, S. Chen, F. Chen, M. Shen, M. Chen, M. Hu, T. He, Y. Su, J. Wang, J. Zhao, Indoxyl sulfate induces platelet hyperactivity and contributes to chronic kidney disease-associated thrombosis in mice. Blood. 129, 2667-2679 (2017).

26. W. Chen, F. Wang, W. Zeng, J. Sun, L. Li, M. Yang, J. Sun, Y. Wu, X. Zhao, C. Zhu, Regulation of Cellular Response Pattern to Phosphorus Ion is a New Target for the Design of Tissue-Engineered Blood Vessel. Adv. Healthc. Mater. 4, 1004-1008 (2015).

27. 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).

(22)

279

28. 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). 29. M. C. Hu, M. Shi, J. Zhang, J. Pastor, T. Nakatani, B. Lanske, M. S. Razzaque, K. P. Rosenblatt, M. G. Baum, M. Kuro-o, O. W. Moe, Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. FASEB J. 24, 3438-3450 (2010).

30. S. K. Cha, B. Ortega, H. Kurosu, K. P. Rosenblatt, M. Kuro-O, C. L. Huang, Removal of sialic acid involving Klotho causes cell-surface retention of TRPV5 channel via binding to galectin-1. Proc. Natl. Acad. Sci. U. S. A. 105, 9805-9810 (2008).

31. Q. Chang, S. Hoefs, A. W. van der Kemp, C. N. Topala, R. J. Bindels, J. G. Hoenderop, The beta-glucuronidase klotho hydrolyzes and activates the TRPV5 channel. Science. 310, 490-493 (2005).

32. R. T. Alexander, T. E. Woudenberg-Vrenken, J. Buurman, H. Dijkman, B. C. van der Eerden, J. P. van Leeuwen, R. J. Bindels, J. G. Hoenderop, Klotho prevents renal calcium loss. J. Am. Soc. Nephrol. 20, 2371-2379 (2009). 33. 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).

34. H. Sugiura, T. Yoshida, S. Shiohira, J. Kohei, M. Mitobe, H. Kurosu, M. Kuro-o, K. Nitta, K. Tsuchiya, Reduced Klotho expression level in kidney aggravates renal interstitial fibrosis. Am. J. Physiol. Renal Physiol. 302, F1252-64 (2012).

35. Y. Lin, M. Kuro-o, Z. Sun, Genetic deficiency of anti-aging gene klotho exacerbates early nephropathy in STZ-induced diabetes in male mice. Endocrinology. 154, 3855-3863 (2013).

36. 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. U. S. A. 107, 19308-19313 (2010).

37. 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).

38. R. Nagai, Y. Saito, Y. Ohyama, H. Aizawa, T. Suga, T. Nakamura, M. Kurabayashi, M. Kuroo, Endothelial dysfunction in the klotho mouse and downregulation of klotho gene expression in various animal models of vascular and metabolic diseases. Cell Mol. Life Sci. 57, 738-746 (2000).

39. 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). 40. 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).

41. 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).

(23)

280

42. 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).

43. 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).

44. 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).

45. T. Nakatani, M. Ohnishi, M. S. Razzaque, Inactivation of klotho function induces hyperphosphatemia even in presence of high serum fibroblast growth factor 23 levels in a genetically engineered hypophosphatemic (Hyp) mouse model. FASEB J. 23, 3702-3711 (2009).

46. T. Nakatani, B. Sarraj, M. Ohnishi, M. J. Densmore, T. Taguchi, R. Goetz, M. Mohammadi, B. Lanske, M. S. Razzaque, In vivo genetic evidence for klotho-dependent, fibroblast growth factor 23 (Fgf23) -mediated regulation of systemic phosphate homeostasis. FASEB J. 23, 433-441 (2009).

47. M. Ohnishi, T. Nakatani, B. Lanske, M. S. Razzaque, In vivo genetic evidence for suppressing vascular and soft-tissue calcification through the reduction of serum phosphate levels, even in the presence of high serum calcium and 1,25-dihydroxyvitamin d levels. Circ. Cardiovasc. Genet. 2, 583-590 (2009).

48. M. Ohnishi, T. Nakatani, B. Lanske, M. S. Razzaque, Reversal of mineral ion homeostasis and soft-tissue calcification of klotho knockout mice by deletion of vitamin D 1alpha-hydroxylase. Kidney Int. 75, 1166-1172 (2009).

49. M. Ohnishi, M. S. Razzaque, Dietary and genetic evidence for phosphate toxicity accelerating mammalian aging. FASEB J. 24, 3562-3571 (2010).

50. H. Olauson, K. Lindberg, R. Amin, T. Jia, A. Wernerson, G. Andersson, T. E. Larsson, Targeted deletion of klotho in kidney distal tubule disrupts mineral metabolism. J. Am. Soc. Nephrol. 23, 1641-1651 (2012).

51. I. Alesutan, J. Voelkl, M. Feger, D. V. Kratschmar, T. Castor, S. Mia, M. Sacherer, R. Viereck, O. Borst, C. Leibrock, M. Gawaz, M. Kuro-O, S. Pilz, A. Tomaschitz, A. Odermatt, B. Pieske, C. A. Wagner, F. Lang, Involvement Of Vascular Aldosterone Synthase In Phosphate-Induced Osteogenic Transformation Of Vascular Smooth Muscle Cells. Sci. Rep. 7, 2059-017-01882-2 (2017).

52. C. L. Bockmeyer, V. Forstmeier, F. Modde, S. Lovric, R. A. Claus, M. Schiffer, P. A. Agustian, C. Grothusen, K. Grote, I. Birschmann, K. Theophile, H. H. Kreipe, V. Brocker, J. U. Becker, ADAMTS13--marker of contractile phenotype of arterial smooth muscle cells lost in benign nephrosclerosis. Nephrol. Dial. Transplant. 26, 1871-1881 (2011).

53. C. L. Bockmeyer, D. S. Kern, V. Forstmeier, S. Lovric, F. Modde, P. A. Agustian, S. Steffens, I. Birschmann, J. Traeder, M. E. Dammrich, A. Schwarz, H. H. Kreipe, V. Brocker, J. U. Becker, Arteriolar vascular smooth muscle cell differentiation in benign nephrosclerosis. Nephrol. Dial. Transplant. 27, 3493-3501 (2012).

(24)

281

54. S. S. Fischer, D. S. Kempe, C. B. Leibrock, R. Rexhepaj, B. Siraskar, K. M. Boini, T. F. Ackermann, M. Foller, B. Hocher, K. P. Rosenblatt, M. Kuro-O, F. Lang, Hyperaldosteronism in Klotho-deficient mice. Am. J. Physiol. Renal Physiol. 299, F1171-7 (2010).

55. O. Andrukhova, S. Slavic, A. Smorodchenko, U. Zeitz, V. Shalhoub, B. Lanske, E. E. Pohl, R. G. Erben, FGF23 regulates renal sodium handling and blood pressure. EMBO Mol. Med. 6, 744-759 (2014).

56. J. L. Gooch, B. R. Roberts, S. L. Cobbs, J. A. Tumlin, Loss of the alpha-isoform of calcineurin is sufficient to induce nephrotoxicity and altered expression of transforming growth factor-beta. Transplantation. 83, 439-447 (2007).

57. V. L. Chiasson, K. A. Jones, S. E. Kopriva, A. Mahajan, K. J. Young, B. M. Mitchell, Endothelial cell transforming growth factor-beta receptor activation causes tacrolimus-induced renal arteriolar hyalinosis. Kidney Int. 82, 857-866 (2012).

58. M. Islam, J. F. Burke Jr, T. A. McGowan, Y. Zhu, S. R. Dunn, P. McCue, J. Kanalas, K. Sharma, Effect of anti-transforming growth factor-beta antibodies in cyclosporine-induced renal dysfunction. Kidney Int. 59, 498-506 (2001).

59. D. H. Han, S. G. Piao, J. H. Song, J. Y. Ghee, H. S. Hwang, B. S. Choi, J. Kim, C. W. Yang, Effect of sirolimus on calcineurin inhibitor-induced nephrotoxicity using renal expression of KLOTHO, an antiaging gene. Transplantation. 90, 135-141 (2010).

60. S. G. Piao, S. H. Kang, S. W. Lim, B. H. Chung, K. C. Doh, S. B. Heo, L. Jin, C. Li, C. W. Yang, Influence of N-acetylcysteine on Klotho expression and its signaling pathway in experimental model of chronic cyclosporine nephropathy in mice. Transplantation. 96, 146-153 (2013).

61. H. E. Yoon, J. Y. Ghee, S. Piao, J. H. Song, D. H. Han, S. Kim, N. Ohashi, H. Kobori, M. Kuro-o, C. W. Yang, Angiotensin II blockade upregulates the expression of Klotho, the anti-ageing gene, in an experimental model of chronic cyclosporine nephropathy. Nephrol. Dial. Transplant. 26, 800-813 (2011).

62. H. E. Yoon, S. W. Lim, S. G. Piao, J. H. Song, J. Kim, C. W. Yang, Statin upregulates the expression of klotho, an anti-aging gene, in experimental cyclosporine nephropathy. Nephron Exp. Nephrol. 120, e123-33 (2012). 63. M. Jin, P. Lv, G. Chen, P. Wang, Z. Zuo, L. Ren, J. Bi, C. W. Yang, X. Mei, D. Han, Klotho ameliorates cyclosporine A-induced nephropathy via PDLIM2/NF-kB p65 signaling pathway. Biochem. Biophys. Res. Commun. Klotho ameliorates cyclosporine A-induced nephropathy via PDLIM2/NF-kB p65 signaling pathway 486, 451-457 (2017). 64. Q. F. Liu, J. M. Ye, L. X. Yu, X. H. Dong, J. H. Feng, Y. Xiong, X. X. Gu, S. S. Li, Klotho mitigates cyclosporine A (CsA)-induced epithelial-mesenchymal transition (EMT) and renal fibrosis in rats. Int. Urol. Nephrol. 49, 345-352 (2017).

(25)

282

Supplemental figure 1. Composition of hyalinous lesions. (A) PAS staining on Klotho-/- kidney showing arteriolar

hyalinosis, (B) Von Kossa staining on the same arteriole, showing no calcification. (C) Positive control for the Von Kossa staining (human placenta). (D) Oil red O staining on Klotho-/- kidney, showing arteriolar hyalinosis, without

a fatty component. (E) Positive control for the Oil red O staining (human carotid artery plaque). Arrows indicate hyalinous lesions. Mouse images were taken at original magnifications of 400×. Human images were taken at original magnifications of 100×.

(26)
(27)

Referenties

GERELATEERDE DOCUMENTEN

We confirmed the expression of both membrane-bound and alternative Klotho mRNA transcripts in human kidney, primary human renal tubular epithelial cells (RTECs), and a human

(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

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