You are on page 1of 50

ToxSci Advance Access published March 9, 2015

Revised manuscript with tracked changes

Title:
Glomerulonephritis-induced changes in urinary and kidney microRNA profiles in rats

Authors:
Mira Pavkovic *,†
(mira_pavkovic@hms.harvard.edu)
Björn Riefke *
(bjoern.riefke@bayer.com)
Anna-Lena Frisk ‡
(anna-lena.frisk@bayer.com)

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Ina Gröticke§
(ina.groeticke@bayer.com)
Heidrun Ellinger-Ziegelbauer *
(heidrun.ellinger-ziegelbauer@bayer.com)

Affiliations:
* Investigational Toxicology, GDD-GED-Toxicology, Bayer Pharma AG, 42096 Wuppertal and
13353 Berlin, Germany.

† current: Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences,


Harvard Medical School, 02115 Boston, Massachusetts, USA.

‡ Pathology, GDD-GED-Toxicology, Bayer Pharma AG, 13353 Berlin, Germany.

§ Indication Expansion, GDD-GCGE-Common Mechanism Research, Bayer Pharma AG, 13353


Berlin, Germany

Running title:
Urine miRNA biomarker and glomerulonephritis

Corresponding author and address:


Dr. Heidrun Ellinger-Ziegelbauer
Aprather Weg 18a, Bldg 514
42096 Wuppertal, Germany
Office: +49 202 36 4396
Email: heidrun.ellinger-ziegelbauer@bayer.com

1
Abstract

MicroRNAs (miRNAs) regulate gene expression post-transcriptionally and thus are involved in

various physiological and pathological states. Due to their stability in biofluids miRNAs have

also been proposed as biomarkers (BMs) for tissue injury. We investigated the usefulness of

urinary miRNAs for detection of site-specific renal damage in an anti-glomerular basement

membrane (GBM) glomerulonephritis (GN) model in rats by comparing GN-induced urinary

miRNAs profiles to traditional and newer protein BMs, and to proximal tubular injury-induced

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


urinary miRNA profiles observed previously after cisplatin treatment.

Male Wistar Kyoto and Sprague Dawley rats were dosed once with 1, 2.5 and 5 ml/kg

nephrotoxic serum (NTS) or 1.5 and 5 ml/kg NTS, respectively. GN and tubular damage were

observed histopathologically in all treated rats after 14 days. Although serum creatinine and

BUN were not changed, several protein BMs and 74 urinary miRNAs were found to be

increased 8 and 14 days after NTS administration. Of these 74 miRNAs, five were identified as

increased after NTS- but not after cisplatin treatment. Using in situ hybridization two of them,

miR-10b and -100, were found to be localized in distal segments of the nephron, potentially

reflecting the tubular injury in those regions.

Furthermore, evaluation of both miRNA and mRNA expression in the kidney revealed possible

miRNA-mRNA interactions mostly associated with fibrotic and transforming growth factor β

signaling.

In conclusion, our investigations support the potential of urinary miRNAs as specific BMs for

kidney injury, and suggest a role of miRNAs in pathological processes during GN in the kidney.

Keywords: urinary microRNAs, biomarker, glomerulonephritis

2
1. Introduction

Detection of kidney injury is a challenge either in preclinical and clinical phases of drug

development, or in patients with acute kidney injury (AKI) caused by other reasons, since

especially in the latter case patients are at high risk to develop chronic or end-stage renal

diseases (Guengerich, 2011; Hsu et al., 2013). As traditional biomarkers for kidney function like

serum creatinine and blood urea nitrogen are associated with low sensitivity, specificity, and

capability for early diagnosis (Vaidya et al., 2008), large efforts were and are still being

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


undertaken in the exploration of new kidney injury biomarkers (BMs) with enhanced

characteristics. Significant success was achieved within the protein BM research leading to

regulatory qualification of new BMs in urine for detection of nephrotoxicity in rats (EMA, 2009).

Although these new protein BMs outperform traditional BMs in sensitivity and specificity, they

are partly limited for instance in terms of antibody availability to ensure translatability across pre-

clinical laboratory animal species and to humans (Jensen, 2004) or in terms of stability in

biofluids (Dieterle and Sistare, 2010). Additionally, none of these is exclusively specific for

glomerular injury.

Recently another molecule class, microRNAs (miRNAs), has entered the BM research field.

Accumulating evidence suggests them as promising new BMs for tissue injury. MiRNAs are

small (approximately 22 nucleotides), non-coding RNAs known to post-transcriptionally regulate

gene expression through binding to 3'-untranslated region of target mRNAs (Bartel, 2004). They

are evolutionarily conserved across a wide range of species and involved in essential

physiological functions, including proliferation, apoptosis and differentiation (Krol et al., 2010).

Not unexpected in light of their wide range of functions, miRNA involvement was identified in

various pathological states like cancer, cardiovascular as well as kidney diseases (Hermeking,

2012; Schena et al., 2014; Small and Olson, 2011). With the discovery of miRNAs in body fluids

(Weber et al., 2010) their features as ideal BMs were recognized: tissue-specificity, less-

3
invasive accessibility, high stability in biofluids, translational potential and sensitive

quantification with real-time PCR (Saikumar et al., 2014, Mall et al., 2013).

Several studies have already reported the BM potential of e.g. plasma miRNAs for the detection

of cancer (Fu et al., 2014), drug induced liver injury (Wang et al., 2014;), or myocardial injury

(Nishimura et al., 2014). Compared to reports on blood-based miRNAs as potential BMs, data

for urinary miRNAs as candidate BMs for kidney damage is limited, yet still promising, as

suggested by studies analyzing urine of patients with AKI or immunoglobulin A nephropathy

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


(Ramachandran et al., 2013; Saikumar et al., 2012; Wang et al., 2011).

Recently the Health and Environmental Science Institute1 (HESI) Biomarkers of Nephrotoxicity

Committee engaged into investigation of the potential of miRNA measurements in urine for

detection of site-specific renal damage, studying compounds with known specific toxic effects

on discrete segments of the nephron in a pre-competitive collaboration. In a first study we

examined the miRNA profile in urine from Wistar rats with cisplatin-induced proximal tubular

injury and found many miRNAs with significantly increased urinary levels upon cisplatin

treatment (Pavkovic et al., 2014a). These results were confirmed by other HESI Committee

members reporting several identical miRNAs to be increased in urine after cisplatin and

gentamicin induced proximal tubular injury in Sprague Dawley rats (Kanki et al., 2014;

Nassirpour et al., 2014).

The objective of the present study was to investigate urinary miRNA levels in a rat injury model

affecting a different part of the nephron i.e. the glomerulus and to identify site-specific miRNAs

by comparing urinary miRNA profiles after glomerular vs. proximal tubular injury. Therefore we

employed nephrotoxic serum (NTS) which contains antibodies against the glomerular basement

membrane (GBM) to induce glomerulonephritis (GN). Anti-GBM GN models in rats are widely

used since its biochemical and histopathological characteristics are similar to crescentic

nephritis and Goodpasture's disease in humans (Pusey, 2003); although, such models are

challenging since tubular damage is a known side effect of antiserum-induced GN models

4
(Salant and Cybulsky, 1988). To our knowledge, this is the first time that urinary miRNA were

profiled in an anti-GBM GN model. Urine was collected 8 and 14 days after a single injection of

1, 2.5 and 5 ml/kg NTS in male Wistar Kyoto rats and of 1.5 and 5 ml/kg NTS in male Sprague

Dawley rats. In addition to analyses of classical endpoints and several newer protein BMs,

urinary miRNA profiles were measured by quantitative real-time PCR. Potential site-specific

localization of two miRNAs apparently specific for the injury present in the GN model was

investigated by in situ hybridization. Furthermore, since different rat strains show differential

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


susceptibility to NTS-induced GN, two different rat strains were chosen for characterization of

molecular processes affected in this model, by investigating gene expression in the kidneys

from both rat strains.

5
2. Materials and Methods

2.1. Animal Studies

All animal experiments were performed according to the German guidelines for care and use of

laboratory animals. Male Wistar Kyoto (WKY) and Sprague Dawley (SD) rats, 8 weeks old, were

purchased from Charles River laboratories (Sulzbach, Germany). Animals were individually

housed in type III makrolon cages, with a 12h light-dark cycle. Standard rodent chow pellets and

water were provided ad libitum.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Rats were randomly grouped (WKY: control group n=3, treatment groups n=5; SD: all groups

n=6) and dosed once iv with 0, 1 (WKY), 1.5 (SD), 2.5 (WKY) or 5 ml/kg nephrotoxic serum

(NTS; Sheep Anti-Rat Glomerular Basement Membrane (GBM) Serum (PTX-001S); Probetex,

San Antonio, TX). Control groups received appropriate volumes of normal saline solution.

Doses were selected based on Probetex recommendations and dose finding studies performed

in house. For urine collection animals were housed in individual metabolism cages (24h) and

had access to food and water. Urine was collected over ice on day 8 and 14, centrifuged

(3000xg 10 min 10°C) and the supernatant was stored at -80°C. Blood was collected on day 14

and resulting serum and plasma were stored at -80°C. On day 14 rats were euthanized and the

kidneys were removed for histopathological examination and RNA isolation. Kidney tissue for

RNA isolation was shock frosted with liquid nitrogen and stored at -80°C.

2.2. Biochemical and histopathological analyses

Blood urea nitrogen and serum creatinine concentrations were measured with an ADVIA 1650

using reagents from Siemens Diagnostics (Fernwald, Germany). Creatinine was determined by

the Jaffe´ method. Urinary total protein was determined quantitatively with an ADVIA 2400 using

reagents from Siemens Diagnostics (Fernwald, Germany). Urinary creatinine was measured

using an enzymatic test on a Roche Cobas cIII.

6
Urinary protein biomarkers were analyzed with two different platforms according to the

manufacturer’s instructions. MesoScale Discovery (MSD; Gaithersburg, MD): urinary albumin

(ALB) was measured with the Kidney Injury Panel 1 Assay Kit. Luminex xMAP (LMX; purchased

from Novagen, EMD Chemicals; Madison, WI): urinary β-2-microglobulin (β2M), glutatione-S-

transferase alpha (αGST), kidney injury molecule-1 (KIM-1), tissue inhibitor of metallopeptidase-

1 (TIMP-1), vascular endothelial growth factor (VEGF), and calbindin (CALB), clusterin (CLU),

cystatin C (CysC), lipocalin-2 (neutrophil gelatinase-associated lipocalin, NGAL), osteopontin

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


(OPN) were measured with the WideScreen Rat Kidney Toxicity Panels 1 and 2, respectively.

Absolute concentrations were normalized to the corresponding urinary creatinine concentration.

Statistical analysis was performed with ANOVA and Dunnett’s test or with the Student’s t-test

using GraphPad Prism 5 (GraphPad Software Inc., USA) between replicates of the treatment

groups and corresponding time-matched control group.

For histopathological examination kidneys were fixed in 10% neutral buffered formalin, trimmed,

embedded, sectioned and stained with hematoxylin and eosin. Histopathological findings

(mesangial hypercellularity, accumulation of glomerular matrix, hyperplasia of Bowman capsule,

hyaline casts, tubular degeneration/regeneration, tubular dilatation, interstitial fibrosis,

mononuclear cell infiltration) were graded separately per parameter using a severity scoring

system: grade 0, absent; grade 1, minimal; grade 2, slight; grade 3, moderate; grade 4, marked;

grade 5, very severe. In order to further characterize the lesion a Periodic acid-Schiff reaction

(PAS) stain was performed, too. Additional findings in the SD rats, included cysts, tubular

basophilia, and pelvic dilation, these findings were interpreted to be spontaneous background

findings.

2.3. RNA extraction

Total RNA including small RNA species (miRNAs) was isolated from 200 µl urine using the

miRNeasy Mini Kit (Qiagen; Hilden, Germany). Each urine sample was mixed with 700 µl

7
QIAzol, and then spiked with 2.5 fmol synthetic control miRNA ath-miR-159a (Sigma Aldrich;

Hamburg, Germany); all following procedures were performed according to the manufacturer’s

instructions. Finally, RNA was eluted with 40 µl RNase-free water.

Total kidney RNA including small RNA species was also isolated with the miRNeasy Mini Kit

from 20 mg kidney tissue whereas total kidney RNA (mRNA) was isolated with the RNeasy Mini

Kit (Qiagen) from 70 mg kidney tissue, both according to the manufacturer’s instructions.

The quality of isolated RNAs was assessed using Bioanalyzer Small or Pico RNA Kits (Agilent;

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Santa Clara, CA) and the quantity was determined with a spectrophotometer.

2.4. Quantitative real-time PCR and data analysis

Urinary miRNA profiles were analyzed with TaqMan® Low Density A Arrays (TaqMan cards,

Applied Biosystems by life technologies; Darmstadt, Germany), representing 373 unique rodent

miRNAs and several small RNAs. A fixed volume of 3 µl of the RNA eluate was employed.

Megaplex reverse transcription, megaplex pre-amplification and final quantitative PCR on the

TaqMan cards were performed according to the manufacturer’s instructions. MiRNA levels were

calculated according to the ‘modified ∆Ct quantification’ approach as described previously

(Pavkovic, et al., 2014a). This approach is based on calculating 230-Ct and normalizing to urinary

creatinine (230-Ct/uCrea) to derive a unit-less absolute amount, corrected for urine dilution. For

selection of affected miRNAs derived from the TaqMan card data, tools implemented in Analyst

software (Genedata; Basel, Switzerland) were employed, including ANOVA with factors

treatment and time (BH-Q<0.05 considered as significant), Student’s t-test (treated vs. control,

p<0.01 considered as significant) and a 2-fold change cutoff between treated and control

groups.

Selected miRNAs were re-analyzed in urine samples with TaqMan® MicroRNA Assays followed

by absolute quantification using standard curves run in parallel, as described previously

(Pavkovic, et al., 2014a). For several miRNAs for which we did not obtain a reliable standard

8
curve with TaqMan® MicroRNA Assays, we used miCURY LNATM PCR assays from Exiqon

(Vedbaek, Denmark). A fixed volume of 2 µl of the RNA eluate was employed for these assays.

Universal reverse transcription followed by a specific SYBER green based quantitative PCR

were conducted according to the manufacturer’s instructions.

Kidney miRNAs were measured with TaqMan cards according to the manufacturer’s

instructions. Relative levels of kidney miRNAs (treated vs. control) were calculated using the

general ∆∆Ct method with small RNA U6 as endogenous control gene: ∆∆Ct = (Cttarget miRNA –

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


CtU6)treated - (Cttarget miRNA – CtU6)control. Tools available via Genedata’s Analyst software, including

Student’s t-tests (treated vs. control, p<0.05 considered as significant) and a 1.5-fold change

cutoff between treated and control groups, were used for selection of deregulated miRNAs.

2.5. Microarray expression profiling

Biotin-labeled copyRNA samples prepared from 500 ng high quality total kidney RNA were

hybridized on GeneChip® Rat genome 230_2.0 arrays (Affymetrix; Santa Clara, CA) according

to the manufacturer’s instructions. After scanning of the fluorescent GeneChip images with the

Affymetrix GeneChip Scanner 3000, raw data image files (dat) were converted into cel-files.

Finally, one intensity value per probe set was derived with Affymetrix Microarray Suite (MAS)

5.0. The GeneChip® Rat genome 230_2.0 array comprises over 31000 probe sets representing

approximately 28700 well annotated rat genes.

Using Genedata’s Analyst, genes encoding mRNAs, whose levels were significantly changed,

were selected separately for each rat strain using ANOVA based on the factor treatment (BH-

Q<0.005 considered as significant), Student’s t-test (p<0.001 considered as significant) and a 2-

fold deregulation cutoff between treated and control groups.

Ingenuity Pathway Analysis (IPA, Ingenuity® Systems, www.ingenuity.com) was employed for

pathway analysis in general and to search for deregulated mRNAs containing sequences

complementary to those present in the deregulated miRNAs, i.e. so-called miRNA target

9
analysis for identification of mRNAs potentially regulated by miRNAs. The following filter criteria

were applied: (1) experimentally observed and/or highly predicted target relation, i.e. sequence

complementarity between a mRNA and a miRNA, (2) opposite direction of expression change

after NTS administration, since miRNAs are mostly negative regulators of the mRNAs they can

bind, and (3) known expression of the miRNAs in kidney.

2.6 in situ Hybridization

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


In a separate experiment the localization of selected miRNAs was investigated in the kidney of a

healthy male Wistar rat. After whole-body perfusion with a fixing solution containing 3% formalin

and 1% glutaraldehyde, kidneys were immediately removed, fixed in neutral buffered formalin,

trimmed and paraffin embedded. In situ hybridization was performed on approximately 5 µm

thick sections using double-digoxigenin (DIG) labeled miRNA probes from Exiqon according to

the manufacturer’s instructions with minor modifications. In brief, sections were first

deparaffinized with xylene and ethanol (100%, 96% and 70%) followed by incubation with

proteinase-K (2.5 µg per slide for 10 min 37°C). After washing with 1xPBS, sections were

dehydrated with ethanol (70%, 96% and 100%), air dried for 15 min and pre-hybridized for 30

min at 55°C with 1xExiqon’s hybridization buffer supplemented with 4.3% salmon sperm DNA

(Sigma-Aldrich; Hamburg, Germany). Hybridization with the miRNA probes (miR-10b:

ACACAAATTCGGTTCTACAGGG, Tm 82°C, 100 nM; miR-100: CACAAG-

TTCGGATCTACGGGTT, Tm 83°C, 100 nM; miR-26a: AGCCTATCCTGGATTACTTGAA, Tm

85°C, 100 nM; miR-192: GGCTGTCAATTCATAGGTCAG, Tm 83°C, 80 nM) was performed for

one hour 30°C under the RNA melting temperature corresponding to the optimal hybridization

temperature. Sections were washed sequentially with pre-heated SSC-buffer (1x, 0.2x, 0.1x) at

55°C and blocked with blocking-buffer (1% BSA, 2% sheep serum in 0.1% PBS-T) for 30 min at

room temperature. Alkaline phosphatase (AP) conjugated anti-DIG (Roche Diagnostics,

Mannheim, Germany) was diluted 1:800 in antibody-dilution-buffer (1% BSA, 1% sheep serum

10
in 0.05% PBS-T) and incubated for one hour at room temperature. After washing with 0.1%

PBS-T, sections were incubated with AP-substrate solution (NBT-BCIP ready-to-use tablets;

Roche Diagnostics, Mannheim, Germany) light-protected over night at room temperature. The

AP reaction was stopped with cold water. For counter staining sections were incubated in 0.01%

eosin for approximately one minute, dehydrated with ethanol (70%, 90% and 100%) and then

xylene. Finally, sections were scanned and analyzed using the MIRAX Scanner and Viewer,

respectively (Zeiss; Jena, Germany). General experimental settings were established with

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Exiqon’s positive controls (U6 probe: abundant nuclear signal; miR126 probe: abundant

endothelial signal) and negative controls (scrambled probe: no complementarity to any known

miRNAs sequence). The pictures shown are representative for at least three independent in situ

hybridization experiments.

11
3. Results

3.1 Histopathology and clinical chemistry

Wistar Kyoto (WKY) and Sprague Dawley (SD) rats were treated once with different doses of

nephrotoxic serum (NTS). The WKY kidneys were histologically examined after 14 days.

Glomerulonephritis (GN), characterized by mesangial hypercellularity, glomerular matrix

accumulation and Bowman’s capsule hyperplasia, was diagnosed in all NTS-treated rats (Fig.1).

Further, an accumulation of PAS positive material in the mesangium was seen in both rat

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


strains (Suppl.Fig.2). Although fully formed glomerular crescents were not detected, the

activation/hyperplasia of the Bowman’s capsule epithelia (hyperplasia) might represent dose

dependent initial lesions of crescent formation and/or the hypercellularity might

develop/contribute to crescent formations in a later stage.

All findings increased dose-dependently from minimal to moderate severity scores, and were

not observed in control rats. In addition to glomerular changes, tubular degeneration and

regeneration was observed in the cortex including the corticomedullary junction and extending

to the inner stripe of the outer medulla. Treated SD rats, examined 8 and 14 days after NTS

injection, showed comparable findings, but glomerular changes were less severe than in WKY

rats, while tubular damage and intra group variability were larger. All histopathological findings

are listed in Suppl.Tab.1.

Traditional kidney injury biomarkers like blood urea nitrogen and serum creatinine were not

increased after NTS treatment (Suppl.Fig.1 A and B), but severe proteinuria was detected. 8

and 14 days after NTS administration total protein levels were significantly and dose-

dependently increased in urine from WKY and SD rats (Fig. 2).

3.2 Evaluation of urinary protein biomarkers for kidney injury

For measurement of newly qualified, recently endorsed, or exploratory protein biomarkers (BMs)

urine was collected 8 and 14 days after NTS treatment. Corresponding to severe proteinuria a

12
large increase of albumin (ALB) was measured in urine from all treated rats of both strains at

both time points (Tab.1). β-2-microglobuline (β2M), kidney injury molecule (KIM-1), clusterin

(CLU) and cystatin C (CysC) showed similar urinary profiles: no increase after treatment with

the lowest doses in both rat strains on both days, an increase after treatment of WKY rats with

the middle dose of 2.5 ml/kg NTS on both days and a significant increase after treatment with

the high dose (5 ml/kg) in both rat strains on both days (Tab.1). None of the other BMs, i.e.

neutrophil gelatinase-associated lipocalin (NGAL), osteopontin (OPN), α-glutathione-S-

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


transferase (αGST), tissue inhibitor of metallopeptidase-1 (TIMP-1), or vascular endothelial

growth factor (VEGF), were increased in urine after treatment with the lowest doses in both rat

strains, and after treatment of WKY rats with the middle dose of 2.5 ml/kg NTS; only NGAL and

VEGF were significantly increased on day 14 and 8 NTS (Tab.1 and Suppl.Tab.2), respectively.

Treatment with 5 ml/kg NTS increased urinary levels of all other BMs, except calbindin (CALB)

which showed no significant changes in urinary levels at any time point. Overall, the NTS-

induced increases of all urinary BM levels were dose-dependent, larger on day 8 than on day 14

per dose group and higher in WKY rats than in SD rats. Interestingly, despite both ALB and

KIM-1 showing a strong treatment-dependent increase in urine, the treatment vs. control ratios

were clearly higher for ALB than for KIM-1, e.g. approximately 140-fold higher 14 days after

treatment of WKY rats with 5 ml/kg NTS.

3.3 Comparison of urinary miRNAs increased by GN-associated vs. cisplatin-induced proximal

tubular injury

To explore miRNAs increased or decreased in urine with GN-associated kidney damage, we

measured over 350 miRNAs in urine samples from control and NTS (5 ml/kg) groups on day 8

and 14 using TaqMan cards. After statistical analysis, in total 74 miRNAs were found showing

significantly increased levels in urine from WKY (72 miRNAs) and SD (30 miRNAs) rats after

NTS treatment (Suppl.Fig.4). The miRNA increases were higher and displayed less intra-group

13
variability in urine from WKY than from SD rats (Suppl.Tab.3). For further analysis we selected

12 miRNAs: miR-10a, -10b, -21, -26a, -34a, -34b, -100, -184, -192, -197, -211 and -486. The

selection was based on two main considerations: known expression in the glomerulus or at least

in the kidney and either increased or not increased in urine compared to previously reported

urinary miRNA profiles after cisplatin (Cp) induced proximal tubular injury (Pavkovic, et al.,

2014a). The latter consideration refers to the identification of miRNAs potentially specific for one

kidney etiology, here proximal glomerular vs. tubular injury. Candidates were derived from a

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


comparison of GN- and Cp-affected urinary miRNA groups (Fig.3), which were essentially all

increased in urine by the respective treatment (Supp Fig. 2 and Pavkovic, et al., 2014a). In the

Cp study we identified in total 136 significantly affected miRNA in urine after a single

administration of 3 mg/kg with analysis 3, 5, 8, 15 and 24 days thereafter. Maximal necrosis had

been detected histopathologically 8 days after Cp treatment. 68 urinary miRNAs of the 74 NTS-

and 136 Cp- increased urinary miRNAs were identical. Although then 68 miRNAs appeared to

be specifically increased by Cp, a qualitative examination of the profiles of these miRNAs after

NTS treatment revealed that they were also increased in the GN rat models (data not shown).

Still, five of the six miRNAs increased in a significant manner only by NTS (miR-10a, -10b, -100,

-211 and -486) remained as apparently specific for NTS-induced injury after inspection of the

profile of these miRNAs in urine from Cp-treated rats, i.e. appeared not consistently affected by

Cp. They were thus selected for further analysis together with seven other miRNAs. For

verification of their profiles derived from TaqMan card PCR, absolute urinary concentrations

were measured using single miRNA-specific TaqMan or Exiqon assays with standard curves

run in parallel which currently represents a reliable and accepted method for urinary miRNA

quantification. Applying standard curve quantification most of the 12 urinary miRNA increases

after 5 ml/kg NTS seen before with TaqMan card measurement could be verified. For nine

miRNAs the increases were also statistically significant in WKY rats after treatment with the

highest dose of NTS on either day 8 (seven miRNAs) or 14 (4 miRNAs), with two miRNAs

14
significantly increased at both time points. In SD rats only miR-21 was also found significantly

increased 8 days after treatment (Tab.2). Comparison with our Cp data showed that four of

these 12 selected miRNAs, miR-21, -34a, -184, -192, were clearly increased after NTS and Cp

treatment based on the measurement of absolute urinary concentrations in both studies. The

increase of the five miRNAs, miR-10a, -10b, -100, -211, -486, which seemed to be specifically

increased after NTS treatment only based on TaqMan card data, could also be verified with

standard curve quantification for WKY rats after NTS treatment.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


We additionally measured the selected miRNAs in urine samples from rats treated with lower

NTS doses. In urine from WKY rats treated with 2.5 ml/kg NTS only miR-10a (1.4-fold) and miR-

21 (4.6-fold) were significantly increased on day 8 (Tab.2). MiR-10b and -100, although not

significant in three out of four cases, appeared strongly increased in urine from WKY rats

treated with 2.5 and 1 ml/kg NTS after 14 days. In urine from SD rats treated with 1.5 ml/kg NTS

clearly increased miRNAs were not detected (Tab.2). The increase of urinary microRNAs after

lower NTS doses correlates with increased levels of several protein BMs. Except for ALB, none

of the protein BMs was significantly increased after the lowest doses of 1 and 1.5 ml/kg NTS in

WKY and SD rat urine, respectively. Yet a few did show higher levels after 2.5 ml/kg NTS in

urine from WKY rats (Tab.1).

3.4 Distinct localization of miR-10b and 100 in the rat kidney

In situ hybridization (ISH) experiments were performed to investigate where in the normal

kidney those miRNAs are expressed which were found to be specifically increased by NTS-

induced GN but not Cp-induced proximal tubular injury. We used untreated and not treated

kidney, since the latter may be of insufficient quality for detection of ISH signals, and since the

knowledge of the localization of a miRNA in normal kidney should allow to infer the damage

location if such a miRNA is increased in urine after a toxic treatment, provided that this miRNA

is not induced at the expression level in another location by the toxic treatment. Based on this

15
observation we hypothesized the five miRNAs listed above, i.e. miR-10a, -10b, -100, -211 and -

486 to be localized in glomeruli within the kidney, since this should be the primary damage site

impacted by NTS treatment. For initial evaluation we selected miR-10b and -100 since they

were most increased in urine after NTS treatment (Tab.2) but were not changed in the kidney

tissue (Suppl.Tab.6). With U6 and miR-126 as positive controls, we observed the expected

nuclear and endothelial staining, respectively (Fig.4 A and B), while no staining was observed

with the negative control (Fig.4 E), establishing that the protocol used can detect site-specific

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


localization of a miRNA. Furthermore, miR-26a and -192, which are known to be expressed in

kidney, were both detected in glomeruli and proximal tubules (Fig.4 C and D). Unexpectedly, in

the normal rat kidney miR-10b and -100 expression was localized in distal segments of the

nephron, more precisely in the descending thin limb of the loop of Henle, the distal convoluted

tubule, the connecting tubule and the cortical collecting duct; but not in glomeruli (Fig.5).

3.5 mRNA and miRNA patterns associated with rat kidney glomerulonephritis

For elucidation of molecular pathways which may play a role or may be associated with NTS-

induced GN and to potentially identify a molecular basis for different strain susceptibility, mRNA

and miRNA levels were examined in the kidney 14 days after NTS administration in both rat

strains. Gene i.e. mRNA expression measured with Affymetrix GeneChip microarrays resulted

in 428 and 40 significantly deregulated mRNAs in kidneys from WKY and SD rats, respectively;

yielding in total 439 mRNAs deregulated in both strains together (data not shown). Gene

expression profiles were comparable within the two rat strains in terms of direction of

deregulation and dose dependency (Suppl.Fig.4). 253 of the 439 deregulated genes could be

functionally assigned to categories like inflammation, proliferation, regeneration, structural

remodeling and others (Suppl.Tab.4). The categories with the highest numbers of genes

assigned to them were inflammation with mRNAs coding for e.g. interleukin-24 (Il-24) or CD53,

and regeneration with mRNAs coding for e.g. different collagens like Col1a1 or Col4a1. Two

16
mRNAs were oppositely changed by NTS treatment in the two rat strains, being decreased in

kidneys from SD rats but increased in kidneys from WKY rats. Both proteins encoded by these

mRNAs are associated with immune response/ inflammation: RT1 class II, locus Ba (Rt1-Bb)

and complement component 6 (C6) (Suppl.Fig.4 and Suppl.Tab.4).

Since NTS effects were more prominent in WKY than in SD rats, we focused on WKY rats for

miRNA analysis. Kidney miRNA levels measured with TaqMan cards resulted in 80 significantly

deregulated miRNAs after NTS administration (Suppl.Tab.5). Some of the miRNAs increased in

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


urine were found to be increased (miR-21, -34b, -146b, -197) or decreased (miR-192, -193b) in

kidneys after NTS treatment. To identify possible regulatory influences of miRNAs on mRNAs in

the kidney during GN, we employed tools implemented in the Ingenuity Pathway analysis (IPA)

software. Within the pool of 439 significantly deregulated mRNAs in the kidney after NTS

treatment, we identified 69 mRNAs that could potentially be targeted by 44 of the 80

deregulated kidney miRNAs (Suppl.Tab.7). These mRNAs encode proteins associated with e.g.

fibrosis or the TGFβ-pathway and the corresponding miRNA-mRNA pairs mostly showed the

expected opposite direction of change (Fig.6 A and B). For example, increased levels of miR-

197 and -132 could be associated with decreased levels of insulin-like growth factor binding

protein 3 (Igfbp3) and matrix metallopeptidase 9 (Mmp9) mRNAs , respectively, in the kidney

after NTS-induced GN (Fig.6A).

17
4. Discussion

To investigate whether miRNAs could complement or enhance urinary protein biomarkers (BMs)

with respect to diagnosis of glomerular kidney injury, urinary miRNA profiles were examined in a

glomerulonephritis (GN) model in two rat strains. The study encompassed administration of

Wistar Kyoto (WKY) and Sprague Dawley (SD) rats with different doses of nephrotoxic serum

(NTS), followed by measurement of traditional and newer protein BMs 8 and 14 days after

treatment, and histological examination of the kidneys 8 (SD only) and 14 days after treatment.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Urinary miRNA profiles obtained with this GN model were then compared with previously

reported urinary miRNA profiles after cisplatin (Cp)-induced proximal tubular injury to search for

miRNAs specifically associated with glomerular damage. To reveal potential involvement of

miRNA-mRNA pairs in GN-induced kidney damage, miRNA and mRNA expression was

examined in the kidney.

Although traditional BMs did not indicate kidney damage, confirming their insensitivity (Urbschat

et al., 2011), both glomerular and tubular damage were observed histopathologically on day 14.

Tubular damage was a secondary response to glomerular changes, since the latter can lead to

protein overload in primary urine and thus to injury in exposed tubules (Togashi et al.,

2013).The severity of tubular damage indicates potential primary tubular injury, possibly due to

reabsorption of the NTS or cross-reactivity of the polyclonal antibodies in the NTS with tubular

structures. We suspect onset of tubular damage already on day 8, as seen for SD rats and as

KIM-1, a qualified protein BM for proximal tubular injury (Vaidya et al., 2010), was even higher in

urine on day 8 compared to day 14 in WKY rats. Tubular damage was probably also the reason

for increased levels of other protein BMs most of which are qualified for proximal tubular injury

(Fuchs and Hewitt, 2011).

In parallel to proteinuria, albumin (ALB) was the only protein BM in urine clearly increased after

all NTS doses in both rat strains. ALB indicates loss of glomerular integrity and/or impaired

18
proximal tubular function (Swain et al., 2011). It was increased after both Cp-induced tubular

injury reported earlier (Pavkovic et al., 2014b) and NTS-induced GN reported here, indicating

that as expected urinary ALB alone is not sufficient for differentiation between tubular vs.

glomerular injury. We still suggest that treatment-induced increases of urinary ALB in relation to

that of proximal tubular specific urinary KIM-1 can serve to localize the primary site of injury,

since in our data ALB vs. KIM-1 ratios were much higher in the GN study associated with

glomerular (and tubular) injury compared to the Cp study with tubular injury, only.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Using a PCR profiling approach (Pavkovic, et al., 2014a) 74 miRNAs were identified with

significantly increased urinary levels 8 and/or 14 days after high dose NTS treatment in both rat

strains. For about one third of these miRNAs kidney expression has already been described

(Landgraf et al., 2007; Tian et al., 2008). Corresponding to the more pronounced pathological

findings in WKY rats, a higher number of significantly changed miRNAs associated with higher

fold-changes were found in urine from WKY compared to SD rats. Slightly increased urinary

levels at lower NTS doses were observed for miR-10b, -21, -34b, -100 and -192 in WKY rats, in

parallel to minimal changes seen histopathologically in the corresponding kidneys and

increased urinary ALB levels, indicating that they may represent potentially quite sensitive BMs.

Nevertheless, none of the urinary miRNA significantly outperformed all protein BMs, thus at

least in this study their usefulness as BMs is rather associated with specific information about

the localization of injury than with increased sensitivity.

The majority of the urinary miRNAs affected by NTS treatment overlapped with miRNAs

described previously to be affected by Cp-induced proximal injury (Pavkovic, et al., 2014a). One

explanation is that many identical miRNAs are expressed in different kidney compartments

(Kriegel et al., 2013). As described above, a second obvious but not mutually exclusive

explanation is that apart from glomerular injury, tubular injury was also present our GN model.

Although Cp is an acute toxicant leading to early injury development, we still suggest that the

19
comparison to NTS-induced GN is reasonable since the even the maximal dose of 3 mg/kg Cp

caused maximal necrosis on day 8 only. Nevertheless, five miRNAs (miR-10a, -10b, -100, -211

and -486) seemed to be NTS-specific.

Since these five were particularly interesting in terms of finding miRNAs specific for glomerular

injury, the localization of two of them, miR-10b and -100, was investigated within the normal rat

kidney by in situ hybridization. Contrary to our expectations that they would be expressed in

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


glomeruli, these two were found to be localized in the descending thin limb of the loop of Henle,

the distal convoluted tubule, the connecting tubule and the cortical collecting duct. Yet

interestingly, their location partially overlapped with that of the observed tubular lesions. An

impairment of these portions of the nephron, in addition to glomerular damage, corresponds to

findings by Iida et al. (2014) describing distal nephron segment injuries in rats with anti-GBM

GN as well as in patients with Ig A nephropathy (Iida et al., 2014). Therefore, increased miR-

10b and miR-100 levels in urine and detectable but not altered expression in kidney tissue after

NTS, instead of indicating direct glomerular injury, may rather indicate leakage out of damaged

distal nephron regions, for which qualified and specific BMs are also still missing (Huang et al.,

2014). Further, this would be in line with findings for miR-100 being increased in urine after

gentamicin-induced injury affecting proximal and distal convoluted tubules (Nassirpour, et al.,

2014) but not after Cp-induced proximal tubular injury (Kanki, et al., 2014; Pavkovic, et al.,

2014a). Ichii et al.(2014) found miR-10b to be abundantly expressed in mouse glomeruli. This

apparent contrasting result could be due to species differences or to the fact that their

measurement was based on isolated glomeruli only. The same group found miR-26a and -126

as the two most abundantly expressed miRNAs in mouse glomeruli. They also found miR-26a

elevated in urine from a murine GN model and from patients with lupus nephritis. We found

increased urinary miR-26a levels in both our GN model here, and in our previously reported

proximal tubule injury model (Pavkovic, et al., 2014a). Our in situ hybridization experiments as

20
well as PCR data from Ichii et al. (2014) indicate that miR-26a is not exclusively expressed in

glomeruli but also in proximal tubules. On the other hand, miR-126 is reported as specifically

expressed in endothelial cells (Wang et al., 2008), which we confirmed by in situ hybridization.

We did not observe increased levels of miR-34c, as found by Church et al. (2014) after

Doxorubicin-induced glomerular injury. As miR-34-family members can be induced by the DNA

damage responsive transcription factor p53, one explanation may be that the measured miR-

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


34c increase by Church et al. may rather be due to DNA damage induced by Doxorubicin, by its

action as inhibitor of Topoisomerase 2 (Zunino et al., 1990), and not by glomerular damage in

general. This would correspond to an increase of miR-34a in urine and kidney we observed

after Cp treatment, which apart from proximal tubular damage induces DNA strand breaks. MiR-

34c and miR-34a are two miR-34 family members with the same seed sequence. We did

observe, though, an increase of miR-34b in urine and kidneys from WKY rats after NTS

treatment. MiR-34b is known to be clustered with miR-34c (miRbase.org), yet which has a

different seed sequence for target mRNAs than miR-34c and miR-34a. Besides p53, the miR-34

family is also regulated by other transcription factors like Myc or FoxO3a and involved in cell

cycle regulation, apoptosis and epithelial-mesenchymal transition (EMT) (Rokavec et al., 2014).

Therefore it is possible that different members of the miR-34 family are involved in various

functions, including DNA damage response regulation or glomerular injury. Further specific

investigations are needed to clarify details.

With respect to miRNAs indicating kidney injury in general, our GN study corroborates previous

findings suggesting urinary miR-21 and -192 as promising BMs. Both have conserved

sequences in mice, rats, dogs, rhesus monkeys and humans and are expressed in the kidney

(miRBase, 2013). MiR-21 was previously reported as increased in urine form rats with

gentamicin-induced kidney injury and renal ischemia-reperfusion injury (Nassirpour, et al., 2014;

Saikumar, et al., 2012) as well as in urine from patients suffering from nephrotic syndrome,

21
diabetic nephropathy or AKI (Konta et al., 2014; Ramachandran, et al., 2013). Both were found

increased early in urine from rats after Cp-induced kidney injury (Kanki, et al., 2014; Pavkovic,

et al., 2014a). In contrast, these two miRNAs were not increased in urine from rats after

methapyrilene-induced liver injury (Pavkovic, et al., 2014a), suggesting specificity concerning

kidney vs. liver damage.

To obtain insight into molecular processes during GN pathogenesis, mRNA and miRNA

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


expression was investigated in the kidneys after NTS treatment. The immunopathological

processes during GN are still not fully understood and likely involve both innate and adaptive

immunity (Henique et al., 2014). In the present study, several hundred mRNAs were found

deregulated, which functionally were mostly associated with inflammation and regeneration.

The data discussed in this publication have been deposited in NCBI's Gene Expression

Omnibus (Edgar et al., 2002) and are accessible through GEO Series accession number

GSE64265 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE64265).

The β-chain of the major histocompatibility complex class II RT1.B (Rt1-Bb) and complement

component 6 (C6) were identified as two mRNAs differentially expressed between WKY and SD

rat strains which could be related to known different susceptibilities to NTS of different rat

strains (Kawasaki et al., 1992); both were increased in WKY and decreased in SD rats.

Increased Rt1-Bb expression in WKY rats could indicate a stronger and more persistent cellular

reaction of the adaptive immune system in this strain, in line with findings indicating adaptive

immune reactions during GN (Huang et al., 1997; Tam et al., 1999). The complement cascade

is also known to be essential for GN development, especially terminal cascade products like C6

(Nangaku et al., 1997).

The other major functional category indicated to be affected, regeneration, describes mainly

increased expression of genes encoding proteins involved in amino acid synthesis, protein

translation, protein transport and cell adhesion. Some of these mRNAs, which include potential

22
targets for deregulated miRNAs, are reported to be involved in fibrotic pathways. Fibrosis is

described to follow glomerular inflammation in a GN model similar to the one used here (Zhou et

al., 2010). Transforming growth factor β (TGFβ) is known as the main cytokine in renal fibrosis

(Stahl and Felsen, 2001), and several mRNAs related to TGFβ signaling including those coding

for Tgfb2, as well as typical TGFβ down-stream targets like transgelin (Tagln, a.k.a. smooth

muscle protein 22α) and collagen genes, were increased by NTS treatment. Further, TGFβ-

dependent EMT was described in proximal tubular and glomerular epithelia cells which could

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


contribute to the development of fibroblasts during GN (Abbate et al., 2002; Marshall et al.,

2011). In line with this, vimentin (Vim) an activated fibroblasts marker, also found in renal

biopsies from patients with glomerulonephritis (Bob et al., 2014), was increased after NTS

treatment while predicted miRNA-regulators of Vim mRNA, miR-382-5p and let-7g, were

decreased. A further potential regulatory interaction was found here for decreased matrix

metalloproteinase 9 (Mmp9) mRNA and increased miR-132-3p. The MMP9 protein is an

important regulator of extracellular matrix (ECM) degradation, and Kuroda et al. (2004)

described that Mmp9 was decreased after NTS treatment as seen here, leading to increased

ECM. A regulatory connection between Mmp9 and miR-132-3p was also suggested in an in

vitro model for cardiac fibrosis (Jiang et al., 2013).

Increased miR-21 levels in our GN model may also be associated with fibrotic changes in the

kidneys. In a murine model of diabetic nephropathy, also characterized by ECM accumulation,

miR-21 was increased in the kidney and its expression correlated positively with collagen 4

(Col4) and negatively with Mmp9 (Wang et al., 2013) as seen in our GN model.

In summary, we found a considerable number of urinary miRNAs increased after NTS-induced

GN with stronger changes overall in WKY vs. SD rats, confirming the former as more

appropriate to study GN. By comparison of GN-associated urinary miRNA profiles with those

after Cp-induced tubular injury, we found that most had similar profiles, due to also observed

23
tubular injury in our GN model and many miRNAs likely being expressed broadly in the kidney.

Still, five miRNAs could be identified as being potentially GN-specific miRNAs. Investigation of

the localization of two of these, miR-10b and -100, by in situ hybridization in the normal kidney

revealed their expression within distal nephron segments. Thus, although appearance of these

two miRNAs in urine apparently does not indicate glomerular damage, they may represent BMs

for distal tubular injury. Finally, potential mRNA-miRNA regulatory interactions associated with

GN are suggested from their expression profiles in kidney.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015

24
Supplementary figure legends

Supplementary Figure 1 Blood urea nitrogen (BUN; A) and in serum creatinine (sCrea; B)

concentrations in Wistar Kyoto (WKY) and Sprague Dawley (SD) rats 14 days after a single

injection of the indicated nephrotoxic serum (NTS) amount in ml/kg. Shown are means (WKY

control n=3, treated n=5; SD n=6) with standard deviation. Statistical analysis was performed

with ANOVA and Dunnett’s test. Statistical significance is indicated by * p<0.05, ** p<0.01 and

*** p<0.001 compared to time matched control groups.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Supplementary Figure 2 Representative Periodic acid–Schiff (PAS) stained kidney sections of

Wistar Kyoto (WKY) rats 14 days after injection of saline (control) or 5 ml/kg nephrotoxic serum

(NTS). Scale bars: 50 µm.

Supplementary Figure 3 Hierarchical clustering of 74 miRNAs affected by treatment with

nephrotoxic serum (NTS) in urine samples 8 and 14 days. Shown are treated vs. control ratios

derived from TaqMan card data quantified according to the ‘modified ∆Ct approach’. WKY:

Wistar Kyoto, SD: Sprague Dawley.

Supplementary Figure 4 Hierarchical clustering of 253 significantly changed and categorized

mRNAs in kidney samples 14 days after the treatment with NTS. Shown are treated vs. control

ratios derived from measurement with Affymetrix GeneChip arrays. Categories, to which these

deregulated genes were assigned, are indicated on the right. WKY: Wistar Kyoto, SD: Sprague

Dawley, Rt1-Bb: RT1 class II, locus Ba, C6: complement component 6.

Supplementary table legends (all tables are in one Excel file)

Supplementary Table 1 Histopathological findings of kidneys from Wistar Kyoto (WKY) 14

days after and Sprague Dawley (SD) rats 8 and 14 days after a single injection of nephrotoxic

25
serum (NTS). Shown is the number of animals with positive findings from each dose group

(WKY control n=3, treated n=5; SD n=6). Severity score system: grade 0, absent; grade 1,

minimal; grade 2, slight; grade 3, moderate; grade 4, marked; grade 5, very severe. N/E: not

examined.

Supplementary Table 2 Mean ± Standard deviation of protein biomarker concentrations

normalized to urinary creatinine (µg/mmol urinary creatinine). These values were used for fold-

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


change calculations and statistical analysis presented in Tab. 1. WKY: Wistar Kyoto, SD:

Sprague Dawley; N/A: not available; Q: qualified; E: recently endorsed for qualification.

Supplementary Table 3 74 nephrotoxic serum (NTS) affected miRNAs in urine samples 8 and

14 days after treatment. Shown are treated vs. control ratios derived from TaqMan card data

quantified according to the ‘modified ∆Ct approach’ and corresponding Student’st-test p-values;

bold p-values are <0.05 and were considered as significant. WKY: Wistar Kyoto, SD: Sprague

Dawley, rno: Rattus norvegicus. The table corresponds to the heatmap shown in Supplementary

Figure 2.

Supplementary Table 4 253 significantly changed mRNAs in kidney samples 14 days after the

treatment with nephrotoxic serum (NTS). Shown are treated vs. control ratios derived from

measurement with Affymetrix GeneChip arrays and assigned categories for mechanistical

analysis. WKY: Wistar Kyoto, SD: Sprague Dawley. The table corresponds to the heatmap

shown in Supplementary Figure 3.

Supplementary Table 5 80 significantly changed miRNAs in kidney samples 14 days after the

treatment with nephrotoxic serum (NTS). Shown are treated vs. control ratios derived from

26
TaqMan card data quantified according to the ∆∆Ct method with U6 as endogenous control.

WKY: Wistar Kyoto, rno: Rattus norvegicus.

Supplementary Table 6 Expression levels in kidney tissue of the five potentially specific

miRNAs are indicated as mean copies/ng kidney RNA used (with standard deviation) as derived

from miRNA standard curves run in parallel. Results for Wistar Kyoto (WKY) and Sprague

Dawley (SD) are shown (WKY control n=3, treated n=5; SD control and treated n=6). Statistical

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


analysis was performed with Student’s t-test and statistical significance (p<0.05) compared to

time matched control groups is indicated with bold means. N/M: not measured, EQ: measured

with Exiqon’s PCR assays, TM: measured with TaqMan PCR assays.

Supplementary Table 7 Identified target mRNAs of the affected miRNAs in the pool of altered

mRNAs after NTS-induced glomerulonephritis using Ingenuity Pathway Analysis. rno: Rattus

norvegicus. The table corresponds partially to the networks shown in Figure 4.

27
Acknowledgements

We would like to thank Sabine Michel-Kaulmann, Kerstin Noklies, and Brigitte Poschmann for

technical assistance. We especially thank Dr. Ute Bach and Dr. Matthias Rinke for histological

examination of the in situ hybridization data. We furthermore would like to acknowledge the

continuous support by Dr. Thomas Steger-Hartmann and all members of the HESI Biomarkers

of Nephrotoxicity Project Committee for valuable advice and discussion. Finally, we thank Vishal

S. Vaidya, PhD (Harvard Medical School) for helpful discussions during the manuscript

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


preparation.

28
References

Abbate, M., Zoja, C., Rottoli, D., Corna, D., Tomasoni, S., and Remuzzi, G. (2002). Proximal

tubular cells promote fibrogenesis by TGF-beta1-mediated induction of peritubular

myofibroblasts. Kidney Int 61(6), 2066-77.

Bartel, D. P. (2004). MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116(2),

281-97.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Bob, F., Gluhovschi, G., Herman, D., Petrica, L., Bozdog, G., Gluhovschi, C., Velciov, S.,

Gadalean, F., Timar, R., Potencz, E., Dema, A., and Schiller, A. (2014). Immunohistochemical

study of tubular epithelial cells and vascular endothelial cells in glomerulonephritis. Renal failure

36(8), 1208-14.

Church, R. J., McDuffie, J. E., Sonee, M., Otieno, M., Ma, J. Y., Liu, X., Watkinsa, P. B., and

Harrilla, A. H. (2014). MicroRNA-34c-3p is an early predictive biomarker for doxorubicin-induced

glomerular injury progression in male Sprague-Dawley rats. Toxicology Research 3(5), 384-394.

Dieterle, F., Perentes, E., Cordier, A., Roth, D. R., Verdes, P., Grenet, O., Pantano, S., Moulin,

P., Wahl, D., Mahl, A., End, P., Staedtler, F., Legay, F., Carl, K., Laurie, D., Chibout, S. D.,

Vonderscher, J., and Maurer, G. (2010). Urinary clusterin, cystatin C, beta2-microglobulin and

total protein as markers to detect drug-induced kidney injury. Nat Biotechnol 28(5), 463-9.

EMA (2009). Final conclusions of the pilot joint EMEA/FDA VXDA experience on qualification of

nephrotoxicity biomarkers. www.emea.europa.eu Doc.ref. EMEA/679719/2008 Rev.

1(Committee for medicinal products for human use).

29
Dieterle, F., and Sistare, F. (2010). Biomarkers of Acute Kidney Injury. In Biomarkers: In

Medicine, Drug Discovery, and Enviromental Health (V. S. Vaidya, and J. V. Bonventre, Eds.)

doi, pp. 237-263. Wiley & Sons, Inc.

Edgar, R., Domrachev, M., and Lash, A. E. (2002). Gene Expression Omnibus: NCBI gene

expression and hybridization array data repository. Nucleic acids research 30(1), 207-10.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Fu, Z., Qian, F., Yang, X., Jiang, H., Chen, Y., and Liu, S. (2014). Circulating miR-222 in plasma

and its potential diagnostic and prognostic value in gastric cancer. Medical oncology 31(9), 164.

Fuchs, T. C., and Hewitt, P. (2011). Preclinical perspective of urinary biomarkers for the

detection of nephrotoxicity: what we know and what we need to know. Biomarkers in medicine

5(6), 763-79.

Guengerich, F. P. (2011). Mechanisms of drug toxicity and relevance to pharmaceutical

development. Drug Metab Pharmacokinet 26(1), 3-14.

Hartner, A., Hilgers, K. F., Bitzer, M., Veelken, R., and Schocklmann, H. O. (2003). Dynamic

expression patterns of transforming growth factor-beta(2) and transforming growth factor-beta

receptors in experimental glomerulonephritis. J Mol Med (Berl) 81(1), 32-42.

Henique, C., Papista, C., Guyonnet, L., Lenoir, O., and Tharaux, P. L. (2014). Update on

crescentic glomerulonephritis. Seminars in immunopathology 36(4), 479-90.

Hermeking, H. (2012). MicroRNAs in the p53 network: micromanagement of tumour

suppression. Nat Rev Cancer 12(9), 613-26.

30
Ho, J., Ng, K. H., Rosen, S., Dostal, A., Gregory, R. I., and Kreidberg, J. A. (2008). Podocyte-

specific loss of functional microRNAs leads to rapid glomerular and tubular injury. Journal of the

American Society of Nephrology : JASN 19(11), 2069-75.

Hsu, R. K., McCulloch, C. E., Dudley, R. A., Lo, L. J., and Hsu, C. Y. (2013). Temporal changes

in incidence of dialysis-requiring AKI. Journal of the American Society of Nephrology : JASN

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


24(1), 37-42.

Huang, J. X., Blaskovich, M. A., and Cooper, M. A. (2014). Cell- and biomarker-based assays

for predicting nephrotoxicity. Expert opinion on drug metabolism & toxicology 10(12), 1621-35.

Huang, X. R., Tipping, P. G., Apostolopoulos, J., Oettinger, C., D'Souza, M., Milton, G., and

Holdsworth, S. R. (1997). Mechanisms of T cell-induced glomerular injury in anti-glomerular

basement membrane (GBM) glomerulonephritis in rats. Clin Exp Immunol 109(1), 134-42.

Ichii, O., Otsuka-Kanazawa, S., Horino, T., Kimura, J., Nakamura, T., Matsumoto, M., Toi, M.,

and Kon, Y. (2014). Decreased miR-26a expression correlates with the progression of podocyte

injury in autoimmune glomerulonephritis. PloS one 9(10), e110383.

Iida, T., Fujinaka, H., Xu, B., Zhang, Y., Magdeldin, S., Nameta, M., Liu, Z., Yoshida, Y., Yaoita,

E., Tomizawa, S., Saito, A., and Yamamoto, T. (2014). Decreased urinary calbindin 1 levels in

proteinuric rats and humans with distal nephron segment injuries. Clinical and experimental

nephrology 18(3), 432-43.

31
Jensen, O. N. (2004). Modification-specific proteomics: characterization of post-translational

modifications by mass spectrometry. Current opinion in chemical biology 8(1), 33-41.

Jiang, X., Ning, Q., and Wang, J. (2013). Angiotensin II induced differentially expressed

microRNAs in adult rat cardiac fibroblasts. The journal of physiological sciences : JPS 63(1), 31-

8.

Kanki, M., Moriguchi, A., Sasaki, D., Mitori, H., Yamada, A., Unami, A., and Miyamae, Y. (2014).

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Identification of urinary miRNA biomarkers for detecting cisplatin-induced proximal tubular injury

in rats. Toxicology 324, 158-68.

Kanno, K., Okumura, F., Toriumi, W., Ishiyama, N., Nishiyama, S., and Naito, K. (1998).

Nephrotoxic serum-induced nephritis in Wistar-Kyoto rats: a model to evaluate antinephritic

agents. Jpn J Pharmacol 77(2), 129-35.

Kawasaki, K., Yaoita, E., Yamamoto, T., and Kihara, I. (1992). Depletion of CD8 positive cells in

nephrotoxic serum nephritis of WKY rats. Kidney Int 41(6), 1517-26.

Konta, T., Ichikawa, K., Suzuki, K., Kudo, K., Satoh, H., Kamei, K., Nishidate, E., and Kubota, I.

(2014). A microarray analysis of urinary microRNAs in renal diseases. Clinical and experimental

nephrology 18(5), 711-7.

Kriegel, A. J., Liu, Y., Liu, P., Baker, M. A., Hodges, M. R., Hua, X., and Liang, M. (2013).

Characteristics of microRNAs enriched in specific cell types and primary tissue types in solid

organs. Physiol Genomics 45(23), 1144-56.

32
Krol, J., Loedige, I., and Filipowicz, W. (2010). The widespread regulation of microRNA

biogenesis, function and decay. Nat Rev Genet 11(9), 597-610.

Kuroda, T., Yoshida, Y., Kamiie, J., Kovalenko, P., Nameta, M., Fujinaka, H., Yaoita, E., Endo,

T., Ishizuka, S., Nakabayashi, K., Yamada, A., Nagasawa, T., and Yamamoto, T. (2004).

Expression of MMP-9 in mesangial cells and its changes in anti-GBM glomerulonephritis in

WKY rats. Clinical and experimental nephrology 8(3), 206-15.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Lai, J. Y., Luo, J., O'Connor, C., Jing, X., Nair, V., Ju, W., Randolph, A., Ben-Dov, I. Z., Matar,

R. N., Briskin, D., Zavadil, J., Nelson, R. G., Tuschl, T., Brosius, F. C., 3rd, Kretzler, M., and

Bitzer, M. (2014). MicroRNA-21 in Glomerular Injury. Journal of the American Society of

Nephrology : JASN doi: 10.1681/ASN.2013121274.

Landgraf, P., Rusu, M., Sheridan, R., Sewer, A., Iovino, N., Aravin, A., Pfeffer, S., Rice, A.,

Kamphorst, A. O., Landthaler, M., Lin, C., Socci, N. D., Hermida, L., Fulci, V., Chiaretti, S., Foa,

R., Schliwka, J., Fuchs, U., Novosel, A., Muller, R. U., Schermer, B., Bissels, U., Inman, J.,

Phan, Q., Chien, M., Weir, D. B., Choksi, R., De Vita, G., Frezzetti, D., Trompeter, H. I.,

Hornung, V., Teng, G., Hartmann, G., Palkovits, M., Di Lauro, R., Wernet, P., Macino, G.,

Rogler, C. E., Nagle, J. W., Ju, J., Papavasiliou, F. N., Benzing, T., Lichter, P., Tam, W.,

Brownstein, M. J., Bosio, A., Borkhardt, A., Russo, J. J., Sander, C., Zavolan, M., and Tuschl, T.

(2007). A mammalian microRNA expression atlas based on small RNA library sequencing. Cell

129(7), 1401-14.

Mall, C., Rocke, D. M., Durbin-Johnson, B., and Weiss, R. H. (2013). Stability of miRNA in

human urine supports its biomarker potential. Biomarkers in medicine 7(4), 623-31.

33
Marshall, C. B., Krofft, R. D., Blonski, M. J., Kowalewska, J., Logar, C. M., Pippin, J. W., Kim, F.,

Feil, R., Alpers, C. E., and Shankland, S. J. (2011). Role of smooth muscle protein SM22alpha

in glomerular epithelial cell injury. American journal of physiology. Renal physiology 300(4),

F1026-42.

miRBase (2013). http://mirbase.org/. In (Vol. 20. Faculty of Life Sciences at the University of

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Manchester with funding from the BBSRC, and was previously hosted and supported by the

Wellcome Trust Sanger Institute.

Nangaku, M., Alpers, C. E., Pippin, J., Shankland, S. J., Kurokawa, K., Adler, S., Johnson, R. J.,

and Couser, W. G. (1997). Renal microvascular injury induced by antibody to glomerular

endothelial cells is mediated by C5b-9. Kidney Int 52(6), 1570-8.

Nassirpour, R., Mathur, S., Gosink, M. M., Li, Y., Shoieb, A. M., Wood, J., O'Neil, S. P., Homer,

B. L., and Whiteley, L. O. (2014). Identification of tubular injury microRNA biomarkers in urine:

comparison of next-generation sequencing and qPCR-based profiling platforms. BMC genomics

15, 485.

Nishimura, Y., Kondo, C., Morikawa, Y., Tonomura, Y., Torii, M., Yamate, J., and Uehara, T.

(2014). Plasma miR-208 as a useful biomarker for drug-induced cardiotoxicity in rats. Journal of

applied toxicology : JAT doi: 10.1002/jat.3044.

Pavkovic, M., Riefke, B., and Ellinger-Ziegelbauer, H. (2014a). Urinary microRNA profiling for

identification of biomarkers after cisplatin-induced kidney injury. Toxicology 324, 147-57.

34
Pavkovic, M., Riefke, B., Gutberlet, K., Raschke, M., and Ellinger-Ziegelbauer, H. (2014b).

Comparison of the MesoScale Discovery and Luminex multiplex platforms for measurement of

urinary biomarkers in a cisplatin rat kidney injury model. J Pharmacol Toxicol Methods 69(2),

196-204.

Pusey, C. D. (2003). Anti-glomerular basement membrane disease. Kidney Int 64(4), 1535-50.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Ramachandran, K., Saikumar, J., Bijol, V., Koyner, J. L., Qian, J., Betensky, R. A., Waikar, S.

S., and Vaidya, V. S. (2013). Human miRNome profiling identifies microRNAs differentially

present in the urine after kidney injury. Clin Chem 59(12), 1742-52.

Rokavec, M., Li, H., Jiang, L., and Hermeking, H. (2014). The p53/miR-34 axis in development

and disease. Journal of molecular cell biology 6(3), 214-30.

Saikumar, J., Hoffmann, D., Kim, T. M., Gonzalez, V. R., Zhang, Q., Goering, P. L., Brown, R.

P., Bijol, V., Park, P. J., Waikar, S. S., and Vaidya, V. S. (2012). Expression, circulation, and

excretion profile of microRNA-21, -155, and -18a following acute kidney injury. Toxicol Sci

129(2), 256-67.

Saikumar, J., Ramachandran, K., and Vaidya, V. S. (2014). Noninvasive micromarkers. Clin

Chem 60(9), 1158-73.

Salant, D. J., and Cybulsky, A. V. (1988). Experimental glomerulonephritis. Methods Enzymol

162, 421-61.

35
Schena, F. P., Serino, G., and Sallustio, F. (2014). MicroRNAs in kidney diseases: new

promising biomarkers for diagnosis and monitoring. Nephrology, dialysis, transplantation :

official publication of the European Dialysis and Transplant Association - European Renal

Association 29(4), 755-63.

Small, E. M., and Olson, E. N. (2011). Pervasive roles of microRNAs in cardiovascular biology.

Nature 469(7330), 336-42.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Stahl, P. J., and Felsen, D. (2001). Transforming growth factor-beta, basement membrane, and

epithelial-mesenchymal transdifferentiation: implications for fibrosis in kidney disease. Am J

Pathol 159(4), 1187-92.

Swain, A., Turton, J., Scudamore, C. L., Pereira, I., Viswanathan, N., Smyth, R., Munday, M.,

McClure, F., Gandhi, M., Sondh, S., and York, M. (2011). Urinary biomarkers in hexachloro-1:3-

butadiene-induced acute kidney injury in the female Hanover Wistar rat; correlation of alpha-

glutathione S-transferase, albumin and kidney injury molecule-1 with histopathology and gene

expression. Journal of applied toxicology : JAT 31(4), 366-77.

Tam, F. W., Smith, J., Morel, D., Karkar, A. M., Thompson, E. M., Cook, H. T., and Pusey, C. D.

(1999). Development of scarring and renal failure in a rat model of crescentic

glomerulonephritis. Nephrology, dialysis, transplantation : official publication of the European

Dialysis and Transplant Association - European Renal Association 14(7), 1658-66.

Tian, Z., Greene, A. S., Pietrusz, J. L., Matus, I. R., and Liang, M. (2008). MicroRNA-target pairs

in the rat kidney identified by microRNA microarray, proteomic, and bioinformatic analysis.

Genome Res 18(3), 404-11.

36
Togashi, Y., Imura, N., and Miyamoto, Y. (2013). Urinary cystatin C as a renal biomarker and its

immunohistochemical localization in anti-GBM glomerulonephritis rats. Exp Toxicol Pathol 65,

1137-1143.

Urbschat, A., Obermuller, N., and Haferkamp, A. (2011). Biomarkers of kidney injury.

Biomarkers 16 Suppl 1, S22-30.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Vaidya, V. S., Ferguson, M. A., and Bonventre, J. V. (2008). Biomarkers of acute kidney injury.

Annu Rev Pharmacol Toxicol 48, 463-93.

Vaidya, V. S., Ozer, J. S., Dieterle, F., Collings, F. B., Ramirez, V., Troth, S., Muniappa, N.,

Thudium, D., Gerhold, D., Holder, D. J., Bobadilla, N. A., Marrer, E., Perentes, E., Cordier, A.,

Vonderscher, J., Maurer, G., Goering, P. L., Sistare, F. D., and Bonventre, J. V. (2010). Kidney

injury molecule-1 outperforms traditional biomarkers of kidney injury in preclinical biomarker

qualification studies. Nat Biotechnol 28(5), 478-85.

Wang, G., Tam, L. S., Li, E. K., Kwan, B. C., Chow, K. M., Luk, C. C., Li, P. K., and Szeto, C. C.

(2011). Serum and urinary free microRNA level in patients with systemic lupus erythematosus.

Lupus 20(5), 493-500.

Wang, J., Gao, Y., Ma, M., Li, M., Zou, D., Yang, J., Zhu, Z., and Zhao, X. (2013). Effect of miR-

21 on Renal Fibrosis by Regulating MMP-9 and TIMP1 in kk-ay Diabetic Nephropathy Mice. Cell

Biochem Biophys 67(2), 537-546.

37
Wang, S., Aurora, A. B., Johnson, B. A., Qi, X., McAnally, J., Hill, J. A., Richardson, J. A.,

Bassel-Duby, R., and Olson, E. N. (2008). The endothelial-specific microRNA miR-126 governs

vascular integrity and angiogenesis. Developmental cell 15(2), 261-71.

Wang, Y., Chen, T., and Tong, W. (2014). miRNAs and their application in drug-induced liver

injury. Biomarkers in medicine 8(2), 161-72.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Weber, J. A., Baxter, D. H., Zhang, S., Huang, D. Y., Huang, K. H., Lee, M. J., Galas, D. J., and

Wang, K. (2010). The microRNA spectrum in 12 body fluids. Clin Chem 56(11), 1733-41.

Zhou, C., Wu, J., Torres, L., Hicks, J. M., Bartkowiak, T., Parker, K., and Lou, Y. H. (2010).

Blockade of osteopontin inhibits glomerular fibrosis in a model of anti-glomerular basement

membrane glomerulonephritis. American journal of nephrology 32(4), 324-31.

Zunino, F., and Capranico, G. (1990). DNA topoisomerase II as the primary target of anti-tumor

anthracyclines. Anti-cancer drug design 5(4), 307-17.

38
Footnote
1
The Health and Environmental Sciences Institute (HESI) is a nonprofit institution whose

mission is to engage scientists from academia, government and industry to identify and resolve

global health and environmental issues.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015

39
Figure legends

Figure 1 Histopathological changes in the kidney of rats administered nephrotoxic serum (NTS).

14 days after the injection of 5 ml/kg NTS, enlarged glomeruli with hypercellularity, increased

amounts of eosinophilic glomerular matrix (white arrow) and hyperplasia of the Bowman’s

capsule (black arrow) were observed in animals give NTS (B), but not in rats given vehicle (A).

Shown are representative haematoxilin and eosin stained kidney sections from Wistar Kyoto

rats. Scale bars: 50 µm.

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Figure 2 Increased urinary total protein levels in Wistar Kyoto (WKY) and Sprague Dawley (SD)

rats 8 and 14 days after administration of different doses of NTS. Shown are the means (WKY

control n=3, treated n=5; SD n=6) with standard deviation from urinary creatinine (uCrea)

normalized concentrations. Statistical analysis was performed with ANOVA and Dunnett’s test.

Statistical significance is indicated by * p<0.05, ** p<0.01 and *** p<0.001 compared to time

matched control groups.

Figure 3 Venn diagram of urinary miRNAs selected to be significantly affected after treatment of

nephrotoxic serum or cisplatin.

Figure 4 U6 as general positive control (A), miR-126 as positive control for endothelia cells (B),

miR-26a (C) and miR-192 (D) expression observed by in situ hybridization in the kidney from a

healthy male Wistar rat. Using the scrambled probe as negative control (E) no staining was

observed. Blue: positive probe signal, pink: eosin counter staining, black arrows point to

positively stained glomerular cells, white arrows point to positively stained proximal tubular cells,

the grey arrow points to positively stained endothelial cells.

40
Figure 5 Localization of miR-10b (A and C) and miR-100 (B and D) expression observed by in

situ hybridization in the kidney from a healthy male Wistar rat. C and D are magnifications of A

and B, respectively. Blue: positive probe signal, pink: eosin counter staining, black arrows point

to positive staining in distal tubules.

Figure 6 Mechanistic analysis of miRNAs and mRNAs deregulated in the kidney after NTS

administration. Using Ingenuity Pathway Analysis target mRNAs of the affected miRNAs were

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


identified in the pool of deregulated mRNAs after NTS-induced glomerulonephritis. A) miRNAs

and potential target mRNAs associated with fibrosis. B) miRNAs and potential target mRNAs

associated with the TGFβ-pathway. The color represents NTS-induced increase (red shading)

and decrease (green shading) in kidneys of Wistar Kyoto rats 14 days after NTS administration.

Black and gray arrows represent miRNA-mRNA interaction showing and not showing inverse

changes in expression levels after NTS treatment, respectively.

41
Table 1 Protein biomarker measurement in rat urine after administration of nephrotoxic serum (NTS).

WKY SD

1 ml/kg NTS 2.5 ml/kg NTS 5 ml/kg NTS 1.5 ml/kg NTS 5 ml/kg NTS

Biomarker Day 8 Day 14 Day 8 Day 14 Day 8 Day 14 Day 8 Day 14 Day 8 Day 14

ALBQ 353 ** (t) 201 N/A 4204 ** N/A 2836 * 100 105 3216 ** (t) N/A

β2MQ 6.5 2.7 41 88.1 *** 121 *** 119 *** 1.1 1 38.4 ** 66 ***

KIM-1Q 1.2 0.9 73 6.6 * 200 ** 19.6 *** 2.4 1.3 50.9 * 13 **

CLUQ 1.5 2 237 55.4 1230 *** 349 ** 2 1.5 472 136

CysCQ 1.2 1.3 15 ** 5.6 *** 31.4 *** 8.2 *** 1.4 1.2 9.9 *** 4.3 **

NGALE 1.1 2.4 4.3 5.3 *** 12 *** 9.2 *** 3.8 4.4 6.7 ** 6.3 *

OPNE 0.7 1.9 1.2 1.5 29.7 * 4.8 2.1 2 9.5 3.3

aGST 1.7 1 4.6 1.9 19.2 *** 3.8 *** 3.7 1.9 8.3 * 0.7

TIMP-1 2.3 1.5 111 46.1 549 ** 58.1 3.2 1.2 34.3 * 2.8

VEGF 1.8 1.1 4.4 *** 2.1 8.3 *** 3.5 ** 2.1 1.3 3.7 ** 1.5 *

CALB 0.8 1.2 0.9 1 1.1 1.4 1.3 1.3 0.8 1.2

Biomarker levels urinary creatinine normalized and relative to the mean of time-matched control groups measured in urine from
Wistar Kyoto (WKY) and Sprague Dawley (SD) rats after a single injection of NTS. Statistical analysis: ANOVA and Dunnett’s test or
Student’s t-test (t) with * p<0.05, ** p<0.01 and *** p<0.001 compared to time matched control groups. N/A: not available; Q:
qualified; E: recently endorsed for qualification.

42

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Table 2 Profiles of 12 selected miRNAs using standard curve quantification.
WKY SD

1 ml/kg NTS 2.5 ml/kg NTS 5 ml/kg NTS 1.5 ml/kg NTS 5 ml/kg NTS

miRNA Day 8 Day 14 Day 8 Day 14 Day 8 Day 14 Day 8 Day 14 Day 8 Day 14

#,TM
miR-10a 0.6 ± 0.1* 0.3 ± 0.1 1.4 ± 0.6* 1.1 ± 0.7 3.5 ± 2.4* 1.1 ± 0.7 0.78 ± 0.73 0.89 ± 0.28 1.15 ± 0.55 2.02 ± 2.12
#,EQ
miR-10b 1.0 ± 0.3 63.9 ± 16.8* 0.0 ± 0.0 66.1 ± 129.4 63.4 ± 118.4* 0.7 ± 0.5
#,EQ
miR-100 0.9 ± 0.3 37.6 ± 12.9 0.0 ± 0.0 102.5 ± 143.0 38.5 ± 68.7* 0.8 ± 0.9 N/M
#,EQ
miR-486 0.8 ± 0.2 12.4 ± 3.2 0.0 ± 0.0 9.7 ± 18.2 15.4 ± 25.3* 0.7 ± 0.2
TM
miR-197 1.3 ± 0.2 1.9 ± 0.3 0.6 ± 0.2* 3.0 ± 4.5 4.9 ± 2.0** 1.5 ± 0.4 0.73 ± 0.05 1.55 ± 1.63 1.25 ± 0.57 1.60 ± 0.64
#,TM
miR-211 1.4 ± 0.3 1.6 ± 0.4 1.0 ± 0.4* 2.6 ± 3.0 4.8 ± 1.8** 1.8 ± 0.4* 0.88 ± 0.09 1.94 ± 2.86 1.76 ± 1.01 1.80 ± 0.73*
TM
miR-21 2.2 ± 1.5 1.1 ± 0.7 4.6 ± 4.7* 1.2 ± 0.5 4.2 ± 1.1* 2.8 ± 0.6** 0.78 ± 0.25 1.02 ± 0.41 5.81 ± 4.40** 2.26 ± 1.64
TM
miR-192 1.6 ± 1.4 2.3 ± 2.0 1.9 ± 1.7 1.8 ± 0.5 1.7 ± 0.6 3.9 ± 2.1* 0.31 ± 0.18** 0.84 ± 0.59 2.80 ± 3.04 2.12 ± 1.46
EQ
miR-26a 1.5 ± 0.5 1.3 ± 0.4 1.9 ± 0.7 1.2 ± 0.7 1.0 ± 0.3 1.6 ± 0.2** N/M
TM
miR-34a 4.5 ± 3.3 5.4 ± 3.9 1.1 ± 1.1 2.0 ± 2.8 4.0 ± 2.6 2.0 ± 0.3 0.58 ± 0.37 1.37 ± 0.81 0.59 ± 0.46 1.16 ± 0.79
TM
miR-34b 5.3 ± 7.3 2.5 ± 3.4 2.8 ± 3.6 0.5 ± 0.2 4.2 ± 2.6 1.3 ± 0.5 0.43 ± 0.52 0.94 ± 0.61 4.75 ± 3.69 1.78 ± 1.76
TM
miR-184 1.9 ± 1.6 5.8 ± 5.1 1.0 ± 1.0 2.8 ± 2.7 2.7 ± 1.5 6.0 ± 7.0 0.21 ± 0.13** 0.60 ± 0.32 0.73 ± 0.48 2.52 ± 2.96

Shown are treated vs. control ratio means ± standard deviation of urinary miRNA levels after injection of Wistar Kyoto (WKY) and
Sprague Dawley (SD) with NTS (WKY control n=3, treated n=5; SD n=6) derived from urinary creatinine normalized absolute miRNA
concentrations. Statistical analysis was performed with Student’s t-test and statistical significance is indicated by * p<0.05, ** p<0.01

43

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


and *** p<0.001 compared to time matched control groups. #: apparently NTS-specific, N/M: not measured, E: measured with
Exiqon’s PCR assays, T: measured with TaqMan PCR assays.

44

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015
Fig.1. Histopathological changes in the kidney of rats administered nephrotoxic serum (NTS). 14 days after
the injection of 5 ml/kg NTS, enlarged glomeruli with hypercellularity, increased amounts of eosinophilic
glomerular matrix (white arrow) and hyperplasia of the Bowman’s capsule (black arrow) were observed in
animals give NTS (B), but not in rats given vehicle (A). Shown are representative haematoxilin and eosin
stained kidney sections from Wistar Kyoto rats. Scale bars: 50 µm
87x33mm (300 x 300 DPI)
1

Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015


Fig. 2. Increased urinary total protein levels in Wistar Kyoto (WKY) and Sprague Dawley (SD) rats 8 and 14
days after administration of different doses of NTS. Shown are the means (WKY control n=3, treated n=5;
SD n=6) with standard deviation from urinary creatinine (uCrea) normalized concentrations. Statistical
analysis was performed with ANOVA and Dunnett’s test. Statistical significance is indicated by * p<0.05, **
p<0.01 and *** p<0.001 compared to time matched control groups.
61x46mm (300 x 300 DPI)
Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015
Fig.3. Venn diagram of urinary miRNAs selected to be significantly affected after treatment of nephrotoxic
serum or cisplatin
70x55mm (300 x 300 DPI)
Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015
Fig.4. U6 as general positive control (A), miR-126 as positive control for endothelia cells (B), miR-26a (C)
and miR-192 (D) expression observed by in situ hybridization in the kidney from a healthy male Wistar rat.
Using the scrambled probe as negative control (E) no staining was observed. Blue: positive probe signal,
pink: eosin counter staining, black arrows point to positively stained glomerular cells, white arrows point to
positively stained proximal tubular cells, the grey arrow points to positively stained endothelial cells.
103x102mm (300 x 300 DPI)
Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015
Fig.5. Localization of miR-10b (A and C) and miR-100 (B and D) expression observed by in situ hybridization
in the kidney from a healthy male Wistar rat. C and D are magnifications of A and B, respectively. Blue:
positive probe signal, pink: eosin counter staining, black arrows point to positive staining in distal tubules.
106x72mm (300 x 300 DPI)
Downloaded from http://toxsci.oxfordjournals.org/ at University of Toledo on March 15, 2015
Fig.6. Mechanistic analysis of miRNAs and mRNAs deregulated in the kidney after NTS administration. Using
Ingenuity Pathway Analysis target mRNAs of the affected miRNAs were identified in the pool of deregulated
mRNAs after NTS-induced glomerulonephritis. A) miRNAs and potential target mRNAs associated with
fibrosis. B) miRNAs and potential target mRNAs associated with the TGFβ-pathway. The color represents
NTS-induced increase (red shading) and decrease (green shading) in kidneys of Wistar Kyoto rats 14 days
after NTS administration. Black and gray arrows represent miRNA-mRNA interaction showing and not
showing inverse changes in expression levels after NTS treatment, respectively.
103x63mm (300 x 300 DPI)

You might also like