You are on page 1of 29

Veterinary Clinical Pathology ISSN 0275-6382

REVIEW ARTICLE

Renal biomarkers in domestic species


Jessica A. Hokamp, Mary B. Nabity
Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA

Key Words Abstract: Current conventional tests of kidney damage and function in
Cats, dogs, enzymes, kidney, SDMA blood (serum creatinine and urea nitrogen) and urine (urine protein crea-
tinine ratio and urine specific gravity) are widely used for diagnosis and
Correspondence
monitoring of kidney disease. However, they all have important limita-
J.A. Hokamp, Department of Veterinary
Pathobiology, College of Veterinary Medicine
tions, and additional markers of glomerular filtration rate and glomerular
and Biomedical Sciences, Texas A&M and tubular damage are desirable, particularly for earlier detection of renal
University, 4467 TAMU, College Station, disease when therapy is most effective. Additionally, urinary markers of
TX 77843, USA kidney damage and function may help localize damage to the affected
E-mail: jhokamp@cvm.tamu.edu portion of the kidney. In general, the presence of high- and intermediate-
molecular weight proteins in the urine are indicative of glomerular
DOI:10.1111/vcp.12333 damage, while low-molecular weight proteins and enzymes in the urine
suggest tubular damage due to decreased reabsorption of proteins, direct
tubular damage, or both. This review aims to discuss many of these new
blood and urinary biomarkers in domestic veterinary species, focusing
primarily on dogs and cats, how they may be used for diagnosis of renal
disease, and their limitations. Additionally, a brief discussion of serum
creatinine is presented, highlighting its limitations and important consider-
ations for its improved interpretation in domestic species based on past
literature and recent studies.

Markers of Glomerular Damage/Dysfunction


I. Introduction a. Immunoglobulins
II. Renal Biomarkers: Blood i. Background
Endogenous markers of glomerular filtration rate (GFR) ii. Measurement and Stability
a. Creatinine iii. Values in Healthy Animals
i. Nephron Mass vs. Nephron Function iv. Non-Renal Influences
ii. Value of Population-Specific Reference Intervals v. Renal Disease in Veterinary Medicine
iii. Trending of Serum Creatinine 1. Acute Kidney Injury (AKI)
iv. Non-Renal Influences 2. Chronic Kidney Disease (CKD)
v. Analytic Challenges b. Additional Urinary Proteins Indicating Glomerular Damage/Dysfunction
b. Symmetric Dimethylarginine (SDMA) i. Albumin
i. Background ii. C-Reactive Protein
ii. Measurement and Stability Markers of Tubular Damage/Dysfunction
iii. Reference Limit a. Retinol Binding Protein (RBP)
iv. Non-Renal Influences i. Background
v. Renal Disease in Veterinary Medicine ii. Measurement and Stability
Markers of altered metabolism iii. Values in Healthy Animals
c. Fibroblast Growth Factor-23 (FGF-23) iv. Non-Renal Influences
i. Background v. Renal Disease in Veterinary Medicine
ii. Measurement, Stability, and Reference Range 1. AKI
iii. Non-renal Influences 2. CKD
iv. Renal Disease in Veterinary Medicine d. Neutrophil Gelatinase-Associated Lipocalin (NGAL)
III. Renal Biomarkers: Urine i. Background
Renal Protein Handling ii. Measurement and Stability

(continued) (continued)

28 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

iii. Non-Renal Influences Renal Biomarkers: Blood


iv. Renal Disease in Veterinary Medicine
1. AKI Endogenous markers of GFR
2. CKD
e. Additional Urinary Proteins Indicating Tubular Damage/Dysfunction
Creatinine
i. Tamm-Horsfall Protein
ii. Cystatin C Serum creatinine is the most widely used endogenous
f. Urinary Enzymes marker for estimating GFR, and its metabolism, mea-
i. Background
surement, and diagnostic significance in dogs have
ii. Measurement and Stability
previously been reviewed.4 While the present review
iii. Values in Healthy Animals
iv. Non-Renal Influences focuses primarily on new tests of renal function, it is
v. Renal Disease in Veterinary Medicine important to consider factors that can either enhance
1. AKI or limit the clinical use of sCr in order to optimally help
2. CKD clinicians and clinical pathologists interpret the value
i. Cauxin of this conventional test. In particular, accurate inter-
IV. Conclusions
pretation of the literature, population-specific refer-
V. References
ence intervals, trending of sCr, and consideration of
muscle mass influence and analytic variability are all
needed to best interpret sCr in dogs and cats. Of note,
although creatinine is referred to as sCr throughout
Introduction
this manuscript, creatinine is also commonly measured
in plasma.
Urinalysis has contributed to medical diagnoses for
thousands of years. However, it was not until the
routine use of clinical chemistry approximately 50– Nephron mass vs nephron function. It is widely accepted
60 years ago that measurement of renal biomarkers that at least 75% of nephron mass must be lost before
became commonplace in human and veterinary sCr increases above the reference limit.4 The original
medicine. This allowed for both an improved under- source for this statement likely originates from partial
standing of the renal system and ability to diagnose nephrectomy studies in dogs. However, the statement is
renal disease. Historically, renal biomarkers have often misinterpreted as 75% loss of renal function vs
focused on kidney function testing, and this is the mass. In partial nephrectomy studies, 3/4th loss of renal
basis for current conventional tests in blood (serum mass corresponded to an approximately 50–60% or 35–
creatinine [sCr], urea or urea nitrogen [UN]) as 45% decrease in renal function based on inulin clearance
endogenous indicators of glomerular filtration rate one month or 13 months post surgery, respectively.5,6
(GFR). We are increasingly recognizing the need to The much lower decrease in function as compared with
identify renal disease at an early stage, when thera- the percentage of nephron loss is due to compensatory
peutic options are most effective. While sCr and UN changes in remaining nephrons (ie, compensatory renal
play an important role in the diagnosis of kidney hypertrophy).5,7 Furthermore, using an age- and
disease, their limitations create poor confidence for breed-specific reference limit (sCr ≥ 106 mmol/L or
their use as early indicators of disease. New markers 1.2 mg/dL) along with frequent monitoring, adolescent
of renal function aim to overcome these limitations. dogs with rapidly progressive kidney disease due to X-
In addition, there are now many urinary markers linked hereditary nephropathy (XLHN) demonstrated
that can detect kidney damage and help localize increased sCr after GFR had decreased an average of
that damage to the compartment of the kidney that 48% (range 39–68%).8 Based on these studies, sCr can
is affected. be more sensitive for detecting decreased renal function
Many excellent and comprehensive reviews of than has been historically assumed.
renal biomarkers have been published focusing on the
human literature, with several recent reviews avail- Value of population-specific reference intervals. While sCr
able in veterinary medicine.1–3 This review aims to is not as poorly sensitive as generally believed, its
comprehensively summarize the field in veterinary inability to consistently detect < 50% decline in kidney
medicine, particularly focusing on recent advances in function at least partly stems from reference intervals
renal biomarker research. that are overly wide for patients with low baseline

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 29
Renal biomarkers review Hokamp and Nabity

sCr. Since current methodologies are highly specific study of geriatric cats, where both sCr and GFR were
for creatinine, the wide reference intervals largely lower in cats > 15 years old as compared with cats
stem from biologic differences in sCr among individ- < 15 years old.22 Therefore, even careful monitoring
uals. Serum creatinine has relatively high individual- of sCr over time in an individual patient is an unreli-
ity in dogs and cats9–11, meaning that variability able indicator of declining GFR if concurrent muscle
between individuals is much higher than the variabil- wasting is present. Additionally, as might be expected
ity observed within a single animal. Serum creatinine with any endogenous marker of GFR, hydration and
is influenced by age12,13 and particularly by breed in blood volume status as well as urinary tract obstruction
dogs14–19 and, to a lesser extent, in cats.20 It might or rupture can influence sCr. Particularly early on,
also be influenced by sex and the veterinary clinic these changes do not necessarily reflect inherent kid-
evaluating the patient.21 Therefore, sCr would bene- ney function, further hampering the sensitivity and
fit from age- and breed-specific reference intervals, specificity of sCr for kidney disease.
ideally (although not practically) for every individual
instrument and laboratory.
Analytic challenges. Finally, sCr is plagued by inconsis-
tencies in its measurement between instruments and
Trending of serum creatinine. Small increases in sCr even laboratories, which can result in markedly different
within the reference interval can reflect significant results. While most reference laboratory instruments
decreases in GFR in an individual patient8, particularly have excellent precision and provide results of similar
since variation in sCr within an individual healthy dog magnitude among instruments23, recent studies illus-
or cat is minimal over weeks to months and even trate the high imprecision and bias possible with some
years.4,9–11 In fact, the critical difference or reference instruments and among different laboratories.23,24 In
change value for detecting a significant increase or normal to mildly azotemic samples, one study using
decrease in sCr is only 23–27 lM/L (0.3 mg/dL) in reference laboratories found differences of up to
clinically healthy dogs10, and 17% (corresponding to 40 lmol/L (0.45 mg/dL) in the same sample measured
similar absolute values as in dogs) in clinically healthy by the same laboratory and up to 50 lmol/L (0.57 mg/
cats.9 Thus, the sensitivity of sCr for detecting early dL) among different laboratories, even when exclud-
kidney disease can be improved by evaluating serial ing 2 laboratories (out of 10) with extreme outliers.23
fasted sCr measurements in an individual animal Larger differences were noted in moderately to mark-
(trending) to look for increases that likely reflect wors- edly azotemic samples: excluding outlier results from 3
ening renal function. This concept of detecting small laboratories, up to a 68 lmol/L (0.77 mg/dL) differ-
but clinically relevant increases in sCr is actively being ence was observed within the same laboratory, and
adopted in cases of acute kidney injury (AKI), illus- 117 lmol/L (1.32 mg/dL) difference between labora-
trated by the International Renal Interest Society tories.23 A still larger variation in sCr measurement
(IRIS) Grading of AKI. In this grading scheme, an was observed among 99 veterinary practices, where
increase in sCr ≥ 26 lmol/L (0.3 mg/dL) within a 48- results ranged from 80 to 200 lmol/L (0.9–2.26 mg/
hour period is a criterion for identifying Grade I and dL) for a single sample spiked with 112 lmol/L
Grade II AKI (www.IRIS-Kidney.com). Furthermore, (1.25 mg/dL) creatinine.24
in adolescent dogs with XLHN, trending of sCr detected These results highlight that many instruments/
an average of 27% (range 5–49%) decrease in GFR.8 laboratories are performing below the total allowable
Despite heightened awareness of small, but clinically error (TEa) guideline for sCr (TEa ≤ 20%) set by the
relevant increases in sCr over a short time frame, more Quality Assurance and Laboratory Standards Commit-
recognition is needed with slowly progressive CKD, in tee of the American Society for Veterinary Clinical
which small increases might occur over many months Pathology (ASVCP).25 Even with TEa ≤ 20%, analytic
or years. variability alone (instrument precision and bias) could
account for an increase or decrease in sCr ≥ 18 mmol/
Nonrenal influences. It is well known that an inherent L (0.2 mg/dL) in normal to mildly azotemic samples,
limitation of sCr is its dependence on muscle mass. which is approaching the critical difference for detect-
Serum creatinine can overestimate renal function in ing a significant change in sCr in clinically healthy dogs
cachectic, geriatric, and very young patients as well as and cats.9,10 To minimize this analytic variability, it is
in small-breed dogs. Additionally, sCr increases might particularly important that serial determinations of sCr
be offset by ongoing muscle wasting in animals with are measured on a single instrument that is subjected
CKD and in elderly animals, as supported by a recent to a strict quality assurance program.

30 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

In summary, sCr can be a sensitive marker of precision, although accuracy was not assessed.42 An
declining renal function if careful monitoring and/or accurate and precise liquid chromatography–mass
appropriate reference intervals are used in animals spectrometry (LC–MS/MS) assay has recently been
with relatively stable muscle mass. However, because developed for SDMA measurement in dogs and
of both inherent biologic and current analytic limita- cats.8,43 In addition, a high-throughput immunoassay
tions of sCr, additional endogenous markers of GFR with similar performance to the LC–MS/MS method
would aid in the early diagnosis of renal disease. Two has been developed and validated44, allowing SDMA
recently studied markers are cystatin C and symmetric to become a standard analyte on chemistry panels
dimethylarginine (SDMA). An excellent recent review offered by IDEXX Laboratories, Inc. Symmetric
of cystatin C is available.26 Therefore, only SDMA will dimethylarginine is stable in canine and feline serum
be further discussed below. and plasma for at least 7 days at room temperature,
and 14 days at 4°C and with up to 3 freeze–thaw cycles
Symmetric dimethylarginine (SDMA) (between 80°C and room temperature).8,45 While no
Symmetric dimethylarginine is a methylated amino published studies are available regarding the long-term
acid (arginine) of similar size to creatinine (SDMA: stability of SDMA in frozen samples, anecdotal evi-
202 daltons [Da]; creatinine: 113 Da). Symmetric dence supports that it is stable for at least 5 years when
dimethylarginine is formed by posttranslational frozen at 20°C or 80°C (IDEXX, Laboratories, Inc.,
methylation of arginine by type 2 protein arginine Personal Communication).
methyltransferases27, and it is released into circulation
following proteolysis. Symmetric dimethylarginine Reference limit. In clinically healthy animals, SDMA
was originally discovered 45 years ago in human concentrations are similar across studies and species,
urine.28 Concentrations of Symmetric dimethylargi- despite differences in methodology and animal popula-
nine in protein fractions from various organs in rats tions. Two studies in healthy human subjects deter-
demonstrated SDMA to be highest in the brain and rel- mined the reference interval using LC–MS/MS as
atively high in the liver, lung, kidney, spleen, and 0.32–0.65 lmol/L (6.5–13.1 lg/dL)46 and 0.225–
small intestine as compared with heart, muscle, skin, 0.533 lmol/L (4.5–10.8 lg/dL).47 Using serum from
and blood.29 The kidneys are the major source of 120 healthy adult dogs of varying ages and breeds, a
SDMA excretion28,30–32, and SDMA does not appear to 95% reference interval for SDMA was calculated as 6–
be reabsorbed by the tubules for reutilization.28 Sym- 13 lg/dL48, and in both dogs and cats, the upper refer-
metric dimethylarginine was first shown to be ence limit using the LC–MS/MS methodology was set
increased in human patients with CKD over 20 years at < 14 lg/dl (0.69 lmol/L). Furthermore, a range of
ago33; however, SDMA has only recently gained 7.3–12.4 lg/dL (0.36–0.61 lmol/L) was observed in
prominence as an endogenous marker of GFR in peo- 21 healthy geriatric cats in a recent study.43 However,
ple34–36, correlating strongly with renal function based in juvenile dogs, SDMA might occasionally reach or
on inulin clearance.36 Symmetric dimethylarginine exceed the 14 lg/dL reference limit.8 In older studies
has largely been believed to be inert, reducing nitric using high-performance LC, SDMA in healthy dogs
oxide synthesis indirectly by competing with L-argi- ranged from 0.22 to 0.61 lmol/L (4.4–12.3 lg/dL),49–
nine uptake in cells.35,37 However, a recent study 51
although values up to 14.1 lg/dL (0.7 lmol/L) were
demonstrated a direct effect of SDMA, in which observed in a group consisting of both healthy dogs
uncoupling of endothelial nitric oxide synthase and dogs with mitral regurgitation.52
resulted in superoxide anion production in glomerular
endothelial cells.38 In addition, a recent review high-
lights several studies that support a proinflammatory Nonrenal influences. Currently, SDMA’s rate of produc-
role of SDMA.39 However, chronic infusion of SDMA tion is thought to be relatively constant, although in
in healthy mice had no evident effect on renal struc- theory, altered arginine methylation and/or protein
ture or function as well as cardiac function.40 breakdown could contribute to increased or decreased
serum concentrations. Furthermore, SDMA does not
appear to be highly protein bound53, and its clearance
Measurement and stability. Mass spectrometry is the gold is similar to that of creatinine, supporting that it is
standard for SDMA measurement, as it uniquely and freely filtered through the glomerulus.54 It is largely
accurately detects the molecule.41 However, recent renally excreted; however, hepatic clearance of SDMA
studies in people have used a commercially available was observed in people55, and a small degree of uptake
ELISA with at least one study demonstrating good by the gut was observed in rats.56 Unlike asymmetric

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 31
Renal biomarkers review Hokamp and Nabity

dimethylarginine (ADMA), which is largely degraded of angiotensin-converting-enzyme inhibitors.49,50,52


by dimethylarginine dimethylaminohydrolase In one study, SDMA demonstrated a mild but statisti-
(DDAH)57, enzymatic degradation does not seem to be cally significant correlation with adjusted body weight
a major factor in SDMA elimination. However, some only when excluding creatinine as an explanatory
degree of enzymatic degradation of SDMA is possi- variable, and the authors speculated that this was due
ble55,58, and in particular, alanine-glyoxylate amino- to lower GFR in the larger dogs.52
transferase 2 (Agxt2), which is largely present in the
liver but also kidney, can metabolize SDMA.59 Sym-
metric dimethylarginine does not appear to be signifi- Renal disease in veterinary medicine. Several recently
cantly metabolized by renal tubules.30 No studies were published studies of SDMA in dogs and cats have
identified that determined whether or not SDMA can shown strong evidence for SDMA as an endogenous
be secreted from the tubules or reabsorbed for recircu- surrogate marker for GFR, particularly in animals with
lation, although these processes seem unlikely.28 renal disease. In dogs, SDMA correlated strongly with
Various studies, typically measuring SDMA as an inulin clearance in a partial nephrectomy model of
afterthought to ADMA, have found a variety of CKD (r = .851)51 and with iohexol clearance using
external factors to show a lack of statistically signifi- serial measurements in adolescent dogs with XLHN
cant and/or clinically relevant influence on SDMA (r = .95).8 In studies combining both healthy cats
concentrations in people or rats, including weight and cats with CKD, correlations of SDMA or 1/SDMA
loss60, inflammation61, diabetes62, and estrogen ther- with iohexol clearance were r = .7943 and .8271,
apy.63,64 Diet also does not appear to influence respectively. These were similar to correlations of sCr
SDMA in people, even with lysine and arginine load- with clearance. Furthermore, in healthy geriatric
ing.28,65,66 Furthermore, SDMA in people is mini- cats, the correlation of SDMA with iohexol clearance
mally influenced by obesity67, gender, age68, and was higher (r = -0.72) than that observed for sCr
polycystic ovary syndrome.69 (r = .50).22 Symmetric dimethylarginine correla-
In cats and dogs, several nonrenal influences have tion with sCr was also strong in both dogs and
been investigated. Notably, SDMA is not influenced by cats when including those with kidney disease
muscle mass, the major nonrenal influence on sCr. In (r = .72–.95)8,43,71,72, whereas in clinically healthy
healthy geriatric cats and adult dogs, SDMA did not dogs and cats, the correlation between SDMA and sCr
correlate with lean body mass.43,70 Furthermore, in was much lower (r = .32–.46).43,52,70
healthy growing dogs from 2 months to approximately Importantly, SDMA appears to detect a decrease
one year of age, SDMA did not correlate with age, in GFR prior to sCr when based on reference limits
weight, or body condition score, whereas sCr strongly in cats and dogs. In 21 geriatric laboratory cats with
correlated with these variables.8 Intriguingly, SDMA naturally occurring CKD, SDMA increased by the
did not increase as GFR (normalized to body weight) time azotemia (sCr > 2.1 mg/dL) developed in all
decreased in these growing dogs.8 While this could cats, and SDMA increased above its reference limit
suggest a nonrenal influence on SDMA in adolescent an average of 17 months earlier (up to 48 months
dogs, the normalization method for clearance tech- earlier) than sCr.43 Symmetric dimethylarginine also
niques needs further investigation in young, growing shows promise as a sensitive screening test for CKD
dogs before ascribing clinical significance to stable in cats. Using iohexol clearance to estimate GFR and
SDMA concentrations during growth. In addition, in > 30% decrease from the median of clinically healthy
healthy dogs participating in a dietary trial (comparing controls as the gold standard for decreased GFR,
a renal diet with or without supplementation of L-car- SDMA had perfect sensitivity and negative predictive
nitine and fish oil), SDMA significantly decreased over value (100%), indicating that cats with SDMA
6 months (from an average of 13.3 to 6.5 lg/dL).70 < 14 lg/dL did not have decreased GFR.43 Specificity
While the authors speculate that this decrease could be and positive predictive value (PPV) were slightly
due to improved renal function secondary to a dietary lower (91% and 86%, respectively), due to 2 cases
effect, no clearance techniques were performed in where SDMA was mildly increased with a GFR only
these dogs, and nonrenal causes for this decrease can- 25% lower than the median.43 However, this could
not be ruled out. indicate that SDMA can detect < 30% decrease in
In studies evaluating additional nonrenal vari- GFR in cats. In comparison, the upper reference limit
ables in dogs, SDMA does not appear to be influenced of sCr provided perfect specificity and PPV (100%),
by breed, body size, sex, age (in adults), diurnal varia- indicating that cats with sCr above the reference
tion, white-coat effect, or short-term administration interval (sCr > 2.1 mg/dL) all had > 30% decline in

32 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

GFR. However, sensitivity was quite poor (17%), FGF-23 in CKD and secondary renal hyperparathy-
indicating that many cats with decreased GFR are roidism, few studies are available in domestic ani-
undetected when using this reference limit.43 Since mals, and these have focused on feline CKD in
specificity and sensitivity depend on the reference geriatric (> 9 years) cats.78–80
limit used, certainly use of a lower reference limit for
sCr would have improved its sensitivity in this study,
Measurement, stability, and reference range. A human
particularly since sCr in the clinically healthy cats
FGF-23 ELISA has been validated for the detection of
was ≤ 1.6 mg/dL.
plasma FGF-23 concentrations in cats, with acceptable
In dogs with rapidly progressive CKD due to
precision, reproducibility, and dilutional linearity.
XLHN, SDMA increased an average of 4–5 weeks prior
Plasma FGF-23 was stable for up to 7 days at 22°C and
to an increase in sCr and decrease in GFR, based on
14 days at 20°C as well as up to 4 freeze–thaw
their respective reference limits.8 However, when
cycles.80 A reference range determined for geriatric
trending both sCr and SDMA in individual dogs,
cats (56–700 pg/mL) was higher and broader than that
SDMA increased an average of only 2 weeks prior to
used in adult people, which could be due to subclinical
an increase in sCr.8 Notably, SDMA identified ≤ 34%
CKD in the cats used to establish the reference range,
(range, 6–34%) decrease in GFR when compared
diet, or species-specific factors.80
with the GFR of unaffected, age-matched dogs in this
colony, regardless of whether the increase in SDMA
was based on the reference limit, trending, or compar- Renal disease in veterinary medicine. Plasma FGF-23 con-
ison with healthy controls. In some dogs, SDMA centration was significantly increased at baseline in
increased even before iohexol clearance was below geriatric cats that developed azotemia within the
that observed in unaffected, age-matched dogs.8 This following 12 months compared with those that
was in contrast to sCr, which detected a 5–68% remained nonazotemic78, and plasma FGF-23
decrease in GFR. increased significantly with increasing IRIS stage of
In summary, SDMA appears to be a useful endoge- CKD.80 Additionally, the presence of hyperphos-
nous marker of GFR and is particularly promising as a phatemia was associated with greater plasma FGF-23
screening test for early detection of decreased renal compared with normophosphatemic cats within the
function. Furthermore, because it is not influenced by same IRIS stage.80 Plasma phosphate concentration
lean body mass and is less variable among different dog was shown to be an independent predictor of FGF-23
breeds, SDMA could prove especially useful in those concentration.80 Furthermore, plasma FGF-23 con-
patients with poor muscle mass or ongoing muscle loss, centrations decreased significantly from baseline in
where sCr would provide an unreliable estimate of geriatric cats with stable azotemic CKD fed a reduced
GFR. Further studies are needed in veterinary medi- phosphate renal diet for 4–8 weeks. This FGF-23
cine to determine possible extra-renal influences on decrease was observed even in cats that were nor-
SDMA concentration. mophosphatemic (plasma phosphate ≤ 4.5 mg/dL),
although phosphate and PTH concentrations
remained unchanged.79 Hyperthyroid cats with azote-
Markers of altered metabolism
mic CKD or that developed azotemia after becoming
euthyroid had significantly higher baseline plasma
Fibroblast growth factor-23 (FGF-23)
FGF-23 than hyperthyroid cats that remained nona-
Fibroblast growth factor-23 (FGF-23), recently zotemic after treatment. However, after treatment for
reviewed in dogs and cats with CKD73, is a phospha- hyperthyroidism, plasma FGF-23 increased both in
turic hormone that is secreted by osteoblasts and cats that remained non-azotemic and those that
osteocytes into the blood in response to increased became azotemic, and overall baseline plasma FGF-23
serum phosphorus.73–75 Fibroblast growth factor-23 concentration was not a predictor for the develop-
results in increased urinary phosphorus excretion by ment of azotemia post treatment in hyperthyroid cats.81
inhibiting sodium- dependent phosphorus reabsorp- Other renal biomarkers that have been evaluated
tion in the proximal tubule, and it decreases intesti- in the serum and plasma of domestic species include
nal reabsorption of phosphorus via diminished cystatin C in dogs and cats82–90 and neutrophil gelati-
calcitriol.73,76,77 Blood concentration of FGF-23 has nase-associated lipocalin (NGAL)91–93 and homocys-
also been shown to increase as GFR decreases in teine94–96 in dogs as endogenous markers of GFR, and
both people and cats.78 While studies in human C-reactive protein97 and Big-Endothelin-194 as mark-
medicine have evaluated the pathogenesis of plasma ers of inflammation in dogs (Table 1).

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 33
34
Table 1. Summary of serum and/or plasma renal biomarkers in dogs and cats.

Location of Validation/ Values in Healthy Affected in AKI, CKD,


Renal Biomarker Production Type of Protein/Molecule Type of Biomarker Species Animals or Both Nonrenal Influences
Renal biomarkers review

226
Creatinine Muscle Cyclic derivative of creatine Endogenous indirect Dogs Dogs: 0.5–1.5 mg/dL Both Muscle mass; meat diet;
GFR marker Cats Cats: 0.8–1.8 mg/dL226 hydration status
(but depends on
breed/species/
instrument)
Cystatin C All nucleated cells LMW protein and Endogenous indirect Dogs96,227–229 Dogs: < 2.28 CKD: Dogs83,86–89,227–229, Obesity and weight
proteinase inhibitor GFR marker mg/L83,86–89,227–229 cats82,85,90 loss in dogs96
Cats82,84 Cats: < 1.95 (Presumably also AKI)
mg/L82,84,85,90
SDMA All nucleated cells Methylated amino acid Endogenous indirect Dogs8,44,45 Dogs and cats: CKD: Dogs8,51,
(arginine) GFR marker Cats43–45 < 14 µg/dL8,43,48–52 cats43,71,72
(Presumably also AKI)
FGF-23 Osteocytes and Phosphaturic hormone Marker of altered Cats80 Cats: 56–700 pg/mL80 CKD: Cats78–81 Dietary phosphorus;79
osteoblasts phosphorus hyperthyroidism81
metabolism
NGAL Neutrophils, kidney, LMW glycoprotein Endogenous indirect Dogs93 Dogs: < 21.2 ng/mL91–93 AKI: Dogs92,93 Inflammation230
bronchus, stomach, GFR marker CKD: Dogs91–93
small intestine,
pancreas, prostate
gland, thymus
C-reactive Hepatocytes Positive acute phase Inflammatory marker Dogs97 Dogs: 3.21 mg/L Renal diseases in dogs
protein protein (range: 2.09–8.60 (AKI vs CKD not
mg/L)97 specified)97
Homocysteine All nucleated cells Amino acid; Intermediate Endogenous indirect Dogs94–96 Dogs: 4.35  2.69 µmol/L94 AKI: Dogs95 Obesity and weight loss
product of methionine GFR marker CKD: Dogs94,95 in dogs96; icterus,
metabolism severe hemolysis and
lipemia95; cardiac
disease95
Big-Endothelin-1 Blood vessels, lung, Precursor to endothelin-1, a Inflammation Dogs94 Dogs: 6.51  1.86 CKD: Dogs94 Hypertension and
other tissues vasoconstrictor peptide pg/mL94 systemic inflammation94
including kidney
medulla

AKI indicates acute kidney injury; CKD, chronic kidney disease; FGF-23, fibroblast growth factor-23; GFR, glomerular filtration rate; LMW, low molecular weight; NGAL, neutrophil gelatinase-associated
lipocalin; SDMA, symmetric dimethylarginine.
Hokamp and Nabity

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

Renal Biomarkers: Urine row, and are involved in antibody-mediated


defense.98,102 The molecular weight of immunoglobu-
Renal protein handling lins G (IgG) and M (IgM) are 150 kDa98,102,103 and
900 kDa98,102, respectively. Serum immunoglobulin A
Several publications are available that review the mech-
(IgA) is present in monomeric, dimeric, and polymeric
anisms of protein handling by the kidney in health and
forms, with the monomeric form having a molecular
disease.98–100 In brief, proteins with a molecular weight
weight of 160 kDa.104 Thus, IgG, IgM, and IgA are
< 40 kilodaltons (kDa) (low-molecular weight [LMW]
HMW proteins that cannot pass through the glomeru-
proteins) are able to freely pass through the glomerular
lar filtration barrier in the healthy kidney. However,
filtration barrier, while intermediate-molecular weight
with glomerular damage, they may pass into the uri-
(IMW) proteins, those approximately the size of albu-
nary filtrate; thus, they are considered markers of
min, face increased charge and size restrictions, and
glomerular damage.98
high-molecular weight (HMW) proteins (> 100 kDa)
are generally completely restricted due to their large
size.98–100 Healthy tubules reabsorb proteins that are fil- Measurement and stability. Species-specific ELISAs are
tered into the tubular space via receptor-mediated the most common and preferred method for detec-
endocytosis.98,100 Glomerular damage increases the per- tion of urinary immunoglobulins105,106; however,
meability of the glomerular filtration barrier, typically thus far, detection of IgA in canine urine has only
resulting in marked proteinuria, while tubular damage been reported using Western blot.107–109 Canine-spe-
results in mild proteinuria due to decreased reabsorp- cific ELISA assays for IgG, IgM, and IgA are avail-
tion of proteins, leakage of proteins from the damaged able (Bethyl Laboratories, Montgomery, TX, USA
tubular epithelial cells, and upregulation of proteins and Immunology Consultants Laboratories [ICL],
involved in injury and repair.98,99 Portland, OR, USA), and IgG and IgM ELISA assays
When protein biomarkers are quantified in urine, have been validated showing acceptable mean intra-
their concentration is often indexed to urine creatinine assay and inter-assay variabilities105,106,110,111, spik-
(eg, urinary biomarker divided by urine creatinine) or ing recovery106,110, and dilutional linearity.106,110
urine specific gravity. Indexing urinary biomarkers to True stability testing of IgG in urine has not been
urine creatinine assumes that the excretion of urine evaluated; however, uIgG/c in canine urine samples
creatinine is constant between and within individuals, stored for 8 years at 80°C were similar to those
and that both urinary biomarkers and creatinine are stored for 2 years at 80°C.110
inversely proportional to urinary flow rate. When
these assumptions are met, an increased or decreased
Values in healthy animals. Mean urinary IgG/
biomarker/creatinine ratio will reflect increased or
creatinine (uIgG/c) is generally < 3 mg/g, with
decreased biomarker excretion.101 However, when an
maximum values < 10 mg/g observed in all but one
animal is not in steady state (ie, when renal function is
study (Table 2).105,110,112–116 Published studies cur-
rapidly changing), the assumption of constant urine
rently are not available describing the urine concen-
creatinine excretion may not be correct, as demon-
tration of IgM in healthy animals; however, in the
strated in a study of human patients with AKI and post
authors’ personal experience, urinary IgM concen-
kidney transplantation.101 Because of this, the practice
tration is low in clinically healthy dogs. IgA was
of indexing to urine creatinine in cases of AKI is ques-
undetectable in the urine of healthy dogs using
tionable. Therefore, some authors have reported con-
Western blot (Table 2).107–109
centrations of biomarkers without indexing to
creatinine. For purposes of this review, biomarkers
that are indexed to urine creatinine are denoted as Nonrenal influences. A few studies found that urinary
uBiomarker/c, and those that are unindexed concen- IgG concentration is not significantly altered by
trations in the urine are denoted as uBiomarker. hematuria/hemoglobinuria105,115,116 or pyuria/uri-
nary tract infection.105 However, in the authors’
experience in dogs with primarily proteinuric CKD,
Markers of glomerular damage/dysfunction
uIgG/c was significantly higher in dogs with hema-
turia, and fractional excretion of IgG (IgG_FE) was
Immunoglobulins
higher in dogs with pyuria/bacteriuria compared
Immunoglobulins are large glycoproteins made by with dogs with inactive urine sediment (J.A.H.,
plasma cells in the spleen, lymph nodes, and bone mar- M.B.N., unpublished data). Similarly, urinary IgM/

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 35
Renal biomarkers review Hokamp and Nabity

Table 2. Summary of urinary biomarkers of glomerular damage/dysfunction in small animals.

Renal Location of Type of Protein/ Validation/ Values in Healthy Affected in AKI, Nonrenal
Biomarker Production Biomarker Species Animals CKD, or Both Influences

Albumin Hepatocytes Negative acute Dogs123 Dogs: uAlb/c: AKI: Treatment with
phase protein < 296.5 mg/g114–116,124,146; Dogs113–115,125,126 hydrocortisone
Alb_FE: 0%124 CKD: (dogs)111
Cats: uAlb: Dogs,116–119,123,124
11.2  8.4 mg/dL127 cats127,134
C-reactive Hepatocytes Positive acute Dogs105,132 Dogs: uCRP/c: AKI:
protein phase protein 1.06 µg/g;132 below Dogs105,112–115,132
detection limit of CKD:
immunoassay105,112–115,123 Dogs105,132
IgA Plasma cells in Antibody; Not detectable in AKI: Dogs107–109
spleen, lymph HMW protein healthy dog urine
nodes, bone (Western blot)107–109
marrow
IgG Plasma cells in Antibody; Dogs105,110,116 Dogs: uIgG/c: AKI: Hematuria*; Pyuria/
spleen, lymph HMW protein < 10 mg/g105,110,112–115 Dogs105,107–109,112–115 bacteriuria*;
nodes, bone CKD: Treatment with
marrow Dogs106,110,116–119 hydrocortisone111
IgM Plasma cells in Antibody; Dogs106 CKD: Dogs106 Hematuria*; pyuria/
spleen, lymph HMW protein bacteriuria*
nodes, bone
marrow
TXB2 Glomerular Cycloxygenase lipid Dogs105 Dogs: uTXB2/c: AKI: dogs114,115
mesangial cells metabolite; marker < 4.7 µg/g105,114,115
and podocytes of altered intra-renal
hemodynamics
Transferrin Primarily liver; Iron transport protein Cats: uTf: 0.09  CKD: Dogs,117,118
other 0.42 mg/dL127 Cats127,134
tissues as well

AKI indicates acute kidney injury; Alb_FE, fractional excretion of albumin; CKD, chronic kidney disease; HMW, high molecular weight; IgA, immunoglobu-
lin A; IgG, immunoglobulin G; IgM, immunoglobulin M; TXB2, thromboxane B2; uALB, urinary albumin concentration; uALB/c, urinary albumin/urinary
creatinine; uCRP/c, urinary C-reactive protein/urinary creatinine; uIgG/c, urinary immunoglobulin G/urinary creatinine; uTf, urinary transferrin concentra-
tion; uTXB2/c, urinary thromboxane B2/urinary creatinine.
*Personal observations.

creatinine (uIgM/c) and fractional excretion of IgM despite significantly increased uIgG/c vs control dogs
(IgM_FE) were both significantly increased with at baseline and 24 hours later.113
hematuria and pyuria/bacteriuria in these dogs Pyometra can cause significant increases in
(J.A.H., M.B.N., unpublished data). uIgG/c and UPC, with a positive correlation between
UPC and uIgG/c.109,114,115 This increase is typically
Renal disease in veterinary medicine. Acute kidney injury transient, decreasing significantly after ovariohys-
(AKI): Urinary IgG is the main immunoglobulin evalu- terectomy, and in some cases, uIgG/c returns to val-
ated in diseases known to cause AKI, with fewer stud- ues that are not significantly different from healthy
ies evaluating urinary IgA. Dogs with AKI due to a dogs.114 A low proportion of bitches with pyometra
variety of causes, including Babesia rossi, leishmaniasis, also had detectable urinary IgA.109 These studies
and leptospirosis, have demonstrated increases in support that altered glomerular permselectivity can
uIgG/c105,112,113 or IgG and IgA on Western be present in dogs with pyometra, and indeed,
blot105,107,108,112,113, supporting glomerular damage. histopathology demonstrated glomerulosclerosis as
However, as expected, canine leptospirosis resulted the most common glomerular lesion in these dogs.
primarily in increased LMW proteins on sodium dode- However, tubular atrophy, interstitial inflammation,
cyl sulfate polyacrylamide gel electrophoresis, consis- and fibrosis were often present as well, and the role
tent with interstitial nephritis.108 Interestingly, in dogs of pyometra vs previously underlying renal disease
with snake envenomation, UPC was not increased in these older dogs is uncertain.115

36 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

Currently there are no studies evaluating urinary Additional urinary proteins indicating glomerular damage/
IgM in companion animals with AKI. dysfunction

106,110,117,118 Albumin. The use of urinary albumin (Alb) in dogs as a


Chronic kidney disease (CKD): Urinary IgG
106 renal biomarker has recently been reviewed.2 Albumin
and IgM have been shown to increase in dogs
is a negative acute phase, IMW (approximately
with CKD. In dogs with CKD due to X-linked hered-
65 kDa) serum protein synthesized primarily by hepa-
itary nephropathy (XLHN), uIgG/c increased in early
tocytes. It is considered primarily to be a marker of
stages of renal disease while uIgG/c remained low in
glomerular damage, but uAlb can also be present with
healthy age-matched littermates.110,118 Urinary IgG/c
tubular or vascular damage.98,120–122 Measurement of
often increased before UPC and continued to increase
uAlb has been validated in dogs123, and uAlb/c is ele-
in mid to late stages of disease progression.110 Further-
vated in both AKI and CKD117,118,123,124, including
more, uIgG/c was moderately to highly positively cor-
renal damage due to nephrotoxic drugs (methyl can-
related with most glomerular and tubulointerstitial
tharadimide tablets and gentamicin)125,126, snake
(TI) lesions based on histopathology.110 Urinary bio-
envenomation113, pyometra114,115, and hypercorti-
marker concentrations and their fractional excretions
solism.111,116,119 Pyometra caused transient albumin-
were measured in dogs with naturally occurring CKD
uria that significantly decreased or returned to values
to correlate biomarkers with types of renal damage
not significantly different from healthy dogs after ovar-
(glomerular vs TI) and their association with sur-
iohysterectomy.114,115 Cats with stage I CKD had sig-
vival.106 Immunoglobulin G (uIgG/c and IgG_FE) and
nificantly higher concentrations of uAlb than normal
IgM (uIgM/c and IgM_FE) demonstrated moderate,
cats, and uAlb concentration had higher sensitivity
positive correlations with glomerular damage based on
and specificity for detection of CKD compared with
light and electron microscopy (r = .44–.58 and
plasma creatinine.127
r = .41–.58), respectively), which were similar to that
observed for UPC (r = .45–.57). Immunoglobulin
M_FE also correlated moderately well with TI damage C-reactive protein. C-reactive protein (CRP) is a positive
(r = .49). Markedly increased uIgM/c and uIgG/c were acute phase protein in dogs, made primarily by hepato-
associated with immune complex-mediated glomeru- cytes.128,129 The MW of CRP ranges from 110 to
lonephritis (ICGN), while lower uIgM/c was observed 144 kDa130,131, therefore, its presence in the urine
in juvenile nephropathies, nonimmune complex- supports glomerular damage. In dogs with various
mediated glomerulonephropathies, and primary tubu- causes of both AKI and CKD, including renal damage
lar disease. Both IgM_FE and IgG_FE were signifi- due to leishmaniasis132, babesiosis112, snake enveno-
cantly associated with faster time to death due to renal mation (24 hours post envenomation)113, and pyome-
disease in these dogs.106 tra114,115, uCRP/c increases.105 Dogs with pyometra
Urinary IgG has been evaluated in several stud- treated by ovariohysterectomy had uCRP/c values that
ies of dogs with increased cortisol due to either decreased to values not significantly different from
exogenous or endogenous sources, since excess cor- healthy dogs.114,115 In contrast, a significant difference
tisol causes proteinuria, likely by altering the was not found in uCRP/c between healthy dogs and
glomerular filtration barrier. In aged Beagles treated those with CKD in one study, although uCRP was
over 24 weeks with hydrocortisone, uIgG/c and UPC undetectable in the healthy dogs while increased in 3
progressively increased, while tapering and cessation of 10 dogs with CKD.123 Age does not seem to have an
of hydrocortisone treatment resulted in decreased effect on uCRP/c in healthy dogs.123
uIgG/c and UPC.111 In dogs with hyperadrenocorti- Thus far, only one study evaluated the concentra-
cism, uIgG/c was significantly higher than in clini- tion of serum CRP in dogs with naturally occurring
cally healthy controls, supporting glomerular renal disease. In this study, serum CRP was signifi-
dysfunction.116 Finally, in dogs treated with trilos- cantly increased in dogs with reduced GFR, and the
tane or hypophysectomy for ACTH-dependent authors suggested that stimulation of the inflamma-
hyperadrenocorticism, uIgG/c decreased up to 15- tory acute phase response may be implicated in the
fold posttreatment. However, uIgG/c did not com- pathogenesis of renal disease in dogs.97
pletely return to levels comparable to healthy dogs Other urinary protein markers of glomerular dam-
in all cases, consistent with persistence of protein- age not listed in Table 2 that have been shown to
uria in 38% of dogs 12 months posttreatment.119 increase in CKD include transthyretin117, adiponec-

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 37
Renal biomarkers review Hokamp and Nabity

tin133, and ferritin89 in dogs, and apolipoprotein-H in 80°C.110,146 All but one study146 normalized urinary
cats.134 RBP concentration to urinary creatinine (uRBP/c
[mg/g]).105,106,110–116,119,123,124,145,147–149
Markers of tubular damage/dysfunction
Values in healthy animals. In healthy dogs and cats,
The proteins discussed in the next section are abnor-
RBP is generally undetectable or minimally detect-
mally present in the urine due to decreased tubular
able in the urine by Western blot.145,150,151 Using
reabsorption (retinol-binding protein (RBP), neu-
immunoassays to quantify urinary RBP in healthy
trophil gelatinase-associated lipocalin (NGAL), cys-
dogs, the highest reported uRBP/c was 0.9 mg/g116,
tatin C), upregulation of proteins involved in injury
with most studies reporting means and medians
and repair (NGAL), and decreased production by
< 0.15 mg/g regardless of assay used
damaged tubules (Tamm–Horsfall protein [THP]).
(Table 3).105,110–116,119,123,124,147 In healthy cats, uri-
Proteins present due to release from damaged tubular
nary RBP was below the limit of assay detec-
epithelial cells are discussed in the urinary enzyme
tion145,149, and it was not detectable in healthy
section. Additional examples of urinary biomarkers
sheep by 2-dimensional gel electrophoresis and mass
present due to these 4 mechanisms are presented in
spectrometry.152
Table 3.
Retinol-binding protein (RBP)
Nonrenal influences. In dogs, age does not appear to
Retinol-binding protein is a 21-kDa lipocalin that acts have a major influence on uRBP/c, as no significant
as the transport protein for retinol in plasma.135 Reti- differences were found for uRBP/c between healthy
nol-binding protein is primarily produced in the liver young and older dogs.110,123 However, a mild but sta-
but also in the kidney, lungs, spleen, brain, stomach, tistically significant negative correlation with age was
heart, and skeletal muscle.136–138 Retinol-binding pro- found in young adolescent dogs, likely due to low crea-
tein circulates in a complex with transthyretin (TTR), tinine excretion in very young dogs.110 Pyuria, bacteri-
which has a molecular weight of 54 kDa.135,139,140 uria or positive urine culture, and at least mild to
Retinol-binding protein by itself is a LMW protein that moderate hematuria and hemoglobinuria do not seem
can freely pass through the glomerular filtration bar- to significantly affect uRBP146 or uRBP/c105,112; how-
rier; however, the TTR-RBP complex is too large to pass ever, in one study, a mild increase in uRBP was seen in
through the glomerular filtration barrier in the healthy markedly hematuric samples.146 In the authors’ expe-
kidney.139 Retinol-binding protein in the renal filtrate rience, fractional excretion of RBP (RBP_FE) was sig-
is reabsorbed by tubular epithelial cells.141 Tubular nificantly higher in pyuric/bacteriuric samples vs
damage and/or competition for reabsorption by the inactive sediments from dogs with proteinuric CKD
presence of abnormally large amounts of protein (ie, (J.A.H., M.B.N., unpublished data). To the authors’
with glomerular damage) results in decreased reab- knowledge, nonrenal influences on urinary RBP have
sorption of RBP with subsequent loss of RBP into the not been published in cats.
urine.142,143 Glomerular damage could also contribute
to urinary RBP due to loss of the TTR-RBP complex.
Renal disease in veterinary medicine. AKI: Naturally
occurring AKI (eg, pyometra, babesiosis due to Babesia
Measurement and stability. Human RBP immunoassays rossi, and envenomation by cytotoxic and neurotoxic
have been validated for dogs and cats, with adequate snakes) transiently increases uRBP and uRBP/c in
intra-assay and inter-assay variabilities (canine urine dogs, presumptively indicating tubular dysfunc-
and plasma105,110,144 and feline urine145), spiking tion.105,113,115,151 Histologic confirmation of tubular
recovery (canine urine)110, and dilutional linearity damage in dogs with pyometra found that dogs with
along a specific range of the standard curve (canine severe TI lesions on histopathology had higher uRBP/c
urine).105,110 A canine-specific RBP ELISA was (in the 75th percentile) compared with dogs demon-
recently marketed (ICL); however, validation and use strating milder lesions.115 Typically, uRBP/c decreased
of this kit has not yet been published. Retinol-binding significantly after ovariohysterectomy in these dogs,
protein concentration is similar in cystocentesis vs often to values comparable to healthy dogs.105,115
voided urine samples from clinically healthy dogs.146 Urinary RBP has also been detected with 2-dimen-
Retinol-binding protein appears to be relatively stable sional gel electrophoresis in the urine of sheep with
in canine urine samples when frozen, ideally at AKI due to ketoprofen overdose.152

38 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Table 3. Summary of urinary protein and enzyme biomarkers of tubular damage/dysfunction in small and large animals.

Renal Location of Type of Mechanism Validation / Values in


Biomarker Production Protein/ for Altered species Healthy Animals Affected in AKI, CKD, or Both Nonrenal Influences
Hokamp and Nabity

Biomarker Excretion

Urinary proteins
Clusterin Renal tubules Glycoprotein Increased Dogs231 Dogs: uClu/c: 0.27 AKI: Dogs125,231
production (unitless) 231;
uClu: 38.5 ng/mL231
Cystatin C All nucleated cells LMW protein Decreased Dogs232 Cats: below limit AKI: Dogs126 Diabetes in cats233
and proteinase reabsorption Cats26 of quantification CKD: Dogs89,232, cats26,233
inhibitor of assay233
KIM-1 Renal tubules Glycoprotein Increased Undetectable Both, but mostly
production in healthy cats234 with acute processes
AKI: Dogs125, cats234
a lamb176
NGAL Neutrophils, LMW Decreased Dogs93,106,110,164 Dogs: uNGAL/c: AKI: Inflammation/urinary
kidney, bronchus, glycoprotein reabsorption < 6 µg/g;110,164,165 Dogs92,93,125,126,161,165–170 tract infection/pyuria
stomach, small and increased uNGAL: < 21.2 CKD: Dogs91–93,106,110,161,165 (dogs);161,164,165
intestine, pancreas, production ng/mL91–93,161,164 uNGAL/c decreased
prostate gland, thymus with body mass

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
(dogs);164 Age
(dogs)110,164
RBP Primarily liver; Vitamin A Decreased Dogs105,110,144 Dogs: uRBP/c: AKI: Dogs105,113,115,151, Marked hematuria;146
also other organs carrier reabsorption Cats145 < 0.15 mg/ sheep152; Pyuria/bacteriuria
(kidney, lungs, spleen, protein g105,110–116,119,123,124,147; CKD: Dogs105,106,110,116–119,124,150,151, (RBP_FE)*; Negative
brain, stomach, Cats: Undetectable145 cats134,145,148,149 correlation with age in
heart, skeletal muscle) Sheep: Undetectable152 young adolescent
dogs110;Treatment with
hydrocortisone (dogs)111
THP Epithelial cells of Glycoprotein Decreased Cats235 Dogs: uTHP/c: CKD: Dogs117,144,151,174,175, Urolithiasis (cats)235
thick ascending production 10–65 mg/g174 cats134
limb of loop of Cats: uTHP: 49.2 
Henle and distal 35.5 µg/mL235
convoluted tubule
Urinary enzymes
AAP Renal tubular Enzyme Released Dogs191 Dogs: uAAP/c: AKI: Dogs125
brush border from brush 0.7–9.0 U/g191
enzyme border

(continued)
Renal biomarkers review

39
Table 3 (continued)

40
ALP Renal tubular Enzyme Released Dogs: uALP/c: 0.25  AKI: Dogs147,213,215,
brush border from brush 1.17 U/g236;uALP: 5.8 sheep206
enzyme, highest border (0.4–12) U/L/mmol215
in renal proximal Horses: uALP/c:
convoluted tubule; 6.7  3.9 IU/g237;
Also in tubular
Renal biomarkers review

uALP: 10.2  4.0


lysosomes IU/L237; < 3.55
U/mmol225,238
Cauxin Proximal straight Carboxylesterase Uncertain Cats224 CKD: Cats134,223,224
tubular cells in cats protein whether
increased or
decreased with
tubular damage
GGT Renal tubular Enzyme Released from Horses193 Dogs: uGGT/c: < 42 U/ AKI: Dogs147,198,203,213,215,216,
brush border brush border g;192,195,208,210,216,236 horses205,207,
enzyme, highest 3.4 (0.6–6.0) U/L/mmol215 sheep206
in renal proximal uGGT: 6-112 U/L216
straight tubule Cats: uGGT/c: < 36.4 U/g199
Horses:uGGT/c: < 36.54 U/
g193,204,237,238; 0.33  0.8 U/
L/mmol200uGGT: 3.3  3.0 IU/
L;237 < 1.85 IU/mmol193,225,238
Calves: uGGT: 3.2 
2.6 U/µmol217
NAG Lysosomal enzyme Enzyme Cellular leakage Dogs110,123,146,191 Dogs and cats: AKI: Alkaline urine (reduced
in proximal renal (although and cats190 uNAG/c: Dogs114,115,125,126,167,168,194,197,198, activity);199 Hematuria and
120,191,195,199
tubules and associated with < 11 U/g cats185, sheep206 hemoglobinuria (inaccurate
other tissues; glomerular CKD: Dogs106,110,116,123, measurement due to similar
2 renal forms: damage, cats185 color of hemoglobin to m-
NAG-A and NAG-B; possibly due to crescol purple released by
circulates in lysosomal some substrates) 199;
the serum turnover Gender (higher in male
and/or leakage dogs)191,195
through
glomerular
filtration barrier)

AAP, alanine aminopeptidase; AKI, acute kidney injury; ALP, alkaline phosphatase; CKD, chronic kidney disease; GGT, gamma glutamyl-transpeptidase; KIM-1, kidney injury molecule-1; LMW, low molecular
weight; NAG, N-acetyl-b-D-glucosaminidase; NGAL, neutrophil gelatinase-associated lipocalin; RBP, retinol-binding protein; RBP_FE, fractional excretion of retinol-binding protein; THP, Tamm–Horsfall
protein; uAAP/c, urinary alanine aminopeptidase/urinary creatinine; uALP, urinary alkaline phosphatase concentration; uALP/c, urinary alkaline phosphatase/urinary creatinine; uClu, urinary clusterin
concentration; uClu/c, urinary clusterin/urinary creatinine; uGGT, urinary gamma glutamyl-transpeptidase; uGGT/c, urinary gamma glutamyl-transpeptidase/urinary creatinine; uNAG/c, urinary N-acetyl-b-
D-glucosaminidase/urinary creatinine; uNGAL, urinary neutrophil gelatinase-associated lipocalin concentration; uNGAL/c, urinary neutrophil gelatinase-associated lipocalin/urinary creatinine; uRBP/c,
urinary retinol-binding protein/urinary creatinine; uTHP, urinary Tamm–Horsfall protein concentration; uTHP/c, urinary Tamm–Horsfall protein/urinary creatinine
Hokamp and Nabity

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
*Personal observations
Hokamp and Nabity Renal biomarkers review

CKD: More studies have evaluated uRBP for detection that posttreatment median uRBP/c values were within
of tubular dysfunction in dogs and cats with CKD than the range reported for healthy dogs. Urinary RBP/c
with AKI, and dogs with CKD have significantly also decreased after treatment with trilostane; how-
increased uRBP151, uRBP/c105,110,117,118,123,124,144,150, ever, the decrease was not significant, and several dogs
and RBP_FE124,144 compared with healthy dogs. How- had persistent proteinuria.119 These cumulative results
ever, whether this increase is primarily present due to suggest that ACTH-dependent hyperadrenocorticism
tubular damage as opposed to presence of proteinuria results in decreased tubular protein reabsorption; how-
is controversial, as discussed below. ever, the degree of reversibility and posttreatment val-
In dogs with CKD due to XLHN, uRBP/c was ues for uRBP/c depend on the type of treatment
increased prior to the onset of azotemia but after (hypophysectomy vs trilostane) and persistence of pro-
onset of proteinuria, and urinary RBP increased with teinuria.119 Finally, in aged dogs treated with hydro-
disease progression throughout all disease stages cortisone, uRBP/c increased with treatment and
(based on sCr), with the most pronounced increase decreased posttreatment.111
in mid to late stages of renal disease.110,118,150 Fur- In cats, urinary RBP evaluation has focused on
thermore, uRBP/c had the strongest correlation with those with CKD, hyperthyroidism, or both. These stud-
both glomerular and TI lesions compared with other ies have shown that uRBP/c is significantly increased
biomarkers of renal function, and it correlated in cats with either CKD134,145 or untreated hyperthy-
most strongly with conventional measures of disease roidism145,149 compared with healthy cats. These find-
severity (sCr, GFR, and interstitial fibrosis).110 ings suggest that hyperthyroidism causes tubular
Despite this, uRBP/c was not a significant indepen- dysfunction in cats, and this tubular dysfunction may
dent predictor of GFR based on multivariate analysis. be at least partly reversible, as uRBP/c often decreased
It was concluded that uRBP/c might be useful for significantly (although was still detectable, unlike in
detecting early tubular damage before an obvious healthy cats) in cats treated with radioiodine.148,149
increase in sCr.110 However, because hyperthyroidism can mask CKD in
When biomarkers were correlated with histologi- cats, and CKD may be revealed after treatment for
cally proven renal damage in dogs with naturally hyperthyroidism, increased uRBP/c in hyperthyroid
occurring CKD due to a variety of causes, uRBP/c and cats could reflect CKD in some animals. Supporting
RBP_FE were moderately correlated with glomerular this theory, uRBP/c decreased significantly in cats that
and TI damage, with RBP_FE having the second stron- did not develop azotemia post treatment for hyperthy-
gest correlation with TI damage following sCr.106 roidism, but it remained elevated in cats that did
RBP_FE also increased significantly with each increase develop azotemia posttreatment. Thus, in hyperthy-
in IRIS stage, while uRBP/c only increased signifi- roid cats, it was suggested that uRBP/c might be a mar-
cantly in IRIS stages 3 and 4. Both uRBP/c and RBP_FE ker of reversible tubular dysfunction in healthy
were significantly associated with time to death due to kidneys, but also a marker of irreversible damage in
renal disease when evaluated individually; however, cats with preexisting CKD.148
in a multivariate analysis with other biomarkers, nei-
ther uRBP/c nor RBP_FE was significantly associated
Neutrophil gelatinase-associated lipocalin (NGAL)
with survival.106
Despite the promise of uRBP for early detection Neutrophil gelatinase-associated lipocalin is a mem-
of CKD and monitoring of progression, another study ber of the family of lipocalin-binding proteins origi-
concluded that proteinuria influenced uRBP/c more nally isolated from the specific granules of human
than decreased renal function based on sCr and neutrophils153,154, but it is also present in many nor-
plasma creatinine clearance. This conclusion was mal tissues including the kidney.155 Neutrophil gela-
based on finding uRBP/c to be significantly greater in tinase-associated lipocalin is upregulated in epithelial
dogs with proteinuria and borderline proteinuria cells in neoplastic155,156 and inflammatory156 pro-
compared with azotemic, nonproteinuric dogs, and cesses. The original proposed function of NGAL was
the inability of uRBP/c to detect reduced GFR.124 as a bacteriostatic agent that binds bacterial sidero-
Dogs with hyperadrenocorticism had higher phores to prevent iron acquisition.157 Neutrophil
uRBP/c compared with control dogs; however, UPC gelatinase-associated lipocalin has since been found
was also significantly higher in these dogs compared to to be involved in many cellular mechanisms includ-
controls.116 Additionally, in dogs with ACTH-depen- ing renoprotection158, and NGAL’s many functions
dent hyperadrenocorticism, uRBP/c decreased signifi- have been reviewed.159 Neutrophil gelatinase-asso-
cantly after treatment with hypophysectomy119, such ciated lipocalin is a LMW protein that freely passes

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 41
Renal biomarkers review Hokamp and Nabity

through the glomerular filtration barrier and is reab- eral WBC count does not correlate with overall
sorbed almost completely by the proximal tubules in serum NGAL (sNGAL)92; however, dogs with mono-
the healthy kidney.158 With renal damage and pro- meric uNGAL had significantly higher peripheral
tein overload, reabsorption of NGAL in the proximal WBC and neutrophil counts than dogs without the
tubules is impaired. In addition, there is increased uNGAL monomer.161
synthesis of NGAL by damaged tubular epithelial Pyuria and urinary tract infections (UTI) can
cells.160 Three different molecular weight forms of markedly influence urinary NGAL concentrations.
NGAL are present in canine urine161, including a Several studies showed that both uNGAL and uNGAL/
25 kDa monomeric protein that appears to originate c were significantly higher in dogs with pyuria, UTI,
from renal tubular epithelial cells and is associated and other lower urinary tract diseases (such as transi-
with renal damage, a 45–50 kDa dimeric protein that tional cell carcinoma and calcium oxalate urolithiasis),
is the predominant form released by neutrophils and both with and without azotemia, compared with
appears most often with pyuria, and an NGAL/matrix healthy controls.161,164,165 In the authors’ experience
metalloproteinase (MMP) 9 heterodimer complex in dogs with proteinuric CKD, uNGAL/c was higher in
that occurs with both renal injury and pyuria and dogs with active vs inactive sediments (J.A.H., M.B.N.,
hematuria.153,161,162 Although ELISAs that distin- unpublished data). Although dogs with lower urinary
guish between different molecular forms of human tract diseases had increased uNGAL/c compared with
NGAL are available, canine NGAL ELISAs are cur- control dogs, values were still lower than dogs with
rently unable to make this discrimination.163 renal disease.161,165 A uNGAL/c cutoff of > 2.57 lg/g
and a uNGAL cutoff of > 3.38 ng/ml had sensitivities
and specificities in the 70–80% range for dogs with vs
Measurement and stability. In domestic species, NGAL
without a UTI.164 Furthermore, the 50 kDa dimer form
has been evaluated in urine, serum, and plasma in
of NGAL was shown to be more common in urine from
dogs, and it has been partially validated using canine-
dogs with pyuria, while the NGAL/MMP-9 complex
specific ELISAs with acceptable intra-assay and
was found in dogs with pyuria and hematuria.161
inter-assay variabilities, dilutional linearity, and spik-
Other nonrenal diseases (gastritis, protein losing
ing recovery in canine urine and plasma.93,106,110,164
enteropathy, hepatic disease, enteritis, portal shunt,
Neutrophil gelatinase-associated lipocalin has also
bone fracture, intervertebral disk disease) do not
been validated in canine serum, but results were not
appear to affect uNGAL.161
published.91 Neutrophil gelatinase-associated lipocalin
appears to be relatively stable in canine urine, as there
were minimal effects on uNGAL after 4 freeze–thaw Renal disease in veterinary medicine. AKI: While studies
cycles, and no significant differences in uNGAL/c were of drug-induced AKI consistently demonstrate
observed in samples collected up to 8 years apart, increases in urinary NGAL, mixed results are reported
stored at 80°C.110 No difference in uNGAL/c was with regard to timing of this increase, which could at
apparent between cystocentesis and voided samples165 least partly be due to differing protocols, particularly
or between 2-hour spot urine samples and 15-hour with varying concentrations of gentamicin used. Two
collection samples.166 The 3 different molecular weight separate studies in dogs given gentamicin found that
forms of NGAL can be differentiated in canine urine uNGAL/c increased early (as early as day 1 post admin-
samples using Western blot.161 Values in healthy dogs istration) compared with other markers of nephrotoxi-
are reported in Table 3. city (such as sCr and UN) and was correlated with the
severity of tubular damage, including tubular cell
Nonrenal influences. In one study, healthy dogs necrosis, degeneration and regeneration, tubular cell
< 4 months of age often had much higher uNGAL/c hyaline droplet formation, and hyaline casts.166,167 It
than dogs > 4 months of age, but it was suggested was concluded that uNGAL/c was a sensitive and pre-
that this was likely due to preputial neutrophil con- dictive marker of gentamicin-induced nephrotoxic-
tamination from voided urine samples from the ity.166 However, a third study found that although
youngest dogs.110 In another study, there was no uNGAL/c was significantly correlated with GFR and
correlation of uNGAL/c with age in adult dogs, increased at the first time point evaluated (4 days after
although uNGAL (not normalized) was weakly but gentamicin administration), the increase was not sta-
positively correlated with age. Furthermore, this tistically significant until the second time point, at
same study found that both uNGAL and uNGAL/c 8 days post administration. Thus, in this study,
decreased with body mass.164 Interestingly, periph- uNGAL/c was not superior to more traditional markers

42 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

of renal function.126 In AKI induced by administration repeatable in a second study of dogs with CKD that
of intravenous polymyxin B for 7 days, a dose-depen- were predominantly proteinuric but included both
dent increase in uNGAL/c was observed, with signifi- azotemic and nonazotemic dogs.106
cantly increased uNGAL/c in the mid- and high-dose
Additional urinary proteins indicating tubular damage/dys-
groups on day 2.168 Finally, Beagle dogs administered
function
methyl cantharidimide tablets over 30 days demon-
strated significant increases in uNGAL in the high-dose Although typically much more limited in their evalu-
group, while middle- and low-dose groups did not ation, numerous other urinary proteins have been
have significant changes in uNGAL.125 studied as tubular biomarkers in domestic animals.
Repeatedly, studies have shown promise for NGAL Several of these are presented briefly below, while
as a marker of naturally occurring AKI in dogs. Urinary others with minimal references are listed in Table 3.
NGAL, uNGAL/c, and plasma NGAL (pNGAL) were all
significantly greater in dogs with azotemia from natu- Tamm–Horsfall Protein. Tamm–Horsfall protein (THP,
ral causes (whether due to AKI or CKD) vs healthy otherwise called uromodulin) is a 100-kDa pro-
control dogs92,93,161,165, and extremely high pNGAL tein171 present in the thick ascending limb of the
differentiated AKI from CKD in one study93 while loop of Henle and the distal convoluted
extremely elevated uNGAL/c was seen in dogs with tubule.171,172 Tamm–Horsfall protein is one of the
AKI compared to both CKD and lower urinary tract major urinary proteins present in healthy dogs.171
diseases in another study.165 Furthermore, uNGAL/c The biologic function of the protein is still not fully
was increased in nonazotemic (IRIS Grade I) AKI, indi- understood, but it is believed to have roles in water
cating the presence of kidney injury before sCr and electrolyte balance in the thick ascending limb
increased outside of the reference interval.165 Simi- of Henle’s loop, defense against UTI, prevention
larly, in dogs that developed AKI post operatively, against the formation of kidney stones, and in
median uNGAL was significantly greater 12 hours post innate immunity of the kidney.173 Normal urine
surgery and remained elevated for at least 3 days, has high concentrations of THP, and significantly
while sCr did not show a significant increase until reduced urinary THP concentrations and uTHP/c are
24 hours post surgery.169 Urinary NGAL and uNGAL/c seen in dogs and cats with renal disease, including
were also shown to be significantly increased (average CKD.117,134,144,151,174,175 Furthermore, uTHP/c corre-
of 24-fold and 41-fold, respectively) from baseline lates negatively with plasma creatinine concentra-
within 2 hours of reperfusion using a hemorrhage and tion and UPC.174 Therefore, reduced urinary THP
colloid fluid resuscitation model of renal injury in might be a marker of distal tubular damage in dogs
Greyhounds.170 Thus far, uNGAL/c, uNGAL, and and cats.
sNGAL have not been shown to be predictors of sur-
vival in dogs with AKI.92,165
Cystatin C. Cystatin C is a LMW protein (13 kDa), and
its presence in urine is a marker of proximal tubular
CKD: Dogs with CKD have demonstrated increased damage. A recent review includes studies of urinary
uNGAL/c, uNGAL, sNGAL, and pNGAL compared with cystatin C26; therefore, only key information is pro-
healthy dogs and dogs with UTI or other lower urinary vided in Table 3.
tract diseases.91–93,110,161,165 Similar to findings in AKI Other urinary protein markers of tubular dam-
studies, increases in uNGAL/c occurred early in the age and dysfunction not listed in Table 3 that have
development of CKD in dogs with XLHN.110 Serum been shown to increase in AKI include calbindin-
NGAL and fractional excretion of NGAL (NGAL_FE) D28K152, CD1d152, and heat shock protein 1B176 in
also increased with increasing IRIS stages in dogs with sheep, and interleukins 2, 7, and 8, monocyte
proteinuric CKD.106 Furthermore, uNGAL/c and chemoattractant protein-1, granulocyte macrophage
NGAL_FE correlated moderately to strongly with both colony-stimulating factor, and keratinocyte-derived
glomerular and TI lesions in dogs with CKD.106,110 chemokine in dogs.177 Urinary proteins that were
Finally, higher sNGAL and NGAL_FE are associated increased in dogs with CKD include vitamin
with shorter survival compared with lower values in D-binding protein117,118, a1-microglobulin118,
dogs with CKD.92,106 Serum NGAL concentration was b2-microglobulin 110,118
, apolipoprotein A1118,
118 118
actually concluded in one study to be a better predictor megalin , and cubilin. In cats with CKD, uri-
of clinical outcomes than sCr for dogs with CKD that nary cystatin M134, transforming growth factor-
were already azotemic;92 however, this was not b178,179 and interleukin-8179 were increased while

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 43
Renal biomarkers review Hokamp and Nabity

vascular endothelial growth factor was at 4°C for at least 4 days in dogs192, in equine urine,
decreased.179 uGGT was not stable after storage at 20°C, 4°C, and
25°C by 72 hours, with the greatest decrease in
Urinary enzymes enzyme activity occurring after freezing ( 20°C).193
Renal tubular epithelial cells contain enzymes which
have been explored as biomarkers of tubular damage Values in healthy animals. Urinary NAG, GGT, and ALP
(Table 1). The most commonly studied enzymes in activities are generally present at low levels in healthy
domestic animals include N-acetyl-b-D-glucosamini- domestic animals (Table 3). In the urine of healthy
dase (NAG), a lysosomal enzyme180, and gamma-glu- cats, the NAG-A isoenzyme activity was 2–3 times
tamyl transpeptidase (GGT) and alkaline phosphatase higher than NAG-B.185
(ALP), which are brush border enzymes.181 Cauxin
has also been studied in cats, and a brief discussion
Nonrenal Influences. Age does not appear to affect
of this marker will be included at the end of the
uNAG/c in dogs.146,194 However, 3 of 4 studies found
enzyme section. Two isoenzymes of NAG occur in
uNAG/c was significantly higher in males than
the kidney: NAG-A and NAG-B. In people, only
females.191,195–197 Additionally, uNAG/c decreased after
NAG-A can be detected in urine from patients with-
castration of male dogs; therefore, the increase in NAG
out renal disease, whereas renal damage and disease
activity in males may be due to the enzyme content of
results primarily in increased excretion of NAG-
sperm.191,196 In cats, no significant difference in uNAG/
B.182–184 Of note, both NAG-A and B are detectable
c or NAG isoenzymes was found between sexes.185,190
in healthy cats.185 Although present in the serum
Spot measurements of uGGT/c and uNAG/c were
and other tissues and cells, urinary NAG, GGT, and
significantly correlated with 24-hours urine GGT and
ALP originate from the renal tubules in the absence
uNAG excretion, respectively.192,198 However, large
of glomerular damage.186–189
intra- and inter-individual variability was noted for
uNAG/c.191 In cats, there was no apparent circadian
Measurement and stability. Activity of urinary enzymes variation in NAG or GGT excretion, and while short
is expressed as units per liter, and an activity index is (4 hours) urine collection periods estimated 24-hour
typically calculated by normalizing to urinary crea- uNAG/c and uGGT/c, there was greater variability in
tinine concentration. Validation of assays for urinary enzyme indices over the shorter collection time per-
NAG activity (uNAG) has been performed in dogs and iod.199
cats; however, to the authors’ knowledge, validation In both dogs and cats, hematuria, pyuria, and
of assays for urinary GGT (uGGT) and ALP (uALP) bacteriuria/UTI without concomitant pyelonephritis
activity has not been performed in domestic ani- does not appear to affect uNAG or uNAG/c.146,185,197
mals.110,190,191 In canine urine, overall intra-assay and However, dogs with lower UTI accompanied by
inter-assay variabilities, dilutional linearity, and spik- pyelonephritis had markedly increased uNAG/c val-
ing recovery for NAG were acceptable.110,123,191 How- ues, likely indicating the presence of tubular dam-
ever, canine and feline urine samples with lower age.197 Further studies are needed to determine if
concentrations of NAG tended to show higher coeffi- uNAG/c can accurately detect pyelonephritis. While
cients of variation.110,190 NAG activity is relatively changes in urine pH did not seem to affect uNAG/c
stable in urine at 80°C for at least one year; however in dogs195, in cats, NAG activity was decreased with
enzyme degradation, which is not prevented by addi- alkaline urine pH199 or demonstrated a weak nega-
tion of a protease inhibitor, is still possible.110,146,190,191 tive association with urine pH.190 Furthermore,
Studies assessing stability at room temperature, 4°C, induction of muscular inflammation with injection of
and 20°C have shown contrasting findings, although Freund’s complete adjuvant resulted in decreased
NAG appears to be stable for at least one month at uGGT/c in horses, and it was suggested that the pres-
20°C in both feline and canine urine.110,146,190,191 Up ence of inflammation may affect the reliability of
to 4–5 freeze–thaw cycles have been shown to signifi- uGGT/c as an index of renal damage.200 One study
cantly affect urinary NAG activity in dogs and reported that uGGT/c was increased after the admin-
cats.110,190 Thus, it is recommended to keep urine sam- istration of a nonnephrotoxic agent in dogs and con-
ples frozen at -80°C and to limit the number of freeze– cluded that this reflected false positive results.201
thaw cycles. No significant difference in uNAG activity However, mild tubular damage might still have been
was seen between voided and cystocentesis samples missed on ultrastructural examination given the ten-
from dogs.146 While uGGT activity was relatively stable dency for such lesions to be focally distributed.

44 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

Renal disease in veterinary medicine. AKI: Urinary strated increased uNAG/c, uGGT, uGGT/c, uALP, and
enzymes, particularly NAG and GGT, have been most uALP/c that, in several studies, decreased back into the
extensively studied as early markers of AKI by chemi- range of healthy dogs after ovariohysterectomy, sup-
cal (drug) induction in domestic species. Gentamicin, porting transient AKI in these dogs.114,115,194,197,213,214
especially high doses, significantly increased uNAG/c Furthermore, markedly increased uNAG/c was associ-
and uGGT/c in dogs, and uNAG/c increased as early as ated with severe TI lesions and reduced GFR in dogs
24 hours post administration, and continued to with pyometra.115,194 In client-owned dogs with renal
increase throughout the study period.126,167,198,202 disease, uALP/c was a significant indicator of AKI com-
Even therapeutic doses of gentamicin in dogs resulted pared to dogs with CKD or healthy dogs (all confirmed
in a 2-fold increase in uGGT/c, although without statis- histologically), while uGGT/c was not shown to be as
tically significant changes in sCr, UPC, or USG.203 Sim- useful for distinction.215 Significant correlations were
ilarly, horses treated with a therapeutic dose of not detected between urinary enzyme levels and
gentamicin had significantly increased uGGT/c with- extent of morphological kidney damage.215 Transient
out increased sCr during the treatment period.204 increases in uGGT and/or uGGT/c were seen with
Another study found that uNAG/c correlated with the anesthesia and routine surgery in healthy dogs216 and
severity of renal lesions and had high accuracy and diarrheic calves217, and uGGT/c and uALP/c were
sensitivity for detection of renal injury compared with increased in dogs with European Adder envenoma-
UN.167 These and other studies support that urinary tion.147
enzymes are more sensitive and reliable for detecting The studies in both drug-induced and naturally
acute renal tubular damage induced by gentamicin occurring AKI support that tubular enzymes can be
than markers of GFR.202 sensitive markers of tubular damage. However, incon-
Other nephrotoxic drugs evaluated in the context sistent results are present regarding the magnitude of
of tubular enzymes include polymyxin B (dogs)168, enzymuria, whether histologic changes are observed,
methyl cantharidimide (dogs)125, sulfonamide (a and whether or not azotemia develops following the
cat)185, neomycin (horses)205, ketoprofen (sheep)206, insult. Therefore, the usefulness of these markers in
and mercuric chloride and potassium dichromate predicting clinically significant tubular damage is
(ponies)207, all of which resulted in early increases in unknown.
uNAG/c, uGGT/c, or both. One study reported early
increases in uNAG/c and uGGT/c in dogs administered
CKD: Of the tubular enzymes, only NAG has been eval-
a therapeutic dose of ketoprofen long-term, but noted
uated in dogs and cats with CKD. Urinary NAG/c
that these enzyme indices returned to clinically normal
appears to be increased in most dogs and cats with
values in both dogs despite continued drug administra-
CKD as compared to healthy controls.110,123,197 In dogs
tion.208 Overall, administration of nonsteroidal anti-
with XLHN, mild increases were observed as one of the
inflammatory drugs (NSAID) at a therapeutic dose did
earliest findings, even before increased UPC; however,
not appear to significantly increase uGGT/c, uNAG/c,
it did not continue to increase with disease progression
or uALP/c in dogs.208–212 In horses treated with neo-
beyond mid-stage disease.110 Furthermore, uNAG/c
mycin, no histologic evidence of renal damage was
was inconsistently associated with IRIS staging in dogs
observed despite increases in uGGT/c.205 However, the
with proteinuric CKD.106,123 Interestingly, in dogs
study does not indicate what types of histologic studies
with XLHN, uNAG/c correlated moderately to strongly
were performed, and therefore, it is possible that renal
with both glomerular and TI damage lesions.110 In con-
lesions may have been missed. Additionally, dogs
trast, uNAG/c at the time of biopsy in dogs with natu-
given intravenous polymyxin B for 7 days demon-
rally occurring CKD (mostly proteinuric and variably
strated dose-dependent increases in uNAG/c, with sig-
azotemic) correlated moderately well with glomerular
nificantly increased uNAG/c occurring in the mid- and
damage alone and did not correlate with TI damage.106
high-dose groups on day 2.168 In one cat given sulfon-
Additionally, uNAG/c correlated moderately to
amide to induce acute renal failure, uNAG/c increased
strongly with UPC and uIgG/c in both studies.106,110
within 24 hours and continued to increase to very
These findings support that uNAG/c might be a better
high values several days after administration. N-
indicator of glomerular rather than tubular damage in
acetyl-b-D-glucosaminidase-A and NAG-B also both
canine proteinuric CKD.
increased, with a greater increase in NAG-B.185
Several studies in cats similarly suggest that
Few studies are available evaluating urinary
uNAG/c trends better with proteinuria than with tubu-
enzyme activity in cases of naturally occurring AKI in
lar damage. One study found that when geriatric cats
domestic animals. Dogs with pyometra have demon-

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 45
Renal biomarkers review Hokamp and Nabity

were grouped according to plasma creatinine concen- proximal tubular cell damage. However, when
tration, there was no significant difference in uNAG/c glomerular proteinuria is present, it is reasonable that
between groups, and uNAG/c was not correlated with increased uNAG/c at least partially represents loss of
creatinine. However, when cats were grouped based NAG from the blood due to altered glomerular perme-
on proteinuria, cats with borderline proteinuria and ability.
overt proteinuria had significantly higher uNAG/c
than nonproteinuric cats, and uNAG/c was moderately
correlated with UPC.190 uNAG/c was also unable to Cauxin. Cauxin, a 70 kDa enzyme220, is constitutively
distinguish azotemic from nonazotemic euthyroid secreted from the proximal straight221 and distal220
cats.218 Furthermore, uNAG/c was not significantly tubular epithelial cells in the urine of domestic (partic-
associated with development of azotemia when evalu- ularly intact males)220,221 and big222 cats. Progressive
ated in a multivariate model, as uNAG/c did not pro- tubular damage due to CKD caused decreased tissue
vide any benefit over UPC for predicting development expression223 and low urine excretion of cauxin134 in
of azotemia.219 In contrast, one study found that, cats, suggesting a decreased number of functional
although uNAG/c increased in cats with CKD com- tubular epithelial cells. However, when normalized to
pared to healthy cats, there was no correlation urinary creatinine concentration, decreased cauxin
between uNAG/c and severity of proteinuria, although was not observed in cats with CKD, and in fact,
this was based on unindexed, semi-quantitative mea- increased urinary cauxin/creatinine values were pre-
surement of proteinuria using a dipstick.185 dictive of development of azotemia in geriatric cats.224
Endocrine diseases may also influence urinary Other urinary enzymes noted to be increased
NAG activity, presumably due to renal damage. in AKI but not included in Table 3 include: glu-
Dogs with hyperadrenocorticism had significantly tathione-S-transferase176, lactate dehydrogenase206,
higher uNAG/c compared with control dogs.116 Fur- acid phosphatase206, MMP/pro-MMP 2206 in sheep,
thermore, while several other biomarkers (uALB/c, and MMP/pro-MMP 9 in horses.225
uIgG/c, uRBP/c, and UPC) demonstrated significant
decreases after treatment for hyperadrenocorticism,
uNAG/c did not decrease significantly even after Conclusions
treatment with trilostane and hypophysectomy.119
However, several dogs that were treated for hypera- The number of widely available tests of kidney func-
drenocorticism had persistent proteinuria.119 Despite tion and damage is currently limited, and our interpre-
the increases in uNAG/c in dogs with naturally tation of these tests, particularly creatinine, needs
increased cortisol due to hyperadrenocorticism, the improvement. However, there has recently been sig-
same effect was not seen in dogs treated with nificant progress in the evaluation of many new
hydrocortisone, both in comparison to the control biomarkers in dogs and cats with kidney disease. Most
group and within the treatment group itself.111 In of these biomarkers require further investigation as
dogs with diabetes mellitus, increased uNAG/c was well as test availability using a reliable, high-through-
observed when the disease was not controlled (ie, put, and commercially available platform that has been
hyperglycemia, glucosuria, and ketonuria were pre- validated in the sample of interest before their wide-
sent), whereas uNAG/c values were comparable to spread use can be recommended and achieved. Still,
healthy dogs when blood glucose was controlled.197 studies to date show promise that several of these
Finally, azotemic and nonazotemic cats with hyper- markers can and will aid in early detection, monitor-
thyroidism had higher uNAG/c than healthy cats, ing, and assessment of prognosis in patients with kid-
and uNAG/c decreased with treatment in both ney disease. They can also help localize damage to the
groups.218 In this study, urine microalbumin con- kidney.
centration in hyperthyroid cats was statistically simi- The evidence for the role of urinary biomarkers in
lar among healthy, azotemic, and nonazotemic the early detection of tubular damage in AKI is particu-
cats.218 larly compelling, whereas in animals with proteinuric
The majority of studies show that urinary NAG lar- kidney disease, the influence of filtered proteins (origi-
gely trends with proteinuria. For both dogs and cats nating from the blood) must be considered. There is
with CKD, a possible reason behind the stronger corre- also the potential for nonrenal influences, such as
lation of urinary NAG with glomerular damage/pro- hematuria and pyuria, to alter biomarker concentra-
teinuria as compared with TI damage/azotemia could tions. For plasma-derived proteins serving as indicators
be increased lysosomal activity as opposed to active of tubular reabsorptive function (eg, RBP, cystatin C)

46 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

these limitations are not likely to be overcome. How- other endogenous markers of glomerular filtration rate
ever, for indicators of tubular damage, we should in healthy dogs. J Vet Intern Med. 2007;21:936–942.
strive to use biomarkers that are either exclusively 11. Ruaux CG, Carney PC, Suchodolski JS, Steiner JM.
produced by the renal tubules or have renal tubular- Estimates of biological variation in routinely measured
specific forms that can be distinguished from circulat- biochemical analytes in clinically healthy dogs. Vet Clin
ing forms of the protein. Without such specificity, Pathol. 2012;41:541–547.
early, noninvasive detection of tubular damage will 12. Rosset E, Rannou B, Casseleux G, Chalvet-Monfray K,
remain challenging. Finally, no single biomarker is Buff S. Age-related changes in biochemical and hema-
likely to be sufficient for even one component of tologic variables in Borzoi and Beagle puppies from
altered kidney function or damage, and a panel of birth to 8 weeks. Vet Clin Pathol. 2012;41:272–282.
tests will be needed in the future for comprehensive 13. Rørtveit R, Saevik BK, Eggertsd
ottir AV, et al. Age-
evaluation to best determine renal status in an indi- related changes in hematologic and serum biochemical
vidual animal. variables in dogs aged 16-60 days. Vet Clin Pathol.
2015;44:47–57.
Disclosure: Dr. Nabity serves on the Idexx Renal Advi- 14. Misbach C, Chetboul V, Concordet D, et al. Basal
sory Board. plasma concentrations of routine variables and packed
cell volume in clinically healthy adult small-sized dogs:
effect of breed, body weight, age, and gender, and
establishment of reference intervals. Vet Clin Pathol.
References
2014;43:371–380.
1. Cobrin AR, Blois SL, Kruth SA, Abrams-Ogg AC, 15. Zaldıvar-L
opez S, Marın LM, Iazbik MC, et al. Clinical
Dewey C. Biomarkers in the assessment of acute and pathology of Greyhounds and other sighthounds. Vet
chronic kidney diseases in the dog and cat. J Small Clin Pathol. 2011;40:414–425.
Anim Pract. 2013;54:647–655.
16. Medaille C, Trumel C, Concordet D, Vergez F, Braun
2. De Loor J, Daminet S, Smets P, Maddens B, Meyer E. JP. Comparison of plasma/serum urea and creatinine
Urinary biomarkers for acute kidney injury in dogs. J concentrations in the dog: a 5-year retrospective study
Vet Intern Med. 2013;27:998–1010. in a commercial veterinary clinical pathology labora-
3. Pressler BM. Clinical approach to advanced renal func- tory. J Vet Med A Physiol Pathol Clin Med. 2004;51:119–
tion testing in dogs and cats. Vet Clin North Am Small 123.
Anim Pract. 2013;43:1193–1208, v. 17. Miyagawa Y, Takemura N, Hirose H. Assessments of
4. Braun JP, Lefebvre HP, Watson AD. Creatinine in the factors that affect glomerular filtration rate and indi-
dog: a review. Vet Clin Pathol. 2003;32:162–179. rect markers of renal function in dogs and cats. J Vet
5. Brown SA, Finco DR, Crowell WA, Choat DC, Navar Med Sci. 2010;72:1129–1136.
LG. Single-nephron adaptations to partial renal abla- 18. Dunlop MM, Sanchez-Vazquez MJ, Freeman KP, et al.
tion in the dog. Am J Physiol. 1990;258:F495–F503. Determination of serum biochemistry reference inter-
6. Bovee KC, Kronfeld DS, Ramberg C, Goldschmidt M. vals in a large sample of adult greyhounds. J Small
Long-term measurement of renal function in partially Anim Pract. 2011;52:4–10.
nephrectomized dogs fed 56, 27, or 19% protein. Invest 19. Feeman WE, Couto CG, Gray TL. Serum creatinine
Urol. 1979;16:378–384. concentrations in retired racing Greyhounds. Vet Clin
7. Bricker NS, Klahr S, Rieselbach RE. The functional Pathol. 2003;32:40–42.
adaptation of the diseased kidney. I. Glomerular filtra- 20. Reynolds BS, Concordet D, Germain CA, et al. Breed
tion rate. J Clin Invest. 1964;43:1915–1921. dependency of reference intervals for plasma bio-
8. Nabity MB, Lees GE, Boggess MM. SDMA assay valida- chemical values in cats. J Vet Intern Med. 2010;24:809–
tion, stability, and evaluation as a marker for early 818.
detection of chronic kidney disease in dogs. J Vet Intern 21. Concordet D, Vergez F, Trumel C, et al. A multicentric
Med. 2015;29:1036–1044. retrospective study of serum/plasma urea and crea-
9. Baral RM, Dhand NK, Freeman KP, Krockenberger tinine concentrations in dogs using univariate and
MB, Govendir M. Biological variation and reference multivariate decision rules to evaluate diagnostic effi-
change values of feline plasma biochemistry analytes. ciency. Vet Clin Pathol. 2008;37:96–103.
J Feline Med Surg. 2014;16:317–325. 22. Hall JA, Yerramilli M, Obare E, et al. Comparison of
10. Pagitz M, Frommlet F, Schwendenwein I. Evaluation serum concentrations of symmetric dimethylarginine
of biological variance of cystatin C in comparison with and creatinine as kidney function biomarkers in

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 47
Renal biomarkers review Hokamp and Nabity

healthy geriatric cats fed reduced protein foods 35. Bode-B€ oger SM, Scalera F, Kielstein JT, et al. Symmet-
enriched with fish oil, L-carnitine, and medium-chain rical dimethylarginine: a new combined parameter for
triglycerides. Vet J. 2014;202:588–596. renal function and extent of coronary artery disease. J
23. Ulleberg T, Robben J, Nordahl KM, Ulleberg T, Heiene Am Soc Nephrol. 2006;17:1128–1134.
R. Plasma creatinine in dogs: intra- and inter-labora- 36. Kielstein JT, Salpeter SR, Bode-Boeger SM, Cooke JP,
tory variation in 10 European veterinary laboratories. Fliser D. Symmetric dimethylarginine (SDMA) as
Acta Vet Scand. 2011;53:25. endogenous marker of renal function–a meta-analysis.
24. Braun JP, Cab e E, Geffr
e A, Lefebvre HP, Trumel C. Nephrol Dial Transplant. 2006;21:2446–2451.
Comparison of plasma creatinine values measured by 37. Ei Closs. Basha FZ, Habermeier A, F€
orstermann U.
different veterinary practices. Vet Rec. 2008;162:215– Interference of L-arginine analogues with L-arginine
216. transport mediated by the y+ carrier hCAT-2B. Nitric
25. Harr KE, Flatland B, Nabity M, Freeman KP; ASVCP. Oxide. 1997;1:65–73.
ASVCP guidelines: allowable total error guidelines for 38. Feliers D, Lee DY, Gorin Y, Kasinath BS. Symmetric
biochemistry. Vet Clin Pathol. 2013;42:424–236. dimethylarginine alters endothelial nitric oxide activ-
26. Ghys L, Paepe D, Smets P, et al. Cystatin C: a new renal ity in glomerular endothelial cells. Cell Signal.
marker and its potential use in small animal medicine. 2015;27:1–5.
J Vet Intern Med. 2014;28:1152–1164. 39. Schepers E, Speer T, Bode-B€ oger SM, Fliser D, Kiel-
27. Wei H, Mundade R, Lange KC, Lu T. Protein arginine stein JT. Dimethylarginines ADMA and SDMA: the
methylation of non-histone proteins and its role in dis- real water-soluble small toxins? Semin Nephrol.
eases. Cell Cycle. 2014;13:32–41. 2014;34:97–105.
28. Kakimoto Y, Akazawa S. Isolation and identification of 40. Veldink H, Faulhaber-Walter R, Park JK, et al. Effects
N-G, N-G- and N-G, N’-G-dimethyl-arginine, N-epsi- of chronic SDMA infusion on glomerular filtration
lon-mono-, di-, and trimethyllysine, and glucosyl- rate, blood pressure, myocardial function and renal
galactosyl- and galactosyl-delta-hydroxylysine from histology in C57BL6/J mice. Nephrol Dial Transplant.
human urine. J Biol Chem. 1970;245:5751–5758. 2013;28:1434–1439.
29. Nakajima T, Matsuoka Y, Kakimoto Y. Isolation and 41. Schwedhelm E. Quantification of ADMA: analyti-
identification of N-G-monomethyl, N-G, N-G- cal approaches. Vasc Med. 2005;10(Suppl. 1):S89–S95.
dimethyl- and N-G, N’ G-dimethylarginine from the 42. Tenderenda-Banasiuk E, Wasilewska A, Taranta-
hydrolysate of proteins of bovine brain. Biochem Bio- Janusz K, Korzeniecka-Kozerska A. Asymmetric and
phys Acta. 1971;230:212–222. symmetric dimethylarginine in adolescents with hype-
30. Nijveldt RJ, Van-Leeuwen PA, Van-Guldener C, ruricemia. Dis Markers. 2013;35:407–412.
et al. Net renal extraction of asymmetrical (ADMA) 43. Hall JA, Yerramilli M, Obare E, Yerramilli M, Jewell
and symmetrical (SDMA) dimethylarginine in fast- DE. Comparison of serum concentrations of symmetric
ing humans. Nephrol Dial Transplant. 2002;17:1999– dimethylarginine and creatinine as kidney function
2002. biomarkers in cats with chronic kidney disease. J Vet
31. Al-Banchaabouchi M, Marescau B, Possemiers I, et al. Intern Med. 2014;28:1676–1683.
NG, NG-dimethylarginine and NG, NG-dimethylargi- 44. Patch D, Obare E, Xie H. High throughput immunoas-
nine in renal insufficiency. Pflugers Arch. say that correlates to gold standard liquid chromatog-
2000;439:524–531. raphy mass spectrometry (LC-MS) assay for the
32. Nijveldt RJ, Teerlink T, van-Guldener C, et al. Han- chronic kidney disease (CKD) marker symmetric
dling of asymmetrical dimethylarginine and symmetri- dimethylarginine (SDMA) [abstract]. J Vet Intern Med.
cal dimethylarginine by the rat kidney under basal 2015;29:1216.
conditions and during endotoxaemia. Nephrol Dial 45. Yerramilli M, Obare E, Relford R. Stability of SDMA
Transplant. 2003;18:2542–2550. (symmetric dimethylarginine) in canine and feline
33. Vallance P, Leone A, Calver A, Collier J, Moncada S. serum and plasma [abstract]. J Vet Intern Med.
Accumulation of an endogenous inhibitor of nitric 2013;27:737.
oxide synthesis in chronic renal failure. Lancet. 46. El-Khoury JM, Bunch DR, Reineks E, et al. A simple
1992;339:572–575. and fast liquid chromatography-tandem mass spec-
34. Fleck C, Janz A, Schweitzer F, et al. Serum concentra- trometry method for measurement of underivatized L-
tions of asymmetric (ADMA) and symmetric (SDMA) arginine, symmetric dimethylarginine, and asymmet-
dimethylarginine in renal failure patients. Kidney Int. ric dimethylarginine and establishment of the refer-
2001;59:S14–S18. ence ranges. Anal Bioanal Chem. 2012;402:771–779.

48 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

47. Schwedhelm E, Xanthakis V, Maas R, et al. Plasma 60. Krzyzanowska K, Mittermayer F, Kopp HP, Wolzt M,
symmetric dimethylarginine reference limits from the Schernthaner G. Weight loss reduces circulating asym-
Framingham offspring cohort. Clin Chem Lab Med. metrical dimethylarginine concentrations in morbidly
2011;49:1907–1910. obese women. J Clin Endocrinol Metab. 2004;89:6277–
48. Rentko V, Nabity M, Yerramilli M. Determination of 6281.
serum symmetric dimethylarginine reference limit in 61. Blackwell S, O’Reilly DS, Reid D, Talwar D. Plasma
clinically healthy dogs [abstract]. J Vet Intern Med. dimethylarginines during the acute inflammatory
2013;27:750. response. Eur J Clin Invest. 2011;41:635–641.
49. Moesgaard SG, Holte AV, Mogensen T, et al. Effects of 62. Onozato ML, Tojo A, Leiper J, et al. Expression of NG,
breed, gender, exercise and white-coat effect on mark- NG-dimethylarginine dimethylaminohydrolase and
ers of endothelial function in dogs. Res Vet Sci. protein arginine N-methyltransferase isoforms in dia-
2007;82:409–415. betic rat kidney: effects of angiotensin II receptor
50. Moesgaard SG, Pedersen LG, Teerlink T, Haggstrom J, blockers. Diabetes. 2008;57:172–180.
Pedersen HD. Neurohormonal and circulatory effects 63. Teerlink T, Neele SJ, de-Jong S, Netelenbos JC, Ste-
of short-term treatment with enalapril and quinapril houwer CD. Oestrogen replacement therapy lowers
in dogs with asymptomatic mitral regurgitation. J Vet plasma levels of asymmetrical dimethylarginine in
Intern Med. 2005;19:712–719. healthy postmenopausal women. Clin Sci (Lond).
51. Tatematsu S, Wakino S, Kanda T, et al. Role of nitric 2003;105:67–71.
oxide-producing and -degrading pathways in coronary 64. Holden DP, Cartwright JE, Nussey SS, Whitley GS.
endothelial dysfunction in chronic kidney disease. J Estrogen stimulates dimethylarginine dimethylamino-
Am Soc Nephrol. 2007;18:741–749. hydrolase activity and the metabolism of asymmetric
52. Pedersen LG, Tarnow I, Olsen LH, Teerlink T, Pedersen dimethylarginine. Circulation. 2003;108:1575–1580.
HD. Body size, but neither age nor asymptomatic 65. P€aiv€
a H, Lehtim€aki T, Laakso J, et al. Dietary composi-
mitral regurgitation, influences plasma concentrations tion as a determinant of plasma asymmetric dimethy-
of dimethylarginines in dogs. Res Vet Sci. 2006;80:336– larginine in subjects with mild hypercholesterolemia.
342. Metabolism. 2004;53:1072–1075.
53. Rifai K, Bode-Boeger SM, Martens-Lobenhoffer J, 66. Rytlewski K, Olszanecki R, Korbut R, Zdebski Z. Effects
et al. Removal of asymmetric dimethylarginine during of prolonged oral supplementation with l-arginine on
artificial liver support using fractionated plasma sepa- blood pressure and nitric oxide synthesis in preeclamp-
ration and adsorption. Scand J Gastroenterol. sia. Eur J Clin Invest. 2005;35:32–37.
2010;45:1110–1115. 67. Marliss EB, Chevalier S, Gougeon R, et al. Elevations
54. MacAllister RJ, Rambausek MH, Vallance P, et al. of plasma methylarginines in obesity and ageing are
Concentration of dimethyl-L-arginine in the plasma of related to insulin sensitivity and rates of protein turn-
patients with end-stage renal failure. Nephrol Dial over. Diabetologia. 2006;49:351–359.
Transplant. 1996;11:2449–2452. 68. Atzler D, Schwedhelm E, Nauck M, et al. Serum refer-
55. Siroen MP, van-der-Sijp JR, Teerlink T, et al. The ence intervals of homoarginine, ADMA, and SDMA in
human liver clears both asymmetric and symmetric the Study of Health in Pomerania. Clin Chem Lab Med.
dimethylarginine. Hepatology. 2005;41:559–565. 2014;52:1835–1842.
56. Nijveldt RJ, Siroen MP, Teerlink T, et al. Gut and liver 69. Lakhani K, Kay AR, Leiper J, Barry JA, Hardiman PJ.
handling of asymmetric and symmetric dimethy- Symmetric dimethylarginine (SDMA) is raised in
larginine in the rat under basal conditions and during women with polycystic ovary syndrome: a pilot study.
endotoxemia. Liver Int. 2004;24:510–518. J Obstet Gynaecol. 2011;31:417–419.
57. Ogawa T, Kimoto M, Sasaoka K. Purification and prop- 70. Hall J, Yerramilli M, Obare E, et al. Relationship
erties of a new enzyme, NG, NG-dimethylarginine between lean body mass and serum renal biomarkers
dimethylaminohydrolase, from rat kidney. J Biol Chem. in healthy dogs. J Vet Intern Med. 2015;29:808–814.
1989;264:10205–10209. 71. Braff J, Obare E, Yerramilli M, Elliott J, Yerramilli M.
58. Schwedhelm E, B€ oger RH. The role of asymmetric and Relationship between serum symmetric dimethy-
symmetric dimethylarginines in renal disease. Nat Rev larginine concentration and glomerular filtration rate
Nephrol. 2011;7:275–285. in cats. J Vet Intern Med. 2014;28:1699–1701.
59. Kittel A, Maas R, K€
onig J, et al. In vivo evidence that 72. Jepson RE, Syme HM, Vallance C, Elliott J. Plasma
Agxt2 can regulate plasma levels of dimethylarginines asymmetric dimethylarginine, symmetric dimethy-
in mice. Biochem Biophys Res Commun. 2013;430:84–89. larginine, l-arginine, and nitrite/nitrate concentra-

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 49
Renal biomarkers review Hokamp and Nabity

tions in cats with chronic kidney disease and hyperten- 86. Pasa S, Bayramli G, Atasoy A, et al. Evaluation of
sion. J Vet Intern Med. 2008;22:317–324. serum cystatin-C in dogs with visceral leishmaniasis.
73. de Brito Galvao JF, Nagode LA, Schenck PA, Chew DJ. Vet Res Commun. 2009;33:529–534.
Calcitriol, calcidiol, parathyroid hormone, and fibrob- 87. Miyagawa Y, Takemura N, Hirose H. Evaluation of the
last growth factor-23 interactions in chronic kidney measurement of serum cystatin C by an enzyme-
disease. J Vet Emerg Crit Care (San Antonio). linked immunosorbent assay for humans as a marker
2013;23:134–162. of the glomerular filtration rate in dogs. J Vet Med Sci.
74. Seiler S, Heine GH, Fliser D. Clinical relevance of FGF- 2009;71:1169–1176.
23 in chronic kidney disease. Kidney Int Suppl. 2009;76: 88. Braun JP, Perxachs A, Pechereau D, De la Farge F.
S34–S42. Plasma cystatin C in the dog: reference values and vari-
75. Liu S, Quarles LD. How fibroblast growth factor 23 ations with renal failure. Comp Clin Pathol. 2002;11:44–
works. J Am Soc Nephrol. 2007;18:1637–1647. 49.
76. Shimada T, Kakitani M, Yamazaki Y, et al. Targeted 89. Garcia-Martinez JD, Martinez-Subiela S, Tvarijonavi-
ablation of Fgf23 demonstrates an essential physiologi- ciute A, Caldin M, Ceron JJ. Urinary ferritin and cys-
cal role of FGF23 in phosphate and vitamin D metabo- tatin C concentrations at different stages of kidney
lism. J Clin Invest. 2004;113:561–568. disease in leishmaniotic dogs. Res Vet Sci. 2015;99:204–
207.
77. Nakai K, Komaba H, Fukagawa M. New insights into
the role of fibroblast growth factor 23 in chronic kid- 90. Poswiatowska-Kazczyszyn I. Usefulness of serum cys-
ney disease. J Nephrol. 2010;23:619–625. tatin C measurement for assessing renal function in
cats. Bull Vet Inst Pulawy. 2012;56:235–239.
78. Finch NC, Geddes RF, Syme HM, Elliott J. Fibroblast
growth factor 23 (FGF-23) concentrations in cats with 91. Ahn HJ, Hyun C. Evaluation of serum neutrophil
early nonazotemic chronic kidney disease (CKD) and gelatinase-associated lipocalin (NGAL) activity in dogs
in healthy geriatric cats. J Vet Intern Med. 2013;27:227– with chronic kidney disease. Vet Rec. 2013;173:452.
233. 92. Hsu WL, Lin YS, Hu YY, et al. Neutrophil gelatinase-
79. Geddes RF, Elliott J, Syme HM. The effect of feeding a associated lipocalin in dogs with naturally occurring
renal diet on plasma fibroblast growth factor 23 con- renal diseases. J Vet Intern Med. 2014;28:437–442.
centrations in cats with stable azotemic chronic kidney 93. Steinbach S, Weis J, Schweighauser A, Francey T, Nei-
disease. J Vet Intern Med. 2013;27:1354–1361. ger R. Plasma and urine neutrophil gelatinase-asso-
80. Geddes RF, Finch NC, Elliott J, Syme HM. Fibroblast ciated lipocalin (NGAL) in dogs with acute kidney
growth factor 23 in feline chronic kidney disease. J Vet injury or chronic kidney disease. J Vet Intern Med.
Intern Med. 2013;27:234–241. 2014;28:264–269.
81. Williams TL, Elliott J, Syme HM. Calcium and phos- 94. Rossi G, Giordano A, Breda S, et al. Big-endothelin
phate homeostasis in hyperthyroid cats: associations 1 (big ET-1) and homocysteine in the serum of
with development of azotaemia and survival time. J dogs with chronic kidney disease. Vet J.
Small Anim Pract. 2012;53:561–571. 2013;198:109–115.
82. Ghys LF, Meyer E, Paepe D, Delanghe J, Daminet S. 95. Rossi S, Rossi G, Giordano A, Paltrinieri S. Homocys-
Analytical validation of a human particle-enhanced teine measurement by an enzymatic method and
nephelometric assay for cystatin C measurement in potential role of homocysteine as a biomarker in dogs.
feline serum and urine. Vet Clin Pathol. 2014;43:226– J Vet Diagn Invest. 2008;20:644–649.
234. 96. Tvarijonaviciute A, Ceron JJ, Holden SL, et al. Effect
83. Gonul R, Kayar A, Or M. Assessment of renal function of weight loss in obese dogs on indicators of renal func-
in dogs with renal disease using serum cystatin C. tion or disease. J Vet Intern Med. 2013;27:31–38.
Indian Vet J. 2004;81:872–874. 97. Raila J, Schweigert FJ, Kohn B. C-reactive protein con-
84. Ghys LF, Paepe D, Duchateau L, et al. Biological vali- centrations in serum of dogs with naturally occurring
dation of feline serum cystatin C: the effect of breed, renal disease. J Vet Diagn Invest. 2011;23:710–715.
age and sex and establishment of a reference interval. 98. D’Amico G, Bazzi C. Pathophysiology of proteinuria.
Vet J. 2015;204:168–173. Kidney Int. 2003;63:809–825.
85. Martin C, Pechereau D, De la Farge F, Braun JP. Cys- 99. Maack T. Renal handling of low molecular weight pro-
tatine C plasmatique chez le chat: Les technique teins. Am J Med. 1975;58:57–64.
actuelles ne permettent pas de l’utiliser comme mar- 100. Zandi-Nejad K, Eddy AA, Glassock RJ, Brenner BM.
queur d’insufficance renale. Rev Med Vet. Why is proteinuria an ominous biomarker of progres-
2002;153:305–310.

50 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

sive kidney disease? Kidney Int Suppl. 2004;66:S76– 114. Maddens B, Daminet S, Smets P, Meyer E. Escherichia
S89. coli pyometra induces transient glomerular and tubu-
101. Waikar SS, Sabbisetti VS, Bonventre JV. Normaliza- lar dysfunction in dogs. J Vet Intern Med.
tion of urinary biomarkers to creatinine during 2010;24:1263–1270.
changes in glomerular filtration rate. Kidney Int. 115. Maddens B, Heiene R, Smets P, et al. Evaluation of
2010;78:486–494. kidney injury in dogs with pyometra based on protein-
102. Tizard IR. Antibodies: Soluble Forms of BCR. 7th ed. uria, renal histomorphology, and urinary biomarkers.
Philadelphia, PA: Saunders; 2004. J Vet Intern Med. 2011;25:1075–1083.
103. Edelman GM, Cunningham BA, Gall WE, et al. The 116. Smets PM, Lefebvre HP, Kooistra HS, et al. Hypercorti-
covalent structure of an entire gammaG immunoglob- solism affects glomerular and tubular function in dogs.
ulin molecule. Proc Natl Acad Sci USA. 1969;63:78–85. Vet J. 2012;192:532–534.
104. Kerr MA. The structure and function of human IgA. 117. Raila J, Aupperle H, Raila G, Schoon HA, Schweigert
Biochem J. 1990;271:285–296. FJ. Renal pathology and urinary protein excretion in a
14-month-old Bernese mountain dog with chronic
105. Maddens BE, Daminet S, Demeyere K, et al. Valida-
renal failure. J Vet Med A Physiol Pathol Clin Med.
tion of immunoassays for the candidate renal markers
2007;54:131–135.
C-reactive protein, immunoglobulin G, thromboxane
B2 and retinol binding protein in canine urine. Vet 118. Vinge L, Lees GE, Nielsen R, et al. The effect of pro-
Immunol Immunopathol. 2010;134:259–264. gressive glomerular disease on megalin-mediated
endocytosis in the kidney. Nephrol Dial Transplant.
106. Hokamp JA, Cianciolo RE, Boggess MM, et al. Corre-
2010;25:2458–2467.
lation of urine and serum biomarkers with renal dam-
age and survival in dogs with naturally occurring 119. Smets PM, Lefebvre HP, Meij BP, et al. Long-term fol-
chronic kidney disease. J Vet Intern Med. 2015; doi: low-up of renal function in dogs after treatment for
10.1111/jvim.13832. [Epub ahead of print] ACTH-dependent hyperadrenocorticism. J Vet Intern
Med. 2012;26:565–574.
107. Zaragoza C, Barrera R, Centeno F, et al. SDS-PAGE
and Western blot of urinary proteins in dogs with 120. Grauer GF. Measurement, interpretation, and impli-
leishmaniasis. Vet Res. 2003;34:137–151. cations of proteinuria and albuminuria. Vet Clin North
Am Small Anim Pract. 2007;37:283–295, vi-vii.
108. Zaragoza C, Barrera R, Centeno F, Tapia JA, Mane MC.
Characterization of renal damage in canine leptospiro- 121. Yaqoob M, McClelland P, Patrick AW, et al. Tubulopa-
sis by sodium dodecyl sulphate-polyacrylamide gel thy with macroalbuminuria due to diabetic nephropa-
electrophoresis (SDS-PAGE) and Western blotting of thy and primary glomerulonephritis. Kidney Int Suppl.
the urinary proteins. J Comp Pathol. 2003;129:169–178. 1994;47:S101–S104.
109. Zaragoza C, Barrera R, Centeno F, Tapia JA, Mane 122. Yaqoob M, McClelland P, Patrick AW, et al. Tubular
MC. Canine pyometra: a study of the urinary proteins damage in microalbuminuric patients with primary
by SDS-PAGE and Western blot. Theriogenology. glomerulonephritis and diabetic nephropathy. Ren
2004;61:1259–1272. Fail. 1995;17:43–49.
110. Nabity MB, Lees GE, Cianciolo R, et al. Urinary 123. Smets PM, Meyer E, Maddens BE, Duchateau L,
biomarkers of renal disease in dogs with X-linked Daminet S. Urinary markers in healthy young and
hereditary nephropathy. J Vet Intern Med. aged dogs and dogs with chronic kidney disease. J Vet
2012;26:282–293. Intern Med. 2010;24:65–72.
111. Smets PM, Lefebvre HP, Aresu L, et al. Renal function 124. Raila J, Brunnberg L, Schweigert FJ, Kohn B. Influ-
and morphology in aged Beagle dogs before and after ence of kidney function on urinary excretion of albu-
hydrocortisone administration. PLoS ONE. 2012;7: min and retinol-binding protein in dogs with naturally
e31702. occurring renal disease. Am J Vet Res. 2010;71:1387–
1394.
112. Defauw P, Schoeman JP, Smets P, et al. Assessment of
renal dysfunction using urinary markers in canine 125. Luo XQ, Yang X, Hu R, et al. [Effects of methyl can-
babesiosis caused by Babesia rossi. Vet Parasitol. tharidimide tablets on urinary protein and enzymes in
2012;190:326–332. Beagle dogs]. Zhongguo Zhong Yao Za Zhi.
2014;39:4426–4429.
113. Hrovat A, Schoeman JP, de Laat B, et al. Evaluation of
snake envenomation-induced renal dysfunction in 126. Sasaki A, Sasaki Y, Iwama R, et al. Comparison of
dogs using early urinary biomarkers of nephrotoxicity. renal biomarkers with glomerular filtration rate in
Vet J. 2013;198:239–244. susceptibility to the detection of gentamicin-induced

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 51
Renal biomarkers review Hokamp and Nabity

acute kidney injury in dogs. J Comp Pathol. 142. Bernard AM, Vyskocil AA, Mahieu P, Lauwerys RR.
2014;151:264–270. Assessment of urinary retinol-binding protein as an
127. Maeda H, Sogawa K, Sakaguchi K, et al. Urinary albu- index of proximal tubular injury. Clin Chem.
min and transferrin as early diagnostic markers of 1987;33:775–779.
chronic kidney disease. J Vet Med Sci. 2015;77:937– 143. Christensen EI, Moskaug JO, Vorum H, et al. Evidence
943. for an essential role of megalin in transepithelial trans-
128. Ceron JJ, Eckersall PD, Martynez-Subiela S. Acute port of retinol. J Am Soc Nephrol. 1999;10:685–695.
phase proteins in dogs and cats: current knowledge 144. Raila J, Forterre S, Kohn B, Brunnberg L, Schweigert
and future perspectives. Vet Clin Pathol. 2005;34:85–99. FJ. Effects of chronic renal disease on the transport of
129. Eckersall PD, Bell R. Acute phase proteins: biomarkers vitamin A in plasma and urine of dogs. Am J Vet Res.
of infection and inflammation in veterinary medicine. 2003;64:874–879.
Vet J. 2010;185:23–27. 145. van Hoek I, Daminet S, Notebaert S, Janssens I, Meyer
130. Gotschlich EC, Edelman GM. C-reactive protein: a E. Immunoassay of urinary retinol binding protein as
molecule composed of subunits. Proc Natl Acad Sci a putative renal marker in cats. J Immunol Methods.
USA. 1965;54:558–566. 2008;329:208–213.
131. Kushner I, Somerville JA. Estimation of the molecular 146. Smets PM, Meyer E, Maddens B, Duchateau L, Dami-
size of C-reactive protein and CX-reactive protein in net S. Effect of sampling method and storage condi-
serum. Biochim Biophys Acta. 1970;207:105–114. tions on albumin, retinol-binding protein, and N-
acetyl-beta-D-glucosaminidase concentrations in
132. Martinez-Subiela S, Garcia-Martinez JD, Tvarijonavi-
canine urine samples. J Vet Diagn Invest. 2010;22:896–
ciute A, et al. Urinary C reactive protein levels in dogs
902.
with leishmaniasis at different stages of renal damage.
Res Vet Sci. 2013;95:924–929. 147. Palviainen M, Raekallio M, Vainionpaa M, Lahtinen
H, Vainio O. Evaluation of renal impairment in dogs
133. Tvarijonaviciute A, Ceron JJ, Martinez-Subiela S, Gar-
after envenomation by the common European adder
cia-Martinez JD. Serum and urinary adiponectin in
(Vipera berus berus). Vet J. 2013;198:723–724.
dogs with renal disease from leishmaniasis. Vet Rec.
2012;171:297. 148. van Hoek I, Lefebvre HP, Peremans K, et al. Short-
and long-term follow-up of glomerular and tubular
134. Ferlizza E, Campos A, Neagu A, et al. The effect of
renal markers of kidney function in hyperthyroid cats
chronic kidney disease on the urine proteome in the
after treatment with radioiodine. Domest Anim Endocri-
domestic cat (Felis catus). Vet J. 2015;204:73–81.
nol. 2009;36:45–56.
135. Kanai M, Raz A, Goodman DS. Retinol-binding pro-
149. van Hoek I, Meyer E, Duchateau L, et al. Retinol-
tein: the transport protein for vitamin A in human
binding protein in serum and urine of hyperthyroid
plasma. J Clin Invest. 1968;47:2025–2044.
cats before and after treatment with radioiodine. J Vet
136. Soprano DR, Soprano KJ, Goodman DS. Retinol-bind- Intern Med. 2009;23:1031–1037.
ing protein and transthyretin mRNA levels in visceral
150. Nabity MB, Lees GE, Dangott LJ, et al. Proteomic anal-
yolk sac and liver during fetal development in the rat.
ysis of urine from male dogs during early stages of
Proc Natl Acad Sci USA. 1986;83:7330–7334.
tubulointerstitial injury in a canine model of progres-
137. Soprano DR, Soprano KJ, Goodman DS. Retinol-bind- sive glomerular disease. Vet Clin Pathol. 2011;40:222–
ing protein messenger RNA levels in the liver and in 236.
extrahepatic tissues of the rat. J Lipid Res.
151. Forterre S, Raila J, Schweigert FJ. Protein profiling of
1986;27:166–171.
urine from dogs with renal disease using ProteinChip
138. Raila J, Buchholz I, Aupperle H, et al. The distribution analysis. J Vet Diagn Invest. 2004;16:271–277.
of vitamin A and retinol-binding protein in the blood
152. Palviainen M, Raekallio M, Rajamaki MM, Linden J,
plasma, urine, liver and kidneys of carnivores. Vet Res.
Vainio O. Kidney-derived proteins in urine as
2000;31:541–551.
biomarkers of induced acute kidney injury in sheep.
139. Monaco HL. The transthyretin-retinol-binding protein Vet J. 2012;193:287–289.
complex. Biochim Biophys Acta. 2000;1482:65–72.
153. Kjeldsen L, Johnsen AH, Sengelov H, Borregaard N.
140. Ingenbleek Y, Young V. Transthyretin (prealbumin) in Isolation and primary structure of NGAL, a novel pro-
health and disease: nutritional implications. Annu Rev tein associated with human neutrophil gelatinase. J
Nutr. 1994;14:495–533. Biol Chem. 1993;268:10425–10432.
141. Christensen EI, Willnow TE. Essential role of megalin 154. Bundgaard JR, Sengelov H, Borregaard N, Kjeldsen
in renal proximal tubule for vitamin homeostasis. J L. Molecular cloning and expression of a cDNA
Am Soc Nephrol. 1999;10:2224–2236.

52 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

encoding NGAL: a lipocalin expressed in human ney injury in beagle dogs. Toxicol Appl Pharmacol.
neutrophils. Biochem Biophys Res Commun. 2014;280:30–35.
1994;202:1468–1475. 168. Burt D, Crowell SJ, Ackley DC, Magee TV,
155. Friedl A, Stoesz SP, Buckley P, Gould MN. Neutrophil Aubrecht J. Application of emerging biomarkers of
gelatinase-associated lipocalin in normal and neoplas- acute kidney injury in development of kidney-
tic human tissues. Cell type-specific pattern of expres- sparing polypeptide-based antibiotics. Drug Chem
sion. Histochem J. 1999;31:433–441. Toxicol. 2014;37:204–212.
156. Nielsen BS, Borregaard N, Bundgaard JR, et al. Induc- 169. Lee YJ, Hu YY, Lin YS, et al. Urine neutrophil
tion of NGAL synthesis in epithelial cells of human gelatinase-associated lipocalin (NGAL) as a biomar-
colorectal neoplasia and inflammatory bowel diseases. ker for acute canine kidney injury. BMC Vet Res.
Gut. 1996;38:414–420. 2012;8:248.
157. Goetz DH, Holmes MA, Borregaard N, et al. The neu- 170. Davis J, Raisis AL, Cianciolo RE, et al. Urinary neu-
trophil lipocalin NGAL is a bacteriostatic agent that trophil gelatinase-associated lipocalin concentration
interferes with siderophore-mediated iron acquisition. changes after acute haemorrhage and colloid-
Mol Cell. 2002;10:1033–1043. mediated reperfusion in anaesthetized dogs. Vet
158. Mori K, Lee HT, Rapoport D, et al. Endocytic delivery Anaesth Analg. 2015; doi: 10.1111/vaa.12311. [Epub
of lipocalin-siderophore-iron complex rescues the kid- ahead of print]
ney from ischemia-reperfusion injury. J Clin Invest. 171. Schweigert FJ, Raila J, Haebel S. Vitamin A excreted
2005;115:610–621. in the urine of canines is associated with a Tamm-
159. Schmidt-Ott KM, Mori K, Li JY, et al. Dual action of Horsfall like protein. Vet Res. 2002;33:299–311.
neutrophil gelatinase-associated lipocalin. J Am Soc 172. Raila J, Neumann U, Schweigert FJ. Immunochemical
Nephrol. 2007;18:407–413. localization of megalin, retinol-binding protein and
160. Kuwabara T, Mori K, Mukoyama M, et al. Urinary Tamm-Horsfall glycoprotein in the kidneys of dogs. Vet
neutrophil gelatinase-associated lipocalin levels reflect Res Commun. 2003;27:125–135.
damage to glomeruli, proximal tubules, and distal 173. Rampoldi L, Scolari F, Amoroso A, Ghiggeri G,
nephrons. Kidney Int. 2009;75:285–294. Devuyst O. The rediscovery of uromodulin (Tamm-
161. Hsu WL, Chiou HC, Tung KC, et al. The different Horsfall protein): from tubulointerstitial nephropa-
molecular forms of urine neutrophil gelatinase-asso- thy to chronic kidney disease. Kidney Int.
ciated lipocalin present in dogs with urinary diseases. 2011;80:338–347.
BMC Vet Res. 2014;10:202. 174. Raila J, Schweigert FJ, Kohn B. Relationship between
162. Cai L, Rubin J, Han W, Venge P, Xu S. The origin of urinary Tamm-Horsfall protein excretion and renal
multiple molecular forms in urine of HNL/NGAL. Clin function in dogs with naturally occurring renal dis-
J Am Soc Nephrol. 2010;5:2229–2235. ease. Vet Clin Pathol. 2014;43:261–265.
163. Bangert K, Rosenkilde N, Jorgensen JP, Baer A, Utten- 175. Mohamaden W, Wang H, Guan H, Meng X, Li J.
thal LO. Immunochemical Determination of Mono- Immunohistochemical localization and mRNA quan-
mer, Homodimer, and Total Neutrophil Gelatinase- tification of osteopontin and Tamm-Horsfall protein in
Associated Lipocalin [abstract]. American Society of canine renal tissue after potassium oxalate injection.
Nephrology, Kidney Week, 2012. BMC Vet Res. 2014;10:70.
164. Daure E, Belanger MC, Beauchamp G, Lapointe C. 176. Cooper TK, Zhong Q, Nabity M, Rosenberg G, Weiss
Elevation of neutrophil gelatinase-associated lipocalin WJ. Use of urinary biomarkers of renal ischemia in a
(NGAL) in non-azotemic dogs with urinary tract infec- lamb preclinical left ventricular assist device model.
tion. Res Vet Sci. 2013;95:1181–1185. Artif Organs. 2012;36:820–824.
165. Segev G, Palm C, LeRoy B, Cowgill LD, Westropp JL. 177. McDuffie JE, Sablad M, Ma J, Snook S. Urinary
Evaluation of neutrophil gelatinase-associated lipoca- parameters predictive of cisplatin-induced acute renal
lin as a marker of kidney injury in dogs. J Vet Intern injury in dogs. Cytokine. 2010;52:156–162.
Med. 2013;27:1362–1367. 178. Arata S, Ohmi A, Mizukoshi F, et al. Urinary trans-
166. Kai K, Yamaguchi T, Yoshimatsu Y, et al. Neutrophil forming growth factor-beta1 in feline chronic renal
gelatinase-associated lipocalin, a sensitive urinary bio- failure. J Vet Med Sci. 2005;67:1253–1255.
marker of acute kidney injury in dogs receiving gen- 179. Habenicht LM, Webb TL, Clauss LA, Dow SW, Quimby
tamicin. J Toxicol Sci. 2013;38:269–277. JM. Urinary cytokine levels in apparently healthy cats
167. Zhou X, Ma B, Lin Z, et al. Evaluation of the useful- and cats with chronic kidney disease. J Feline Med Surg.
ness of novel biomarkers for drug-induced acute kid- 2013;15:99–104.

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 53
Renal biomarkers review Hokamp and Nabity

180. Le Hir M, Dubach UC, Schmidt U. Quantitative distri- gamma-glutamyltranspeptidase in equine urine. Am J
bution of lysosomal hydrolases in the rat nephron. His- Vet Res. 1985;46:147–150.
tochemistry. 1979;63:245–251. 194. Heiene R, Moe L, Molmen G. Calculation of uri-
181. Guder WG, Ross BD. Enzyme distribution along the nary enzyme excretion, with renal structure and
nephron. Kidney Int. 1984;26:101–111. function in dogs with pyometra. Res Vet Sci. 2001;
182. Dance N, Price RG, Robinson D, Stirling JL. Beta- 70:129–137.
galactosidase, beta-glucosidase and N-acetyl-beta-glu- 195. Brunker JD, Ponzio NM, Payton ME. Indices of urine
cosaminidase in human kidney. Clin Chim Acta. N-acetyl-beta-D-glucosaminidase and gamma-gluta-
1969;24:189–197. myl transpeptidase activities in clinically normal adult
183. Dance N, Price RG, Cattell WR, Lansdell J, Richards B. dogs. Am J Vet Res. 2009;70:297–301.
The excretion of N-acetyl-beta-glucosaminidase and 196. Higashiyama N, Nishiyama S, Itoh T, Nakamura M.
beta-galactosidase by patients with renal disease. Clin Effect of castration on urinary N-acetyl-beta-D-gluco-
Chim Acta. 1970;27:87–92. saminidase levels in male beagles. Ren Physiol.
184. Price RG, Dance N, Richards B, Cattell WR. The excre- 1983;6:226–231.
tion of N-acetyl-beta-glucosaminidase and beta-galac- 197. Sato R, Soeta S, Miyazaki M, et al. Clinical availability
tosidase following surgery to the kidney. Clin Chim of urinary N-acetyl-beta-D-glucosaminidase index in
Acta. 1970;27:65–72. dogs with urinary diseases. J Vet Med Sci. 2002;64:361–
185. Sato R, Soeta S, Syuto B, et al. Urinary excretion of N- 365.
acetyl-beta-D-glucosaminidase and its isoenzymes in 198. Grauer GF, Greco DS, Behrend EN, et al. Estimation of
cats with urinary disease. J Vet Med Sci. 2002;64:367– quantitative enzymuria in dogs with gentamicin-
371. induced nephrotoxicosis using urine enzyme/crea-
186. Kuska J, Kokot F. The electrophoretic spectrum of tinine ratios from spot urine samples. J Vet Intern Med.
gamma-glutamyl transpeptidase (GGTP) in the bile 1995;9:324–327.
and urine. Arch Immunol Ther Exp (Warsz). 199. Uechi M, Uechi H, Nakayama T, et al. The circadian
1970;18:185–189. variation of urinary N-acetyl-beta-D-glucosaminidase
187. Pillion DJ, Jeske AH, Leibach FH. Gamma-glutamyl and gamma-glutamyl transpeptidase in clinically
transpeptidase in the urine from an isolated rabbit kid- healthy cats. J Vet Med Sci. 1998;60:1033–1034.
ney perfused with and without DMSO. Biochem Phar- 200. Mills PC, Auer DE, Kramer H, Barry D, Ng JC. Effects
macol. 1976;25:913–918. of inflammation-associated acute-phase response on
188. Butterworth PJ. Human kidney and urinary alkaline hepatic and renal indices in the horse. Aust Vet J.
phosphatases. Biochem J. 1968;107:467–472. 1998;76:187–194.
189. Kuzniar J, Marchewka Z, Lembas-Bogaczyk J, Kuzniar 201. Crivellenti LZ, Mesa JS, Meirelles AE, et al. False posi-
TJ, Klinger M. Etiology of increased enzymuria in dif- tivity of gamma-glutamyl transpeptidase measure-
ferent morphological forms of glomerulonephritis. ment in urine. Ren Fail. 2014;36:581–584.
Nephron Physiol. 2004;98:p8–p14. 202. Greco DS, Turnwald GH, Adams R, et al. Urinary
190. Jepson RE, Vallance C, Syme HM, Elliott J. Assess- gamma-glutamyl transpeptidase activity in dogs with
ment of urinary N-acetyl-beta-D-glucosaminidase gentamicin-induced nephrotoxicity. Am J Vet Res.
activity in geriatric cats with variable plasma crea- 1985;46:2332–2335.
tinine concentrations with and without azotemia. Am 203. Rivers BJ, Walter PA, O’Brien TD, King VL, Polzin DJ.
J Vet Res. 2010;71:241–247. Evaluation of urine gamma-glutamyl transpeptidase-
191. Reusch C, Vochezer R, Weschta E. Enzyme activities to-creatinine ratio as a diagnostic tool in an experi-
of urinary alanine aminopeptidase (AAP) and N- mental model of aminoglycoside-induced acute renal
acetyl-beta-D-glucosaminidase (NAG) in healthy failure in the dog. J Am Anim Hosp Assoc. 1996;32:323–
dogs. Zentralbl Veterinarmed A. 1991;38:90–98. 336.
192. Gossett KA, Turnwald GH, Kearney MT, Greco DS, 204. Rossier Y, Divers TJ, Sweeney RW. Variations in uri-
Cleghorn B. Evaluation of gamma-glutamyl nary gamma glutamyl transferase/urinary creatinine
transpeptidase-to-creatinine ratio from spot samples ratio in horses with or without pleuropneumonia trea-
of urine supernatant, as an indicator of urinary ted with gentamicin. Equine Vet J. 1995;27:217–220.
enzyme excretion in dogs. Am J Vet Res. 1987;48: 205. Fuentes VO, Gonzalez H, Sanchez V, Fuentes P,
455–457. Rosiles R. The effect of neomycin on the kidney func-
193. Adams R, McClure JJ, Gossett KA, Koonce KL, Ezigbo tion of the horse. Zentralbl Veterinarmed A.
C. Evaluation of a technique for measurement of 1997;44:201–205.

54 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology
Hokamp and Nabity Renal biomarkers review

206. Raekallio MR, Saario-Paunio EM, Rajamaki MM, 219. Jepson RE, Brodbelt D, Vallance C, Syme HM, Elliott
et al. Early detection of ketoprofen-induced acute kid- J. Evaluation of predictors of the development of azo-
ney injury in sheep as determined by evaluation of temia in cats. J Vet Intern Med. 2009;23:806–813.
urinary enzyme activities. Am J Vet Res. 2010;71:1246– 220. Miyazaki M, Kamiie K, Soeta S, Taira H, Yamashita T.
1252. Molecular cloning and characterization of a novel car-
207. Bayly WM, Brobst DF, Elfers RS, Reed SM. Serum boxylesterase-like protein that is physiologically pre-
and urinary biochemistry and enzyme changes in sent at high concentrations in the urine of domestic
ponies with acute renal failure. Cornell Vet. 1986; cats (Felis catus). Biochem J. 2003;370:101–110.
76:306–316. 221. Miyazaki M, Yamashita T, Hosokawa M, Taira H,
208. Narita T, Tomizawa N, Sato R, Goryo M, Hara S. Effects Suzuki A. Species-, sex-, and age-dependent urinary
of long-term oral administration of ketoprofen in clini- excretion of cauxin, a mammalian carboxylesterase.
cally healthy beagle dogs. J Vet Med Sci. 2005;67:847– Comp Biochem Physiol B Biochem Mol Biol.
853. 2006;145:270–277.
209. Borges M, Marini Filho R, Laposy CB, et al. Nons- 222. McLean L, Hurst JL, Gaskell CJ, Lewis JC, Beynon RJ.
teroidal anti-inflammatory therapy: changes on renal Characterization of cauxin in the urine of domestic
function of healthy dogs. Acta Cir Bras. 2013;28:842– and big cats. J Chem Ecol. 2007;33:1997–2009.
847. 223. Miyazaki M, Soeta S, Yamagishi N, et al. Tubulointer-
210. Ko JC, Miyabiyashi T, Mandsager RE, Heaton- stitial nephritis causes decreased renal expression and
Jones TG, Mauragis DF. Renal effects of carprofen urinary excretion of cauxin, a major urinary protein of
administered to healthy dogs anesthetized with the domestic cat. Res Vet Sci. 2007;82:76–79.
propofol and isoflurane. J Am Vet Med Assoc. 224. Jepson RE, Syme HM, Markwell P, et al. Measure-
2000;217:346–349. ment of urinary cauxin in geriatric cats with variable
211. Lobetti RG, Joubert KE. Effect of administration of plasma creatinine concentrations and proteinuria and
nonsteroidal anti-inflammatory drugs before surgery evaluation of urine cauxin-to-creatinine concentra-
on renal function in clinically normal dogs. Am J Vet tion ratio as a predictor of developing azotemia. Am J
Res. 2000;61:1501–1507. Vet Res. 2010;71:982–987.
212. Raekallio MR, Hielm-Bjorkman AK, Kejonen J, Salo- 225. Arosalo BM, Raekallio M, Rajamaki M, et al. Detect-
nen HM, Sankari SM. Evaluation of adverse effects of ing early kidney damage in horses with colic by mea-
long-term orally administered carprofen in dogs. J Am suring matrix metalloproteinase -9 and -2, other
Vet Med Assoc. 2006;228:876–880. enzymes, urinary glucose and total proteins. Acta Vet
213. de Schepper J, van der Stock J, Capiau E, de Cock I. Scand. 2007;49:4.
Renal injury in dogs with pyometra. Tijdschr Dierge- 226. Kaneko J, Harvey J, Bruss M. Clinical Biochemistry of
neeskd. 1987;112(Suppl. 1):124S–126S. Domestic Animals. 6th ed. Burlington, MA: Academic
214. de Schepper J, De Cock I, Capiau E. Urinary gamma- Press; 2008.
glutamyl transferase and the degree of renal dysfunc- 227. Almy FS, Christopher MM, King DP, Brown SA. Eval-
tion in 75 bitches with pyometra. Res Vet Sci. uation of cystatin C as an endogenous marker of
1989;46:396–400. glomerular filtration rate in dogs. J Vet Intern Med.
215. Heiene R, Biewenga WJ, Koeman JP. Urinary alkaline 2002;16:45–51.
phosphatase and y-glutamyl transferase as indicators 228. Jensen AL, Bomholt M, Moe L. Preliminary evalua-
of acute renal damage in dogs. J Small Anim Pract. tion of a particle-enhanced turbidimetric immunoas-
1991;32:521–524. say (PETIA) for the determination of serum cystatin C-
216. Lobetti R, Lambrechts N. Effects of general anesthesia like immunoreactivity in dogs. Vet Clin Pathol.
and surgery on renal function in healthy dogs. Am J 2001;30:86–90.
Vet Res. 2000;61:121–124. 229. Wehner A, Hartmann K, Hirschberger J. Utility of
217. Ulutas B, Sahal M. Urinary GGT/creatinine ratio and serum cystatin C as a clinical measure of renal func-
fractional excretion of electrolytes in diarrhoeic calves. tion in dogs. J Am Anim Hosp Assoc. 2008;44:131–138.
Acta Vet Hung. 2005;53:351–359. 230. Oikonomou KA, Kapsoritakis AN, Theodoridou C,
218. Lapointe C, Belanger MC, Dunn M, Moreau M, et al. Neutrophil gelatinase-associated lipocalin
Bedard C. N-acetyl-beta-D-glucosaminidase index as (NGAL) in inflammatory bowel disease: association
an early biomarker for chronic kidney disease in cats with pathophysiology of inflammation, established
with hyperthyroidism. J Vet Intern Med. 2008;22:1103– markers, and disease activity. J Gastroenterol.
1110. 2012;47:519–530.

Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology 55
Renal biomarkers review Hokamp and Nabity

231. Garcia-Martinez JD, Tvarijonaviciute A, Ceron JJ, Cal- 235. Rhodes DC, Hinsman EJ, Rhodes JA, Hawkins EC. Uri-
din M, Martinez-Subiela S. Urinary clusterin as a renal nary Tamm-Horsfall glycoprotein concentrations in
marker in dogs. J Vet Diagn Invest. 2012;24:301–306. normal and urolithiasis-affected male cats determined
232. Monti P, Benchekroun G, Berlato D, Archer J. Initial by an ELISA. Zentralbl Veterinarmed A. 1992;39:621–
evaluation of canine urinary cystatin C as a marker of 634.
renal tubular function. J Small Anim Pract. 236. Lobetti RG, Jacobson LS. Renal involvement in
2012;53:254–259. dogs with babesiosis. J S Afr Vet Assoc. 2001;72:23–
233. Paepe D, Ghys LF, Smets P, et al. Routine kidney vari- 28.
ables, glomerular filtration rate and urinary cystatin C 237. Brobst DF, Carroll RJ, Bayly WM. Urinary enzyme
in cats with diabetes mellitus, cats with chronic kidney concentrations in healthy horses. Cornell Vet.
disease and healthy cats. J Feline Med Surg. 1986;76:299–305.
2014;17:880–888. 238. Edwards DJ, Brownlow MA, Hutchins DR. Indices of
234. Bland SK, Cote O, Clark ME, DeLay J, Bienzle D. Char- renal function: reference values in normal horses. Aust
acterization of kidney injury molecule-1 in cats. J Vet Vet J. 1989;66:60–63.
Intern Med. 2014;28:1454–1464.

56 Vet Clin Pathol 45/1 (2016) 28–56 ©2016 American Society for Veterinary Clinical Pathology

You might also like