• No results found

Inhibition of tyrosine kinase receptor signaling attenuates fibrogenesis in an ex vivo model of human renal fibrosis

N/A
N/A
Protected

Academic year: 2021

Share "Inhibition of tyrosine kinase receptor signaling attenuates fibrogenesis in an ex vivo model of human renal fibrosis"

Copied!
42
0
0

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

Hele tekst

(1)

University of Groningen

Inhibition of tyrosine kinase receptor signaling attenuates fibrogenesis in an ex vivo model of

human renal fibrosis

Bigaeva, Emilia; Stribos, Elisabeth G. D.; Mutsaers, Henricus A. M.; Piersma, Bram; Leliveld,

Anna M.; de Jong, Igle J.; Bank, Ruud A.; Seelen, Marc A.; van Goor, Harry; Wollin, Lutz

Published in:

American journal of physiology-Renal physiology

DOI:

10.1152/ajprenal.00108.2019

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

Final author's version (accepted by publisher, after peer review)

Publication date: 2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Bigaeva, E., Stribos, E. G. D., Mutsaers, H. A. M., Piersma, B., Leliveld, A. M., de Jong, I. J., Bank, R. A., Seelen, M. A., van Goor, H., Wollin, L., Olinga, P., & Boersema, M. (2020). Inhibition of tyrosine kinase receptor signaling attenuates fibrogenesis in an ex vivo model of human renal fibrosis. American journal of physiology-Renal physiology, 318(1), F117-F134. https://doi.org/10.1152/ajprenal.00108.2019

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)

Inhibition of tyrosine kinase receptor signaling attenuates

1

fibrogenesis in an ex vivo model of human renal fibrosis

2 3

Emilia Bigaeva a,1, Elisabeth G.D. Stribos a,b,1, Henricus A.M. Mutsaers a,c, Bram Piersmae, Anna M.

4

Leliveld d, Igle J. de Jong d, Ruud A. Bank e, Marc A. Seelen b, Harry van Goor e, Lutz Wollin f, Peter

5

Olinga a,*, Miriam Boersema a.

6 7

a Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen

8

Research Institute of Pharmacy, the Netherlands 9

b Department of Internal Medicine, Division of Nephrology, University of Groningen, University

10

Medical Center Groningen, the Netherlands 11

c Department of Clinical Medicine, Aarhus University, Denmark

12

d Department of Urology, University of Groningen, University Medical Center Groningen, the

13

Netherlands 14

e Department of Pathology and Medical Biology, University of Groningen, University Medical Center

15

Groningen, the Netherlands 16

f Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany

17 18

1 These authors contributed equally to this work

19

*Corresponding author: 20

Prof. Peter Olinga, Department of Pharmaceutical Technology and Biopharmacy, University of 21

Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands. Tel: +31 50 363 8373. E-22

mail: p.olinga@rug.nl 23

24

Supplemental Material available at 25

URL: https://figshare.com/s/87cd11d7a729a236a8c6 26

DOI: https://doi.org/10.6084/m9.figshare.9999557 27

(3)

ABSTRACT 28

Poor translation from animal studies to human clinical trials is one of the main hurdles in the 29

development of new drugs. Here, we used precision-cut kidney slices (PCKS) as a translational model 30

to study renal fibrosis and to investigate whether inhibition of tyrosine kinase receptors, with the 31

selective inhibitor nintedanib, can halt fibrosis in murine and human PCKS. We used renal tissue of 32

murine and human origin to obtain PCKS. Control slices and slices treated with nintedanib were 33

studied to assess viability, activation of tyrosine kinase receptors, cell proliferation, collagen type I 34

accumulation, gene and protein regulation. During culture, PCKS spontaneously develop a fibrotic 35

response that resembles in vivo fibrogenesis. Nintedanib blocked culture-induced phosphorylation of 36

platelet-derived growth factor receptor and vascular endothelial growth factor receptor. Furthermore, 37

nintedanib inhibited cell proliferation, reduced collagen type I accumulation and expression of 38

fibrosis-related genes in healthy murine and human PCKS. Modulation of extracellular matrix 39

homeostasis was achieved already at 0.1 μM, while high concentrations (1 and 5 μM) elicited possible 40

non-selective effects. In PCKS from human diseased renal tissue, nintedanib showed limited capacity 41

to reverse established fibrosis. In conclusion, nintedanib attenuated the onset of fibrosis in both 42

murine and human PCKS by inhibiting the phosphorylation of tyrosine kinase receptors; however, the 43

reversal of established fibrosis was not achieved. 44

45

KEYWORDS 46

Renal fibrosis; precision-cut kidney slices; tyrosine kinase receptor; nintedanib 47

48 49 50

(4)

INTRODUCTION 51

Renal fibrosis, defined by the progressive deposition of connective tissue, is a hallmark of chronic 52

kidney disease (CKD), which affects an estimated 10% of the population in developed countries (19). 53

CKD progresses to end-stage renal disease (ESRD), that eventually requires replacement therapy — 54

dialysis or transplantation. Current research investigates strategies to halt CKD progression, or even 55

to reverse renal fibrosis (6, 37). Yet, no effective therapy has been clinically implemented. 56

Pathological activation of various receptor tyrosine kinases (RTKs) — such as platelet-derived growth 57

factor (PDGF), fibroblast growth factor (FGF) and epidermal growth factor (EGF) receptors — plays 58

a key role in renal fibrogenesis (3, 5, 29, 36, 44). The PDGF receptor (PDGFR) is an attractive 59

molecular target for antifibrotic therapies (8), since PDGFR signaling is involved in 60

(trans)differentiation of collagen-producing myofibroblasts (7, 9, 18). The receptors PDGFRα and β 61

are expressed in renal tissue mainly by glomerular mesangial cells, interstitial fibroblasts and vascular 62

smooth-muscle cells (4). Several studies reported an increased expression of both receptors in murine 63

and human renal disease (7, 12). The activation of the PDGFR leads to glomerulosclerosis and 64

(tubulo)interstitial fibrosis (29). Therefore, blocking PDGFR signaling is a promising strategy to halt 65

the progression of renal fibrosis. 66

Nintedanib is a small molecule tyrosine kinase inhibitor, approved in several countries worldwide for 67

the treatment of idiopathic pulmonary fibrosis (IPF) and for the second line treatment of non-small-68

cell lung carcinoma with adenocarcinoma histology. Nintedanib affects signaling pathways of 69

multiple growth factors, including vascular endothelial growth factor (VEGF), FGF and PDGF, as 70

well as Lck and Src non-receptor kinases (32, 41). In a phase II randomized clinical trial, nintedanib 71

showed anti-angiogenic effects and had an acceptable safety profile in patients with advanced renal 72

cell carcinoma (10). To our knowledge, the impact of nintedanib on human renal fibrosis has not been 73

published. 74

The lack of translational models of human renal fibrosis hampers the search for effective antifibrotic 75

therapies (33). In vitro models lack cellular heterogeneity, and animal models have limited 76

implications for human disease. To partly overcome these limitations, we used precision-cut kidney 77

slices (PCKS) as an ex vivo model of renal fibrosis (13, 34, 43). PCKS replicate the organotypic 78

(5)

multicellular characteristics, as one slice maintains the complex three-dimensional architecture of the 79

kidney, and have a high translational impact, as both murine and human tissue, healthy and diseased, 80

can be used. In addition, PCKS culture is reproducible and allows for a substantial reduction of 81

animal use, making it a promising preclinical tool for drug development. 82

In this study, we aimed to investigate therapeutic effects of nintedanib in PCKS, and to find whether 83

inhibition of nintedanib’s molecular targets may prevent renal fibrosis in murine and, more 84

importantly, in human kidneys. 85

(6)

MATERIALS AND METHODS 87

Ethics statement 88

This study was approved by the Medical Ethical Committee of the University Medical Centre 89

Groningen (UMCG), according to Dutch legislation and the Code of Conduct for dealing responsibly 90

with human tissue in the context of health research (www.federa.org), forgoing the need of written 91

consent for ‘further use’ of coded-anonymous human tissue. 92

The animal experiments were approved by the Animal Ethics Committee of the University of 93

Groningen (DEC 6416AA-001). 94

95

Renal tissue 96

Macroscopically healthy renal cortical tissue (n=9) was obtained from tumor nephrectomies, and 97

fibrotic renal tissue (n=10) was obtained from ESRD nephrectomies or transplantectomies. Table 1 98

summarizes patient demographics. Renal tissue was stored in ice-cold University of Wisconsin (UW) 99

organ preservation solution, and cold ischemia time was limited to 2-3 hours. 100

Murine tissue was obtained from male C57BL/6 mice, with an average weight of 28.3 g (± 2.4) and 101

12.1 weeks of age (± 2.2). The animals were housed in filter-top cages with free access to water and 102

food. Kidneys were harvested via a terminal procedure performed under isoflurane/O2 anesthesia

103

(Pharmachemie BV, Haarlem, the Netherlands) and stored in ice-cold UW solution until further use. 104

105

Preparation and treatment of precision-cut kidney slices 106

PCKS were prepared according to the protocol by Poosti et al. (mouse; (30)) and Stribos et al. (human; 107

(34)), using a Krumdieck tissue slicer. Slices were incubated in Williams’ Medium E with GlutaMAX 108

(Life Technologies, Carlsbad, California, USA) containing 10 μg/mL ciprofloxacin and 26 mM 109

glucose, at 37°C in a 80% O2/5% CO2 atmosphere while gently shaken. Culture medium was used

110

without serum supplementation to avoid the associated batch-to-batch variability (i.e., to establish 111

fully controlled environment). Nintedanib was kindly provided by Boehringer Ingelheim (Biberach, 112

Germany). We treated murine or human PCKS with nintedanib (0.1 – 10 μM) for 48h. Analyses were 113

(7)

performed using three pooled slices from the same animal/donor (technical replicates) from at least 114

three to five animals or donors (biological replicates). 115

116

Viability of PCKS 117

Viability of the slices was assessed by measuring ATP content using the ATP bioluminescence kit 118

(Roche Diagnostics, Mannheim, Germany), as previously described (39). 119

120

Quantitative real-time PCR and low-density array 121

Total RNA was extracted with the RNeasy mini kit (Qiagen, Venlo, the Netherlands) and reverse 122

transcribed using the Reverse Transcription System (Promega, Leiden, the Netherlands). cDNA was 123

used for quantitative real-time PCR performed with a Viia7 Real-Time PCR system (Applied 124

Biosystems, Bleiswijk, the Netherlands). Gene expression was calculated using the 2-ΔCt method (26)

125

and corrected for GAPDH. The Taqman gene expression assays used in this study are listed in 126

Supplementary Table S1. 127

The expression of 44 genes related to fibrosis (Supplementary Table S2) was examined using a 128

custom-designed low-density array (LDA, Applied Biosystems) (31). cDNA from renal tissue of 129

C57BL/6 control mice that underwent UUO microsurgery was kindly provided by Dr. Bram Piersma. 130

A total of 100 μl reaction mixture containing 6 ng/μl cDNA and 50 μl 2x Taqman Universal PCR 131

Master Mix (Applied Biosystems) was loaded per sample. PCR amplification was performed on a 132

Viia7 Real-Time PCR system (Applied Biosystems). 133

134

Western blotting 135

Total protein was extracted from PCKS with ice-cold RIPA buffer (Thermo Scientific, Waltham, 136

Massachusetts, USA) supplemented with a protease inhibitor cocktail and PhosStop (Sigma-Aldrich, 137

Saint Louis, Missouri, USA). A total of 80-100 μg of protein was separated via SDS-PAGE using 10% 138

polyacrylamide gels and blotted onto polyvinylidene fluoride membranes (Trans-Blot Turbo Transfer 139

System, Bio-Rad, Veenendal, the Netherlands). 2,2,2-trichloroethanol (TCE; Sigma-Aldrich) allowed 140

for visible detection of total protein load (22). Membranes were blocked in 5% non-fat milk/TBST 141

(8)

(Bio-Rad) and incubated with the primary antibody (Supplementary Table S3) overnight at 4˚C, 142

followed by incubation with the appropriate HRP-conjugated secondary antibody. Protein bands were 143

visualized using Clarity Western ECL Substrate (Bio-Rad) and ChemiDoc Touch Imaging System 144

(Bio-Rad). Protein expression was corrected for total protein and expressed as a relative value to the 145

control group. 146

147

Phosphoproteomic analysis of RTKs by multiplex 148

A human RTK phosphoprotein magnetic bead panel (Merck Millipore, Billerica, Massachusetts, 149

USA), was used according to manufacturer’s instructions. Total protein was extracted using the 150

supplied lysis buffer supplemented with protease inhibitor cocktail. Samples were diluted to a 151

concentration of 1.5 μg/μl and passed through a 0.45 μm syringe filter (Whatman, Maidstone, UK). 152

Detection was performed with the MAGPIX multiplexing instrument (Luminex, Austin, Texas, USA). 153

Mean fluorescent intensity (MFI) was used for quantification. 154

155

Histology 156

PCKS were fixed in 4% buffered formalin, embedded in paraffin and sectioned 2 μm thick. Tissue 157

damage and renal fibrosis were assessed by Periodic acid–Schiff (PAS) and Picro Sirius Red (PSR) 158

staining. Additionally, we performed immunohistochemistry for Ki-67, α-SMA and collagen type I. 159

After deparaffinisation and antigen retrieval with 0.1 M Tris-EDTA (pH 9.0) in microwave oven for 160

15 min, tissue sections were blocked with 2% rat serum in PBS/2% BSA for 10 min and then 161

incubated with primary antibodies (Supplementary Table S3) for 1h. The antibodies were localized 162

using the appropriate HRP-conjugated secondary and tertiary antibodies and the ImmPact NovaRed 163

kit (Vector, Burlingame, USA), followed by hematoxylin counterstaining. Stained tissue sections were 164

scanned using a Nanozoomer Digital Pathology Scanner (NDP Scan U10074-01, Hamamatsu 165

Photonics K.K., Japan). To quantify the stained areas, the whole-slide images were processed with 166

Aperio ImageScope v12.3 (Aperio Technologies, Vista, CA) by applying the Positive Pixel Count V9 167

algorithm (hue value set to 0). The intensities were measured as percentages − number of positive and 168

(9)

strong positive pixels divided by the total number of pixels − and expressed as relative values to the 169 control group. 170 171 Statistics 172

The results are expressed as mean ± standard error of mean (SEM) of minimum 3 independent 173

experiments. Statistics were performed using GraphPad Prism 6.0 (GraphPad Software Inc.) by 174

unpaired Student’s t-test or one-way ANOVA followed by Dunnett’s multiple comparisons test. The 175

protein levels of HSP47 and α-SMA were compared using non-parametric Kruskal-Wallis test, 176

followed by Dunn’s multiple comparisons test. Differences between groups were considered to be 177

statistically different when p < 0.05. For the LDA heatmap, average-linkage clustering was performed 178

using Pearson correlation. The heatmap was generated using the online tool Morpheus 179

(https://software.broadinstitute.org/morpheus/). 180

(10)

RESULTS 182

A brief summary of the study workflow is presented in Figure 1A. Murine and human PCKS remained 183

viable during 48h incubation, as reflected by unchanged ATP and total protein content (Figure 1B). In 184

addition, PAS staining revealed typical structural changes in slices due to the culturing in accordance 185

with previously reported results (34, 35). In particular, murine and human PCKS from healthy kidneys 186

showed signs of cellular damage (i.e., pyknosis and anucleosis), tubulointerstitial injury and 187

glomerular injury at 48h. In turn, culturing of human fibrotic PCKS induced further expansion of 188

interstitial ECM, tubular atrophy and glomerular sclerosis. Nintedanib at 5 μM showed no indication 189

towards worsening of the morphology (Figure 1C). 190

191

Nintedanib in murine PCKS 192

Mitigation of fibrosis and inflammation by nintedanib

193

During 48h incubation, spontaneous onset of fibrosis occurred in mPCKS, as reflected by an 194

upregulation of mRNA levels of collagen type I, fibronectin and heat shock protein HSP47 (encoded 195

by Col1a1, Fn1 and Serpinh1, respectively) and by increased protein levels of HSP47 (Figure 2A and 196

B). Nintedanib effectively mitigated fibrogenesis as it clearly reduced Col1a1 gene expression (Figure 197

3A), with an IC50 of 0.7 μM (Supplementary Figure S1A). Furthermore, nintedanib reduced mRNA

198

levels of Fn1 and Acta2 (IC50 of 4.4 μM and 6.2 μM, respectively), while it affected Serpinh1 only at

199

the highest concentration (IC50=5.8 μM). Treatment with 5 μM nintedanib significantly decreased

200

interstitial accumulation of collagen type I and protein expression of HSP47, but did not affect α-SMA 201

(Figure 3B and C). Collagen mRNA and protein levels declined below baseline expression (at 0h) 202

when mPCKS were treated with 5 and 10 μM nintedanib. Spontaneous onset of fibrosis in mPCKS 203

was accompanied by an inflammatory response after 48h (Figure 2C). We observed a significant 204

decrease in mRNA levels of tumor necrosis factor, interleukin-1 beta and interleukin-6 (encoded by 205

Tnf, Il-1β and Il-6, respectively) in the presence of nintedanib, while Cxcl1 (chemokine (C-X-C motif)

206

ligand 1) expression was not affected (Figure 3D). 207

208

Antiproliferative activity of nintedanib in mPCKS

(11)

Previous studies reported the antiproliferative effects of nintedanib (15, 40, 41). We evaluated these 210

effects in mPCKS by Ki-67 immunohistochemistry, a marker of cell proliferation (Figure 3E). We 211

observed a 3.5-fold culture-induced increase in proliferation that was attenuated by 5 μM nintedanib 212

by approximately 30%. 213

214

Low density array for genes related to ECM homeostasis

215

To investigate whether the observed onset of fibrogenesis in mPCKS approximates in vivo 216

fibrogenesis, we measured and compared the expression of 43 genes related to ECM homeostasis in 217

kidneys from mice with renal injury induced by unilateral ureteral obstruction (UUO) and mPCKS. 218

Figure 4A shows that both obstruction of murine kidneys (for 3 or 7 days) and culture of mPCKS 219

introduced considerable changes in the expression of ECM-related genes: the majority of the tested 220

genes were upregulated in the obstructed kidneys and cultured mPCKS. We performed a detailed 221

statistical analysis on a subset of these samples and compared transcriptional changes that occur 222

during the obstruction of murine kidneys for 7 days and during 48h culture of mPCKS (Figure 5). The 223

analysis revealed that 31 out of 43 tested genes (72%) were regulated in the same direction (i.e., 224

upregulated) in the UUO kidneys and mPCKS, while only three genes (Eln, Fmod, Pcolce) were 225

regulated in the opposite direction. Additionally, seven genes were regulated during culture of 226

mPCKS, but not during 7d UUO, and included P4ha1, P4ha3, P4hb, Leprel1, Leprel2, Loxl1 and 227

Mmp13). Among tested transcripts, only expression of Adamts14 was unaltered by the obstruction or

228

culturing. Taken together, culturing of PCKS for 48h induced changes in ECM homeostasis that for 229

the large part mirrored changes observed in UUO kidneys, indicating that PCKS resemble in vivo 230

fibrogenesis. 231

Next, we investigated the impact of nintedanib on mPCKS. Figure 4B shows that nintedanib 232

manifested its inhibitory activity already at the lowest concentration (0.1 μM). A detailed statistical 233

analysis of the subset of these samples showed that 0.1 μM nintedanib significantly reduced 234

expression of 80% of the tested ECM-related genes, including collagen subtypes Col1a1, Col1a2, 235

Col3a1, Col4a1, Col5a1 and Col6a1 and ECM degradation matrix metalloproteinases encoded by

236

Mmp2, Mmp9 and Mmp13 (Figure 6). In turn, treatment of mPCKS with nintedanib at 5 μM

(12)

significantly inhibited only 59% of ECM-related transcripts. This was accompanied by the observation 238

that at high concentrations — 1 and 5 μM — nintedanib regulated different mRNA clusters than at 239

lower concentrations — 0.1 and 0.5 µM. In particular, seven genes (16 %) were regulated significantly 240

differently (i.e., not in the same direction) by nintedanib at 0.1 μM vs. 5 μM (Figure 6C). Among these 241

genes, Adamts3, Pcolce, Slc39a13, Ddr1 and Ddr2 were downregulated by nintedanib at low 242

concentration but not regulated at high concentration. Two transcripts – Dcn and Leprel1 – “switched” 243

their expression from unaltered or downregulated after the treatement with 0.1 μM nintedanib to being 244

upregulated by 5 μM nintedanib. 245

246

Nintedanib in healthy human PCKS 247

Targeted inhibition of gene expression and tyrosine kinase receptor activation

248

Culturing of the slices led to a significant downregulation of VEGFR1, VEGFR2 and FGFR2 mRNA 249

(Figure 7A). Nintedanib reduced expression of PDGFRB, VEGFR1 and VEGFR3 in hPCKS already at 250

0.1 μM (Figure 7B). The treatment did not affect expression of VEGFR2; however, it increased 251

FGFR2 expression at 5 μM.

252

Four phospho-RTKs (RTKs) were upregulated during incubation of hPCKS: PDGFRα, p-253

PDGFRβ, p-VEGFR1, and p-VEGFR2 (Figure 7C). This suggests that the onset of fibrosis in healthy 254

human PCKS was associated with the activation of PDGF and VEGF signaling pathways. Figure 7D 255

shows that nintedanib reduced phosphorylation of p-PDGFRα and p-VEGFR1 at 0.3 μM (by 39.8% 256

and 55%, respectively) and of PDGFRβ and VEGFR2 at 0.1 μM (by 45.3% and 23%). In contrast, the 257

activation of VEGFR3 and FGFR1 in hPCKS was neither affected by 48h incubation, nor by the 258

treatment with nintedanib (Figure 7C and D). 259

260

Mitigation of fibrosis and inflammation markers by nintedanib

261

In line with previously published data (34), fibrogenesis was initiated during incubation of hPCKS, as 262

revealed by the increase in COL1A1 and SERPINH1 transcription and protein levels of HSP47 (Figure 263

8A and B). Expression of ACTA2 significantly dropped after 48h, while FN1 expression remained 264

unchanged. Treatment with nintedanib resulted in a concentration-dependent inhibition of all tested 265

(13)

fibrosis markers, except for ACTA2 (Figure 9A). The IC50 was 0.6 μM for COL1A1, 1.5 μM for

266

SERPINH1 and 1.7 μM for FN1 (Supplementary Figure S1B). Nintedanib at 5 μM reduced the

267

accumulation of collagen type I to the baseline levels (at 0h) and affected protein level of HSP47 268

(Figure 9B and C). In concordance with gene expression, nintedanib had no influence on α-SMA 269

expression. 270

Similar to mPCKS, a substantial increase in mRNA levels of cytokines, such as TNF, IL-1B, IL-6 and 271

CXCL8/IL-8, occurred in hPCKS during culture period (Figure 8C). Nintedanib significantly reduced

272

the expression of these inflammation markers at the highest tested concentration (5 μM) (Figure 9D). 273

Interestingly, IL-1B mRNA level was inhibited at 0.5 μM. 274

275

Antiproliferative activity of nintedanib in hPCKS

276

We observed comparable results of Ki-67 expression in hPCKS, as in mPCKS: expression increased 277

during culture (fold induction of 10.4), and nintedanib reduced proliferation by approximately 73% 278

(Figure 9E). 279

280

Nintedanib in established fibrosis PCKS 281

Characterization of fibrotic human PCKS

282

Fibrotic hPCKS (fhPCKS) showed high basal gene expression of COL1A1, SERPINH1 and FN1, as 283

well as clear inflammatory profile compared to healthy kidneys (Figure 10A and B). Histologic 284

analysis confirmed the fibrotic phenotype by showing an extensive tubular atrophy, ECM 285

accumulation and interstitial fibrosis (Figure 10C and D). 286

To analyze the processes occurring during culture of fhPCKS, we studied viability and gene 287

expression up to 72h. Similar to healthy human PCKS (34), ATP content of fhPCKS increased during 288

first 24h, after which levels plateaued (Figure 10E). As described earlier, healthy PCKS develop a 289

fibrotic response during incubation. We observed a different pattern in fhPCKS: mRNA expression of 290

COL1A1, SERPINH1 and FN1 remained unchanged (Figure 10F). On the other hand, elevated

291

collagen type I deposition and highly increased protein expression of HSP47 might indicate that 292

fibrogenesis was still ongoing during incubation (Figure 10G and H). Similar to hPCKS, ACTA2 293

(14)

expression dropped in fhPCKS during culture, while α-SMA protein expression remained unchanged. 294

Regarding the inflammation markers, fhPCKS showed unaffected gene expression of TNF and IL-1B 295

during culture. Levels of IL-6 and CXCL8/IL-8 increased at 24h and then gradually declined (Figure 296

10I). 297

298

Effect of nintedanib on fibrosis and inflammation markers

299

Treatment of fhPCKS with nintedanib did not affect fibrosis markers on gene expression. Only the 300

highest tested concentration (5 μM) numerically, but not statistically significantly, reduced expression 301

of COL1A1 and SERPINH1 (Figure 11A). We detected non-significant effects on interstitial 302

accumulation of collagen type I and protein expression of α-SMA, however, nintedanib at 5 μM 303

significantly affected HSP47 (Figure 11B and C). Interestingly, 1 and 5 μM nintedanib downregulated 304

IL-1B in fhPCKS by 82.5% and 86.3%, respectively (Figure 11D).

305 306

Antiproliferative activity of nintedanib in fhPCKS

307

Similar to hPCKS, culture for 48h induced Ki-67 expression in fhPCKS (Figure 11E), while 308

nintedanib inhibited cell proliferation by approximately 48%. 309

(15)

DISCUSSION 310

PCKS provide a unique opportunity to translate the obtained results from rodent models to human, 311

which is important for clinical drug development (38). In this study we expanded the experimental 312

application of PCKS: we used healthy renal tissue of murine and human origin, and explored PCKS 313

from human fibrotic renal tissue as a model of established fibrosis. With this translational ex vivo 314

model, we investigated the effects of nintedanib in healthy and diseased tissue. PCKS model replicates 315

some of the main characteristics of CKD, such as cellular damage, tubulointerstitial fibrosis, (local) 316

inflammation, accumulation of ECM proteins and dysregulated matrix turnover, as shown in this study 317

and previously reported by others (30, 34, 35). We demonstrated that spontaneous induction of 318

fibrogenesis in PCKS resembles in vivo fibrogenesis, making this model suitable to study the 319

mechanism of action and efficacy of antifibrotic compounds. We showed that nintedanib blocks the 320

expression and phosphorylation of tyrosine kinase receptors and inhibits cell proliferation. 321

Additionally, nintedanib attenuated the onset of fibrosis not only in murine, but also in human PCKS, 322

although reversal of established fibrosis could not be achieved. 323

Nintedanib engaged its intended targets in human kidneys: it inhibited gene expression of PDGFRβ, 324

VEGFR1 and 3, as well as culture-induced phosphorylation of PDGFRα and β, VEGFR1 and 2 325

starting at 0.1 μM — a concentration that is in the range of its maximum human exposure of 0.07 µM 326

in patients with IPF after standard dosing (11, 28, 42). The attenuation of PDGFR-α and β signaling 327

by nintedanib is of therapeutic interest for renal fibrosis, as PDGF signaling leads to the differentiation 328

of pericytes and resident fibroblasts to profibrotic ECM-producing myofibroblasts (9). The literature 329

presents conflicting results on the role of VEGFR signaling in fibrosis and peritubular capillary 330

restoration (2, 20, 24, 27). Therefore, beneficial inhibition of VEGFR signaling by nintedanib in renal 331

fibrosis is subject to further studies. 332

The inhibitory effects of nintedanib on RTKs were investigated by Liu et al. in a unilateral ureteral 333

obstruction (UUO) mouse model (25). They reported that nintedanib effectively blocked UUO-334

induced phosphorylation of PDGFRβ, VEGFR2, FGFR1 and FGFR2. In human PCKS, both FGFR1 335

(16)

and FGFR2 were not affected by nintedanib, possibly due to the differences in fibrogenic processes in 336

murine and human tissues. 337

PCKS develop an early inflammatory response during culture, followed by the onset of fibrosis (34). 338

Nintedanib exerted anti-inflammatory activity in mPCKS and hPCKS. The observed effects are in line 339

with earlier studies of nintedanib in mouse models of lung fibrosis (23, 40). This anti-inflammatory 340

activity of nintedanib might translate into attenuation of renal injury. 341

Nintedanib also exerted antifibrotic effects in mPCKS and hPCKS, demonstrated by a marked 342

reduction of collagen 1a1 mRNA and protein expression. Furthermore, in mPCKS nintedanib 343

modulates ECM homeostasis even at the lowest concentration. Downregulation of these genes might 344

lead to the altered secretion and fibril formation of collagen, as reported in primary human lung 345

fibroblasts treated with TGF-β (21). High concentrations of nintedanib regulate different mRNA 346

clusters than low concentrations, reflected by a partial switch from inhibitory profile at 0.1 and 0.5 µM 347

to the induction of some ECM related genes at 1 and 5 µM. This could be explained by possible non-348

selective activity of nintedanib at high concentrations, while low concentrations have a more specific 349

kinase inhibitory profile (14). Despite that nintedanib at 1 and 5 µM has an altered impact on ECM 350

homeostasis, its overall effect remains antifibrotic. 351

The demonstrated attenuation of fibrosis concurs with previous results: nintedanib reduced lung 352

fibrosis in bleomycin- or silica-treated mice and rats (1, 40, 41) and showed antifibrotic effects in 353

various mouse models of systemic sclerosis (16, 17). Liu et al. (25) found that administration of 354

nintedanib for 7 days after UUO injury attenuated renal fibrosis. Nintedanib inhibited TGF-β1 induced 355

renal fibroblasts-to-myofibroblasts transition and expression of ECM proteins in vitro in renal 356

interstitial fibroblasts, indicating that nintedanib affects early events of TGF- β signaling. Wollin et al. 357

also reported that nintedanib at higher concentrations possesses anti-TGF-β activity (40, 41). We 358

hypothesize that the observed antifibrotic effects of nintedanib in PCKS might be attributed to a 359

combination of RTK inhibition and, perhaps non-selective, anti-TGF-β activity. 360

The culture of mPCKS, hPCKS and fhPCKS induced a strong spontaneous proliferative response and 361

in line with published data (15, 40, 41), nintedanib effectively inhibited culture-induced cell 362

proliferation in PCKS. 363

(17)

Our newly established translational PCKS model with tissue from fibrotic human kidneys showed a 364

clear fibrotic phenotype compared to renal tissue from control donors. Culture of fhPCKS did not 365

further increase the assessed markers of fibrosis, although an inflammatory peak was observed after 366

the first day of culture. The pre-existing fibrotic phenotype of fhPCKS might explain the difference 367

with healthy PCKS. 368

Nintedanib showed diminished reduction in fibrosis markers in fhPCKS compared to hPCKS, most 369

likely due to an increased interindividual variability (underlying primary renal disease, dialysis time, 370

time since transplantation and medication) in the fibrotic kidney slices. Nintedanib seems to be more 371

effective in preventing or halting the onset of fibrosis in healthy PCKS rather than in reversing the 372

established fibrosis as modeled by fhPCKS. Our data in human diseased tissue is in conflict with the 373

data in murine diseased tissue: delayed administration of nintedanib to UUO mice by Liu et al. (25) 374

resulted in partial reversal of established renal fibrosis. However, even prolonged kidney obstruction 375

in mice is not as severe as the late-stage fibrosis seen in CKD patients, emphasizing the need for 376

models that more closely resemble human pathology. PCKS, especially the culture of fhPCKS, can 377

serve as a model for human CKD. Nevertheless, limitations of the ex vivo PCKS model are: (1) the 378

relatively short culture period (48-72h) is not always sufficient to detect post-translational events; (2) 379

the availability of freshly resected human renal tissue is limited; (3) interorgan interactions cannot be 380

directly assessed; and (4) circulating immune cells that contribute to the fibrogenesis are absent, 381

because slices lack blood circulation, even though PCKS retain native vascular network. Considering 382

the significant role of infiltrating inflammatory cells and associated cytokines in renal fibrosis, a co-383

culture system of PCKS with immune cells (or subsets of immune cells) can be established for future 384

studies, which will better reflect the immunological interactions during the fibrogenic process in renal 385

tissue. 386

Taken together, our results demonstrate the pharmacological effects of nintedanib in an ex vivo model 387

of renal fibrosis, facilitating translation from animal studies to the clinic. Treatment of PCKS with 388

nintedanib inhibited cell proliferation and attenuated the onset of inflammation and fibrosis, although 389

reversal of established fibrosis could not be achieved. Nintedanib successfully inhibited PDGFR and 390

VEGFR phosphorylation, demonstrating the potential use of these receptors as therapeutic targets to 391

(18)

attenuate renal fibrosis. Therefore, along with the benefit of reducing animal use, human PCKS might 392

provide direct and clinically relevant insights into human renal disease and therapeutic strategies. 393

(19)

ACKNOWLEDGEMENTS 394

The authors thank the abdominal transplantation surgeons of the University Medical Center Groningen 395

for providing the human renal tissue. 396

397

CONFLICT OF INTEREST 398

The authors declare the following competing financial interest: Dr. Lutz Wollin is an employee of 399

Boehringer Ingelheim Pharma GmbH & Co. 400

401

AUTHOR CONTRIBUTIONS 402

E.B., E.G.D.S., P.O. and M.B. designed the study; E.B., E.G.D.S. and M.B. carried out experiments 403

and analyzed the data; B.P., R.A.B., H.v.G. and L.W. provided additional analytical tools and 404

chemicals for this study; A.M.L. and I.J.d.J. helped with human tissue procurement; E.B. and E.G.D.S. 405

wrote the manuscript with critical review from H.A.M.M., R.A.B., M.A.S., H.v.G, L.W., P.O. and 406

M.B. All of the authors approved the final version of the manuscript for publication. 407

408

FUNDING 409

This work was kindly supported by ZonMw (the Netherlands Organization for Health Research and 410

Development), grant number 114025003 and by Lundbeckfonden, grant number R231-2016-2344 411

(received by H.A.M.M). 412

(20)

REFERENCES 414

1. Ackermann M, Kim YO, Wagner WL, Schuppan D, Valenzuela CD, Mentzer SJ, Kreuz S, Stiller

415

D, Wollin L, Konerding MA. Effects of nintedanib on the microvascular architecture in a lung fibrosis

416

model. Angiogenesis 20: 359–372, 2017.

417

2. Bábíčková J, Klinkhammer BM, Buhl EM, Djudjaj S, Hoss M, Heymann F, Tacke F, Floege J,

418

Becker JU, Boor P. Regardless of etiology, progressive renal disease causes ultrastructural and

419

functional alterations of peritubular capillaries. Kidney Int 91: 70–85, 2017.

420

3. Beyer C, Distler JHW. Tyrosine kinase signaling in fibrotic disorders. Biochim Biophys Acta - Mol

421

Basis Dis 1832: 897–904, 2013.

422

4. Boor P, Ostendorf T, Floege J. PDGF and the progression of renal disease. Nephrol Dial Transplant

423

29: i45–i54, 2014.

424

5. Buhl EM, Djudjaj S, Babickova J, Klinkhammer BM, Folestad E, Borkham-Kamphorst E,

425

Weiskirchen R, Hudkins K, Alpers CE, Eriksson U, Floege J, Boor P. The role of PDGF-D in

426

healthy and fibrotic kidneys. Kidney Int 89: 848–861, 2016.

427

6. Cernaro V, Trifirò G, Lorenzano G, Lucisano S, Buemi M, Santoro D. New therapeutic strategies

428

under development to halt the progression of renal failure. Expert Opin Investig Drugs 23: 693–709,

429

2014.

430

7. Chen Y-T, Chang F-C, Wu C-F, Chou Y-H, Hsu H-L, Chiang W-C, Shen J, Chen Y-M, Wu K-D,

431

Tsai T-J, Duffield JS, Lin S-L. Platelet-derived growth factor receptor signaling activates pericyte–

432

myofibroblast transition in obstructive and post-ischemic kidney fibrosis. Kidney Int 80: 1170–1181,

433

2011.

434

8. van Dijk F, Olinga P, Poelstra K, Beljaars L. Targeted Therapies in Liver Fibrosis: Combining the

435

Best Parts of Platelet-Derived Growth Factor BB and Interferon Gamma. Front Med 2: 72, 2015.

436

9. Duffield JS. Cellular and molecular mechanisms in kidney fibrosis. J Clin Invest 124: 2299–2306, 2014.

437

10. Eisen T, Loembé A-B, Shparyk Y, MacLeod N, Jones RJ, Mazurkiewicz M, Temple G, Dressler H,

438

Bondarenko I. A randomised, phase II study of nintedanib or sunitinib in previously untreated patients

439

with advanced renal cell cancer: 3-year results. Br J Cancer 113: 1140–1147, 2015.

440

11. Eisen T, Shparyk Y, Macleod N, Jones R, Wallenstein G, Temple G, Khder Y, Dallinger C,

441

Studeny M, Loembe A-B, Bondarenko I. Effect of small angiokinase inhibitor nintedanib (BIBF 1120)

442

on QT interval in patients with previously untreated, advanced renal cell cancer in an open-label, phase

(21)

II study. Invest New Drugs 31: 1283–1293, 2013.

444

12. Floege J, Eitner F, Alpers CE. A New Look at Platelet-Derived Growth Factor in Renal Disease. J Am

445

Soc Nephrol 19: 12–23, 2008.

446

13. Genovese F, Kàrpàti ZS, Nielsen SH, Karsdal MA. Precision-Cut Kidney Slices as a Tool to

447

Understand the Dynamics of Extracellular Matrix Remodeling in Renal Fibrosis. Biomark Insights 11:

448

77–84, 2016.

449

14. Hilberg F, Roth GJ, Krssak M, Kautschitsch S, Sommergruber W, Tontsch-Grunt U, Garin-Chesa

450

P, Bader G, Zoephel A, Quant J, Heckel A, Rettig WJ. BIBF 1120: Triple Angiokinase Inhibitor with

451

Sustained Receptor Blockade and Good Antitumor Efficacy. Cancer Res 68: 4774–4782, 2008.

452

15. Hostettler KE, Zhong J, Papakonstantinou E, Karakiulakis G, Tamm M, Seidel P, Sun Q, Mandal

453

J, Lardinois D, Lambers C, Roth M. Anti-fibrotic effects of nintedanib in lung fibroblasts derived

454

from patients with idiopathic pulmonary fibrosis. Respir Res 15: 157, 2014.

455

16. Huang J, Beyer C, Palumbo-Zerr K, Zhang Y, Ramming A, Distler A, Gelse K, Distler O, Schett

456

G, Wollin L, Distler JHW. Nintedanib inhibits fibroblast activation and ameliorates fibrosis in

457

preclinical models of systemic sclerosis. Ann Rheum Dis 75: 883–890, 2016.

458

17. Huang J, Maier C, Zhang Y, Soare A, Dees C, Beyer C, Harre U, Chen C-W, Distler O, Schett G,

459

Wollin L, Distler JHW. Nintedanib inhibits macrophage activation and ameliorates vascular and

460

fibrotic manifestations in the Fra2 mouse model of systemic sclerosis. Ann Rheum Dis 76: 1941–1948,

461

2017.

462

18. Humphreys BD, Lin S-L, Kobayashi A, Hudson TE, Nowlin BT, Bonventre J V, Valerius MT,

463

McMahon AP, Duffield JS. Fate Tracing Reveals the Pericyte and Not Epithelial Origin of

464

Myofibroblasts in Kidney Fibrosis. Am J Pathol 176: 85–97, 2010.

465

19. James MT, Hemmelgarn BR, Tonelli M. Early recognition and prevention of chronic kidney disease.

466

Lancet 375: 1296–1309, 2010.

467

20. Kang DH, Joly AH, Oh SW, Hugo C, Kerjaschki D, Gordon KL, Mazzali M, Jefferson JA, Hughes

468

J, Madsen KM, Schreiner GF, Johnson RJ. Impaired angiogenesis in the remnant kidney model: I.

469

Potential role of vascular endothelial growth factor and thrombospondin-1. [Online]. J Am Soc Nephrol

470

12: 1434–47, 2001. http://www.ncbi.nlm.nih.gov/pubmed/11423572 [8 Mar. 2019].

471

21. Knüppel L, Ishikawa Y, Aichler M, Heinzelmann K, Hatz R, Behr J, Walch A, Bächinger HP,

472

Eickelberg O, Staab-Weijnitz CA. A Novel Antifibrotic Mechanism of Nintedanib and Pirfenidone.

(22)

Inhibition of Collagen Fibril Assembly. Am J Respir Cell Mol Biol 57: 77–90, 2017.

474

22. Ladner CL, Yang J, Turner RJ, Edwards RA. Visible fluorescent detection of proteins in

475

polyacrylamide gels without staining. Anal Biochem 326: 13–20, 2004.

476

23. Lee HY, Hur J, Kim IK, Kang JY, Yoon HK, Lee SY, Kwon SS, Kim YK, Rhee CK. Effect of

477

nintedanib on airway inflammation and remodeling in a murine chronic asthma model. Exp Lung Res 43:

478

187–196, 2017.

479

24. Lin S-L, Chang F-C, Schrimpf C, Chen Y-T, Wu C-F, Wu V-C, Chiang W-C, Kuhnert F, Kuo CJ,

480

Chen Y-M, Wu K-D, Tsai T-J, Duffield JS. Targeting Endothelium-Pericyte Cross Talk by Inhibiting

481

VEGF Receptor Signaling Attenuates Kidney Microvascular Rarefaction and Fibrosis. Am J Pathol 178:

482

911–923, 2011.

483

25. Liu F, Wang L, Qi H, Wang J, Wang Y, Jiang W, Xu L, Liu N, Zhuang S. Nintedanib, a triple

484

tyrosine kinase inhibitor, attenuates renal fibrosis in chronic kidney disease. Clin Sci 131: 2125–2143,

485

2017.

486

26. Livak KJ, Schmittgen TD. Analysis of Relative Gene Expression Data Using Real-Time Quantitative

487

PCR and the 2−ΔΔCT Method. Methods 25: 402–408, 2001.

488

27. Long DA, Norman JT, Fine LG. Restoring the renal microvasculature to treat chronic kidney disease.

489

Nat Rev Nephrol 8: 244–250, 2012.

490

28. Mross K, Stefanic M, Gmehling D, Frost A, Baas F, Unger C, Strecker R, Henning J,

Gaschler-491

Markefski B, Stopfer P, de Rossi L, Kaiser R. Phase I Study of the Angiogenesis Inhibitor BIBF 1120

492

in Patients with Advanced Solid Tumors. Clin Cancer Res 16: 311–319, 2010.

493

29. Ostendorf T, Eitner F, Floege J. The PDGF family in renal fibrosis. Pediatr Nephrol 27: 1041–1050,

494

2012.

495

30. Poosti F, Pham BT, Oosterhuis D, Poelstra K, van Goor H, Olinga P, Hillebrands J-L. Precision-cut

496

kidney slices (PCKS) to study development of renal fibrosis and efficacy of drug targeting ex vivo. Dis

497

Model Mech 8: 1227–1236, 2015.

498

31. Remst DFG, Blom AB, Vitters EL, Bank RA, van den Berg WB, Blaney Davidson EN, van der

499

Kraan PM. Gene Expression Analysis of Murine and Human Osteoarthritis Synovium Reveals

500

Elevation of Transforming Growth Factor β-Responsive Genes in Osteoarthritis-Related Fibrosis.

501

Arthritis Rheumatol 66: 647–656, 2014.

502

32. Roth GJ, Binder R, Colbatzky F, Dallinger C, Schlenker-Herceg R, Hilberg F, Wollin S-L, Kaiser

(23)

R. Nintedanib: From Discovery to the Clinic. J Med Chem 58: 1053–1063, 2015.

504

33. Stribos EGD, Hillebrands J-L, Olinga P, Mutsaers HAM. Renal fibrosis in precision-cut kidney

505

slices. Eur J Pharmacol 790: 57–61, 2016.

506

34. Stribos EGD, Luangmonkong T, Leliveld AM, de Jong IJ, van Son WJ, Hillebrands J-L, Seelen

507

MA, van Goor H, Olinga P, Mutsaers HAM. Precision-cut human kidney slices as a model to

508

elucidate the process of renal fibrosis. Transl Res 170: 8-16.e1, 2016.

509

35. Stribos EGD, Seelen MA, van Goor H, Olinga P, Mutsaers HAM. Murine Precision-Cut Kidney

510

Slices as an ex vivo Model to Evaluate the Role of Transforming Growth Factor-β1 Signaling in the

511

Onset of Renal Fibrosis. Front Physiol 8: 1026, 2017.

512

36. Strutz F, Zeisberg M, Hemmerlein B, Sattler B, Hummel K, Becker V, Müller GA. Basic fibroblast

513

growth factor expression is increased in human renal fibrogenesis and may mediate autocrine fibroblast

514

proliferation. Kidney Int 57: 1521–1538, 2000.

515

37. Tampe D, Zeisberg M. Potential approaches to reverse or repair renal fibrosis. Nat Rev Nephrol 10:

516

226–237, 2014.

517

38. Westra IM, Mutsaers HAM, Luangmonkong T, Hadi M, Oosterhuis D, de Jong KP, Groothuis

518

GMM, Olinga P. Human precision-cut liver slices as a model to test antifibrotic drugs in the early onset

519

of liver fibrosis. Toxicol Vitr 35: 77–85, 2016.

520

39. Westra IM, Oosterhuis D, Groothuis GMM, Olinga P. Precision-cut liver slices as a model for the

521

early onset of liver fibrosis to test antifibrotic drugs. Toxicol Appl Pharmacol 274: 328–338, 2014.

522

40. Wollin L, Maillet I, Quesniaux V, Holweg A, Ryffel B. Antifibrotic and Anti-inflammatory Activity of

523

the Tyrosine Kinase Inhibitor Nintedanib in Experimental Models of Lung Fibrosis. J Pharmacol Exp

524

Ther 349: 209–220, 2014.

525

41. Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, Kolb M. Mode of action of

526

nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J 45: 1434–1445, 2015.

527

42. Wollin SL, Bonella F, Stowasser S. Idiopathic pulmonary fibrosis: current treatment options and

528

critical appraisal of nintedanib. Drug Des Devel Ther 9: 6407, 2015.

529

43. Zhang S, Liu Q, Xiao J, Lei J, Liu Y, Xu H, Hong Z. Molecular validation of the precision-cut kidney

530

slice (PCKS) model of renal fibrosis through assessment of TGF-β1-induced Smad and p38/ERK

531

signaling. Int Immunopharmacol 34: 32–36, 2016.

532

44. Zhuang S, Liu N. EGFR signaling in renal fibrosis. Kidney Int Suppl 4: 70–74, 2014.

(24)

FIGURE LEGENDS 534

Figure 1. (A) Schematic illustration of the workflow. Renal tissue of murine or human origin was 535

used to obtain cylindrical cores. By placing the tissue cores in Krumdieck tissue slicer, filled with ice-536

cold Krebs-Henseleit buffer, we prepared precision-cut kidney slices with a wet weight of 4-5 mg and 537

estimated thickness of 250-300 µm. Slices were subsequently incubated in 12-well plates (1 slice per 538

well) in culture medium with or without nintedanib for 48h at 37ºC. Medium was refreshed at 24h. At 539

the end of culture period, samples were collected by pooling three slices (from each animal/donor) for 540

each type of analysis. Visualization of kidney slices prepared from healthy tissue is represented by the 541

blue color, from diseased tissue – by the orange color. Same color code is applied to all figures. (B) 542

Viability of murine, human and fibrotic human PCKS treated with nintedanib for 48h was measured 543

by ATP and total protein content. Data are shown as values relative to non-treated control slices at 48h 544

and are expressed as mean (± SEM), n=4-5, *p<0.05. (C) Representative images of Periodic acid– 545

Schiff (PAS) staining of untreated slices at 0h and 48h as well as of slices treated with 5 µM 546

nintedanib (scale bar = 100 µm). 547

548

Figure 2. Spontaneous fibrogenic and inflammatory response in murine PCKS during culture. 549

(A) mRNA expression of fibrosis markers. (B) Protein levels of HSP47 and α-SMA with 550

representative Western blot images. (C) mRNA expression of inflammation markers. Data are 551

expressed as mean (± SEM), n=4-5. Gene expression levels were compared using unpaired Student’s 552

t-test; protein levels were compared using non-parametric Mann-Whitney test, *p<0.05 553

Col1a1, collagen type I alpha 1; Serpinh1, serine proteinase inhibitor clade H (Heat Shock Protein 47) 554

member 1; Fn1, fibronectin 1; Acta2, alpha 2 smooth muscle actin; HSP47, heat shock protein 47; α-555

SMA, alpha smooth muscle actin; Tnf, tumor necrosis factor; Il-1b, interleukin 1 beta; Il-6, interleukin 556

6; Cxcl1, C-X-C motif chemokine ligand 1. 557

558

Figure 3. Antifibrotic, anti-inflammatory and antiproliferative effect of nintedanib in healthy 559

mouse kidney. Murine PCKS were cultured in the presence of nintedanib (1, 5 or 10 µM) for 48h. (A) 560

mRNA expression of fibrosis markers after 48h incubation. (B) Representative sections with 561

(25)

interstitial collagen type I accumulation visualized by immunohistochemistry (scale bar = 50 µm) and 562

quantitative analysis of staining intensity. (C) Protein levels of HSP47 and α-SMA at 48h with 563

representative Western blot images. (D) mRNA expression of inflammation markers after 48h 564

incubation. (E) Expression of cell proliferation marker Ki-67 in murine PCKS during culture and after 565

48h treatment with 5 µM nintedanib was visualized by immunohistochemistry and quantified as 566

relative intensity values. 567

Data are expressed as mean (± SEM), n=3-5, *p<0.05. 568

Col1a1, collagen type I alpha 1; Serpinh1, serine proteinase inhibitor clade H (Heat Shock Protein 47) 569

member 1; Fn1, fibronectin 1; Acta2, alpha 2 smooth muscle actin; HSP47, heat shock protein 47; α-570

SMA, alpha smooth muscle actin; Tnf, tumor necrosis factor; Il-1b, interleukin 1 beta; Il-6, interleukin 571

6; Cxcl1, C-X-C motif chemokine ligand 1; mPCKS, murine precision-cut kidney slices. 572

573

Figure 4. Transcriptional changes in the extracellular matrix homeostasis genes in murine 574

kidney slices as measured by TaqMan low density array. (A) Heatmap illustrating log2 fold

575

changes in the expression of extracellular matrix (ECM) related genes in murine kidneys subjected to 576

3 days or 7 days of ureteral obstruction (UUO) and in murine PCKS during 48h culture. Fold changes 577

are relative to the average expression in the corresponding control group (i.e., 3d UUO relative to 3d 578

SHAM; 7d UUO relative to 7d SHAM; 48h mPCKS relative to 0h mPCKS). (B) Heatmap of ECM 579

modulation profiles in murine PCKS at 0h, 48h and treated with nintedanib (0.1-5 μM) for 48h. Fold 580

changes are relative to the average expression in 0h PCKS group. 581

Red and blue indicate relatively high and low expression, respectively (grey color indicates 582

undetermined values). Average-linkage hierarchical clustering (supervised in A, unsupervised in B) 583

was performed using Pearson correlation. Complementary statistical analyses performed on ΔCt 584

values are shown in Supplementary Figure S3 and S4. Full gene names are listed in Supplementary 585

Table S2. 586

587

Figure 5. Detailed analysis of the transcriptional changes in the extracellular matrix homeostasis 588

determined by TLDA in the UUO mouse kidneys and murine PCKS. (A) Heatmap that illustrates 589

(26)

changes in the gene expression patterns (log2 fold changes) in kidneys from mice subjected to 7 days

590

UUO and in murine PCKS (mPCKS) cultured for 48h, as these groups were included for the statistical 591

analysis. Red and blue indicate relatively high and low expression, respectively (grey color indicates 592

undetermined values). Supervised average-linkage hierarchical clustering was performed using 593

Pearson correlation. (B) Statistical analysis (on the ΔCt values) of the transcriptional changes in ECM 594

homeostasis in the obstructed for 7d murine kidneys and in cultured for 48h PCKS (as determined by 595

TLDA). Fold changes were calculated as relative to the corresponding control (i.e., 7d UUO vs. 7d 596

SHAM; 48h mPCKS vs. 0h mPCKS). Fold changes in red indicate significant upregulation (with p < 597

0.05), fold changes in blue indicate significant downregulation (with p < 0.05), while fold changes in 598

black indicate no significant regulation of a gene. Data are expressed as mean ± SEM; experimental 599

groups (7d UUO vs. 7d SHAM; 48h mPCKS vs. 0h mPCKS) were compared by unpaired two-tailed 600

Student’s t-test. (C and D) Number of genes that were not regulated or statistically significantly 601

altered in expression during 7d UUO or during 48h culture of mPCKS. 602

603

Figure 6. Detailed analysis of the transcriptional changes in the expression of extracellular 604

matrix related genes in murine PCKS treated with nintedanib for 48h. (A) Heatmap that 605

illustrates changes in the gene expression patterns (log2 fold changes) in mPCKS cultured in the

606

absence of nintedanib, or treated with nintedanib at 0.1 μM or 5 μM for 48h, as these groups were 607

included for the statistical analysis. Fold changes were calculated as relative to the average expression 608

in the untreated PCKS (48h). Red and blue indicate relatively high and low expression, respectively 609

(grey color indicates undetermined values). Unsupervised average-linkage hierarchical clustering was 610

performed using Pearson correlation. (B) Statistical analysis (on the ΔCt values) of the transcriptional 611

changes in the expression of ECM-related genes in mPCKS after treatment with nintedanib at the 612

lowest (0.1 μM) and the highest (5 μM) tested concentration (as determined by TLDA). Fold changes 613

were calculated as relative to the untreated slices (48h). Fold changes in blue indicate significant 614

downregulation (with p < 0.05), fold changes in red indicate significant upregulation (with p < 0.05), 615

while fold changes in black indicate no significant change in the expression of a gene. Data are 616

(27)

expressed as mean ± SEM; experimental groups (48h untreated mPCKS, mPCKS treated with 0.1 μM 617

nintedanib and mPCKS treated with 5 μM nintedanib) were compared by one-way ANOVA followed 618

by Dunnett’s multiple comparisons test. While color indicates significance for comparisons: 48h 619

untreated mPCKS vs. nintedanib 0.1 μM and 48h untreated mPCKS vs. nintedanib 5 μM, (*) denotes 620

statistical differences between mPCKS treated with nintedanib 0.1 μM vs. nintedanib 5 μM. (*) 621

colored in red indicate genes that were considered as “switched” in their expression between 0.1 μM 622

and 5 μM nintedanib treatment groups. Ns, not significant; n.a., not available (e.g. due to 623

undetermined values). (C) Number of genes that were statistically significantly inhibited by nintedanib 624

0.1 μM and 5 μM, as well as number of genes that “switched” their expression when mPCKS were 625

exposed to the highest tested concentration of nintedanib compared to the lowest tested concentration. 626

627

Figure 7. Tyrosine kinase receptors (RTKs) expression and activation in human PCKS after 48h 628

culture and after nintedanib treatment. (A) RTKs mRNA expression in hPCKS after 48h culture, 629

as measured by rt-qPCR. (B) RTKs mRNA expression in hPCKS after 48h treatment with nintedanib 630

(0.1 – 5 µM). (C) Phosphorylation of RTKs in hPCKS was measured by multiplex magnetic bead 631

assay and expressed as relative mean fluorescence intensity (MFI) to 0h control. (D) Phosphorylation 632

of RTKs in hPCKS treated with nintedanib (0.1 – 5 µM) for 48h, shown as mean MFI relative to the 633

untreated hPCKS. Data are expressed as mean (± SEM), n=4-5, *p<0.05 634

FLT1, Fms related tyrosine kinase 1; FLT4, Fms related tyrosine kinase 4; KDR, kinase insert domain 635

receptor; PDGFRB, platelet derived growth factor receptor beta; PDGFRA, platelet derived growth 636

factor receptor alpha; VEGFR1, vascular endothelial growth factor receptor 1; VEGFR2, vascular 637

endothelial growth factor receptor 2; VEGFR3, vascular endothelial growth factor receptor 3; FGFR1, 638

fibroblast growth factor receptor 1; FGFR2, fibroblast growth factor receptor 2. 639

640

Figure 8. Spontaneous fibrogenic and inflammatory response in human PCKS during culture. 641

(A) mRNA expression of fibrosis markers. (B) Protein levels of HSP47 and α-SMA with 642

representative Western blot images. (C) mRNA expression of inflammation markers. Data are 643

(28)

expressed as mean (± SEM), n=4-5. Gene expression levels were compared using unpaired Student’s 644

t-test; protein levels were compared using non-parametric Mann-Whitney test, *p<0.05 645

COL1A1, collagen type I alpha 1; SERPINH1, serine proteinase inhibitor clade H (Heat Shock Protein 646

47) member 1; FN1, fibronectin 1; ACTA2, alpha 2 smooth muscle actin; HSP47, heat shock protein 647

47; α-SMA, alpha smooth muscle actin; TNF, tumor necrosis factor; IL-1B, interleukin 1 beta; IL-6, 648

interleukin 6; CXCL8, C-X-C motif chemokine ligand 8 (IL-8, interleukin 8). 649

650

Figure 9. Effects of nintedanib in healthy human kidney. Human PCKS were cultured in the 651

presence of nintedanib (0.1 – 5 µM) for 48h. (A) mRNA expression of fibrosis markers after 48h 652

incubation. (B) Representative sections with interstitial collagen type I accumulation visualized by 653

immunohistochemistry (scale bar = 100 µm) and quantitative analysis of staining intensity. (C) Protein 654

levels HSP47 and α-SMA at 48h with representative Western blot images. (D) mRNA expression of 655

inflammation markers after 48h incubation. (E) Expression of cell proliferation marker Ki-67 in 656

human PCKS during culture and after 48h treatment with 5 µM nintedanib was visualized by 657

immunohistochemistry and quantified as relative intensity values. 658

Data are expressed as mean (± SEM), n=4-5, *p<0.05. 659

COL1A1, collagen type I alpha 1; SERPINH1, serine proteinase inhibitor clade H (Heat Shock Protein 660

47) member 1; FN1, fibronectin 1; ACTA2, alpha 2 smooth muscle actin; HSP47, heat shock protein 661

47; α-SMA, alpha smooth muscle actin; TNF, tumor necrosis factor; IL-1B, interleukin 1 beta; IL-6, 662

interleukin 6; CXCL8, C-X-C motif chemokine ligand 8 (IL-8, interleukin 8); hPCKS, human 663

precision-cut kidney slices. 664

665

Figure 10. Characterization of fibrotic human precision-cut kidney slices. PCKS were prepared 666

from fibrotic human renal tissue obtained during ESRD nephrectomies or transplantectomies. (A) 667

Baseline (prior incubation) mRNA expression of fibrosis markers in fibrotic compared to healthy 668

tissue slices. (B) Baseline (prior incubation) mRNA expression of inflammation markers in fibrotic 669

compared to healthy tissue slices. (C) Representative photomicrographs of human healthy and fibrotic 670

PCKS prior incubation (scale bar = 250 μm). Histologic analyses by PAS and PSR staining showing 671

(29)

extensive tubular atrophy and interstitial fibrosis, while α-SMA and collagen type I 672

immunohistochemistry further confirmed the fibrotic phenotype. (D) Quantitative analysis of collagen 673

type I immunohistochemistry in healthy and fibrotic PCKS (prior incubation). (E) Viability of fibrotic 674

PCKS during incubation presented as the average of pmol ATP per μg total protein. (F) Effect of 675

incubation on mRNA expression of fibrosis markers. (G) Representative images of 676

immunohistochemistry of collagen type I (scale bar = 100 μm) in fibrotic PCKS during 72h culture 677

with quantitative analysis. (H) Protein levels of HSP47 and α-SMA (n=5) during incubation with 678

representative Western blot images. (I) Effect of incubation on mRNA expression of inflammation 679

markers in fhPCKS. Data are expressed as mean (± SEM), n=9 for healthy PCKS and n=8-9 for 680

fibrotic PCKS, *p<0.05. 681

COL1A1, collagen type I alpha 1; SERPINH1, serine proteinase inhibitor clade H (Heat Shock Protein 682

47) member 1; FN1, fibronectin 1; ACTA2, alpha 2 smooth muscle actin; HSP47, heat shock protein 683

47; α-SMA, alpha smooth muscle actin; TNF, tumor necrosis factor; IL-1B, interleukin 1 beta; IL-6, 684

interleukin 6; CXCL8, C-X-C motif chemokine ligand 8 (IL-8, interleukin 8); PAS, periodic acid-685

Schiff; PSR, Picro Sirius red. 686

687

Figure 11. Antifibrotic and anti-inflammatory effect of nintedanib in fibrotic human precision-688

cut kidney slices. Fibrotic human PCKS were cultured in the presence of nintedanib (0.1 – 5 μM) for 689

48h. (A) mRNA expression of fibrosis markers after 48h incubation. (B) Representative sections with 690

interstitial collagen type I accumulation visualized by immunohistochemistry (scale bar = 100 µm) 691

and quantitative analysis of staining intensity. (C) Protein levels HSP47 and α-SMA at 48h with 692

representative Western blot images. (D) mRNA expression levels of inflammation markers after 48h 693

incubation. (e) Expression of cell proliferation marker Ki-67 in fibrotic human PCKS during culture 694

and after 48h treatment with 5 µM nintedanib was visualized by immunohistochemistry and quantified 695

as relative intensity values. 696

Data are expressed as mean (± SEM), n=4-5, *p<0.05. 697

COL1A1, collagen type I alpha 1; SERPINH1, serine proteinase inhibitor clade H (Heat Shock Protein 698

47) member 1; FN1, fibronectin 1; ACTA2, alpha 2 smooth muscle actin; HSP47, heat shock protein 699

(30)

47; α-SMA, alpha smooth muscle actin; TNF, tumor necrosis factor; IL-1B, interleukin 1 beta; IL-6, 700

interleukin 6; CXCL8, C-X-C. 701

702

Supplementary Figure S1. IC20 and IC50 of nintedanib in murine and human PCKS. 703

Nintedanib-induced inhibition of mRNA expression of fibrosis markers after 48h incubation in murine 704

PCKS (A) and in human PCKS (B). IC20 and IC50 values were calculated by fitting the data to a 705

four-parameter log(inhibitor) vs response curve. Data points are the mean (± SEM), n=3-5. 706

(31)

Table 1. Patient demographics

Healthy renal

tissue (n=9) Fibrotic renal tissue (n=10)

Gender (% male) 67 40

Age (in years) 66 ± 8 47 ± 15

Nephrectomy side (% left) 37.5 50

Creatinine before

nephrectomy (μmol/L) 81 ± 13 545 ± 403

eGFR before nephrectomy

(ml/min/1.73m²)* 81 ± 9 NA

Time on dialysis (mean in

months) NA 116 ± 136

Time since (first) transplantation (mean in months)

NA 132 ± 150

Type renal tissue NA Non-functioning

kidney allograft (n=4), kidney allograft with infected abscess (n=1), non-functioning native ESRD kidney (n=5).

*calculated using the Modification of Diet in Renal Disease (MDRD) formula. Values are presented as the mean ± standard deviation or otherwise if indicated.

(32)

A B 0h cont rol 48h cont rol 48h ni nt edani b 5 μM mPCKS hPCKS fhPCKS 0 1 5 10 0.0 0.5 1.0 1.5 Nintedanib [µM] Re la ti v e AT P v a lu e Viability mPCKS 0 0.1 0.3 0.5 1 5 0.0 0.5 1.0 1.5 Nintedanib [µM] Re la ti v e AT P v a lu e Viability hPCKS 0 0.1 0.3 1 5 0.0 0.5 1.0 1.5 Nintedanib [µM] Re la ti v e AT P v a lu e Viability fhPCKS 0 1 5 10 0.0 0.5 1.0 1.5 Nintedanib [µM] Re la ti v e p ro te in c o n te n t Protein mPCKS 0 0.1 0.3 0.5 1 5 0.0 0.5 1.0 1.5 Nintedanib [µM] Re la ti v e p ro te in c o n te n t Protein hPCKS 0 0.1 0.3 1 5 0.0 0.5 1.0 1.5 Nintedanib [µM] Re la ti v e p ro te in c o n te n t Protein fhPCKS Krumdieсk Tissue Slicer Treatment: +/- nintedanib Incubation: 1 slice/well for 48 hours;

80% O2/ 5% CO2; 370C; shaking WT C57BL/6 Healthy kidneys Tissue cores (60-200 slices) Analysis Precision-cut kidney slices (PCKS) Healthy kidneys Transplantectomy Fibrotic kidneys 200-300 μm 5 mm C

Referenties

GERELATEERDE DOCUMENTEN

In human PCKS, galunisertib consistently inhibited collagen type I transcription and de novo synthesis, and showed moderate reduction of HSP47, PLOD2 and fibronectin mRNA levels

The work presented in this thesis was conducted at the Greningen Research Institute of Pharmacy, department of Pharmaceutical Technology and Biopharmacy, University of Groningen,

In Chapter 2 we use NGS and IPA to describe the transcriptional changes and biological processes that occur during culture of healthy murine and human PCTS from different

In this study we described the changes in the transcriptional profiles during culture of PCTS obtained from five different organs (liver, kidney, jejunum, ileum and colon) and

Functional pathway analysis of differentially expressed gene sets was performed with QIAGEN Ingenuity® Pathway Analysis software (IPA®, QIAGEN Redwood City, California, USA) for

S1 – The mRNA expression levels of fibrosis, inflammation and fat metabolism markers in mouse PCLS obtained from mice on the control diets of HFD, AMLN and CDAA.. The PCLS

drug (0.1 and 1 μM), showing the importance of this pathway in ECM production. The method of PCTS for preclinical drug testing is highly useful due to several advantages: 1)

PCR of Gpnmb expression in PCLS from murine models of (A) fibrosis and (B) NASH; All PCLS were collected after preparation (no incubation); Fold induction is relative to