You are on page 1of 15

Genomics and Risk Assessment | Mini-Monograph

Identification of Putative Gene-Based Markers of Renal Toxicity


Rupesh P. Amin,1 Alison E. Vickers,2 Frank Sistare,3 Karol L. Thompson,3 Richard J. Roman,4 Michael Lawton,5
Jeffrey Kramer,5 Hisham K. Hamadeh,1,6 Jennifer Collins,1 Sherry Grissom,1 Lee Bennett,1 C. Jeffrey Tucker,1
Stacie Wild,6 Clive Kind,7 Victor Oreffo,7 John W. Davis II,8 Sandra Curtiss,5 Jorge M. Naciff,9 Michael
Cunningham,1 Raymond Tennant,1 James Stevens,10 Bruce Car,11 Timothy A. Bertram,5 and Cynthia A. Afshari1,6
1National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research
Triangle Park, North Carolina, USA; 2Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA; 3Center for Drug
Evaluation and Research, U.S. Food and Drug Administration, Laurel, Maryland, USA; 4Medical College of Wisconsin and Physiogenix
Inc., Milwaukee, Wisconsin, USA; 5Pfizer Inc, St. Louis, Missouri, USA, and Groton, Connecticut, USA; 6Amgen Inc., Thousand Oaks,
California, USA; 7AstraZeneca Inc., Leicestershire, United Kingdom; 8Schering-Plough Research Institute, Lafayette, New Jersey, USA;
9The Procter & Gamble Company, Miami Valley Laboratories, Cincinnati, Ohio, USA; 10Eli Lilly and Company, Indianapolis, Indiana,
USA; 11Bristol-Myers Squibb Company, Wilmington, Delaware, USA

expression profiles derived from kidneys


This study, designed and conducted as part of the International Life Sciences Institute working group exposed to these three nephrotoxicants at
on the Application of Genomics and Proteomics, examined the changes in the expression profile of multiple doses and time points (associated
genes associated with the administration of three different nephrotoxicants—cisplatin, gentamicin, with varying types and severity of toxicity),
and puromycin—to assess the usefulness of microarrays in the understanding of mechanism(s) of we hypothesized that we could identify pat-
nephrotoxicity. Male Sprague-Dawley rats were treated with daily doses of puromycin (5–20 terns of gene expression that reflect differ-
mg/kg/day for 21 days), gentamicin (2–240 mg/kg/day for 7 days), or a single dose of cisplatin ent types of nephrotoxicity. An important
(0.1–5 mg/kg). Groups of rats were sacrificed at various times after administration of these com- outcome is the identification of potential
pounds for standard clinical chemistry, urine analysis, and histological evaluation of the kidney. RNA gene-based markers of nephrotoxicity and
was extracted from the kidney for microarray analysis. Principal component analysis and gene expres- evidence that one can detect region-specific
sion–based clustering of compound effects confirmed sample separation based on dose, time, and renal toxicity using microarray technology.
degree of renal toxicity. In addition, analysis of the profile components revealed some novel changes Additional functional genomics studies may
in the expression of genes that appeared to be associated with injury in specific portions of the afford the opportunity to validate the pro-
nephron and reflected the mechanism of action of these various nephrotoxicants. For example, posed novel gene-based markers of nephro-
although puromycin is thought to specifically promote injury of the podocytes in the glomerulus, the toxicity, which in part may improve current
changes in gene expression after chronic exposure of this compound suggested a pattern similar to the sensitivity to assessing xenobiotic-induced
known proximal tubular nephrotoxicants cisplatin and gentamicin; this prediction was confirmed his- nephrotoxicity.
tologically. We conclude that renal gene expression profiling coupled with analysis of classical end
points affords promising opportunities to reveal potential new mechanistic markers of renal toxicity. Materials and Methods
Key words: biomarkers, cisplatin, gentamicin, microarrays, nephrotoxicity, proximal tubule, General
puromycin, toxicogenomics. Environ Health Perspect 112:465–479 (2004). doi:10.1289/txg.6683
available via http://dx.doi.org/ [Online 15 January 2004] This study was conducted in accordance
with Good Laboratory Practices. All
animals were handled and treated in accor-
Renal toxicity commonly occurs after admin- Recent application of microarray-based dance to the NIH Guide for the Care
istration of xenobiotics and a variety of phar- gene profiling has provided some unique and Use of Laboratory Animals (NIH
maceutical agents. The process is typically insights into compound-specific and 1999). Experiments were performed on
initiated by a toxic injury to tubular epithelial toxicity-related gene expression changes in
cells in various nephron segments or by the liver (Hamadeh et al. 2002a, 2002b; This article is part of the mini-monograph
injury to specific cell types in the glomerulus. Waring et al. 2001a, 2001b), and several “Application of Genomics to Mechanism-Based
The initial injury is often followed by cellular laboratories have applied similar techniques Risk Assessment.”
proliferation and repair that attempts to to identify gene expression changes related Address correspondence to C. Afshari, Amgen
restore normal renal function (Toback 1992). to nephrotoxicity (Huang et al. 2001; Inc., One Amgen Center Dr., MS 5-1-A, Thousand
Changes in the expression of mRNA specifi- Nagasawa et al. 2001; Yoshida et al. 2002). Oaks, CA 91320 USA. Telephone: (805) 447-3537.
Fax: (805) 449-4687. E-mail: cafshari@amgen.com
cally expressed in the injured kidney cells are This study, conducted as a consortium We would like to thank the following people: J.
some of the earliest events that accompany collaboration with the International Life Bonventure for donation of antibody reagents, and
renal injury. This is accompanied by changes Sciences Institute (ILSI) Health and S. Pettit, S. Susanne, P. Bushel, and R. Paules for
in the expression of other genes that con- Environmental Sciences Institute (HESI) maintenance of and posting of data to the
tribute either to cellular repair or recovery of (Kramer et al. 2004; Pennie et al. 2004), International Life Sciences Institute (ILSI) and the
renal function or in those that mediate fibro- compares kidney gene expression profiles National Institute of Environmental Health
Sciences web sites. Our appreciation to B. Pennie,
sis and further pathology of the kidney induced by cisplatin and gentamicin, com- D. Robinson, and R. Tyler for their leadership role
(Matejka 1998; Norman et al. 1988; pounds that injure the proximal tubule, in the establishment of this ILSI working group.
Safirstein et al. 1990). For example, eleva- and puromycin, which produces protein- We thank members of the Health and
tions in the expression of heme oxygenase I uria by selectively damaging podocytes, a Environmental Health Sciences (HESI) Technical
(HO-1), kidney injury molecule-1 (KIM-1), key component of the glomerular filtration Committee on Application of Genomics to
clusterin, thymosin beta 4, osteopontin, and barrier to plasma proteins. Conventional Mechanism-Based Risk Assessment for critically
reviewing this manuscript prior to submission.
several growth factors have been reported in clinical chemistry and histopathological or The authors declare they have no competing
various models of renal injury (Hammerman ultrastructural findings confirmed the financial interests.
1998a, 1998b; Huang et al. 2001; Ichimura induction of tubular or glomerular injury in Received 18 August 2003; accepted 14 January
et al. 1998; Yoshida et al. 2002). these studies. Through comparison of the 2004.

Environmental Health Perspectives • VOLUME 112 | NUMBER 4 | March 2004 465


Mini-Monograph | Amin et al.

approximately 8- to 9-week-old male Tissue Collection Duncan’s multiple range post hoc analysis
(approximately 250–300 g) Sprague- After CO2 anesthesia, a sample of blood was (more than two groups) or a Student t test
Dawley rats Crl:CD (SD)IGS BR VAF+ collected from the vena cava for standard for comparison between two mean values.
(abbreviated SD) purchased from Charles clinical chemistry analysis. The kidneys were
River Breeding Laboratories (Raleigh, NC). collected, weighed, and stored frozen. Half RNA Extraction
Food [Purina Rodent Chow no. 5002 the kidney and the left lobe of the liver were Homogenized kidney samples (one of two
(Purina Milling Inc., St. Louis, MO), or placed in 10% neutral buffered formalin for 15-mL samples per animal) were processed
Certified Rodent Diet 18% no. 5LG3, histology, and the other half was flash-frozen using a Qiagen RNeasy Maxi kit (Qiagen)
(PMI Feeds, Richmond, IN)], except for in liquid nitrogen and stored at –80°C for according to the manufacturer instructions.
study-defined fasting procedures, and water Western blot or immunohistochemical Final RNA product was quantified from the
were available ad libitum. analysis. The right kidney was minced, ultraviolet absorbance at 260 nm (Beckman
transferred to 5 mL RLT with β-mercapto- DU520UV/Vis Spectrophotometer;
Test Article and Treatment ethanol (+ BME) buffer (Qiagen, Valencia, Beckman Coulter Inc., Fullerton, CA). The
Cisplatin (CAS no. 15663-27-1; lot CA) in a 50-mL centrifuge tube and quality of the RNA was evaluated by mea-
059H3657), gentamicin sulfate (CAS no. homogenized for 60–120 sec. Additional suring the 260-/280-nm ratios and con-
1405410; lot 50K0924), and puromycin RLT (+ BME) buffer was added to achieve a firmed by formaldehyde agarose gels or
dihydrochloride hydrate (CAS no. 58-58-2; final volume of 30 mL, which was divided analysis of the sample on a Bioanalyzer
Lot 080K4031) were purchased from into 15-mL aliquots and stored at –80°C (Agilent Technologies, Palo Alto, CA).
Sigma Chemical Company (St. Louis, until the RNA was extracted. RNA was deemed of a suitable quality for
MO). Doses were selected based on previ- microarray analysis if the 260/280 ratio was
ous reports that documented histopatholog- Histology between 1.6 and 2.0 and the gel and
ical changes in the kidney by the 7-day time The tissues were fixed for a maximum of Bioanalyzer traces showed no visible degra-
point or later. The rats received a single ip 48 hr and then transferred to 70% ethanol dation products lower than the 18S riboso-
injection of vehicle (0.9% NaCl solution) for up to 10 days before being placed in mal band. An equal amount of RNA
or cisplatin at a dose of 0.3, 1, or 5 mg/kg paraffin. The tissues were embedded in extracted from the kidney of all rats in a
(20 per group). Cisplatin- and vehicle- paraffin blocks, cut into 5-µm sections, and treatment group was pooled into single
treated rats were sacrificed by inhalation of stained with hematoxylin and eosin samples. In select instances RNA from kid-
carbon dioxide 4, 24, 48, and 144 hr after (H&E). Ultrastructural analysis using elec- ney of individual animals was also analyzed
injection of vehicle or cisplatin. This por- tron microscopy was performed on kidney by microarray and/or by RT–PCR. Samples
tion of the study was conducted at Pfizer tissue obtained from animals treated with were stored at –80°C until hybridization.
Inc (Groton, CT). gentamicin. In this case the kidney tissue
Gentamicin was dissolved in a sterile was fixed with paraformaldehyde rather cDNA Microarray Manufacturing
0.9% (w/v) NaCl solution and the animals than formalin. Gene expression analysis was conducted on
were treated with daily ip injections of the same RNA samples using several plat-
vehicle or gentamicin at doses of 2, 20, 80 Clinical Chemistry and Urinalysis forms including Affymetrix (Santa Clara,
or 240 mg/kg/day for up to 7 days. A At scheduled necropsies, blood (fasted, vena CA) and NIEHS custom cDNA microar-
group of rats (three to five per group) were cava bleeding; approximately 2–3 mL) was rays. The analysis of samples using the
sacrificed at 4, 24, 48, and 144 hr after the collected into serum separation collection Affymetrix platform was performed accord-
initial administration of gentamicin. This tubes and allowed to clot. Serum was col- ing to the manufacturer details (Affymetrix
portion of the study was conducted at lected (centrifugation at approximately 4°C, 2003; Lockhart et al. 1996) and is detailed
Novartis Pharmaceuticals (East Hanover, 1,500 × g, 10 min) and stored frozen at –70 in the accompanying paper (Thompson
NJ) and the Medical College of Wisconsin to –80°C for routine serum biochemistry et al. 2004).
(Madison, WI). analysis. Albumin, alkaline phosphatase, The analysis using the custom cDNA
Puromycin was dissolved in sterile alanine amino transferase, aspartate amino microarray manufactured at NIEHS is as
0.9% (w/v) NaCl solution. The rats transferase, bilirubin, blood urea nitrogen follows: Sequence-verified rat cDNA clones
received daily ip injections of vehicle or (BUN), calcium (Ca2+), chloride, creati- that covered the 3´ end of the gene and
puromycin at doses of 5 or 20 mg/kg/day nine, gamma glutamyltransferase, glucose, ranged in size from 500 to 2,000 bp
for up to 21 days. Puromycin-treated and magnesium, phosphate, 5´-nucleotidase, (Research Genetics, Huntsville, AL) were
time-matched vehicle control animals were potassium, sorbitol dehydrogenase, sodium, printed on glass slides. M13 primers were
sacrificed at 4, 24, 48, and 144 hr, and 21 total cholesterol, and total protein were used to amplify insert cDNAs from puri-
days after the first exposure to the com- among the parameters measured using a fied plasmid DNA in a 100-µL polymerase
pound. This portion of the study was co- standard clinical chemistry analyzer. chain reaction (PCR) reaction mixture. A
ordinated by staff from the National Urinary creatinine, total protein, glucose, sample of the PCR products (10 µL) was
Institute of Environmental Health Sciences calcium, sodium, potassium, chloride, mag- separated on 2% agarose gels to ensure
(NIEHS; Research Triangle Park, NC). nesium, and phosphorus concentrations quality of the amplifications. The remain-
were also analyzed. ing PCR products were purified by ethanol
Urine Collection precipitation, resuspended in ArrayIt buffer
Animals were placed in standard metabolic Statistical Analysis (TeleChem Intl. Inc., Sunnyvale, CA), and
cages overnight (approximately 12–14 hr) Mean values ± 1 standard error (SE) are spotted onto poly- L -lysine–coated glass
prior to necropsy. Food but not water was presented for serum biochemistry, urine slides using a modified robotic DNA
removed from the animals during this chemistry, and organ and body weight meas- arrayer (Beecher Instruments, Bethesda
time. Urine volumes were recorded and the urements. Significance of the differences in MD) with TeleChem pins to produce the
urine samples were frozen and stored at mean values were evaluated using either a NIEHS rat chip containing > 7,000 clones.
–80°C for subsequent analysis. one-way analysis of variance followed by a A list of clones on this chip is available on

466 VOLUME 112 | NUMBER 4 | March 2004 • Environmental Health Perspectives


Mini-Monograph | Gene-based markers of renal toxicity

the NIEHS web site (NIEHS Microarray Inc., St. Charles, MO), was used to further FAM-MGB–labeled probe. Taqman one-
Group 2003a). Methods were adapted elucidate discriminate gene sets using step PCR master mix reagent kit (Applied
from DeRisi et al. (1996) and are avail- principal component analysis (PCA). Biosystems) was used for all reactions
able on the NIEHS web site (NIEHS according to manufacturer’s specifications.
Microarray Group 2003b) Real-Time Quantitative PCR All reactions were carried out using the ABI
RNA samples representing single animals Prism 7700 Sequence Detection System
cDNA Microarray Hybridization treated with cisplatin or gentamicin were (Applied Biosystems). Transcript levels
and Analysis also used to validate the expression profile were measured using the comparative CT
For microarray hybridizations, each total of several genes identified using cDNA methods described in User Bulletin No. 2
RNA sample (35–75 µg) was labeled with microarray data including osteopontin, inos- (Applied Biosystems 1997).
Cyanine 3 (Cy3)- or Cyanine 5 (Cy5)-con- itol polyphospate multikinase, L-arginine The following primers, listed in the 5´
jugated dUTP (Amersham, Piscataway, glycine amidinotransferase, prosaposin, to 3´ direction, were used for reverse
NJ) by a reverse transcription reaction lipocalin, synaptogyrin 2, kallikrein, KIM-1, transcription-PCR (RT–PCR) verification
using the reverse transcriptase SuperScript and an expressed sequence tag (EST) of gentamicin-induced gene expression
(Invitrogen, Carlsbad, CA) and the primer (GenBank accession no. AA957270). The changes: kallikrein (GenBank accession no.
Oligo dT (Amersham). Control samples primers for these genes were designed using M19647) forward = GCACCGGCTT
were labeled with Cy3, whereas samples Primer Express (Applied Biosystems, Foster GTCAGTCAA, probe = CCCTCACC
derived from chemically exposed animals City, CA) and custom made (Bioserve CTGACTAC, reverse = TCGGGTGTG
were labeled with Cy5. The fluorescently Biotechnologies, Laurel, MD) except for GTTCCTCATG; prosaposin (GenBank
labeled cDNAs were mixed and hybridized renal kallikrein primers and probe that was accession no. NM_013013) forward =
simultaneously to the cDNA microarray purchased from Applied Biosystems assay by AAGAGCCCAAGCAGTCTGCAT,
chip. Each RNA pair was labeled and design service. probe = CGCCCATGTGCCTC, reverse =
hybridized independently in duplicate or Real-time PCR was performed using the TGTTCCAAATAGATGACCAGCTT
quadruplicate to a total of two to four ABI Prism 7700 Sequence Detection System CT; osteopontin (GenBank accession no.
arrays. The cDNA chips were scanned with (Applied Biosystems) according to the man- M99252) forward = CCAGCACACAAG
an Axon Scanner (Axon Instruments, ufacturer instructions. Primer and probe CAGACGTTT, probe = CAGTCGATGT
Foster City, CA) using independent laser sequences are listed later in this section. CCCTGAC, reverse = CAGTCCGTAA
excitation of the two fluors at 532 and 635 Primer and probe concentrations were opti- GCCAAGCTATCA; KIM-1 (AF35963)
nm wavelengths for the Cy3 and Cy5 mized such that the amplification efficiency forward = CGCAGAGAAACCCGAC
labels, respectively. Image analysis, back- of the endogenous reference was similar to TAAG, reverse = CAAAGCTCAGAGAG
ground subtraction, normalization, and that of the target gene. The endogenous ref- CCCATC; glyceraldehyde-3-phosphate-
determination of significant gene changes erence used for samples was actin RNA. For dehydrogenase (GAPDH) (GenBank acces-
were conducted using the ArraySuite, ver- the cisplatin samples the SYBR Green I sion no. M17701) forward = AGATGG
sion 2.0 extensions of the IPLab image pro- labeling kit (Applied Biosystems) was used TGAAGGTCGGTGTC, reverse = ACTG
cessing software package (Scanalytics, according to the manufacturer instructions TGGTCATGAGCCCTTC; cyclophilin B
Fairfax, VA) (Chen et al. 1997) as previ- to detect double-stranded DNA generated (GenBank accession no. AF071225) for-
ously described (Bushel et al. 2001, 2002; during PCR amplification. Reverse transcrip- ward = CAAGCCACTGAAGGATGTCA,
Hamadeh et al. 2002a, 2002b, 2002c). tion and PCR reactions were performed at reverse = AAAATCAGGCCTGTGG
Hierarchical cluster analysis was carried the same time in a 50-µL reaction containing AATG; Cytb1 (GenBank accession no.
out with the Cluster, version 2.12, and 4 mM magnesium chloride, 0.8 mM of each J01435) forward = CGTCACAGCCCA
TreeView, version 1.6, package (Eisen et al. dNTP, 100 ng total RNA, 0.4 µM reverse TGCATTCG, reverse = CTGTTCATCC
1998). The cDNA data set is available primer, 0.4 µM forward primer, 0.4 U/µL TGTTCCAGCTC.
from NIEHS (NIEHS Microarray Group RNasin, 0.025 U/µL AmpliTaq Gold DNA The following primers, listed in the
2003c). The complete data set is currently polymerase (Roche, Basel, Switzerland), and 5´ to 3´ direction, were used for RT–PCR
being submitted to ArrayExpress (EMBL- 0.25 U/µL MulV reverse transcriptase verification of cisplatin-induced gene
European Bioinformatics Institute, (Roche). Amplification reactions were car- expression changes: Mipmk (GenBank
Hinxton, U.K.; http://www.ebi.ac.uk/ ried out using the following temperature accession no. NM_134417) forward =
arrayexpress) and will be available for pub- profile: 48°C, 30 min; 95°C, 10 min; 95°C, TGCCTGTCCACACTACATACA
lic download by the second quarter of 15 sec; 60°C, 1 min) for 40 cycles. GATG, reverse = CGGCATTTAGGAAT
2004. Accession numbers referencing this Fluorescence emission was detected for each CCGTTCT; L-arginine glycine amidino-
data set will be available on the HESI web PCR cycle and the threshold cycle (CT) val- transferase (GenBank accession no.
site (http://hesi.ilsi.org/index.cfm?pubenti- ues were determined. The C T value was U07971) forward = CGAGGCGGTGC
tyid=120). GeneSpring, version 4.2 defined as the actual PCR cycle when the ACTACATC, reverse = GCACAGGAAT
(Silicon Genetics, Redwood City, CA), fluorescence signal increased above the back- TTTGGGAGGAA; lipocalin/EST
Cluster, and TreeView software (Eisen et ground threshold. Induction or repression of (GenBank accession no. NM_130741) for-
al. 1998) were used to perform the self- a gene in a treated sample relative to control ward = GCAGTGGCCTGATGGTTCA,
organizing maps (SOM) procedure and/or was calculated as follows: fold increase/ reverse = GCACAGGAATTTTGGGA
visualize sets of genes constituting the decrease = 2 – [C T(exposed) – C T(control) ]. GGAA; synaptogyrin 2 (GenBank acces-
union of all significant gene changes from Values are reported as an average of triplicate sion no. AI058493) forward = TGCTCG
each dose and time point. SOM is a neural analyses. For the gentamicin samples, reverse GCTCCCCACTT, reverse = CTGGAG
network architecture that categorized simi- transcription and PCR were carried out in a ACGGTTGGCACAGT; Prosaposin
lar trends in the expression pattern of genes 25-µL reaction volume using 10 ng total (GenBank accession no. M19936) forward
across multiple doses and time points. In RNA in triplicate per reaction, 5 nM = ACTGCTGCCCGATGCAAT, reverse
addition, Partek Pro, version 5.02 (Partek forward and reverse primer, and 25 nM = AAGTAGGCGACTTCTGCAAGCT.

Environmental Health Perspectives • VOLUME 112 | NUMBER 4 | March 2004 467


Mini-Monograph | Amin et al.

Western Blots permeabilized with hyaluronidase (1 mg/mL) Results


Samples of renal tissue were homogenized in a sodium acetate buffer. The sections
in 10 mM potassium homogenization were placed in an antigen-retrieval solution
Clinical Chemistry and Urinalysis
buffer containing 250 mM sucrose, at 95°C for 45 min (Dako Corp., Cisplatin. A summary of the clinical chem-
1 mM EDTA, and 0.1 mM phenylmethyl- Carpenteria, CA) and nonspecific binding istry data for all three nephrotoxicants is
sulfonyl fluoride, adjusted to pH 7.6. was blocked with bovine serum albumin. presented in Table 1. The low dose
Homogenates were centrifuged at 3,000 × The sections were incubated with a 1:200 (0.3 and 1 mg/kg) of cisplatin produced
g for 5 min and 9,000 × g for 15 min. The dilution of a KIM-1 primary antibody only minor changes in the histologic
renal homogenates (50 µg) were separated (provided by J. Bonventre) or vimentin appearance of the kidney or clinical chem-
by electrophoresis on an 8% sodium dode- antibody, washed with Tris-buffered saline istry at any time point during the experi-
cyl sulfate–polyacrylamide gel elec- and exposed to a biotinylated second anti- ment. The high dose (5 mg/kg) of cisplatin
trophoresis gel and transferred to a body for 1 hr. The sections were developed had minimal effect on urine or serum
nitrocellulose membrane in a transfer using an avidin-biotin-peroxidase complex, chemistry relative to the values seen in the
buffer (25 mM Tris, 192 mM glycine, and counterstained with H&E, and viewed time-matched control animals injected
20% methanol) at 100 volts at 4°C for using a 60× oil objective. with vehicle at 4, 24, and 48 hr (data not
2 hr. The membranes were blocked in a
TBST-20 buffer (10 mM Tris, 150 mM Table 2. Summary of observations indicative of cisplatin-, gentamicin-, and puromycin-induced renal
NaCl, and 10% nonfat dry milk) overnight dysfunction.a
and incubated with a 1:1000 dilution of Cisplatin Gentamicin Puromycin
primary antibodies raised against heme Renal tubular dysfunction
oxygenase 1 (StressGen, Vancover, Altered serum electrolyte/mineral concentration + + –/+
Canada) or KIM-1 (kindly provided by Calcuria + + +
J. Bonventre, Massachusetts General Tubular injury
Hospital, Boston, MA) for 2 hr. The mem- Proteinuria + + +
brane was washed with the TBST-20 solu- Single-cell necrosis + – –
Tubular degeneration + + –/+
tion and incubated for 1 hr with a 1:2000 Tubular regeneration + – –/+
dilution of a horseradish peroxidase–cou- Apoptosis – + –
pled second antibody. The membrane was Mononuclear/lymphocytic infiltrates + + +
washed with TBST-20 and then developed Myelin figures (electron microscopy) NE + NE
using enhanced chemiluminescence (Pierce Proximal tubular necrosis + + (high dose) +
Chemical Co., Rockford, IL). Relative Protein casts + + +
Focal segmental glomerulosclerosis – – +
intensity of the bands was determined Glomerular injury or other indicators of renal injury
using UnScanit software (Silk Scientific, Altered serum creatinine + + (high dose) –
Inc., Orem, UT). Glomerular thickening of basement membrane + – +
Altered serum urea nitrogen + + (high dose) –
Immunohistochemistry
Abbreviations: NE, not evaluated; +, end point observed; –, end point not observed; –/+, end point observed in some
Paraffin sections 3 µm thin were prepared instances.
aCisplatin (5 mg/kg), gentamicin (80 or 240 mg/kg/day), and puromycin (20 mg/kg/day). For cisplatin-treated rats, micro-
and mounted on glass slides. The sections
scopic alterations were evident at day 1.
were deparaffinized, rehydrated, and
Table 1. Changes in serum and urine chemistry and organ and body weights 7 days after cisplatin and gentamicin treatment and 21 days after puromycin treatment.a
Cisplatin Serum Protein
dose Body weight Kwt Cr BUN Urine excretion
(mg/kg) (g) (g) (mg/dL) (mg/dL) Glucose/Cr Prot/Cr Ca/Cr (mg/day)
Vehicle 279 ± 7 2.5 ± 0.1 0.30 ± 0.01 15 ± 1 0.23 ± 0.01 1.0 ± 0.1 0.07 ± 0.01 39 ± 3
0.3 288 ± 8 2.7 ± 0.1 0.24 ± 0.02 14 ± 1 0.17 ± 0.06 1.1 ± 0.1 0.11 ± 0.02 28 ± 3
1 289 ± 10 2.7 ± 0.1 0.22 ± 0.04 11 ± 1 0.15 ± 0.04 1.5 ± 0.2 0.12 ± 0.03 39 ± 4
5 252 ± 3* 2.7 ± 0.3 1.4 ± 0.4* 63 ± 20* 6.1 ± 2.0* 1.8 ± 0.3* 0.23 ± 0.08* 66 ± 10

Gentamicin Serum Protein


dose Body weight Kwt Cr BUN Urine excretion
(mg/kg/day) (g) (g) (mg/dL) (mg/dL) Glucose/Cr Prot/Cr Ca/Cr (mg/day)
Vehicle 269 ± 7 2.0 ± 0.1 0.19 ± 0.02 16 ± 1 0.19 ± 0.02 0.5 ± 0.1 0.07 ± 0.01 5±1
2 278 ± 8 2.1 ± 0.1 0.15 ± 0.02 13 ± 1 0.16 ± 0.02 0.4 ± 0.1 0.06 ± 0.01 4±1
10 280 ± 6 2.1 ± 0.1 0.16 ± 0.03 13 ± 1 0.20 ± 0.02 0.5 ± 0.1 0.09 ± 0.02 5±1
80 264 ± 7 2.2 ± 0.2 0.16 ± 0.03 15 ± 2 0.23 ± 0.02 1.3 ± 0.1* 0.14 ± 0.07* 14 ± 1*
240 252 ± 8 3.2 ± 0.2* 1.9 ± 0.1* 127 ± 41 0.49 ± 0.01* 4.9 ± 0.3* 0.26 ± 0.07* 142 ± 31*

Puromycin Protein
dose Body weight Kwt Serum Urine excretion
(mg/kg) (g) (g) Cr BUN Glucose/Cr Prot/Cr Ca/Cr (mg/day)
Vehicle 269 ± 7 2.0 ± 0.1 0.19 ± 0.02 16 ± 1 0.19 ± 0.02 0.5 ± 0.1 0.07 ± 0.01 91.7 ± 17
5 278 ± 8 2.1 ± 0.1 0.15 ± 0.02 13 ± 1 0.16 ± 0.02 0.4 ± 0.1 0.06 ± 0.01 99.5 ± 44
10 280 ± 6 2.1 ± 0.1 0.16 ± 0.03 13 ± 1 0.20 ± 0.02 0.5 ± 0.1 0.09 ± 0.02 1891 ± 1728
Abbreviations: Ca, calcium; Cr, creatinine; Kwt, total kidney weight in grams; Prot, protein.
aMean values ± 1 SE from five rats per group are presented. *p < 0.05 from control.

468 VOLUME 112 | NUMBER 4 | March 2004 • Environmental Health Perspectives


Mini-Monograph | Gene-based markers of renal toxicity

shown). However, 7 days after injection of and some glomeruli exhibited thickening of tubular basophilia, tubular casts, increased
the 5 mg/kg dose of cisplatin, plasma crea- the basement membrane. mitotic figures, and mononuclear cellular
tinine and serum BUN concentration were Gentamicin. Rats treated with gentam- infiltrates. Ultrastructural analysis revealed
elevated by approximately 5-fold. Rats icin at 2 and 10 mg/kg/day exhibited no that one control animal had essentially
treated with the high dose of cisplatin discernable renal pathology. Even the rats normal proximal tubular epithelium,
exhibited glucosuria (elevated glucose/crea- treated with the higher dose of gentamicin whereas another had hyaline droplets of
tinine concentration ratio) indicative of (80 mg/kg/day) exhibited only subtle com- different densities (Figure 1E). The proxi-
proximal tubular injury. They also exhib- pound-related changes in the histology of mal tubular epithelium of two treated ani-
ited a 2-fold increase in urine protein and the kidney after 3 and 7 days of exposure mals exhibited large lysosomes of varying
calcium concentration. (Table 2; Figure 1C, D). These changes density that accumulated lamellar material
Gentamicin. Daily treatment of the rats included minimal degeneration of some forming typical myeloid bodies (Figure 1F).
with 2 and 10 mg/kg doses of gentamicin proximal convoluted tubules, apoptosis, In addition, all of the animals treated with
produced very little change in serum or
urine clinical chemistry compared with the
values seen in time-matched vehicle control
animals at day 7 (Table 1). Urinary total
protein was significantly increased (p <
0.001) in rats dosed at 80 mg/kg/day at
4 hr and at 7 days. They also exhibited an
increase in urinary Ca2+ concentration at 4
hr (p < 0.01) and 2, 3, and 7 days (p <
0.01) after administration of gentamicin
(data not shown). Because the degree of
renal injury was minimal in the strain of rat
SD-ICS (Charles River Labs) used in the
present studies, we repeated the study in
three additional rats using a higher dose of
gentamicin (240 mg/kg/day). This dose
produced the expected severe renal injury
as reflected by the marked increase in BUN
and creatinine. These rats also exhibited a
pronounced proteinuria and elevated levels
of glucose and Ca2+ in the urine (Table 1).
Puromycin. Rats treated with
puromycin (20 mg/kg/day) for 21 days
exhibited marked proteinuria (Table 1) and
an elevation in urinary Ca2+. The chronic
loss of protein in the urine was reflected by
a dramatic fall in serum protein and albu-
min levels at day 21 in rats chronically
exposed to 20 mg/kg/day puromycin.
Despite the marked proteinuria, BUN and
creatinine were not significantly elevated at
any of the time points in the study, indicat-
ing that the glomerular filtration rate
(GFR) remained relatively normal in
these rats.
Renal Histopathology
Cisplatin. The low dose (0.3 mg/kg) of cis-
platin had very little effect on the histology
of the kidney. Four of the five animals
treated with the 1 mg/kg dose of cisplatin
exhibited some degree of single-cell tubular
necrosis and some mild tubular degenera-
tion at 48 and 144 hr after administration
of the compound. Rats treated with the
high dose (5 mg/kg) of cisplatin exhibited
severe proximal tubular necrosis and the Figure 1. Histopathological evidence of cisplatin- and gentamicin-induced proximal tubular toxicity.
formation of protein casts, especially in the Representative H&E-stained sections histological sections from kidney of (A) vehicle-treated (magnifica-
tion 20×), (B) cisplatin-treated rats (5 mg/kg, 6 days) (magnification 20×), (C) vehicle-treated rats (magnifi-
pars recta of proximal tubules (Table 2;
cation 200×), and (D) gentamicin-treated rats (80 mg/kg/day; 7 days) (magnification 200×). Representative
Figure 1A, B). There was also evidence of electron micrographs from (E) vehicle-treated rats (magnification 3,000×) and (F) gentamicin-treated rats
mild interstitial inflammation and edema (80 mg/kg/day, day 7) (magnification 4,500×) . Increased apoptosis and cellular infiltrates and myelin fig-
in both the renal cortex and outer medulla, ures were observed in rats treated with gentamicin (80 mg/kg/day, day 7).

Environmental Health Perspectives • VOLUME 112 | NUMBER 4 | March 2004 469


Mini-Monograph | Amin et al.

240 mg/kg/day gentamicin for 7 days lumen. However, the majority of the to nephron segment-specific toxicity as well
exhibited severe proximal tubular necrosis glomeruli appeared normal at the light as gene expression alterations reflecting
and formation of protein casts in the microscopic level. histopathological changes.
proximal tubule. Hierarchical clustering revealed two
Puromycin. There were minimal back- Identification of Tubular Toxicity distinct separations in experimental groups
ground findings in the control and treated Based on Gene Expression Changes that correlated with the severity of renal
groups, defined as less than three foci of For each treatment (compound, dose, and toxicity. As shown in Figure 3A, the hierar-
the following: interstitial lymphocytic time), genes with statistically significant chical order of the clustering tree indicated
inflammatory infiltrates, interstitial fibrosis, expression changes were identified using the several correlations and allowed sample
tubular mineralization, and tubular cysts in approach described by Bushel et al. (2001). separation based on severity, region, and
the cortex or medulla. There was a dose- Hierarchical clustering was used to aid in type of toxicity. For instance, the hierarchi-
related minimal increase in severity of one visualization and biological interpretation of cal ordering of gene expression changes by
or more of the following lesions: interstitial this extensive data set, and in particular, to cisplatin (5 mg/kg/day, 24 and 144 hr),
lymphocytic cellular infiltrate, glomerular identify correlated expression patterns that gentamicin (2, 10, 80, and 240 mg/kg/day,
sclerosis, lymphocytic perivascular cellular reflect potential biological/toxicological day 7), and puromycin (5 mg/kg/day, day
infiltrate in the renal capsule, and tubular processes occurring in the renal tissues of 7; 20 mg/kg/day, days 1 and 7) fall within
regeneration in one of five animals from the animals treated with cisplatin, gentam- one node and are much more closely corre-
each time point (Figure 2; Table 2). The icin, or puromycin at varying doses and lated compared with the remaining lower-
most significant findings were dilation of time points. Hierarchical clustering, dose, earlier time point samples. This
renal tubules, protein casts, expansion of described by Eisen et al. (1998), was separation in the expression patterns
the mesangial matrix material in the applied across the various treatment groups, appeared to be correlated with the severity
glomerulus, and focal segmental glomeru- doses, and time points, using a combined of the renal injury (based on changes in
losclerosis in the rats treated with list of genes identified to be altered statisti- serum biochemistry and/or urinalysis
puromycin (20 mg/kg/day) for 21 days. In cally significantly in at least one of the sam- changes or histopathology) and are linked
two of five rats there were multiple areas of ples studied relative to control (Figure 3A, to the treatment, dose, time, and severity of
involvement in the renal cortex, which B). Using this approach, it was possible to each of these compounds. The second
occupied approximately 25–35% of the extract correlated patterns and natural major node separating the remaining treat-
kidney. Sloughed tubular epithelial cells classes present in the data set that could be ment groups consists of samples associated
were sometimes present in the tubular correlated with biological processes relevant with less prominent renal toxicity.

Figure 2. Representative H&E-stained histological sections from kidney of (A, B) vehicle-treated and (C, D) puromycin-treated rats. (A, B) Intact renal tubules and
glomeruli and uniform nuclei are observed in controls. In addition, uniform tubules with single layer of epithelium lining are observed in renal cortex of control
rats. (C) Increased protein casts and necrosis of renal tubular cells observed in rats treated with puromycin (20 mg/kg/day, day 21). (D) Thickened glomerular
membranes and affected tubules observed in rats treated with puromycin (20 mg/kg/day, day 21). Magnification 40×.

470 VOLUME 112 | NUMBER 4 | March 2004 • Environmental Health Perspectives


Mini-Monograph | Gene-based markers of renal toxicity

C Figure 3. Hierarchical clustering, as described


Severe Puromycin
by Eisen et al. (1998), was applied across the
glomerular 20 mg/kg/day various treatment groups, doses, and time
toxicity points, using a combined list of genes identi-
fied to be altered statistically significantly in
Acute tubular toxicity at least one of the samples studied relative to
control. Gent, gentamicin. (A) Separation of
experimental groups. The A and B notation
Regional separation

behind the high-dose cisplatin indicates two


biological replicates. Gene expression
Gentamicin 80 mg/kg day 1 changes were determined using 2–4 replicate
hybridizations from pooled kidney samples
and include fluor-reversal. (B) Node contain-
Cisplatin 5 mg/kg 24 hr (B) ing several putative markers of renal toxicity.
Green indicates genes that are downregu-
Cisplatin 1 mg/kg 24 hr lated; red indicates genes that are upregu-
Severe proximal lated in treated animals relative to control
tubular toxicity
animals. Accession numbers are from
GenBank (http://www.ncbi.nih.gov/GenBank/).
Cisplatin 5 mg/kg 144 hr (B)
Cisplatin 5 mg/kg 144 hr (A) (C) PCA of cisplatin-induced (red), gentam-
icin-induced (green), and puromycin-induced
(yellow) gene expression changes at varying
doses and time points. The labels of severity
and region of toxicity were overlaid from the
Severity of toxicity clinical pathology data.

Environmental Health Perspectives • VOLUME 112 | NUMBER 4 | March 2004 471


Mini-Monograph | Amin et al.

Clustering based on gene expression compared with the results obtained using and threshold changes in the urinary excre-
profiling of samples obtained from inde- the 80-mg/kg dose for 7 days in the initial tion of protein, glucose, and Ca2+ observed
pendent replicate experiments performed experiment. The higher dose (240 mg/kg) after treatment with 80 mg/kg gentamicin
using the same compounds and protocols resulted in the expected severe proximal (Figure 4).
can reveal if there are variations in gene tubular necrosis and proteinuria. Although The power of microarray analysis is
expression changes across studies. In Figure not identical, the overall changes in renal that it allows elucidation of groups of genes
3A, cisplatin 5 mg/kg, 144 hr (A) and cis- gene expression observed after treatment that correlate with certain expression pat-
platin 5 mg/kg, 144 hr (B) represent renal with 240 mg/kg compared with 80 mg/kg terns that accompany particular types of
gene expression changes observed based on gentamicin for 7 days were still closely cor- biological effects. Analysis of groups or
two independent experiments performed related and hence clustered together despite clusters of genes that contribute to similar
with cisplatin in different groups of ani- the less severe changes in renal histology pathways/functions may be more robust
mals (A and B denote the first and second
biological experiments, respectively). The
gene expression changes observed in these
two different experiments performed with
cisplatin at 5 mg/kg/day were highly corre-
lated, indicating minimal overall variability
in gene expression changes across two inde-
pendent in vivo experiments, and hence the
profiles clustered together.
Multidimensional visualization by
PCA using all of the statistically altered

Gent 4016 80 mg/kg D7


Gent 4019 80 mg/kg D7
Gent 4017 80 mg/kg D7
Gent 4020 80 mg/kg D7
Gent 781 240 mg/kg D6
Gent 783 240 mg/kg D6
Gent 779 240 mg/kg D6
Gent 4018 80mg/kg D7
renal gene expression changes was used to
observe the spatial distribution of the
treatment groups in multidimensional
space (Figure 3C). This approach offers an
opportunity to visualize expression patterns
that can reflect similarities in biological AA874916
responses. The visualization of high-dimen- AA818502
AA955861
sional data in two- or three-dimensional AA956164
AA899737
principal components reveals unsupervised AA818118
clusters within the data. Samples can be AA818709
AA900947
scored using the results of a PCA on known AA874997
AA925982
samples, and these scores may place the AA955326
AA899769
unknown into one of the previously identi- AA899814
AA901183
fied clusters where gene expression changes AA858957
are clearly linked to biological/pathological AA850744
AA819818
events. As shown in Figure 3C, three major AA900860
AA899630
separations of the various treatment groups AA900992
AA956907
were evident. The first includes treatment AA901095
AA899725
groups in which severe renal proximal AA858477
tubular toxicity was observed (cisplatin AA818442
AA818152
5 mg/kg, 24 and 144 hr). The data from AA819093
AA818640
these groups were clearly separated from AA899629
AA819424
early time points after exposure to cisplatin AA818434
and from the low-dose samples where no AA818247
AA955091
histologic evidence of tubular injury were AA858746
AA900207
observed. In addition, the second principal AA875070
AA859846
component shows separation of the high- AA818795
AA899586
dose cisplatin data with tubular injury from AA955185
the puromycin data that exhibited marked AA900570
AA899558
proteinuria and glomerular toxicity.
The samples analyzed by microarrays in
these experiments consisted of pooled sam-
ples. However, we further characterized the
response of individual animals at one
dose/time point for cisplatin (Thompson et
al. 2004) and for gentamicin (day 7) and
compared the response of these animals to
the animals from a related, biologically
Figure 4. Clustering of gentamicin-treated individual animals at 80 and 240 mg/kg after 7 days of exposure.
replicated experiment. In the gentamicin These two biological exposures were conducted independently, and the RNA from each experiment was
replicate experiment, the results from ani- run on the same chip and analyzed using the same analytical approach. Green indicates genes that are
mals dosed with 240 mg/kg of gentamicin downregulated; red indicates genes that are upregulated in treated animals relative to control animals.
for 7 days (to produce severe injury) were Accession numbers are from GenBank (http://www.ncbi.nih.gov/GenBank/).

472 VOLUME 112 | NUMBER 4 | March 2004 • Environmental Health Perspectives


Mini-Monograph | Gene-based markers of renal toxicity

than focusing on a single gene. In addition, the less prominent tubular toxicity was toxicity/repair, including KIM-1, osteo-
gene clusters may be used to identify observed in the rats receiving the higher pontin, vimentin, and several ESTs
known and novel putative gene-based dose of puromycin for 21 days (Figure 4). (Figure 6).
markers of renal toxicity and can poten- Multiple approaches were used to
tially direct future research to validate these Elucidation and Verification robustly determine and confirm the expres-
putative gene-based markers in urine/ of Putative Biomarkers sion changes observed in the present
serum using animal and clinical models. One of the intended outcomes of these microarray studies. First, the genes that
Figure 5 shows a group of genes strongly studies was the elucidation of putative new were differentially modulated were defined
downregulated in samples that exhibited sensitive biomarkers of nephrotoxicity. We using the approach of Chen et al. (1997)
proximal tubular necrosis. Many of these examined the significantly changed genes that employs a specified confidence level
genes are functionally localized to the prox- for those that displayed induction in a (95% in the present study) for determining
imal tubules, further strengthening the dose- and time-dependent manner for fur- differentially expressed genes. Second, to
hypothesis that regional-specific toxicity ther verification in this study. Figure 6 reduce false positives, replicate hybridiza-
may be discerned through analysis of gene shows a grouping of genes after high-dose tions (typically n = 4, with reverse labeling)
expression patterns (Table 3). Interestingly, cisplatin and gentamicin treatment that were performed using the pooled samples
the puromycin samples showed similar appear to be upregulated in a dose- and and/or biological replicates, and a binomial
downregulation of some of these genes, time-dependent fashion. Overlap of these distribution was used to model the results
indicating that tubular toxicity may have genes on clusters derived from both the of the analyses at given confidence levels.
occurred from this treatment. Indeed, NIEHS and Affymetrix microarrays indi- Third, dye reversal was used to avoid false
upon further histopathological evaluation, cate several potential biomarkers of renal positives due to biases in Cy-dye–specific
Gent 2mg/kg D3
Gent 80mg/kg D3
Gent 10mg/kg D3
Gent 10mg/kg D1
Gent 80mg/kg D1
Cisplatin, low, 24h
Cisplatin, mid, 24h
Gent 2mg/kg D1
Gent 80mg/kg D2
Gent 10mg/kg D2
Gent 2mg/kg D2
Gent 10mg/kg D7
Gent 80mg/kg D7
Puromycin, 20mg/kg D7
Puromycin, 20mg/kg D1
Gent 2mg/kg D7
Cisplatin, high 144h (B)
Cisplatin, high, 144h (A)
Cisplatin, high, 24h (B)

AA925935 EST
AA899443 Rattus norvegicus CDK110mRNA
AA818361 EST
AA964628 Glucose-6-phosphatase
AA924904 EST
AI 137907 R. norvegicus alpha-1-macroglobulin mRNA, complete cds
AA900766 EST
AA997886 Rattus norvegicus cytochrome P458 2D18 mRNA, complete cds
AI 070884 Kidney androgen-regulated protein
AA924834 EST
AI 059765 ZAP 36/annexia IV
AA818636 Rat beta-galactoside-alpha 2.6-sialytransferase mRNA
AA998607 kvnurenine aminotransferase II
AI 385145 R. norvegicus mRNA for ras-related GTPase Rab29
AA866385 EST
AA818680 ornithine aminotransferase
AA925553 EST
AA901327 Epidermal growth factor
AA819745 Growth hormone receptor
AA818579 cystosolic cysteine dioxygenase 1
AA860013 ornithine decarboxylase
AA925145 EST
AA899969 EST
AA818440 beta-alanine-pyruvate aminotransferase
AA818741 EST
AA819087 EST
AA899732 EST
AA819309 Guanidinoacetate methyltransferase
AA818855 solute carrier family 15 (H+/peptide transporter), member 2
AI 059692 Eukarvotic elongation factor 2 kinase
AA957263 EST
AA858892 EST
AA998808 alanyl (membrane) aminopeptidase
AA819550 EST
AA955093 EST
AI 059021 Pancreatic lipase
AA92432 EST
AA925724 Transcription factor 2. Hepatic: LF-B3: variant hepatic nuclear factor
AA817972 Proteasome (prosome, macropain) 256S subunit, ATFase
AA900944 EST
AA925291 Nerve growth factor, gamma polypeptide
AA86601 EST
AA818463 EST
AA859075
AA819477 serum amyloid P-component
AA819101 EST

Figure 5. Evidence for puromycin-induced tubular toxicity based on gene expression profiling. A number of enzymatic and transporter functions that map to the
proximal tubular region of the kidney were shown to be downregulated in this cluster of genes. The strong downregulation of these genes in the puromycin-
exposed animals led to the hypothesis that puromycin also induced proximal tubular necrosis. Green indicates genes that are downregulated; red indicates
genes that are upregulated in treated animals relative to control animals. Accession numbers are from GenBank (http://www.ncbi.nih.gov/GenBank/).

Environmental Health Perspectives • VOLUME 112 | NUMBER 4 | March 2004 473


Mini-Monograph | Amin et al.

Table 3. Downregulated gene expression changes correlated with regional specific damage.a incorporation for specific genes, and genes
Renal distribution and enzyme Distributionb Cisplatin Gentamicin with highly variable expression changes
across hybridizations were flagged because
Glomerulus
Adenosine-deaminasec of a large coefficient of variation or a modi-
Proximal tubule fied z-score computation to detect outliers
Glucose-6-phosphatased S1 > S2 > S3 √ (Bushel et al. 2001, 2002). Therefore,
Fructose-1,6-bisphosphatased S1 < S2 > S3 examination of the data from these
Phosphoenolpyruvate carboxylkinased S1 > S2 > S3
Fructokinased S1 = S2 < S3
repeated hybridizations yielded lists of
Fructose-1 phosphate aldolased S1 = S2 > S3 genes that were statistically validated as dif-
Glycerokinased S1 = S2 > S3 ferentially expressed.
Glycerol 3-phosphate dehydrogenased S1 = S2 = S3 In addition, several of the observed
Glutamine synthetase S3 √ √ gene expression changes identified in the
Alanine aminotransferase S1 = S2 < S3 √
Ornithine aminotransferase S1 = S2 < S3 present study were confirmed using alter-
Gamma glutamyltranspeptidasee S1 < S2 < S3 nate approaches, including RT–PCR,
Gamma glutamyl cysteine synthetase S3 in situ hybridization, immunohistochem-
Gluthathione S-transferasee S1 = S2 = S3 √√ istry, and/or Western blot analysis. As
Cytochrome P450e S1 = S2 = S3 √ √√
L-Hydroxyacid oxidasee S1 = S2 < S3 √ shown in Table 4, RT–PCR analysis was
Peroxisomes (D-amino acid oxidase/catalase)e S1 = S2 < S3 √ performed using kidney RNA isolated from
Aminopeptidasesf S1 < S2 < S3 √ rats treated with cisplatin for 24 hr or
Alkaline phosphatase S1 = S2 = S3 144 hr or gentamicin exposed for 7 days.
Fatty acyl-CoA oxidase S1 = S2 < S3
Choline oxidase S1 < S2 = S3 Genes changed with microarray were gen-
25(OH)-D3-1α-hydroxylaseg S1 = S2 < S3 √ erally in qualitative agreement when deter-
Vitamin D binding proteing √ mined using RT–PCR. The expression of
Solute carrier family 15 (H+/peptide) √ genes known to be implicated in the
Isocitrate dehydrogenase 1 √
mechanism(s) of renal toxicity that were
Abbreviations: √, change in gene expression observed; √√, change in gene expression observed in more than 1 gene in confirmed include kallikrein, heme oxyge-
this family. a Information modified from WHO (1991). bS1, S2, and S3 denote renal tubular subregions. cPurine metabolism nase 1, clusterin, osteopontin, and KIM-1
related. dSugar metabolism related. eRole in xenobiotic metabolism. fPeptide/amino acid metabolism. gVitamin D related.
(Table 4; Figure 7A). In addition, several
3.5
3.0 Cisplatin, 5 mg/kg, day 6
Gentamicin, 80 mg/kg, day 7
2.5
Log2 ratio gene expression

2.0
(treated/control)

1.5
1.0
0.5
0
–0.5
–1.0
–1.5
–2.0
Isocitrate dehydrogenase 1
Beta-alanine-pyruvate
aminotransferase
AA819244 EST

Vitamin D binding protein

Class I beta-tubulin

Sialoprotein (osteopontin)

BH S-methyltransferase

AA925142 EST

AA925957 EST

AA957270 EST
Vimentin

HO-1

Thymosin beta 4

Nerve growth factor

Synaptonygrin 2

AA819209 EST

AA819101 EST

AA858892 EST

AA899472 EST

AA899737 EST

AA901025 EST

Inositol polyphosphate
multikinase (Ipmk)

AA925553 EST

(BHMT)
RBP

IGF-BP3

Sgp 2

P4502D18
UDPGT

EGF

Figure 6. Putative gene-based markers of nephrotoxicity. Graph represents level of change relevant to control animals The level of expression is shown as the
log2 of the ratio of the expression in treated samples versus vehicle-treated samples. The values for the expression in cisplatin-treated animals is shown in
closed bars, and the open bars represent the expression in gentamicin-exposed animals.

Table 4. Comparison of cDNA microarray and RT–PCR measurements.


Cisplatina Gentamicin
GenBank RT–PCR cDNA microarray Day 2 RT–PCR Day 7 RT–PCR
accession no.b Gene (5 mg/kg) (5 mg/kg) (10 mg/kg/day) (80 mg/kg/day) (10 mg/kg/day) (80 mg/kg/day)
AA964431 Osteopontin 5.2 4.1 0.9 1.1 1.4 3.9
AA901117 Inositol polyphosphate kinase 0.5 0.7 – – – –
AA920287 L-Arginine-glycine amidinotransferase 0.3 0.5 – – – –
AA858514 Prosaposin 1.07 1.05 1.0 1.0 0.9 0.9
AI058493 Synaptogyrin 2 0.5 1.8 – – – –
AA957270 EST 3.5 2.9 – – – –
M19647 Kallikrein – – 1.01 1.0 0.5 0.3
AA946503 Lipocalin 2 7.7 –c – – – –
–, not done or not on platform. aAll cisplatin values are from 144-hr time point except L-arginine glycine amidinotransferase, which is from the 24-hr time point. bFrom GenBank
(http://www.ncbi.nih.gov/GenBank/). cSequence not present on cDNA microarray. Observed based on similarity to KIM-1 expression on Affymetrix platform (data not shown).

474 VOLUME 112 | NUMBER 4 | March 2004 • Environmental Health Perspectives


Mini-Monograph | Gene-based markers of renal toxicity

genes for which there is limited under- For example, we detected a reduction in However, they are relatively insensitive
standing of a role in renal toxicity were the expression of L -arginine-glycine markers of glomerular injury, as typically
validated. These include inositol polyphos- amidinotransferase and guanidinoacetate up to 75% of nephrons have to be non-
phate multikinase and L-arginine-glycine methyltransferase at days 1 and 6, respec- functional before there are significant eleva-
amidinotransferase. In addition to tively, after treatment with 5 mg/kg cis- tions in serum levels of BUN or creatinine.
RT–PCR verification, heme oxygenase and platin, a dose that induced renal lesions. Although ours is the first study to demon-
KIM-1 protein expression in rat kidney Mapping of these gene products to a bio- strate the modulation in the expression of
after cisplatin treatment was confirmed by chemical pathway points to a role for them these genes in rat kidney after cisplatin
Western blot analysis (Figure 7A–D). in the formation of creatinine from L-argi- treatment, Lee et al. (1998) demonstrated
Using in situ hybridization, expression of nine (see map in Kramer et al. 2004). that cisplatin alters the expression of L-argi-
KIM-1 was confirmed to be increased in Creatinine is formed from creatine and nine-glycine amidinotransferase and guani-
the proximal tubule after exposure to cis- excreted by the kidneys. Traditionally, mea- dinoacetate methyltransferase in the male
platin (Figure 8B). Increased vimentin surements of increased serum urea and crea- rat reproductive tract. Using Western blot
expression was also confirmed by immuno- tinine are used clinically as indices of analysis, Lee et al. (1998) demonstrated
histochemistry (Figure 8D). changes in glomerular filtration rate. that L-arginine-glycine amidinotransferase is
RT–PCR verification of two ESTs
identified to be induced in rat kidney after A C
exposure to 5 mg/kg cisplatin for 144 hr Control Cisplatin
was also performed. Lipocalin 2 (LCN2) 42 kDa
(GenBank accession no. AA946503) 50 kDa
expression was initially identified in cis-
31 kDa
platin gene expression data generated with C C 4 hr 4 hr 24 h 24 hr
the same RNA on an Affymetrix platform
(data not shown). In fact, the expression of B D 500

Intensity of HO-1 mRNA


LCN2 was similar to that observed for 400
KIM-1. Induction of KIM-1 expression
GAPDH 300
after cisplatin treatment was confirmed by
in situ hybridization, RT–PCR, and 200
Western blot analysis (Figure 7). The Cyclo
Cytb1 100
increased expression of another EST
(GenBank accession no. AA957270; now KIM-1 0
C 4 hr 24 hr
known as a tumor necrosis factor (TNF) Cisplatin treatment
receptor superfamily, member 12a, or (144 hr, 5 mg/kg)
Tweak receptor) was verified using Figure 7. (A) Western blot confirms KIM-1 expression in rat kidney after cisplatin treatment (144 hr,
RT–PCR (Table 4). The role of Tweak 5 mg/kg) in vehicle control and cisplatin-treated animals. n = 5. (B) RT–PCR of KIM-1 mRNA, cyclophilin B
receptor in mediating cisplatin renal toxic- (cyclo, cytb1, and GAPDH were used as controls). (C) Western blot of heme oxygenase. (D) Quantitation
ity remains to be determined. of heme oxygenase mRNA shows that increased mRNA expression at 4 hr precedes peak of protein
expression (24 hr).
Discussion
In this study, we used cDNA microarrays
to examine temporal changes in gene
expression patterns in the kidney after
treatment of rats with cisplatin, gentam-
icin, and puromycin, three nephrotoxicants
that primarily injure the proximal tubule
(cisplatin and gentamicin) or the glomeru-
lus (puromycin) via different mechanisms
of action. The results of our microarray
study are consistent with numerous reports
of the modulation in expression of various
mRNA after renal injury induced by cis-
platin, gentamicin, or puromycin (Girton
et al. 2002; Huang et al. 2001). For instance,
using data from cisplatin treatment, it was
possible for us to functionally annotate gene
expression changes to various previously
published and novel categories. These cate-
gories include biochemical pathways related
to creatinine biosynthesis, osmoregulation,
kinase signaling, cell cycle–related genes,
renal transporters, renal injury, and regener-
ative responses, as well as gene expression Figure 8. In situ hybridization confirms KIM-1 expression is very low in a kidney from a control animal (A)
changes related to drug metabolism, detoxi- and highly induced in a cisplatin-exposed kidney (B). Immunohistochemistry shows basal expression of
fication, and drug resistance. vimentin expression in the kidney of a control rat (C) and induction in an animal treated with cisplatin (D).

Environmental Health Perspectives • VOLUME 112 | NUMBER 4 | March 2004 475


Mini-Monograph | Amin et al.

also expressed in the rat kidney and may contribute to renal damage because of member 4 (SLC4A4; GenBank accession
guanidoacetate methyltransferase mRNA is excessive buildup of NO, which has been no. AI144995), macrophage inflammatory
expressed in the kidneys, testes, and epi- implicated in numerous renal pathologies protein 1-alpha (Mip-1-alpha; GenBank
didymis of rats. More recently Yasuda et al. (Valdivielso and Blantz 2002). NO is an accession no. AA924105), interferon, alpha
(2000) demonstrated decreases in urinary important regulator of renal vascular tone inducible protein 27-like (GenBank acces-
concentrations of guanidinoacetic acid, cre- and can modulate renal blood flow, sion no. AA955996), retinol binding pro-
atinine, and creatine after cisplatin treat- glomerular hemodynamics, and the contrac- tein (GenBank accession no. AA860061),
ment. These studies also suggested that tility of mesangial cells. In addition, NOS is and glucose-6-phosphatase (GenBank acces-
production of guanidinoacetic acid, a pre- present in the renal tubular segments and sion no. AA964628). An increase in expres-
cursor of creatinine that is affected by meta- the juxtaglomerular apparatus and may sion of transporter genes may function to
bolic disturbance when kidney function is enhance the renal damage incurred after facilitate reuptake of solutes and small mole-
damaged, may be a marker of renal damage. nephrotoxicant administration. cules passing through the damaged glomeru-
Our studies revealed that there was a The expression of kallikrein decreased in lus. Solute carrier family 4, for example, is
marked decrease in the expression of the kidney after gentamicin (80 mg/kg/day, involved in the coupled movement of
L-arginine-glycine amidinotransferase in the 7 days) (Table 4). Kallikrein is a serine pro- sodium and bicarbonate; the expression of
kidney as early as day 1 after cisplatin treat- tease that cleaves kininogen to produce this gene may be among the renal responses
ment. The biochemical outcome expected kinin, a vasoactive and natriuretic peptide for maintaining solute homeostasis during
from inhibition of L -arginine-glycine (Clements 1989; Schmaier 2003). Kallikrein renal injury. Cathepsins are primarily cys-
amidinotransferase would be a decrease in is produced and secreted in distal nephron teine-dependent lysosomal proteases and
the formation of guanidoacetic acid. It is segments and has been implicated in the play an important role in protein degrada-
possible that this renal molecular response, control of sodium and water excretion and tion and turnover. The authors speculate
in part, may precede or may correlate with the long-term control of arterial pressure. that the upregulation of cathepsin genes
the decrease in urinary excretion of More recently, kallikrein has also been may be a compensatory response to allow
guanidoacetic acid reported by Yasuda et al. implicated in determining the pathogenesis for protein degradation as the nephron com-
(2000). Decreased renal expression of L - of renal injury after administration of pensates for proteinuria by increasing pro-
arginine-glycine amidinotransferase in rats nephrotoxicants. For example, Murakami tein reuptake associated with glomerular
treated with 1 mg/kg, a dose of cisplatin et al. (1998) reported that systemic transfec- toxicity. In addition to the genes described
that resulted in mild pathological changes, tion of the kidney with an adenovirus above, retinol-binding protein was upregu-
was evident at day 6 but not at day 1, pre- expressing kallikrein attenuates the develop- lated in the present study and retinol-bind-
sumably reflecting a later onset of toxicity. ment of gentamicin-induced nephrotoxicity ing protein mRNA is known to be expressed
In contrast, treatment with 5 mg/kg cis- in rats. Murakami et al. (1998) suggest that in rat proximal tubules (Makover et al.
platin decreased the expression of L-argi- the renoprotective effect of kallikrein gene 1989). It is likely that the induction of
nine-glycine amidino transferase expression delivery may be related to the ability of retinol-binding protein may be in response
at day 1, and downregulation of guanidi- kinins to activate phospholipase A and pre- to increased urinary loss associated with
noacetate methyltransferase was most vent phospholipidosis, which is characteris- glomerular and/or tubular toxicity. Recent
prominent after 6 days. The temporal dif- tic of gentamicin toxicity. In addition, studies demonstrate that urinary retinol-
ferences in the modulation of two genes in recent studies have revealed that urinary binding protein has been investigated as a
the same biochemical pathway is unclear; kallikrein levels are reduced and there are prognostic marker of proximal tubular dys-
however, it may be indicative of progression polymorphisms in the kallikrein gene in function in patients with glomerulopathies
of toxicity and biochemical feedback mech- patients with renal disease (Yu et al. 2000, (Kirsztajn et al. 2002).
anisms to compensate for altered creatinine 2002). These results suggest the potential In contrast to the observed upregulation
clearance. L-arginine-glycine amidinotrans- utility of gene expression profiling to iden- of dentin sialophosphoprotein at 24 hr after
ferase was also downregulated in renal tissue tify genes and pathways that may play a role treatment with 5 mg/kg/day puromycin,
of rats treated with gentamicin and puro- in determining the genetic susceptibility to dentin sialophosphoprotein was among some
mycin, further implicating the potential nephrotoxicity (toxicogenomics). of the more downregulated genes at 24 hr
importance of this pathway in overall Puromycin treatment, in the present after treatment with 20 mg/kg puromycin.
response to renal toxicity. Altered expres- studies, was associated primarily with Acidic nuclear phosphoprotein 32 (GenBank
sion of these genes is likely to reflect an glomerular toxicity, in addition to the less accession no. AI070967), L-glycine arginine
altered protein product (not determined in prominent proximal tubular toxicity. amidinotransferase (GenBank accession no.
the present studies). This suggests that inhi- Numerous genes were identified as upregu- AA900287, discussed earlier), and cytosolic
bition of these enzymes may lead to an lated or downregulated at various doses and epoxide hydrolase (GenBank accession no.
increase in levels of arginine, an immediate time points after puromycin treatment. AA819830) were downregulated at 24 hr
substrate for nitric oxide synthase (NOS), Examples of genes upregulated primarily at after treatment with 20 mg/kg puromycin.
producing a nitric oxide (NO) precursor 24 hr after treatment with 5 mg/kg puro- Although the role of these genes in the mol-
that can lead to the formation of NO. A mycin include serum amyloid p-component ecular responses to glomerular toxicity is
role for NO has been implicated in models (GenBank accession no. AA819447) and unclear, the downregulation of some of
of renal injury (Srivastava et al. 1996), and dentin sialophosphoprotein (GenBank these genes (i.e., cytosolic epoxide hydrolase,
L -arginine has been demonstrated to be accession no. AA899472; discussed below), acidic nuclear phosphoprotein 32) was
both protective (Andoh et al. 1997) and dihydropyrimidinase (GenBank accession observed at both 24 hr and after 7 days of
harmful (Tome et al. 1999) in models of no. AI111911), cathepsin H (GenBank treatment with 20 mg/kg/day of puromycin.
renal injury. The authors speculate that accession no. AA819336), cathepsin B Hydrolases have been identified in rat
although the initial effects of NO formation (GenBank accession no. AA963225), alco- glomerular mesangial cells and have been
after renal injury may be beneficial, the pro- hol dehydrogenase (GenBank accession no. implicated in glomerulonephritis (Nakao
longed inhibition of the L-arginine pathway AA875140), solute carrier) family 4, et al. 1999).

476 VOLUME 112 | NUMBER 4 | March 2004 • Environmental Health Perspectives


Mini-Monograph | Gene-based markers of renal toxicity

Renal lesions induced by cisplatin, gen- to truly validate these as biomarkers of (Hoffmann et al. 1993; Nagata et al. 1991),
tamicin, and puromycin generally occur in nephrotoxicity. Studies to investigate and elevated serum and urinary levels of
discrete anatomical regions (e.g., tubular or reversibility, specificity, and sensitivity will β-trace (L-PGDS) have been observed in
glomerular components), although as be needed as part of this validation (Kramer patients with renal failure (Hoffmann et al.
observed in this study, injury can diffuse to et al. 2004). One of these gene products, 1997; Melegos et al. 1999). L-PGDS is a
additional regions of the nephron when KIM-1, is a membrane-spanning protein secretory glycoprotein that catalyzes the iso-
toxicity is severe. Discrete toxicity to an cleaved on the extracellular surface after merization of a precursor of prostanoids,
organ with site-selective functional capacity tubular injury (Bailly et al. 2002). In addi- prostaglandin (PG) H2, to produce
suggests that observed changes of the bio- tion, antibodies to KIM-1 are now being prostaglandin D-2 (Urade and Hayaishi
chemical properties of the affected renal used to measure protein levels in human 2000). Oda et al. (2002) developed and
region can provide pertinent information urine after renal ischemic injury (Han et al. evaluated an enzyme-linked immunosorbent
regarding the molecular outcome of the 2002). This study indicates that KIM-1 assay (ELISA) method to screen for human
lesion. To take advantage of this in facili- may indeed be a specific marker of tubular urinary L-PGDS. Using this method, Oda
tating the interpretation of mRNA expres- injury, which could provide a more sensi- et al. determined that L-PGDS excretion
sion changes in our study, we used the tive indication of damage compared with was significantly increased in patients with
published literature related to enzymology traditional clinical measurements (Han renal disease, even in the absence of
of kidney tissue to compile information et al. 2002). As mentioned earlier, RT–PCR increases in serum creatinine. Therefore, its
regarding the expression of specific genes verification of two ESTs induced after expo- ability to be monitored in urine of patients
within specific regions of the nephron, par- sure to 5 mg/kg cisplatin for 144 hr was per- (Yan et al. 2001) and, in particular, the abil-
ticularly within various tubular segments formed. Tweak receptor, or TNF receptor ity to monitor L-PGDS (a member of the
(Table 3) (Mattenheimmer 1968; WHO superfamily, member 12 (GenBank acces- lipocalin family) in urine of patients with
1991). As shown in Figure 4, decreased sion no. AA957270), belongs to the TNF epithelial renal disease warrants its further
expression of several mRNAs was detected receptor superfamily. Death ligands and evaluation in models of renal injury. This
by microarray profiling, particularly in kid- receptors such as TNF participate in apop- premise is confirmed in these studies. In
neys of rats treated with cisplatin (5 mg/kg, tosis regulation in the course of renal injury addition to the ESTs described above, the
day 6) and to a lesser extent after gentam- (Ortiz et al. 2001), and its induction is likely expression of several other ESTs (e.g.,
icin treatment (80 mg/kg/day, day 7). One involved in apoptotic signaling and regula- GenBank accession nos. AA819209,
explanation could be that the loss of expres- tory mechanisms contributing to cisplatin- AA899472, and AA899737) were com-
sion of these RNAs could, in part, reflect induced renal injury. LCN2 (GenBank monly modulated by cisplatin and gentam-
massive injury and death of proximal tubu- accession no. AA946503) was identified as a icin at doses that caused proximal tubular
lar cells. Although necrosis of tubular seg- putative biomarker of kidney injury based toxicity. In most instances, the expression of
ments was confirmed histologically after on the similarity of its expression pattern these ESTs correlated with severity of proxi-
treatment with each of the three com- to KIM-1. mal tubular injury, and hence, expression
pounds, it varied in severity (cisplatin > The human homolog of LCN2 is was more robust in cisplatin-treated com-
gentamicin > puromycin). One important neutrophil-associated lipocalin (NGAL) pared with gentamicin-treated rats with
point to highlight is that the large grouping and the mouse homolog is 24P3. LCN2 tubular toxicity. For instance, one of the
of genes that are downregulated and has been implicated in the regulation of ESTs (GenBank accession no. AA899472)
grouped together in the clustering diagram cell homeostasis and immune response and appears to have weak similarity to dentin
(Figure 5) could, as a group, serve as a sen- can function as a carrier protein for the phosphoprotein precursor/dentin sialopro-
sitive diagnostic indicator of proximal tubu- general clearance of endogenous and exoge- tein (DSP), whose role in response to renal
lar damage. We confirmed this by using this nous compounds (Flower 1996; Flower et tubular injury has not been previously stud-
set of genes to correctly predict puromycin- al. 2000; Yang et al. 2003). Lipocalin prod- ied. DSP is a 53-kDa protein that has an
induced proximal tubular injury before ucts have been found in neutrophilic gran- overall composition similar to that of the
histopathological analysis was conducted. ules and may play a role in apoptosis sialoprotein osteopontin (Ritchie et al.
Further work will be needed to validate the (Devireddy et al. 2001; Yang et al. 2003). 1994). An increase in level of osteopontin
regulation of the expression of these genes NGAL/24p3 expression has also been mRNA expression in the present studies was
during regenerative/repair processes. detected in inflamed epithelia (Nielsen observed with cisplatin and gentamicin
This work further highlights the appli- et al. 1996), perhaps explaining that the treatment (Figure 3B) [for RT–PCR valida-
cation of microarray in identifying putative appearance of this transcript may be related tion data, refer to Thompson et al. (2004)].
biomarkers of injury. For practical consider- to the damage occurring in the renal Although little is known about the func-
ations in detection assays, it may be most tubules and the accompanying infiltrates tion/role of DSP in response to renal injury,
useful to consider those genes that have low observed histopathologically. Relevant to an increase in the expression of osteopontin
expression in control tissue but are induced/ the present studies is the role implicated for in proximal tubular epithelium has been
upregulated with damage. In this article we lipocalin superfamily members (i.e., demonstrated in human and animal models
have identified several potential biomarkers NGAL/24p3) in inducing the formation of of renal injury and can be associated with
that appear to be upregulated in a dose- and kidney epithelia (Yang et al. 2003). monocyte or macrophage infiltration
time-dependent manner upon injury. These However, the underlying molecular signal- (Hudkins et al. 2001; Magil et al. 1997;
include KIM-1, osteopontin, clusterin, and ing mechanism(s) are not fully known. Pichler et al. 1995). Khan et al. (2002)
several ESTs, including two that were NGAL protein has been identified as a reported concomitant renal expression and
recently defined, LCN2 and TNF receptor complex with matrix metalloproteinases in urinary excretion of osteopontin in a rat
superfamily, member 12a (Tweak receptor). urine of patients with cancer (Yan et al. model of ethylene glycol–induced calcium
Although our study confirmed that the 2001). Lipocalin-type prostaglandin D syn- oxalate nephrolithiasis. Expression of osteo-
expression levels of these genes did increase thase (also known as L-PGDS or β-trace) is pontin protein and mRNA in proximal
in the kidney, further work will be needed another member of the lipocalin family tubular epithelium has been reported in

Environmental Health Perspectives • VOLUME 112 | NUMBER 4 | March 2004 477


Mini-Monograph | Amin et al.

the absence of prominent macrophage foundation for opportunities that exist to Cluster analysis and display of genome-wide
influx in both pretransplant donor biopsies apply gene expression profiling to begin expression patterns. Proc Natl Acad Sci USA
and biopsies with cyclosporine toxicity identifying novel markers of renal toxicity. 95:14863–14868.
Flower DR. 1996. The lipocalin protein family: struc-
(Hudkins et al. 1999, 2001). Further expansion of these analyses may
ture and function. Biochem J. 318:1–14.
Induction of clusterin expression in lead to the identification of groups of genes Flower DR, North AC, Sansom CE. 2000. The lipocalin
these studies was observed after cisplatin- that could ultimately result in the detection protein family: structural and sequence
and gentamicin-induced renal injury and of sensitive markers of renal damage. overview. Biochim Biophys Acta 1482:9–24.
has great potential to be used as a bio- Traditional measures of renal damage, such Girton RA, Sundin DP, Rosenberg ME. 2002. Clusterin
marker of nephrotoxicity. Clusterin is a as proteinuria, creatinine clearance, protects renal tubular epithelial cells from gen-
tamicin-mediated cytotoxicity. Am J Physiol
secreted heterodimeric glycoprotein that and elevated BUN, usually occur after sig-
Renal Physiol 282: 703–709.
circulates in blood at concentrations of nificant kidney damage has occurred. Hamadeh HK, Bushel PR, Jayadev S, DiSorbo O,
50–300 µg/mL (Rosenberg and Silkensen Elucidation and validation of new sensitive Bennett L, Li L, et al. 2002a. Prediction of com-
1995a, 1995b). Clusterin has been induced markers could help clinicians more effi- pound signature using high density gene expres-
in a variety of models of renal tubular ciently monitor patients who are adminis- sion profiling. Toxicol Sci 67:232–240.
injury and/or remodeling after treatment tered potentially nephrotoxic drugs. Hamadeh HK, Bushel PR, Jayadev S, Martin K,
DiSorbo O, Sieber S, et al. 2002b. Gene expres-
with cisplatin (Huang et al. 2001;
sion analysis reveals chemical-specific profiles.
Silkensen et al. 1997) as well as in tubular REFERENCES Toxicol Sci 67:219–231.
epithelial cells, in response to proteinuria, Hamadeh HK, Knight BL, Haugen AC, Sieber S,
in kidneys of nephrotic rats (Correa-Rotter Affymetrix 2003. Technical Documentation Manuals. Amin RP, Bushel PR, et al. 2002. Methapyrilene
et al. 1998) after puromycin treatment. Santa Clara, CA:Affymetrix Inc. Available: toxicity: anchorage of pathologic observations to
http://www.affymetrix.com/support/technical/ma gene expression alterations. Toxicol Pathol
Depletion of clusterin enhances immune nuals.affx [accessed 5 September 2003]. 30:470–482.
glomerular injury in the isolated perfused Andoh TF, Gardner MP, Bennett WM. 1997. Hammerman MR. 1998a. Potential role of growth fac-
kidney (Saunders et al. 1994), and mice Protective effects of dietary L-arginine supple- tors in the prophylaxis and treatment of acute
deficient in clusterin are more susceptible mentation on chronic cyclosporine nephrotoxic- renal failure. Kidney Int Suppl 64:S19–S22.
to injury due to immunocomplexes ity. Transplantation 64:1236–1240. ———. 1998b. Growth factors and apoptosis in
(Rosenberg et al. 2002). Urinary and Applied Biosystems. 1997. User Bulletin #2: Relative acute renal injury. Curr Opin Nephrol Hypertens
Quantitation of Gene Expression. Foster City, 7:419–424.
serum clusterin levels have been studied in CA:Applied Biosystems. Han WK, Bailly V, Abichandani R, Thadhani R,
various models of renal injury (Aulitzky Aulitzky WK, Schlegel PN, Wu DF, Cheng CY, Chen Bonventre JV. 2002. Kidney injury molecule-1
et al. 1992) and may play a role in early CL, Li PS, et al. 1992. Measurement of urinary (KIM-1): a novel biomarker for human renal proxi-
detection of renal injury. Clearly, the clusterin as an index of nephrotoxicity. Proc Soc mal tubule injury. Kidney Int 62:237–244.
potential use of clusterin as a biomarker of Exp Biol Med 199:93–96. Hoffmann A, Conradt HS, Gross G, Nimtz M,
nephrotoxicity has to be confirmed. Bailly V, Zhang Z, Meier W, Cate R, Sanicola M, Lottspeich F, Wurster U. 1993. Purification and
Bonventre JV. 2002. Shedding of kidney injury mol- chemical characterization of beta-trace protein
Numerous efforts are ongoing to iden- ecule-1, a putative adhesion protein involved in from human cerebrospinal fluid: its identification
tify and validate markers of renal function renal regeneration. J Biol Chem 277:39739–39748. as prostaglandin D synthase. J Neurochem
and injury in rodent and human models. Bushel PR, Hamadeh HK, Bennett L, Green J, 61:451–456.
Muramatsu et al. (2002) demonstrated that Ableson A, Misener S, et al. 2002. Computational Hoffmann A, Nimtz M, Conradt HS. 1997. Molecular
cysteine-rich protein 61, a secreted growth selection of distinct class- and subclass-specific characterization of beta-trace protein in human
factor–inducible immediate early gene, is gene expression signatures. J Biomed Inform serum and urine: a potential diagnostic marker
35:160–170. for renal diseases. Glycobiology 7:499–506.
induced in proximal straight tubules of Bushel PR, Hamadeh H, Bennett L, Sieber S, Martin Huang Q, Dunn RT 2nd, Jayadev S, DiSorbo O,
rodents within 2 hr of renal ischemia and K, Nuwaysir EF, et al. 2001. MAPS: a microarray Pack FD, Farr SB, et al. 2001. Assessment of cis-
can be detected in rat urine as early as project system for gene expression experiment platin-induced nephrotoxicity by microarray
3–6 hr after renal injury. Cystatin C is a information and data validation. Bioinformatics technology. Toxicol Sci 63:196–207.
13-kDa plasma protein that inhibits cys- 17:564–565. Hudkins KL, Giachelli CM, Cui Y, Couser WG,
teine proteases and is freely filtered at the Chen Y, Dougherty ER, Bittner ML. 1997. Ratio-based Johnson RJ, Alpers CE. 1999. Osteopontin
decisions and the quantitative analysis of cDNA expression in fetal and mature human kidney. J
glomerulus. Cystatin C appears to have microarray images. J Biomed Opt 2:364–374. Am Soc Nephrol 10:444–457.
improved diagnostic accuracy compared Christensson A, Ekberg J, Grubb A, Ekberg H, Hudkins KL, Le QC, Segerer S, Johnson RJ, Davis CL,
with creatinine measurements as a serum Lindstrom V, Lilja H. 2003. Serum cystatin C is a Giachelli CM, et al. 2001. Osteopontin expression
marker of GFR (Christensson et al. 2003; more sensitive and more accurate marker of in human cyclosporine toxicity. Kidney Int
Laterza et al. 2002; Newman 2002). Oda glomerular filtration rate than enzymatic mea- 60:635–640.
et al. (2002) have demonstrated the utility surements of creatinine in renal transplantation. Ichimura T, Bonventre JV, Bailly V, Wei H,
Nephron Physiol 94:19–27. Hession CA, Cate RL, et al. 1998. Kidney injury
of monitoring lipocalin-type urinary Clements, JA. 1989. The glandular kallikrein family of molecule-1 (KIM-1), a putative epithelial cell
β-trace in patients with renal disease. Star enzymes: tissue-specific expression and hor- adhesion molecule containing a novel
et al. (2002) noted that the approach monal regulation. Endocr Rev 10:393–419. immunoglobulin domain, is up-regulated in renal
described in the article by Oda et al. Correa-Rotter R, Ibarra-Rubio ME, Schwochau G, cells after injury. J Biol Chem 273:4135–4142.
(2002) for developing and evaluating an Cruz C, Silkensen JR, Pedraza-Chaverri J, et al. Khan SR, Johnson JM, Peck AB, Cornelius JG,
ELISA-based approach for lipocalin-type 1998. Induction of clusterin in tubules of Glenton PA. 2002. Expression of osteopontin in
nephrotic rats. J Am Soc Nephrol 9:33–37.
urinary β-trace provides valuable insight DeRisi J, Penland L, Brown PO, Bittner ML,
rat kidneys: induction during ethylene glycol
induced calcium oxalate nephrolithiasis. J Urol
into efforts that need to be undertaken to Meltzer PS, Ray M, et al. 1996. Use of a cDNA 168:1173–1181.
translate a novel marker into a valuable microarray to analyze gene expression patterns Kirsztajn GM, Nishida SK, Silva MS, Ajzen H,
-laboratory assay. in human cancer. Nat Genet 14:457–460. Moura LA, Pereira AB. 2002. Urinary retinol-bind-
In conclusion, this study indicates that Devireddy LR, Teodoro JG, Richard FA, Green MR. ing protein as a prognostic marker in glomeru-
gene expression arrays may be used to dis- 2001. Induction of apoptosis by a secreted lopathies. Nephron 90:424–431.
lipocalin that is transcriptionally regulated by IL-3 Kramer JA, Petit SD, Amin RP, Bertram TA, Car B,
cern regional specific damage of nephro- deprivation. Science 293:829–834. Cunningham M, et al. 2004. Overview of the
toxicity. Additionally, this study provides a Eisen MB, Spellman PT, Brown PO, Botstein D. 1998.

478 VOLUME 112 | NUMBER 4 | March 2004 • Environmental Health Perspectives


Mini-Monograph | Gene-based markers of renal toxicity

application of transcription profiling using ———. 2003c. ILSI Datasets. Research Triangle Hasan SK, Husain MM. 1996. Evidence for the
selected nephrotoxicants for toxicology assess- Park, NC:National Center for Toxicogenomics, involvement of nitric oxide in cisplatin-induced
ment. Environ Health Perspect 112:460–464. National Institute of Environmental Health toxicity in rats. Biometals 9:139–142.
Laterza OF, Price CP, Scott MG. 2002. Cystatin C: an Sciences. Available: http://dir.niehs.nih.gov/ Star R, Hostetter T, Hortin GL. 2002. New markers for
improved estimator of glomerular filtration rate? microarray/ilsi-datasets/home.htm [accessed 5 kidney disease. Clin Chem 48:1375–1376.
Clin Chem 48:699–707. September 2003]. Thompson KL, Afshari CA, Amin RP, Bertram TA,
Lee H, Kim JH, Chae YJ, Ogawa H, Lee MH, NIH. 1999. NIH Guide for the Care and Use of Car B, Cunningham M, et al. 2004. Identification
Gerton GL. 1998. Creatine synthesis and transport Laboratory Animals. Bethesda, MD:National of platform-independent gene expression mark-
systems in the male rat reproductive tract. Biol Institutes of Health. ers of cisplatin nephrotoxicity. Environ Health
Reprod 58:1437–1444. Newman DJ. 2002. Cystatin C. Ann Clin Biochem Perspect 112:488–496.
Lockhart DJ, Dong H, Byrne MC, Follettie MT, 39:89–104. Toback FG. 1992. Regeneration after acute tubular
Gallo MV, Chee MS, et al. 1996. Expression moni- Nielsen BS, Borregaard N, Bundgaard JR, Timshel S, necrosis. Kidney Int 41:226–246.
toring by hybridization to high-density oligonu- Sehested M, Kjeldsen L. 1996. Induction of NGAL Tome LA, Yu L, de Castro I, Campos SB, Seguro AC.
cleotide arrays. Nat Biotechnol 14:1675–1680. synthesis in epithelial cells of human colorectal 1999. Beneficial and harmful effects of L-arginine
Magil AB, Pichler RH, Johnson RJ. 1997. Osteopontin neoplasia and inflammatory bowel diseases. Gut on renal ischaemia. Nephrol Dial Transplant
in chronic puromycin aminonucleoside nephro- 38:414–420. 14:1139–1145.
sis. J Am Soc Nephrol 8:1383–1390. Norman JT, Bohman RE, Fischmann G, Bowen JW, Urade Y, Hayaishi O. 2000. Prostaglandin D synthase:
Makover A, Soprano DR, Wyatt ML, Goodman DS. McDonough A, Slamon D, et al. 1988. Patterns of structure and function. Vitam Horm 58:89–120.
1989. Localization of retinol-binding protein mes- mRNA expression during early cell growth differ Valdivielso JM, Blantz RC. 2002. Acute renal failure:
senger RNA in the rat kidney and in perinephric in kidney epithelial cells destined to undergo is nitric oxide the bad guy? Antioxid Redox Signal
fat tissue. J Lipid Res 30:171–180. compensatory hypertrophy versus regenerative 4:925–934.
Matejka GL. 1998. Expression of GH receptor, IGF-I hyperplasia. Proc Natl Acad Sci USA Waring JF, Ciurlionis R, Jolly RA, Heindel M,
receptor and IGF-I mRNA in the kidney and liver 85:6768–6772. Ulrich RG. 2001a. Microarray analysis of hepato-
of rats recovering from unilateral renal ischemia. Oda H, Shiina Y, Seiki K, Sato N, Eguchi N, Urade Y. toxins in vitro reveals a correlation between
Growth Horm IGF Res 8:77–82. 2002. Development and evaluation of a practical gene expression profiles and mechanisms of tox-
Mattenheimer H. 1968. Enzymology of kidney tissue. ELISA for human urinary lipocalin-type icity. Toxicol Lett 120:359–368.
Curr Probl Clin Biochem 2:119–145. prostaglandin D synthase. Clin Chem 48:1445–1453. Waring JF, Jolly RA, Ciurlionis R, Lum PY,
Melegos DN, Grass L, Pierratos A, Diamandis EP. Ortiz A, Lorz C, Justo P, Catalan MP, Egido J. 2001. Praestgaard JT, Morfitt DC, et al. 2001b.
1999. Highly elevated levels of prostaglandin D Contribution of apoptotic cell death to renal Clustering of hepatotoxins based on mechanism
synthase in the serum of patients with renal fail- injury. J Cell Mol Med 5:18–32. of toxicity using gene expression profiles. Toxicol
ure. Urology 53:32–37. Pennie WD, Pettit SD, Lord PG. 2004. Toxicogenomics Appl Pharmacol 175:28–42.
Murakami H, Yayama K, Chao L, Chao J. 1998. Human in risk assessment: an overview of an HESI col- WHO. 1991. Principles and Methods for the
kallikrein gene delivery protects against gen- laborative research program. Environ Health Assessment of Nephrotoxicity Associated with
tamycin-induced nephrotoxicity in rats. Kidney Perspect 112:417–419. Exposure to Chemicals. Environmental Health
Int 53:1305–1313. Pichler RH, Franceschini N, Young BA, Hugo C, Criteria 119. Geneva:World Health Organization,
Muramatsu Y, Tsujie M, Kohda Y, Pham B, Andoh TF, Burdmann EA, et al. 1995. Pathogenesis International Programme on Chemical Safety.
Perantoni AO, Zhao H, et al. 2002. Early detection of cyclosporine nephropathy: roles of angiotensin Yan L, Borregaard N, Kjeldsen L, Moses MA. 2001.
of cysteine rich protein 61 (CYR61, CCN1) in urine II and osteopontin. J Am Soc Nephrol 6:1186–1196. The high molecular weight urinary matrix metal-
following renal ischemic reperfusion injury. Ritchie HH, Hou H, Veis A, Butler WT. 1994. Cloning loproteinase (MMP) activity is a complex of
Kidney Int 62:1601–1610. and sequence determination of rat dentin sialo- gelatinase B/MMP-9 and neutrophil gelatinase-
Nagasawa Y, Takenaka M, Kaimori J, Matsuoka Y, protein, a novel dentin protein. J Biol Chem associated lipocalin (NGAL). Modulation of
Akagi Y, Tsujie M, et al. 2001. Rapid and diverse 269:3698–3702. MMP-9 activity by NGAL. J Biol Chem
changes of gene expression in the kidneys of Rosenberg ME, Girton R, Finkel D, Chmielewski D, 276:37258–37265.
protein-overload proteinuria mice detected by Barrie A 3rd, Witte DP, et al. 2002. Apolipoprotein Yang J, Mori K, Li JY, Barasch J. 2003. Iron, lipocalin,
microarray analysis. Nephrol Dial Transplant J/clusterin prevents a progressive glomerulopa- and kidney epithelia. Am J Physiol Renal Physiol
16:923–931. thy of aging. Mol Cell Biol 22:1893–1902. 285:F9–F18.
Nagata A, Suzuki Y, Igarashi M, Eguchi N, Toh H, Rosenberg ME, Silkensen J. 1995. Clusterin and the Yasuda M, Sugahara K, Zhang J, Shuin T, Kodama H.
Urade Y, et al. 1991. Human brain prostaglandin D kidney. Exp Nephrol 3:9–14. 2000. Effect of cisplatin treatment on the urinary
synthase has been evolutionarily differentiated ———. 1995. Clusterin: physiologic and pathophysio- excretion of guanidinoacetic acid, creatinine and
from lipophilic-ligand carrier proteins. Proc Natl logic considerations. Int J Biochem Cell Biol creatine in patients with urinary tract neoplasm,
Acad Sci USA 88:4020–4024. 27:633–645. and on superoxide generation in human neu-
Nakao A, Watanabe T, Ohishi N, Toda A, Asano K, Safirstein R, Price PM, Saggi SJ, Harris RC. 1990. trophils. Physiol Chem Phys Med NMR
Taniguchi S, et al. 1999. Ubiquitous localization of Changes in gene expression after temporary 32:119–125.
leukotriene A4 hydrolase in the rat nephron. renal ischemia. Kidney Int 37:1515–1521. Yoshida T, Kurella M, Beato F, Min H, Ingelfinger JR,
Kidney Int 55:100–108. Saunders JR, Aminian A, McRae JL, O’Farrell KA, Stears RL, et al. 2002. Monitoring changes in
NIEHS Microarray Group. 2003a. NIEHS Chips and Adam WR, Murphy BF. 1994. Clusterin depletion gene expression in renal ischemia-reperfusion in
Clones. Research Triangle Park, NC:National enhances immune glomerular injury in the iso- the rat. Kidney Int 61:1646–1654.
Center for Toxicogenomics, National Institute of lated perfused kidney. Kidney Int 45:817–827. Yu H, Anderson PJ, Freedman BI, Rich SS,
Environmental Health Sciences. Available: Schmaier AH. 2003. The kallikrein-kinin and the renin- Bowden DW. 2000. Genomic structure of the
http://dir.niehs.nih.gov/microarray/chips.htm angiotensin systems have a multilayered interac- human plasma prekallikrein gene, identification
[accessed 5 September 2003]. tion. Am J Physiol Regul Integr Comp Physiol of allelic variants, and analysis in end-stage
———. 2003b. Methods and Protocols. Research 285:R1–R13. renal disease. Genomics 69:225–234.
Triangle Park, NC:National Center for Silkensen JR, Agarwal A, Nath KA, Manivel JC, Yu H, Song Q, Freedman BI, Chao J, Chao L, Rich SS,
Toxicogenomics, National Institute of Rosenberg ME. 1997. Temporal induction of clus- et al. 2002. Association of the tissue kallikrein
Environmental Health Sciences. Available: terin in cisplatin nephrotoxicity. J Am Soc gene promoter with ESRD and hypertension.
http://dir.niehs.nih.gov/microarray/methods.htm Nephrol 8:302–305. Kidney Int 61:1030–1039.
[accessed 5 September 2003]. Srivastava RC, Farookh A, Ahmad N, Misra M,

Environmental Health Perspectives • VOLUME 112 | NUMBER 4 | March 2004 479

You might also like