Sie sind auf Seite 1von 8

Clinica Chimica Acta 438 (2015) 350357

Contents lists available at ScienceDirect

Clinica Chimica Acta


journal homepage: www.elsevier.com/locate/clinchim

Invited critical review

Biomarkers of renal function, which and when?


Michael E. Wasung a, Lakhmir S. Chawla b, Magdalena Madero a,
a
Division of Nephrology, Department of Medicine, Instituto Nacional de Cardiologa Ignacio Chvez, Mexico City, Mexico
b
Department of Medicine, Division of Intensive Care Medicine, and the Division of Nephrology, Washington, DC, Veterans Affairs Medical Center, and the Department of Anesthesiology and Critical
Care Medicine, George Washington University

a r t i c l e i n f o a b s t r a c t

Article history: Purpose of the review: Acute kidney injury (AKI) and chronic kidney disease (CKD) are conditions that substantial-
Received 2 April 2014 ly increase morbidity and mortality. Although novel biomarkers are being used in practice, the diagnosis of AKI
Received in revised form 27 August 2014 and CKD is still made with surrogate markers of GFR, such as serum creatinine (SCr), urine output and creatinine
Accepted 29 August 2014 based estimating equations. SCr is limited as a marker of kidney dysfunction in both settings and may be inaccu-
Available online 3 September 2014
rate in several situations, such as in patients with low muscle mass or with uid overload. New biomarkers have
the potential to identify earlier patients with AKI and CKD and in the future potentially intervene to modify out-
Keywords:
Biomarkers
comes.
Acute kidney injury Recent ndings: In particular KIM-1 and NGAL are considered excellent biomarkers in urine and plasma for the
Chronic kidney disease early prediction of AKI; however cycle arrest biomarkers have emerged as novel markers for risk stratication
of AKI. Urine TIMP-2 and IGFBP7 performed better than any other biomarkers reported to date for predicting
the development of moderate or severe AKI. Biomarker combinations are required to increase diagnostic accura-
cy in an acute setting. NGAL, cystatin C, and FGF-23 are promising and accurate biomarkers for CKD detection.
Equations combining cystatin C and SCr perform better than the equations using either cystatin C or SCr alone,
especially in situations where CKD needs to be conrmed. Combining creatinine, cystatin C and urine albumin
to creatinine ratio improves risk stratication for kidney disease progression and mortality.
Summary: Recent advances in molecular biology have resulted in promising biomarkers for AKI and CKD diagnoses;
however more research is necessary to implement them successfully into clinical practice in order to facilitate early
diagnosis, guide interventions and monitor disease progression. The following review describes the most important
biomarkers studied in kidney disease and will discuss the use and the value of these biomarkers in different clinical
settings.
2014 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 351
2. Biomarkers in acute kidney injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 351
2.1. Biomarkers for the early diagnosis of AKI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 351
2.2. Prognosis of AKI severity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 352
2.3. Differential diagnosis of AKI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 353
2.4. Discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 353
3. Biomarkers in chronic kidney disease (CKD) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 353
3.1. Biomarkers for glomerular ltration rate (GFR) measurement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 353
3.2. Cardiovascular disease prediction in CKD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 354
3.3. CKD progression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 354
3.4. Inammatory and brotic markers associated with CKD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 354
3.5. Discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355

Abbreviations: AKI, acute kidney injury; CKD, chronic kidney disease; SCr, serum creatinine; KIM-1, kidney injury molecule-1; NGAL, neutrophil gelatinase-associated lipocalin; TIMP-2,
tissue inhibitor of metalloproteinases-2; IGFBP7, insulin-like growth factor-binding protein; FGF-23, broblast growth factor-23.
Corresponding author at: Juan Badiano No. 1, Tlalpan 14000 Mxico, D.F., Mexico. Tel.: +52 55 55 73 69 02; fax: +52 55 55 73 77 16.
E-mail address: madero.magdalena@gmail.com (M. Madero).

http://dx.doi.org/10.1016/j.cca.2014.08.039
0009-8981/ 2014 Elsevier B.V. All rights reserved.
M.E. Wasung et al. / Clinica Chimica Acta 438 (2015) 350357 351

1. Introduction is still poor. In-hospital mortality has not changed signicantly in the
past 50 years [1,13] and AKI is now considered an independent risk fac-
The incidence of acute kidney injury (AKI) and chronic kidney dis- tor for long-term mortality and for the future development of CKD [68,
ease (CKD) is increasing worldwide and it is associated with increased 14]. One possible reason for the poor outcome of severe AKI in the ICU
morbidity and mortality. Identifying patients early is of paramount im- may be delay in the diagnosis of AKI.
portance in order to offer a prompt intervention and to improve the The current practice to diagnose AKI involves measuring the surro-
prognosis in both settings. A biomarker is dened as a parameter of gate markers of glomerular ltration rate (GFR) such as an increase in
structural, biochemical, physiologic or genetic change that indicates SCr and/or a decrease in urine output with the RIFLE and/or AKIN
the presence, severity or progress of a disease [1]. The measurement criteria. However, the measurement of SCr is limited as a diagnostic
of kidney function is commonly made using serum creatinine concen- test in the early stages of AKI for several reasons. First, SCr can increase
tration (SCr), blood urea nitrogen (BUN) level and urinalysis; however in cases of prerenal azotemia when there is no tubular injury [15]. Sec-
accumulating evidence has shown that these biomarkers are subopti- ond, many non-renal factors can inuence its levels (body weight, mus-
mal to detect kidney disease in early stages [24]. cle metabolism, total body volume and drugs) [12]. Third, SCr in AKI
Kidney injury begins by inducing biological and molecular changes rises up to 72 h after the initial insult to the kidney [12]; fourth, renal in-
that ultimately evolve into cellular damage [5] allowing a wide array jury can exist with no change in SCr due to renal reserve or tubular se-
of molecules to be measured. Biomarker research has increased sub- cretion of creatinine [16]. Urine chemistry such as the fractional
stantially, and each year new potential markers are introduced. Even excretion rate of sodium or urea may differentiate prerenal azotemia
though there is better understanding of which biomarkers are useful from acute tubular necrosis (ATN) in selected patients; however, results
in kidney diseases there is still doubt toward when they should be may be confounded easily with diuretic use and volume status. Urine
measured. Before we begin our review, we outline the characteristics microscopy with evaluation of the urine sediment for renal tubular
of an ideal biomarker in Table 1. As we will see throughout the review, casts is helpful in differentiating prerenal azotemia from ATN [17]; how-
the presence of biomarkers in kidney have diagnostic and prognostic ever it requires expertise from the examiner.
implications in AKI and CKD, however there is currently not enough AKI is characterized by vascular and tubular alterations that express
data regarding the role of biomarkers on decision making. Clinical judg- proteins that can ultimately be measured in urine and in plasma. In AKI,
ment is still critical, biomarkers should not be measured indistinctly and cells produce biomarkers up to 3 days before the clinical syndrome de-
their measurement must be considered in situations that add value to velops [12] and therefore a biomarker should be measured in this peri-
current available markers. od. Table 2 outlines the most promising biomarkers. Biomarkers have
One of the most considerable limitations of biomarkers in kidney been tested in various AKI settings and in diverse populations; in the
disease is the discriminatory value between AKI and CKD. AKI and following section we will discuss some clinical situations wherein
CKD are not distinct syndromes, and actually share common grounds biomarker measurement has demonstrated to be useful.
[6]. Clinical studies have demonstrated that AKI increases the risk for
CKD development [7,8], and CKD has repeatedly shown to be a risk fac- 2.1. Biomarkers for the early diagnosis of AKI
tor for AKI [911]. Some mechanisms explaining CKD progression after
AKI include nephron loss, glomerular hypertrophy, interstitial inam- A considerable amount of evidence demonstrated that plasma and
mation and brosis, endothelial injury and cell cycle and maladaptive urine neutrophil gelatinase-associated lipocalin (NGAL), urinary cystatin
repair [6]. AKI and CKD share biomarkers, reecting the integrative C, urinary kidney injury molecule (KIM-1), urinary interleukin-18 (IL-
pathophysiology between both entities. 18), and glutathione-S-transferase are early diagnostic biomarkers of
We review the current status of the most important biomarkers in AKI in different settings. These biomarkers have been studied extensively
AKI and CKD and mention the possible settings in which their measure- in different situations such as in post-cardiac surgery patients, after con-
ment is useful. This review will rst address biomarkers in AKI and, sec- trast material administration and in kidney transplantation. Evidence
ond, biomarkers in CKD. Each section will discuss the use and the value points out that they are present approximately 2 days before the clinical
of biomarkers in different clinical settings. Tables 2 and 3 summarize syndrome of AKI develops [5,12]. Mishra and colleagues demonstrated
some of the most promising biomarkers in AKI and CKD. the clinical value of NGAL as a marker of kidney injury in 71 children un-
dergoing cardiopulmonary bypass [18]. The 2 hour NGAL levels in urine
2. Biomarkers in acute kidney injury and plasma were predictors of AKI, with an AUC of 0.998 for the urine
NGAL and 0.91 for the plasma NGAL. Further research, particularly in a
AKI is characterized by a sudden and sustained decrease of renal cohort of adults with a high prevalence of CKD did not have such impres-
function, resulting in retention of nitrogenous and non-nitrogenous sive results as the initial trials [19,20]. Although the follow-up studies
waste products [12]. The incidence of AKI requiring dialysis ranges failed to show the outstanding performance of the rst trials, the perfor-
from 4 to 6% in patients hospitalized in the intensive care unit (ICU) mance of NGAL and its elevation early in AKI remains very good. A meta-
with a mortality rate that varies from 60 to 90% depending on the sever- analysis revealed that in cardiac surgical patients, urinary NGAL had an
ity score of each patient [911]. Even though important advances have AUC of 0.775 when measured within 6 h of cardiopulmonary bypass
been made in critical care medicine and in renal replacement therapies [21].
(RRT), the outcome of critically ill patients with AKI that require dialysis The predictive value of NGAL has shown to be inuenced by several
factors, including baseline renal function, severity of AKI and age [22].
Table 1 NGAL is an acute phase reactant that is released from immune cells
Characteristics of an ideal biomarker. [23] and it is found to be elevated in inammatory conditions. In a
1. Noninvasive study performed by Wagener et al. in 426 adult patients submitted to
2. Highly sensitive and specic cardiac surgery, urinary NGAL levels were found to be elevated in all pa-
3. Increases rapidly and reliably in response to kidney disease. tients after the procedure. Urinary NGAL levels correlated signicantly
4. Correlates with the amount of kidney injury
with cardiopulmonary bypass and aortic cross-clamp times; raising
5. Provides risk stratication and prognostic information
6. Is site specic the possibility that CPB induced inammation led to increased levels
7. Applicable across different populations of NGAL [24]. NGAL levels are also inuenced by other medical condi-
8. Identies possible mechanisms of injury (prerenal, intrarenal, postrenal) tions such as preeclampsia and cancer [2527].
9. Highly stable over time and across different temperatures and PH Urinary NGAL, KIM-1, L-FABP, IL-18 and cystatin C seem to have less
10. Does no interfere with drugs
specicity and sensitivity in patients with comorbid conditions [28]. In a
352 M.E. Wasung et al. / Clinica Chimica Acta 438 (2015) 350357

Table 2
Examples of biomarkers used for the detection of AKI.

Legend Source Physiological action

Tubular enzymes released in the urine


Alpha glutathione S transferase GST Expressed in almost all tissues Released from lysosomes, brush-border and cytoplasm
Kidney: proximal tubular cells of proximal tubular epithelial cells
Pi glutathione S transferase GST Expressed in almost all tissues.
Kidney: distal tubular cells
N-acetyl beta glucosaminidase NAG Several tissues (liver, brain, spleen etc.).
Kidney: proximal tubular cells
Inammatory urinary markers
Interleukin 18 IL-18 Monocytes, dendritic cells, macrophages Filtered freely by the glomeruli, reabsorbed, non-secreted
Interleukin 1, 6, 8 IL-1, -6, -8 and epithelial cells
Low molecular proteins
Urinary cystatin C Cystatin C All nucleated cells Filtered freely by the glomeruli, reabsorbed, non-secreted
Hepatocyte Growth Factor HGF Mesenchymal cells
Alpha 1 microglobulin 1MG Liver
Beta 2 microglobulin 2MG All nucleated cells
Proximal tubule proteins
Kidney injury molecule 1 KIM-1 Kidney: proximal tubular cells Up-regulated in proximal tubular epithelial cells
Neutrophil gelatinase-associated lipocalin NGAL Leucocytes, loop of Henle and collecting ducts
Urinary cystatin C Cystatin C All nucleated cells
Liver-type fatty acid-binding protein L-FABP Hepatocytes, kidney: proximal tubular cells
Cell cycle arrest proteins
Insulin-like growth factor-binding protein 7 IGFBP7 Expressed in almost all epithelial cells Renal tubular cells enter a short period of G1 cell-cycle
Tissue inhibitor of metalloproteinases-2 TIMP-2 Kidney: tubular epithelial cells arrest following AKI.

prospective cohort of 529 patients conducted by Endre and colleagues, follow-up trial, which utilized the same enrolment criteria, TIMP-2
none of the biomarkers evaluated had an AUC above 0.7 [29]. The per- and IGFBP-7 were found to have similar performance. In this trial, AKI
formance of these biomarkers improved when adjustment was made experts in a blinded fashion clinically adjudicated the AKI endpoint.
with the baseline renal function and with the duration of renal injury. Both of these markers performed better than existing markers and the
Cycle arrest biomarkers have emerged as novel markers for risk operating characteristics are well suited for clinical use [31].
stratication of AKI. Kashani and colleagues studied two new bio-
markers, insulin-like growth factor-binding protein 7 (IGFBP7) and tis- 2.2. Prognosis of AKI severity
sue inhibitor of metalloproteinases-2 (TIMP-2), in the urine of patients
at high risk of AKI from multiple causes [30]. Both biomarkers are in- Several of the biomarkers mentioned previously performed well for
ducers of G1 cell-cycle arrest, considered as a key mechanism of AKI the risk stratication and prognosis of AKI. In children undergoing car-
and performed better than any other biomarkers reported to date. The diac surgery, plasma NGAL was a predictive biomarker of AKI and AKI
study consisted of a discovery and validation phase. In the discovery severity. Dent and associates measured plasma NGAL at baseline and
phase, blood and urine samples were obtained from three distinct co- at frequent intervals for 24 h after cardiopulmonary bypass (CPB) in a
horts to identify novel biomarkers for AKI among 340 proteins. The pediatric cohort [32]. Using multivariate analysis it was determined
best biomarkers were then compared in the validation phase to a num- that the plasma NGAL at 2 h after CPB was the most powerful predictor
ber of previously described biomarkers such as NGAL, KIM-1, and IL-18, of AKI, and correlated with the duration of AKI and the length of hospital
among others. Urinary IGFBP7 and TIMP-2 performed the best in both stay. The 12 hour plasma level of NGAL correlated with mortality.
phases. Measuring both biomarkers exhibited an AUC of 0.80 for the de- In the study performed by Liangos and colleagues, urinary KIM-1
velopment of AKI within 12 h and when measured alone, IGFBP7 and and N-acetyl--(D)-glucosaminidase (NAG) levels were able to predict
TIMP-2 exhibited an AUC of 0.76 and 0.79 respectively. Importantly, adverse clinical outcomes such as dialysis and mortality in a cohort of
neither TIMP-2 nor IGFBP-7 was elevated in patients without AKI and 201 patients with AKI of multiple causes [33]. The role of serum cystatin
important co-morbidities such as CKD, diabetes and sepsis. In a C to predict adverse clinical outcomes in AKI is limited; in the study

Table 3
Examples of biomarkers for detection of CKD.

Legend Source Physiological action

Amino acids Impaired clearance or increased production in CKD


Plasma asymmetric dimethylarginine ADMA Endothelial cells
Proteins FGF-23 Osteoblasts
Fibroblast growth factor 23
Chemokines MCP-1 All nucleated cells
Monocyte chemoattractant protein-1 Kidney cells
Tubular injury cell markers
Neutrophil gelatinase-associated lipocalin NGAL Leucocytes, loop of Henle and collecting ducts Released from lysosomes, brush-border and cytoplasm
Urinary cystatin C Cystatin C All nucleated cells of proximal tubular epithelial cells
Liver-type fatty acid-binding protein L-FABP Hepatocytes, kidney: proximal tubular cells
Urinary brosis markers
Connective tissue growth factor CTGF All tissues Excessive production of probrotic growth factors and
Transforming growth factor-1 TGF-1 All tissues extracellular matrix
Collagen IV COLIV Kidney, eye, skins
Glomerular cell injury markers
Plasma cystatin C Cystatin C All nucleated cells Impaired GFR
Podocalyxin PCX Podocytes Podocyte structure defect
Nephrin Nep Podocytes
M.E. Wasung et al. / Clinica Chimica Acta 438 (2015) 350357 353

done by Perianayagum et al. [34], serum cystatin C had an AUC of 0.65 2.4. Discussion
for prediction of death and dialysis in 200 patients with AKI, performing
similarly to the SCr level. Current evidence supports the concept that the measurement of cer-
In the study performed by Siew et al. [35] in 451 critically ill adult tain biomarkers for AKI facilitates early detection, differential diagnosis
patients, urinary IL-18 concentrations did not predict the development and prognosis of AKI. The diagnosis of AKI can be made 48 h earlier
of AKI, but did predict death and the need for short term dialysis. In the using biomarkers such as NGAL than with conventional assessments.
prospective study by Endre et al. [29], urinary NGAL, cystatin C and IL- However their usefulness is still limited in patients with preexisting
18 were the strongest predictors of dialysis (AUC N 0.7). All biomarkers renal disease and with comorbidities [22,23] in which the performance
except KIM-1 were moderately predictive of death within 7 days (AUC for predicting AKI in this setting is still poor.
0.610.69). Doi and colleagues found that the urinary levels L-FABP, Encouraging results have been obtained with cyclic arrest markers
NGAL and IL-18 were able to predict 14 day mortality (L-FABP AUC (IGFBP7 and TIMP-2) for early diagnosis and prognosis of AKI in com-
0.9, NGAL AUC 0.83, IL-18 AUC 0.83) and that the prediction improved parison to other markers. The AUCs of both biomarkers for severe AKI
when urinary L-FABP and NGAL levels were combined (AUC 0.93) prediction within 12 h of sample collection were signicantly higher
[36]. In the study performed by Kashani and colleagues [31], risk for than those of other biomarkers [30]. AKI research will be facilitated
major adverse kidney events (death, dialysis or persistent renal impair- with their use and cases of subclinical AKI may be uncovered. Future
ment) within 30 days elevated sharply for TIMP-2 and IGFBP7 above 0.3 studies will be necessary to guide interventions upon their positivity.
and doubled when values were N2.0. There is still a barrier for clinical translation regarding biomarkers in
Biomarkers are a useful aid for the prediction of renal recovery after AKI since there are not enough studies that have evaluated the impact of
AKI. Srisawat and colleagues conducted a multicenter, prospective ob- biomarker information on decision making. What shall we do when a
servational cohort in 76 patients that had AKI and required RRT in the biomarker results positive? Should we initiate dialysis? What does an
ICU [37]. Renal recovery was dened as being alive and free from dialy- intervention include? Do early interventions decrease hospital and
sis at 60 days from the start of RRT. Patients who recovered had a higher long term mortality in AKI? Future studies should answer these ques-
urinary cystatin C level on day 1, and lower uHGF on days 7 and 14. tions before biomarker panels are routinely measured. However, the
Levels of uNGAL and uHGF that decreased in the rst 14 days were asso- use of these biomarkers to identify patients at risk will give clinicians
ciated with greater odds of renal recovery. the opportunity to deploy the KDIGO consensus guidelines [46]. The im-
pact of the deployment of these guidelines in patients at high-risk for
AKI is unknown, but this represents the next frontier in the treatment
2.3. Differential diagnosis of AKI of AKI research. Simply put, do our consensus guidelines improve out-
comes? Future studies should address this issue with the aid of specic
The performance of NGAL and other biomarkers has been evaluated biomarkers.
in different populations with AKI. A single measurement of urinary It is still not clear whether biomarkers perform better than the clin-
NGAL in the emergency department was found to be highly specic ical expertise of physicians; however they should be regarded as a com-
(99%) and sensitive (90%) for AKI diagnosis [38]. Furthermore, urinary plement to a routine assessment and part of a decision tree. None of the
NGAL was able to distinguish patients with AKI from patients with biomarkers in AKI reviewed here can replace careful clinical reasoning.
CKD and prerenal conditions that had an elevated SCr. NGAL levels
have also been found to be useful in identifying other forms of AKI 3. Biomarkers in chronic kidney disease (CKD)
such as delayed graft function [39] and contrast induced nephropathy
[40]. CKD is characterized by a sustained reduction in GFR along with ev-
Parikh and colleagues measured urinary IL-18 in 72 patients in a idence of structural or functional abnormalities of the kidneys on urinal-
mixed population of renal conditions, including healthy subjects with ysis, biopsy and imaging [47]. CKD is associated with increased
no evidence of renal disease. The authors reported urinary IL-18 levels cardiovascular events, leading to a higher morbidity and mortality
to be signicantly higher in patients diagnosed with ATN than in pa- rate. Other important outcomes include infection, AKI, cognitive and
tients with prerenal azotemia or in healthy control subjects [41]. In physical impairment, and progression of kidney disease [48]. Premature
the study conducted by Siew and colleagues [32], urinary elevations of death can occur as a result of complications involved with CKD, espe-
IL-18 did not predict AKI, but did predict poor outcomes such as death cially in the presence of coexistent proteinuria [49]. Table 3 outlines
and the need for short term dialysis. Elevations in urine IL-18 is specic the most promising biomarkers for CKD. In the following section we
for ATN and is associated with a higher risk of adverse clinical outcomes. shall discuss which biomarkers are useful in CKD and when to consider
KIM-1 excretion in the urine is highly specic for kidney injury be- measuring them. Monitoring renal function in CKD requires biomarkers
cause no other organs have shown to express KIM-1 that could change that provide rapid, non-invasive and specic measurements that corre-
its urinary concentration. KIM-1 is a transmembrane glycoprotein that late well with kidney tissue pathology. Early identication of patients
is upregulated in animal models and in humans in the proximal tubule with CKD is important in order to perform early interventions and re-
cells after ischemic or toxic injury [42,43]. KIM-1 is expressed in kidney duce progression to kidney failure or cardiovascular events.
tissue obtained from biopsies from patients with ATN [44], and is effec-
tive at distinguishing ATN from prerenal azotemia and CKD. In adults 3.1. Biomarkers for glomerular ltration rate (GFR) measurement
submitted to cardiac surgery, urinary KIM-1 achieved the highest AUC
in comparison to other biomarkers (NAG, NGAL, IL-18, cystatin C and Evidence suggests that GFR is the best index of renal function in CKD
1 microglobulin). The AUC for KIM-1 to predict AKI 2 h postoperative- [50] and it is measured as the clearance rate of a ltration marker from
ly was 0.74 (95% CI = 0.640.91) followed by IL-18 and NAG [45]. plasma by the kidneys. Normal values are approximately 130 ml/min/
The Topaz study [31] had the objective of validating the ability of uri- 1.73 m2 in young men and 120 ml/min/1.73 m2 in young women and
nary levels of TIMP-2 and IGFBP7 in identifying patients at high risk for values decline as people age [49]. Urine isotope collections (inulin,
development of moderate to severe AKI within 12 h, using a cutoff value iothalamate, EDTA and iohexol) are considered as gold standards for
of more than 0.3 (ng/ml)2/1000. Critically ill patients with a single mea- measuring GFR, however they are impractical, expensive, time consum-
surement of urinary TIMP-2 and IGFBP7 above this level carried a higher ing and invasive in the clinical setting. For this reason, GFR is estimated
risk of developing AKI of multiple causes. Measuring these markers was with equations that take into account endogenous ltration markers
helpful for distinguishing true AKI in a diverse group of patients in the such as SCr and demographic factors. Creatinine generation is affected
intensive care unit. by age, sex, race and body weight independently from GFR [51]. GFR
354 M.E. Wasung et al. / Clinica Chimica Acta 438 (2015) 350357

estimating equations appear to be a more accurate estimate of GFR than inuence the progression of atherosclerosis and carotid stenosis [71,72].
SCr alone [5255]; however they cannot account for individual differ- Asymmetric dimethylarginine (ADMA) is a nitric oxide synthase (NOS)
ences in muscle mass for a given age, sex or race. Table 4 outlines the inhibitor [73,74] that plays an important role in the vascular tone. The
various validated equations for GFR estimation. Muscle wasting is com- levels of ADMA are particularly high in patients with endothelial dys-
mon in CKD and it is associated with lower creatinine generation there- function, hypertension and proteinuria. ADMA levels increase as kidney
fore, providing bias toward a higher eGFR. function declines because of a reduced clearance [74] and is found to be
Cystatin C is a 13-kDa protein synthesized at a constant rate in all nu- associated with several cardiovascular events. ADMA is considered as
cleated cells [56]. It is freely ltered in the glomerulus and is reabsorbed the missing link between cardiovascular disease and CKD [73] by
and catabolized completely in the proximal tubule with a lack of tubular some authors, and is a strong biomarker that predicts a higher mortality
secretion [56]. Cystatin C is less affected by muscle mass than SCr, and is in CKD [7577] as well as a faster progression of kidney injury [78].
considered to be a better marker of early kidney dysfunction [5761] Counteracting the effects of ADMA in CKD should be explored as a strat-
and a more reliable marker of renal function. In a study of 164 patients egy to prevent cardio-renal complications.
with stage 2 or 3 CKD (GFR of 30 to 89 ml/min/1.73 m2), serum cystatin
C had a higher accuracy in detecting a GFR of less than 60 ml/min/ 3.3. CKD progression
1.73 m2 than SCr, but only in women [61]. Investigators from CKD-EPI
developed 3 GFR estimating equations for cystatin C: using cystatin C NGAL is expressed by tubular epithelial cells in response to injury
alone, cystatin C with demographic factors, and cystatin C with creati- and tubulointerstitial damage that is a common pathway in the progres-
nine and demographic factors. The equation that included cystatin C, sion of most forms of kidney disease [79]. NGAL has performed well as a
creatinine and demographic coefcients provided the most accurate biomarker in CKD in several small studies. In one of them performed in
GFR estimates [62]. 45 children with CKD stages 24, plasma NGAL levels were inversely as-
Inker and colleagues [63] established an international reference sociated with GFR [80]. As kidney function declined under 30 ml/min,
standard for cystatin C to develop a GFR estimating equation based on NGAL outperformed cystatin C as a biomarker of kidney function.
5352 subjects from 13 cohorts. The equations were than validated in Smith and colleagues [81] conducted a prospective observational cohort
1119 participants from 5 different studies. Two new equations that in- in 158 adult patients with stage 3 or 4 CKD. Urinary NGAL to creatinine
volved cystatin C alone and in combination with creatinine were devel- ratio (uNCR) was associated with a higher risk of death and initiation of
oped. The 2012 CKD-EPI cystatin C and 2009 CKD-EPI creatinine-based renal replacement therapy independent of renal and cardiovascular risk
equations had similar performances in this setting for the outcome of factors. The authors concluded that measuring urinary uNCR improves
measured GFR. The best GFR estimation resulted from the combined the prediction of renal progression in this population. Plasma NGAL
creatininecystatin C equation. Near the CKD threshold (GFR of 45 levels also predict progression of CKD in adults. The study performed
75 ml/min/1.73/m2), the combined equation meaningfully improved by Bolignano and colleagues [82] demonstrated that plasma and urinary
staging of CKD (GFR b 60 ml/min/1.73/m2) in comparison to the 2009 NGAL had a predictive power for CKD progression even after adjust-
creatinine equation. The combined creatininecystatin C equation ap- ment for eGFR after an 18.5 month follow-up in a cohort of 96 patients.
pears to be the optimal GFR estimate. NGAL was an independent predictor of CKD progression and reected
the severity of renal disease.
3.2. Cardiovascular disease prediction in CKD Warnock and associates evaluated whether combining creatinine,
cystatin C, and urine albumin to creatinine ratio would improve identi-
Cystatin C levels have been associated with adverse clinical events. cation of risks associated with CKD compared with creatinine alone, in
In prior studies in the general population and in the elderly, cystatin C a prospective cohort study involving 26,643 adults enrolled in the Rea-
has been shown to be a better predictor of mortality and adverse cardio- sons for Geographic and Racial Differences in Stroke (REGARDS) study
vascular events than SCr [6467]. Cystatin C levels are also associated [83]. In this study, the adjusted risk of mortality was threefold higher
with a higher rate of non-cardiovascular disease outcomes such as in patients with CKD dened by both GFR cystatin and GFR creatinine
pulmonary disease, cancer and infection [68]. Endothelial nitric oxide than in those with CKD dened by GFR creatinine alone and approxi-
synthase converts the amino acid L-arginine into L-citrulline and nitric mately sixfold higher in patients with CKD identied by all three
oxide [69]. Nitric oxide (NO) is considered an important vasodilator markers. The authors concluded that adding cystatin C to the measures
that also inhibits several processes involved in vascular diseases, such with SCr and albumincreatinine ratio improved the accuracy for all-
as leucocyte adhesion, platelet aggregation and vascular smooth muscle cause mortality and end-stage renal disease prediction.
cell proliferation [70]. An alteration in vascular NO synthesis is known to Disturbances of mineral metabolism are common in CKD. As kidney
function declines phosphate is retained by the kidney, renal calcitriol
production declines and as a result calcium tends to decrease. FGF-23
Table 4
Estimating equations for glomerular ltration rate. MDRD and CKD-EPI equations incorpo- is a 32-kDa protein secreted by osteoblasts primarily that plays an im-
rating creatinine, cystatin C or both. portant role in controlling phosphate concentration. It acts as an endo-
crine hormone by inhibiting parathyroid hormone secretion, decreasing
MDRD
GFR (ml/min/1.73 m2) = 175 (SCr)1.154 (age)0.203 (0.742 levels of calcitriol, and by inducing phosphaturia [84,85]. Circulating
if female) (1.212 if African American) levels increase progressively as kidney function decreases below
2009 CKD-EPI creatinine 60 ml/min/1.73 m2 [86,87]. A considerable amount of evidence has
GFR = 141 min (SCr / , 1) max(SCr / , 1)1.209 0.993age 1.018 shown that an elevated level of FGF-23 is an independent risk factor
[if female] 1.159 [if Black]
for adverse outcomes in CKD such as a faster progression, higher inci-
2012 CKD-EPI cystatin C
eGFR = 133 min(Scys / 0.8, 1)0.499 max(Scys / 0.8, 1)1.328 0.996age dence of cardiovascular disease and increased mortality [88,89].
[0.932 if female]
2012 CKD-EPI creatinine and cystatin C 3.4. Inammatory and brotic markers associated with CKD
eGFR = 135 min(SCr / k, 1)a max(SCr / k, 1)0.601 min(Scys / 0.8,
1)0.375 max(Scys / 0.8, 1)0.711 0.995age [0.969 if female] [1.08 if
Black] Monocyte chemoattractant protein-1 (MCP-1) is a chemotactic pro-
tein that is expressed by a variety of cells, including endothelial cells, -
SCr is serum creatinine and Scys is serum cystatin C; k is 0.7 for females and 0.9 for males; a
is 0.248 for females and 0.207 for males.
broblasts, mononuclear cells and kidney cells upon a proinammatory
MDRD, modication of diet in renal disease equation. stimulus [9092]. Expression of MCP-1 is up-regulated in kidney dis-
CKD-EPI, chronic kidney disease epidemiology collaboration equation. eases that have a sustained inammatory response, such as in diabetic
M.E. Wasung et al. / Clinica Chimica Acta 438 (2015) 350357 355

nephropathy and lupus [93,94]. Studies have demonstrated that early diagnosis and prognosis of AKI. NGAL is a very useful marker in
podocytes and tubular cells produce MCP-1 in response to high glucose AKI diagnosis; however it does not perform well in patients with pre-
levels and advanced glycosylation end products and that urinary levels existing renal failure. Renal recovery is an interesting topic in AKI
of MCP-1 are signicantly higher in patients with diabetic nephropathy research, and decreasing levels of urinary NGAL and urinary HGF demon-
[95,96]. Urinary levels of MCP-1 are also elevated in patients with lupus strated to be useful in predicting recovery in different populations.
nephritis reecting intrarenal expression [94,97]. Serum concentrations During the last century, SCr has been the most used biomarker to
of MCP-1 are also high in patients with diabetic nephropathy and lupus screen and diagnose kidney disease. Creatinine however has several
nephritis; however, the levels of serum MCP-1 do not correlate with the limitations and should be utilized only in estimating equations
degree of renal activity and damage in both entities [94,98]. (Table 3). The CKD-EPI seems to be more generalizable and accurate
Connective tissue growth factor (CTGF) is a member of the CCN fam- than the MDRD estimating equation. More recently the GFR estimating
ily of matricellular proteins, which is implicated in many cellular func- equation combining SCr and cystatin C performed better than equations
tions, which include cell proliferation, adhesion and wound healing that used either SCr or cystatin C alone and is recommended in specic
[99102]. CTGF is considered an important mediator of tissue brosis conditions, such as when conrmation of CKD is required [108]. Cystatin
and several brotic conditions such as liver brosis and scleroderma C is a valuable biomarker of kidney function in CKD, kidney disease pro-
have been found to overexpress it [103105]. Renal diseases that ex- gression and cardiovascular disease prediction. Emerging biomarkers
press higher levels of CTGF levels include diabetic nephropathy, focal for CKD (NGAL, ADMA, MCP-1, CTGF) can serve as a complement in rou-
and segmental glomerulosclerosis and IgA nephropathy [106,107]. tine assessments in CKD patients.
CTGF has been found to be an independent predictor of ESRD and corre- Biomarkers in AKI and CKD share similarities; both entities have a
lates well with the rate of decline of GFR in diabetic nephropathy [107, reduction in nephron number, vascular insufciency and cell cycle dis-
108]. ruption. For this reason biomarkers such as NGAL, KIM-1 and cystatin-
C are present in both. Kidney diseases are complex, have multiple
3.5. Discussion causes, and may accompany systemic diseases and CKD and AKI may co-
exist; therefore a single biomarker is unlikely to meet all the expecta-
Biomarkers in CKD help us estimate GFR, assess cardiovascular dis- tions and biomarker panels will be necessary. Biomarkers should not
ease, determine metabolic abnormalities associated with CKD and dif- replace clinical assessments of patients with CKD and AKI and should
ferentiate inammatory and brotic conditions of the kidney. The be considered a complement to clinical reasoning. Indiscriminate use
pathophysiology of CKD is complex and involves multiple processes in of biomarker panels can distract physicians from taking adequate deci-
its progression and in some cases an underlying primary renal disease sions affecting outcomes.
is involved. Comorbidity is present in practically all patients with CKD, In conclusion, relevant biomarkers have been developed to diag-
therefore a single biomarker is insufcient to meet all of these nose, predict outcome and stratify risk patients with AKI and CKD. It is
expectations. important to know which biomarkers are useful and when can they
Cystatin C is a very useful biomarker in CKD and used for GFR estima- be measured. Currently, the use of biomarkers for decision making is
tion and for cardiovascular disease assessment. The value of estimating not dened; and still measured bench-work and not bed-side. Al-
GFR with cystatin C over conventional assessments is to improve the though some of the biomarkers are highly sensitive and specic, larger,
specicity of CKD diagnosis. KDIGO guidelines [108] recommend multicenter and long term trials are still necessary to make translation
cystatin C measurement in patients in which CKD would be missed and eventually make decisions upon their positivity.
when using eGFR with SCr alone. Cystatin C based GFR equations are
more accurate than the CKDEPI equation in certain subgroups of pa-
References
tients in which the SCr level may be insensitive to capture reduced kid-
ney function such as hospitalized patients, patients with chronic liver [1] Schif H, Lang SM. Update on biomarkers of acute kidney injury: moving closer to
disease and cirrhosis, frail elders, and patients with HIV infection and clinical impact? Mol Diagn Ther 2012;16(4):199207 [1].
[2] Bjornsson TD. Use of serum creatinine concentrations to determine renal function.
malignancy. Cystatin C is less inuenced by muscle mass than creatinine
Clin Pharmacokinet 1979;4(3):20022.
and may reect a better marker of GFR in these populations. Cystatin C [3] Bosch JP, Saccaggi A, Lauer A, Ronco C, Belledonne M, Glabman S. Renal functional
levels however are affected by inammatory conditions that present reserve in humans. Effect of protein intake on glomerular ltration rate. Am J Med
1983;75(6):94350.
high levels of C reactive protein [57], thyroid disease, glucocorticoid
[4] Waikar SS, Betensky RA, Bonventre JV. Creatinine as the gold standard for kidney
use and obesity, and this should be implemented with caution in injury biomarker studies? Nephrol Dial Transplant 2009;24(11):32635.
these conditions. Combining creatinine, cystatin C and urine albumin [5] Bellomo R, Kellum JA, Ronco C. Dening acute renal failure: physiological princi-
to creatinine ratio improves risk stratication and helps predict progres- ples. Intensive Care Med 2004;30(1):337.
[6] Chawla L, Kimmel P. Acute kidney injury and chronic kidney disease: an integrated
sion to end stage kidney disease and mortality. clinical syndrome. Kidney Int 2012;82:51624.
FGF-23 is an accurate marker of CKD metabolic bone disease and [7] Ishani A, Xue JL, HImmelfarb J, et al. Acute kidney injury increases risk of ESRD
may serve as a complement of CKD assessment. FGF-23 as well as among elderly. J Am Soc Nephrol 2009;20:2238.
[8] Lj Lo, Go AS, Chertow GM, et al. Dialysis-requiring acute renal failure increases the
other biomarkers such as NGAL, ADMA, MCP-1 and CTGF are emerging risk of progressive chronic kidney disease. Kidney Int 2009;76:108997.
biomarkers that may help predict progression and stratify risk in CKD, [9] Metnitz PG, Krenn CG, Steltzer H, et al. Effect of acute renal failure requiring renal
however more research is needed in the eld before these can be imple- replacement therapy on outcome in critically ill patients. Crit Care Med 2002;30:
20518.
mented in clinical practice. [10] Uchino S, Kellum JA, Bellomo R, et al. Acute renal failure in critically ill patients: a
multinational, multicenter study. JAMA 2005;294:8138.
4. Conclusion [11] Mehta R, Pascual M, Soroko S, et al. Spectrum of acute renal failure in the intensive
care unit: The PICARD experience. Kidney Int 2004;66:161321.
[12] Soni S, Ronco C, Katz N, Cruz D. Early diagnosis of acute kidney injury: the promise
Biomarker research has increased signicantly in the past ten years of novel biomarkers. Blood Purif 2009;28:16574.
with the purpose of allowing nephrologists to identify kidney injury ear- [13] Dennen P, Douglas IS, Anderson R. Acute kidney injury in the intensive care unit:
an update and primer for the intensivist. Crit Care Med 2010;38(1):26175.
lier. SCr, eGFR and proteinuria are not sensitive enough, and identify kid-
[14] Coca SG, Parikh CR. Urinary biomarkers for acute kidney injury: perspectives on
ney diseases late when valuable interventions can be applied. Biomarkers translation. Clin J Am Soc Nephrol 2008;3(2):48190.
may revolutionize the diagnosis of AKI and CKD, however they must be [15] Schrier R, Wang W, Poole B, Mitra A. Acute renal failure: denitions, diagnosis,
validated in larger populations with comorbidities and with hard clinical pathogenesis and therapy. J Clin Invest 2004;114(1):514.
[16] Herrera J, Rodrguez-Iturbe B. Stimulation of tubular secretion of creatinine in
end points. Of those reviewed, cycle arrest markers appear as the most health and in conditions associated with reduced nephron mass. Evidence for a
promising for AKI, performing better than other existing biomarkers for tubular functional reserve. Nephrol Dial Transplant 1998;13:6239.
356 M.E. Wasung et al. / Clinica Chimica Acta 438 (2015) 350357

[17] Perazella M, Coca S, Hall I, Iyanam U, Koraishy M, Parikh C. Urine microscopy is as- [49] Astor BC, Matsushita K, Gansevoort RT, et al. Lower estimated glomerular ltration
sociated with severity and worsening of acute kidney injury in hospitalized pa- rate and higher albuminuria are associated with mortality and end-stage renal dis-
tients. Clin J Am Soc Nephrol 2010;5:4028. ease. A collaborative meta-analysis of kidney disease population cohorts. Kidney
[18] Mishra J, Dent C, Tarabishi R, et al. Neutrophil gelatinase-associated lipocalin Int 2011;79(12):133140.
(NGAL) as a biomarker for acute renal injury after cardiac surgery. Lancet 2005; [50] Rahman M, Smith MC. Chronic renal insufciency: a diagnostic and therapeutic ap-
365:12318. proach. Arch Intern Med 1998;158(16):174352.
[19] Zappitelli M, Washburn KK, Arikan AA, et al. Urine neutrophil gelatinase associated [51] Stevens LA, Coresh J, Greene T, Levey AS. Assessing kidney functionmeasured and
lipocalin is an early marker of acute kidney injury in critically ill children: a pro- estimated glomerular ltration rate. N Engl J Med 2006;354(23):247383.
spective cohort study. Crit Care 2007;11:R84. [52] Madero M, Sarnak MJ. Creatinine-based formulae for estimating glomerular ltra-
[20] Wheeler DS, Devarajan P, Ma Q, et al. Serum neutrophil gelatinase-associated tion rate: is it time to change to chronic kidney disease epidemiology collaboration
lipocalin (NGAL) as a marker of acute kidney injury in critically ill children with equation? Curr Opin Nephrol Hypertens 2011 Nov;20(6):62230.
septic shock. Crit Care Med 2008;36:1297303. [53] Stevens LA, Levey AS. Use of the MDRD study equation to estimate kidney function
[21] Haase M, Bellomo R, Devarajan P, et al. Accuracy of neutrophil gelatinase associated for drug dosing. Clin Pharmacol Ther 2009;86:4657.
lipocalin (NGAL) in diagnosis and prognosis in acute kidney injury: a systematic re- [54] Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine.
view and meta-analysis. Am J Kidney Dis 2009;54(6):101224. Nephron 1976;16:3141.
[22] McIlroy DR, Wagener G, Lee HT. Neutrophil gelatinase-associated lipocalin and [55] Levey AS, Stevens LA. Estimating GFR using the CKD epidemiology collaboration
acute kidney injury after cardiac surgery: the effect of baseline renal function on di- (CKD-EPI) creatinine equation: more accurate GFR estimates, lower CKD preva-
agnostic performance. Clin J Am Soc Nephrol 2010;5(2):2119. lence estimates, and better risk predictions. Am J Kidney Dis 2010;55:6227.
[23] Schmidt-Ott K, Mori K, Yi Li J, et al. Dual action of neutrophil gelatinase-associated [56] Abrahamson M, Dalboge H, Olafsson I, Carlsen S, Grubb A. Efcient production of
lipocalin. J Am Soc Nephrol 2007;18:40713. native, biologically active human cystatin C by Escherichia coli. FEBS Lett 1988;
[24] Wagener G, Gubitosa G, Wang S, Borregaard N, Kim M, Lee T. Urinary neutrophil 236:148.
gelatinase-associated lipocalin and acute kidney injury after cardiac surgery. Am J [57] Taglieri N, Koenig W, Kaski JC. Cystatin C and cardiovascular risk. Clin Chem 2009;
Kidney Dis 2008;52:42533. 55(11):193243.
[25] Smith ER, Zurakowski D, Saad A, Scott RM, Moses MA. Urinary biomarkers predict [58] Dharnidharka VR, Kwon C, Stevens G. Serum cystatin C is superior to serum creat-
brain tumor presence and response to therapy. Clin Cancer Res 2008;14:237886. inine as a marker of kidney function: a meta-analysis. Am J Kidney Dis 2002;40(2):
[26] Manfredi MA, Zurakowski D, Rufo PA, Walker TR, Fox VL, Moses MA. Increased 2216.
incidence of urinary matrix metalloproteinases as predictors of disease in [59] Van Deventer HE, Paiker JE, Katz IJ, George JA. A comparison of cystatin C-and cre-
pediatric patients with inammatory bowel disease. Inamm Bowel Dis atinine-based prediction equations for the estimation of glomerular ltration rate
2008;14:10916. in black South Africans. Nephrol Dial Transplant 2011;26(5):15538.
[27] DAnna R, Baviera G, Giordano D, Todarello G, Corrado F, Buemi M. Second trimes- [60] Shlipak MG, Mattes MD, Peralta CA. Update on cystatin C: incorporation into clin-
ter neutrophil gelatinase-associated lipocalin as a potential prediagnostic marker of ical practice. Am J Kidney Dis 2013;62(3):595603.
preeclampsia. Acta Obstet Gynecol Scand 2008;87:13703. [61] Hojs R, Bevc S, Ekart R, Gorenjak M, Puklavec L. Serum cystatin C as an endogenous
[28] Lameire N, Vanholder R, Van Biesen W. How to use biomarkers efciently in acute marker of renal function in patients with mild to moderate impairment of kidney
kidney injury. Kidney Int 2011;79:104750. function. Nephrol Dial Transplant 2006;21(7):185562.
[29] Endre Z, Pickering J, Walker R, et al. Improved performance of urinary biomarkers [62] Stevens LA, Coresh J, Schmid CH, et al. Estimating GFR using serum cystatin C alone
of acute kidney injury in the critically ill by stratication for injury duration and and in combination with serum creatinine: a pooled analysis of 3,418 individuals
baseline renal function. Kidney Int 2011;79:111930. with CKD. Am J Kidney Dis 2008;51:395406.
[30] Kashani K, Al-Khafaji A, Ardiles T, et al. Discovery and validation of cell cycle arrest [63] Inker LA, Schmid CH, Tighiouart H, et al. Estimating glomerular ltration rate from
biomarkers in human acute kidney injury. Crit Care 2013;17:R25. serum creatinine and cystatin C. N Engl J Med 2012;367(1):209.
[31] Bihorac A, Chawla L, Shaw A, et al. Validation of cell-cycle arrest biomarkers for [64] Shlipak MG, Katz R, Sarnak MJ, et al. Cystatin C and prognosis for cardiovascular
acute kidney injury using clinical adjudication. Am J Respir Crit Care Med 2014; and kidney outcomes in elderly persons without chronic kidney disease. Ann In-
189(8):9329. tern Med 2006;145(4):23746.
[32] Dent C, Ma Q, Dastrala S, et al. Plasma neutrophil gelatinase-associated lipocalin [65] Shlipak MG, Sarnak MJ, Katz R, et al. Cystatin C and the risk of death and cardiovas-
predicts acute kidney injury, morbidity and mortality after pediatric cardiac sur- cular events among elderly persons. N Engl J Med 2005;352(20):204960.
gery: a prospective uncontrolled cohort study. Crit Care 2007;11(6):R127. [66] Sarnak MJ, Katz R, Stehman-Breen CO, et al. Cystatin C concentration as a risk factor
[33] Liangos O, Perianayagam MC, Vaidya VS, et al. Urinary N-acetyl-beta-(D) for heart failure in older adults. Ann Intern Med 2005;142(7):497505.
glucosaminidase activity and kidney injury molecule-1 level are associated with [67] Madero M, Wassel CL, Peralta CA, et al. Cystatin C associates with arterial stiffness
adverse outcomes in acute renal failure. J Am Soc Nephrol 2007;18:90412. in older adults. J Am Soc Nephrol 2009;20(5):108693.
[34] Perianayagum MC, Seabra VF, Tighiouart H, Liangos O, Jaber BL. Serum cystatin C [68] Fried LF, Katz R, Sarnak MJ, et al. Kidney function as a predictor of
for prediction of dialysis requirement or death in acute kidney injury: a compara- noncardiovascular mortality. J Am Soc Nephrol 2005;16(12):372835.
tive study. Am J Kidney Dis 2009;54(6):102533. [69] Cooke JP, Dzau VJ. Nitric oxide synthase: role in the genesis of vascular disease.
[35] Siew E, Ikizler T, Gebretsadik T, et al. Elevated urinary IL-18 levels at the time of ICU Annu Rev Med 1997;48:489509.
admission predict adverse clinical outcomes. Clin J Am Soc Nephrol 2010;5: [70] von der Leyen HE, Gibbons GH, Morishita R, et al. Gene therapy inhibiting neointi-
1497505. mal vascular lesion: in vivo transfer of endothelial cell nitric oxide synthase gene.
[36] Doi K, Negishi K, Ishizu T, et al. Evaluation of new acute kidney injury biomarkers in Proc Natl Acad Sci U S A 1995;92:113741.
a mixed intensive care unit. Crit Care Med 2011;39(11):24649. [71] Cooke JP, Singer AH, Tsao P, et al. Anti-atherogenic effects of L-arginine in the hy-
[37] Srisawat N, Wen X, Lee M, et al. Urinary biomarkers and renal recover in critically percholesterolemic rabbit. J Clin Invest 1992;90:116872.
ill patients with renal support. Clin J Am Soc Nephrol 2011;6:181523. [72] Candipan RC, Wang B, Buitrago R, et al. Regression or progression: dependency
[38] Nickolas TL, ORourke MJ, Yang J, et al. Sensitivity and specicity of a single emer- upon vascular nitric oxide. Arterioscler Thromb Vasc Biol 1996;16:445.
gency department measurement of urinary neutrophil gelatinase-associated [73] Ueda S, Yamagishi S, Kaida Y, Okuda S. Asymmetric dimethylarginine may be a
lipocalin for diagnosing acute kidney injury. Ann Intern Med 2008;148:8109. missing link between cardiovascular disease and chronic kidney disease. Nephrol-
[39] Parikh CR, Jani A, Mishra J, et al. Urine NGAL and IL-18 are predictive biomarkers for ogy 2007;12(6):58290.
delayed graft function following kidney transplantation. Am J Transplant 2006 Jul; [74] Ueda S, Yamagishi S, Okuda S. New pathways to renal damage: role of ADMA in
6(7):163945. retarding renal disease progression. J Nephrol 2010;23(4):37786.
[40] Hirsch R, Dent C, Pfriem H, et al. NGAL is an early predictive biomarker of contrast- [75] Vallance P, Leone A, Calver A, Collier J, Moncada S. Accumulation of an endogenous
induced nephropathy in children. Pediatr Nephrol 2007;22(12):208995. inhibitor of nitric oxide synthesis in chronic renal failure. Lancet 1992;339(8793):
[41] Parikh CR, Jani A, Melnikov VY, Faubel S, Edelstein CL. Urinary interleukin-18 is a 5725.
marker of human acute tubular necrosis. Am J Kidney Dis 2004;43(3):40514. [76] Miyazaki H, Matsuoka H, Cooke JP, et al. Endogenous nitric oxide synthase inhibi-
[42] Ichimura T, Hung CC, Yang SA, Stevens JL, Bonventre JV. Kidney injury molecule-1: tor: a novel marker of atherosclerosis. Circulation 1999;99(9):11416.
a tissue and urinary biomarker for nephrotoxicant-induced renal injury. Am J Phys- [77] Tripepi G, Mattace Raso F, Sijbrands E, et al. Inammation and asymmetric
iol Renal Physiol 2004;286(3):F55263. dimethylarginine for predicting death and cardiovascular events in ESRD patients.
[43] Zhang Z, Humphreys BD, Bonventre JV. Shedding of the urinary biomarker kidney Clin J Am Soc Nephrol 2011;6(7):171421.
injury molecule-1 (KIM-1) is regulated by MAP kinases and juxtamembrane re- [78] Fliser D, Kronemberg F, Kielstein J, et al. Asymmetric dimethylarginine and pro-
gion. J Am Soc Nephrol 2007;18:270414. gression of chronic kidney disease: the mild to moderate kidney disease study. J
[44] Han WK, Bailly V, Abichandani R, et al. Kidney injury molecule-1 (KIM-1): a novel Am Soc Nephrol 2005;16(8):245661.
biomarker for human renal proximal tubule injury. Kidney Int 2002;62:23744. [79] Kuncio GS, Neilson EG, Haverty T. Mechanisms of tubulointerstitial brosis. Kidney
[45] Liangos O, Tighiouart H, Perianayagam M, et al. Comparative analysis of urinary Int 1991;39:5506.
biomarkers for early detection of acute kidney injury following cardiopulmonary [80] Mitsnefes MM, Kathman TS, Mishra J, et al. Serum neutrophil gelatinase-associated
bypass. Biomarkers 2009 September;14(6):42331. lipocalin as a marker of renal function in children with chronic kidney disease.
[46] Kidney Disease: Improving Global Outcomes (KDIGO) Acute Kidney Injury Work Pediatr Nephrol 2007;22(1):1018.
Group. KDIGO. Clinical practice guideline for acute kidney injury. Kidney Int [81] Smith ER, Lee D, Cai M, et al. Urinary neutrophil gelatinase-associated lipocalin may
Suppl 2012;2:1138. aid prediction of renal decline in patients with non-proteinuric stages 3 and 4
[47] James MT, Hemmelgarn BR, Tonelli M. Early recognition and prevention of chronic chronic kidney disease. Nephrol Dial Transplant 2013;28:156979.
kidney disease. Lancet 2010;375(9722):1296309. [82] Bolignano D, Lacquaniti A, Coppolino G, et al. Neutrophil gelatinase-associated
[48] Levin A, Stevens PE. Early detection of CKD: the benets, limitations and effects on lipocalin (NGAL) and progression of chronic kidney disease. Clin J Am Soc Nephrol
prognosis. Nat Rev Nephrol 2011;7(8):44657. 2009;4:33744.
M.E. Wasung et al. / Clinica Chimica Acta 438 (2015) 350357 357

[83] Warnock DG, Muntner P, McCullough PA, et al. Kidney function, albuminuria, and [97] Chow F, Ozols E, Nikolic-Paterson DJ, Atkins RC, Tesch GH. Macrophages in mouse
all-cause mortality in the REGARDS (Reasons for Geographic and Racial Differences type 2 diabetic nephropathy: correlation with diabetic state and progressive renal
in Stroke) study. Am J Kidney Dis 2010 Nov;56(5):86171. injury. Kidney Int 2004;65(1):11628.
[84] Shimada T, Hasegawa H, Yamazaki Y, et al. FGF-23 is a potent regulator of vitamin [98] Takebayashi K, Matsumoto S, Aso Y, Inukai T. Aldosterone blockade attenuates uri-
D metabolism and phosphate homeostasis. J Bone Miner Res 2004;19:42935. nary monocyte chemoattractant protein-1 and oxidative stress in patients with
[85] Shimada T, Kakitani M, Yamazaki Y, et al. Targeted ablation of Fgf23 demonstrates type 2 diabetes complicated by diabetic nephropathy. J Clin Endocrinol Metab
an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J 2006;91(6):22147.
Clin Invest 2004;113:5618. [99] Schober JM, Chen N, Grzeszkiewicz TM, et al. Identication of integrin alpha(M)
[86] Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, et al. The parathyroid is a target organ for beta(2) as an adhesion receptor on peripheral blood monocytes for Cyr61
FGF23 in rats. J Clin Invest 2007;117:40038. (CCN1) and connective tissue growth factor (CCN2): immediate-early gene prod-
[87] Gutierrez O, Isakova T, Rhee E, et al. Fibroblast growth factor-23 mitigates ucts expressed in atherosclerotic lesions. Blood 2002;99(12):445765.
hyperphosphatemia but accentuates calcitriol deciency in chronic kidney disease. [100] Kiyici S, Erturk E, Budak F, et al. Serum monocyte chemoattractant protein-1 and
J Am Soc Nephrol 2005;16:220515. monocyte adhesion molecules in type 1 diabetic patients with nephropathy. Arch
[88] Isakova T, Wahl P, Vargas GS, et al. Fibroblast growth factor 23 is elevated before Med Res 2006;37(8):9981003.
parathyroid hormone and phosphate in chronic kidney disease. Kidney Int 2011; [101] Leask A, Abraham DJ. The role of connective tissue growth factor, a multifunctional
79:13708. matricellular protein, in broblast biology. Biochem Cell Biol 2003;81(6):35563.
[89] Isakova T, Xie H, Yang W, et al. Fibroblast growth factor 23 and risks of mortality [102] Chien W, O'Kelly J, Lu D, et al. Expression of connective tissue growth factor (CTGF/
and end-stage renal disease in patients with chronic kidney disease. JAMA 2011; CCN2) in breast cancer cells is associated with increased migration and angiogen-
305(23):24329. esis. Int J Oncol 2011;38(6):17417.
[90] Leonard EJ, Yoshimura T. Human monocyte chemoattractant protein-1 (MCP-1). [103] Gupta S, Clarkson MR, Duggan J, Brady HR. Connective tissue growth factor: poten-
Immunol Today 1990 Mar;11(3):97101. tial role in glomerulosclerosis and tubulointerstitial brosis. Kidney Int 2000;58(4):
[91] Cushing SD, Berliner JA, Valente AJ, et al. Minimally modied low density lipopro- 138999.
tein induces monocyte chemotactic protein 1 in human endothelial cells and [104] Ito Y, Aten J, Bende RJ, et al. Expression of connective tissue growth factor in human
smooth muscle cells. Proc Natl Acad Sci U S A 1990;87(13):51348. renal brosis. Kidney Int 1998;53(4):85361.
[92] Elner SG, Strieter RM, Elner VM, et al. Monocyte chemotactic protein gene expres- [105] Paradis V, Dargere D, Vidaud M, et al. Expression of connective tissue growth factor
sion by cytokine-treated human retinal pigment epithelial cells. Lab Invest 1991; in experimental rat and human liver brosis. Hepatology 1999;30(4):96876.
64(6):81925. [106] Riser BL, Cortes P, DeNichilo M, et al. Urinary CCN2 (CTGF) as a possible predictor of
[93] Wada T, Furuichi K, Sakai N, et al. Up-regulation of monocyte chemoattractant diabetic nephropathy: preliminary report. Kidney Int 2003;64(2):4518.
protein-1 in tubulointerstitial lesions of human diabetic nephropathy. Kidney Int [107] Nguyen TQ, Tarnow L, Andersen S, et al. Urinary connective tissue growth factor
2000;58(4):14929. excretion correlates with clinical markers of renal disease in a large population of
[94] Wada T, Yokoyama H, Su SB, et al. Monitoring urinary levels of monocyte chemo- type 1 diabetic patients with diabetic nephropathy. Diabetes Care 2006;29(1):
tactic and activating factor reects disease activity of lupus nephritis. Kidney Int 838.
1996;49(3):7617. [108] Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group. KDIGO.
[95] Banba N, Nakamura T, Matsumura M, Kuroda H, Hattori Y, Kasai K. Possible rela- Clinical practice guideline for the evaluation and management of chronic kidney
tionship of monocyte chemoattractant protein-1 with diabetic nephropathy. Kid- disease. Kidney Int Suppl 2013;3:1150.
ney Int 2000;58(2):68490.
[96] Chow FY, Nikolic-Paterson DJ, Ozols E, Atkins RC, Rollin BJ, Tesch GH. Monocyte
chemoattractant protein-1 promotes the development of diabetic renal injury in
streptozotocin-treated mice. Kidney Int 2006;69(1):7380.

Das könnte Ihnen auch gefallen