401
|
Chirino YI, Pedraza-Chaverri J. Role of oxidative and nitrosative stress in cisplatin-induced nephrotoxicity. ACTA ACUST UNITED AC 2008; 61:223-42. [PMID: 18986801 DOI: 10.1016/j.etp.2008.09.003] [Citation(s) in RCA: 359] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 09/04/2008] [Accepted: 09/10/2008] [Indexed: 02/07/2023]
Abstract
cis-Diamminedichloroplatinum (II) (cisplatin) is an important chemotherapeutic agent useful in the treatment of several cancers; however, it has several side effects such as nephrotoxicity. The role of the oxidative and nitrosative stress in cisplatin-induced nephrotoxicity is additionally supported by the protective effect of several free radical scavengers and antioxidants. Furthermore, in in vitro experiments, antioxidants or reactive oxygen species (ROS) scavengers have a cytoprotective effect on cells exposed to cisplatin. Recently, the participation of nitrosative stress has been more explored in cisplatin-induced renal damage. The use of a water-soluble Fe(III) porphyrin complex able to metabolize peroxynitrite (ONOO(-)) has demonstrated that this anion contributes to both in vivo and in vitro cisplatin-induced toxicity. ONOO(-) is produced when nitric oxide (NO*) reacts with superoxide anion (O(2)(*-)); currently, there are evidences suggesting alterations in NO* production after cisplatin treatment and the evidence appear to NO* has a toxic effect. This article goes through current evidence of the mechanism by more than a few compounds have beneficial effects on cisplatin-induced nephrotoxicity, contribute to understanding the role of oxidative and nitrosative stress and suggest several points as part of the mechanism of cisplatin toxicity.
Collapse
Affiliation(s)
- Yolanda I Chirino
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Tlalpan, DF, Mexico.
| | | |
Collapse
|
402
|
Chawla LS, Dommu A, Berger A, Shih S, Patel SS. Urinary sediment cast scoring index for acute kidney injury: a pilot study. Nephron Clin Pract 2008; 110:c145-50. [PMID: 18953176 DOI: 10.1159/000166605] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 07/08/2008] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Urine microscopy is a useful diagnostic tool; however, the manner in which nephrologists prepare and examine urinary sediment is variable. We developed an acute kidney injury (AKI) cast scoring index (CSI) in order to standardize urinary microscopy. Further, we sought to assess the precision of this scoring system. METHODS Urine from 30 patients with AKI consistent with the syndrome of acute tubular necrosis were collected. Sample preparation was uniform and standardized. A panel of 3 nephrologists blinded to the sample preparation were instructed to grade each slide using the AKI CSI. Subsequently, the AKI CSI was then tested in another 18 patients with AKI to determine if this score could predict nonrenal recovery. RESULTS The inter-observer agreement index was 99.80%, with a coefficient of variation of 1.24%. Of the 90 paired observations, 98.8% fell within 2 standard deviations of the mean, signifying good agreement. The receiver operator characteristic area under the curve for AKI CSI to predict nonrenal recovery was 0.79. CONCLUSIONS AKI CSI is a simple, novel, reliable scoring system to grade the degree of epithelial cell and granular casts present on urine microscopy. A standardized AKI CSI has the potential to incorporate urinary cast analysis into the advancing field of AKI diagnostics. These preliminary data endorse the notion that the AKI CSI may be useful in predicting renal outcomes.
Collapse
Affiliation(s)
- Lakhmir S Chawla
- Department of Critical Care Medicine and Anesthesiology, George Washington University Medical Center, Washington, DC 20037, USA.
| | | | | | | | | |
Collapse
|
403
|
Savage CJ. Urinary clinical pathologic findings and glomerular filtration rate in the horse. Vet Clin North Am Equine Pract 2008; 24:387-404, vii. [PMID: 18652961 DOI: 10.1016/j.cveq.2008.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Urinary specific gravity (USG) measurements are underused by equine ambulatory veterinarians. Urinary dipstick and USG findings can assist in the diagnosis and prognosis of many disease processes in the horse. Simple methods for measurement of the glomerular filtration rate and urinary biochemical markers can improve equine urinary diagnostic abilities in critical care patients. Fractional excretion of electrolytes and minerals assists in fluid care and in management of nutrition of horses.
Collapse
Affiliation(s)
- Catherine J Savage
- Equine Clinical Services, Equine Centre, University of Melbourne, 250 Princes Highway, Werribee, Victoria 3030, Australia.
| |
Collapse
|
404
|
Waanders F, Vaidya VS, van Goor H, Leuvenink H, Damman K, Hamming I, Bonventre JV, Vogt L, Navis G. Effect of renin-angiotensin-aldosterone system inhibition, dietary sodium restriction, and/or diuretics on urinary kidney injury molecule 1 excretion in nondiabetic proteinuric kidney disease: a post hoc analysis of a randomized controlled trial. Am J Kidney Dis 2008; 53:16-25. [PMID: 18823687 DOI: 10.1053/j.ajkd.2008.07.021] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2007] [Accepted: 07/07/2008] [Indexed: 11/11/2022]
Abstract
BACKGROUND Tubulointerstitial damage plays an important role in chronic kidney disease (CKD) with proteinuria. Urinary kidney injury molecule 1 (KIM-1) reflects tubular KIM-1 and is considered a sensitive biomarker for early tubular damage. We hypothesized that a decrease in proteinuria by using therapeutic interventions is associated with decreased urinary KIM-1 levels. STUDY DESIGN Post hoc analysis of a randomized, double-blind, placebo-controlled, crossover trial. SETTING & PARTICIPANTS 34 proteinuric patients without diabetes from our outpatient renal clinic. INTERVENTION Stepwise 6-week interventions of losartan, sodium restriction (low-sodium [LS] diet), their combination, losartan plus hydrochlorothiazide (HCT), and the latter plus an LS diet. OUTCOMES & MEASUREMENTS Urinary excretion of KIM-1, total protein, and N-acetyl-beta-d-glucosaminidase (NAG) as a positive control for tubular injury. RESULTS Mean baseline urine protein level was 3.8 +/- 0.4 (SE) g/d, and KIM-1 level was 1,706 +/- 498 ng/d (increased compared with healthy controls; 74 ng/d). KIM-1 level was decreased by using placebo/LS (1,201 +/- 388 ng/d; P = 0.04), losartan/high sodium (1,184 +/- 296 ng/d; P = 0.09), losartan/LS (921 +/- 176 ng/d; P = 0.008), losartan/high sodium plus HCT (862 +/- 151 ng/d; P = 0.008) and losartan/LS plus HCT (743 +/- 170 ng/d; P = 0.001). The decrease in urinary KIM-1 levels paralleled the decrease in proteinuria (R = 0.523; P < 0.001), but not blood pressure or creatinine clearance. 16 patients reached target proteinuria with protein less than 1 g/d, whereas KIM-1 levels normalized in only 2 patients. Urinary NAG level was increased at baseline and significantly decreased during the treatment periods of combined losartan plus HCT only. The decrease in urinary NAG levels was not closely related to proteinuria. LIMITATIONS Post hoc analysis. CONCLUSIONS Urinary KIM-1 level was increased in patients with nondiabetic CKD with proteinuria and decreased in parallel with proteinuria by using losartan, sodium restriction, their combination, losartan plus HCT, and the latter plus sodium restriction. These results are consistent with the hypothesis of amelioration of proteinuria-induced tubular damage. Long-term studies are warranted to evaluate whether targeting treatment on KIM-1 can improve outcomes in patients with CKD with proteinuria.
Collapse
Affiliation(s)
- Femke Waanders
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
405
|
Trujillo J, Cruz C, Tovar A, Vaidya V, Zambrano E, Bonventre JV, Gamba G, Torres N, Bobadilla NA. Renoprotective mechanisms of soy protein intake in the obese Zucker rat. Am J Physiol Renal Physiol 2008; 295:F1574-82. [PMID: 18815216 DOI: 10.1152/ajprenal.90385.2008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We previously showed that long-term consumption of a soy protein diet (SoyP) reduces renal damage in obese Zucker (ObeseZ) rats by restoring urinary NO2 and NO3 excretion (UNO2/NO3V), suggesting that nitric oxide (NO) deficiency may contribute to the renal progression observed in this model. In addition, there is compelling evidence that hyperleptinemia produced deleterious effects on the kidney through its interaction with the short leptin receptor (ObRa). This study was designed to evaluate the contribution of the NO/endothelial NO synthase (eNOS) system, renal oxidative stress, and ObRa expression to the renoprotection conferred by the consumption of a SoyP in ObeseZ rats. Ten lean and ten male ObeseZ rats were included. One-half of each group was fed with a 20% SoyP and the other half with a 20% casein protein diet (CasP) over the course of 160 days. eNOS protein levels and phosphorylation, renal lipoperoxidation (rLPO), and antioxidant enzyme activity were assessed. In addition, renal ObRa, TGF-beta, and kidney injury molecule (Kim-1) mRNA levels, as well as urinary Kim-1 levels, were measured. Renal injury observed in ObeseZ rats fed with CasP was not associated with changes in eNOS expression or phosphorylation. However, this group did present with increased rLPO, reduced catalase activity, and upregulation of ObRa, TGF-beta1, and Kim-1. In contrast, ObeseZ rats fed with a SoyP exhibited a reduction in NOS-Thr495 phosphorylation and rLPO, as well as an enhanced catalase activity. These findings were associated with a significant reduction of ObRa, TGF-beta1, and Kim-1 mRNA levels and urinary Kim-1 protein. Our results show that renoprotection by SoyP in ObeseZ rats is in part mediated by increased NO availability secondary to a reduction in eNOS-T495 phosphorylation and oxidative stress, together with a significant reduction in ObRa and TGF-beta expression.
Collapse
Affiliation(s)
- Joyce Trujillo
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
406
|
Waikar SS, Bonventre JV. Biomarkers for the diagnosis of acute kidney injury. Nephron Clin Pract 2008; 109:c192-7. [PMID: 18802367 DOI: 10.1159/000142928] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The identification of acute kidney injury relies on tests like blood urea nitrogen and serum creatinine that were identified and incorporated into clinical practice several decades ago. This review summarizes clinical studies of newer biomarkers that may permit earlier and more accurate identification of acute kidney injury. The urine may contain sensitive and specific markers of kidney injury that are present due to either impaired tubular reabsorption and catabolism of filtered molecules or release of tubular cell proteins in response to ischemic or nephrotoxic injury. Many potential markers have been studied. Promising injury markers in the urine include N-acetyl-beta-D-glucosaminidase, neutrophil gelatinase-associated lipocalin, kidney injury molecule-1, and interleukin-18. New biomarkers of kidney injury hold the promise of substantially improving the diagnostic approach to acute kidney injury. Adequately powered clinical studies of multiple biomarkers are needed to qualify the biomarkers before they can be fully adopted in clinical practice. Once adopted, more sensitive biomarkers of acute kidney injury hold the potential to transform the care of patients with renal disease.
Collapse
Affiliation(s)
- Sushrut S Waikar
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|
407
|
Di Giusto G, Anzai N, Endou H, Torres AM. Oat5 and NaDC1 protein abundance in kidney and urine after renal ischemic reperfusion injury. J Histochem Cytochem 2008; 57:17-27. [PMID: 18796410 DOI: 10.1369/jhc.2008.951582] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The aim of this study was to evaluate the abundance of the organic anion transporter 5 (Oat5) and the sodium-dicarboxylate cotransporter 1 (NaDC1) in kidney and urine after renal ischemic reperfusion injury. Renal injury was induced in male Wistar rats by occlusion of both renal pedicles for 0 (Group Sham), 5 (Group I5R60), or 60 (Group I60R60) min. The studies were performed after 60 min of reperfusion. The expression of Oat5 and NaDC1 was evaluated by IHC and Western blotting. Oat5 and NaDC1 abundance and alkaline phosphatase activity (AP) were assayed in urine. A decreased expression in renal homogenates and apical membranes and an increase in urinary excretion of Oat5 and NaDC1 were observed in I60R60 rats, as well as alterations of other widely used parameters for renal dysfunction and injury (plasma creatinine, urinary AP activity, kidney weight, histological lesions). In contrast, in the I5R60 group, only an increase in urinary excretion of Oat5 and mild histopathological damage was detected. This is the first study on Oat5 and NaDC1 detection in urine. These results suggest that urinary excretion of Oat5 might be an early indicator of renal dysfunction, which is useful for detection of even minor alterations in renal structural and functional integrity.
Collapse
|
408
|
Rees AJ, Kain R. Kim-1/Tim-1: from biomarker to therapeutic target? Nephrol Dial Transplant 2008; 23:3394-6. [PMID: 18769021 DOI: 10.1093/ndt/gfn480] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Andrew J Rees
- Institute of Clinical Pathology, Medical University of Vienna, Vienna, Austria.
| | | |
Collapse
|
409
|
Abstract
During the past half decade there has been a paradigm shift in the view of acute kidney disease that has resulted in a change in nomenclature from the older term, "acute renal failure," to "acute kidney injury" (AKI). This article reviews the new criteria for diagnosis and staging of AKI and summarizes the current understanding of the many causes of AKI and the approach to diagnosis and management.
Collapse
Affiliation(s)
- Patricia Khalil
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, A-919 Sciafe Hall, 3550 Terrace Street, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
410
|
Keyes R, Bagshaw SM. Early diagnosis of acute kidney injury in critically ill patients. Expert Rev Mol Diagn 2008; 8:455-64. [PMID: 18598227 DOI: 10.1586/14737159.8.4.455] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute kidney injury (AKI) is a common and serious problem in critically ill patients. Tests currently used to detect AKI (i.e., serum creatinine, serum urea and various urinary indices) often result in delayed detection of injury--becoming abnormal at 48-72 h after the initial insult. This delayed detection translates into a potential missed opportunity for therapeutic interventions at a time when kidney damage may be limitable or reversible. This may also, in particular, account for the poor clinical outcomes commonly associated with AKI. The development of novel serum and urinary biomarkers capable of detecting AKI at an earlier phase of illness is therefore vital. This article will review the pitfalls of current conventional testing in kidney injury and discuss the emergence of novel biomarkers with the potential to revolutionize the field of critical care nephrology.
Collapse
Affiliation(s)
- Robert Keyes
- Division of Critical Care Medicine, University of Alberta Hospital, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
411
|
Abstract
Acute kidney injury (AKI) is a significant cause of perioperative patient morbidity and mortality. The definition of AKI has recently changed and further research is underway to identify clinically relevant biomarkers to aid in the diagnosis of the syndrome. AKI is often multi-factorial in origin and patients with certain preoperative risk factors are at elevated risk of perioperative AKI. An anesthesiologist's main objective for perioperative renal protection is prevention by maintenance of euvolemia, preservation of adequate renal perfusion, and avoidance of nephrotoxins. This review will address the definition and diagnosis of AKI, identify patients at risk of AKI, and critically appraise management options for perioperative renal protection.
Collapse
|
412
|
Haase M, Bellomo R, Story D, Davenport P, Haase-Fielitz A. Urinary interleukin-18 does not predict acute kidney injury after adult cardiac surgery: a prospective observational cohort study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2008; 12:R96. [PMID: 18673539 PMCID: PMC2575584 DOI: 10.1186/cc6972] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 07/16/2008] [Accepted: 08/01/2008] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Urinary interleukin-18 (IL-18) measured during the immediate postoperative period could be a promising predictor of acute kidney injury following adult cardiac surgery. METHODS In a single-centre prospective observational cohort study, we enrolled 100 adult cardiac surgical patients undergoing cardiopulmonary bypass at a tertiary hospital. We measured the urinary concentration of IL-18 and creatinine preoperatively, on arrival in the intensive care unit, and 24 hours postoperatively. We assessed urinary IL-18 concentration and urinary IL-18/urinary creatinine ratio in relation to the postoperative development of acute kidney injury defined as an increase in serum creatinine of greater than 50% from preoperative to postoperative peak value within 48 hours after surgery. RESULTS Twenty patients developed acute kidney injury. On arrival in the intensive care unit and at 24 hours postoperatively, urinary IL-18 (median [interquartile range]) was not different in patients who subsequently developed acute kidney injury compared with those who did not: on arrival in the intensive care unit (168 [717] versus 104 [256] pg/mL; P = 0.70) and at 24 hours (195 [483] versus 165 [246] pg/mL; P = 0.47). On arrival in the intensive care unit (area under the curve for the receiver operating characteristic curve [AUC-ROCC] 0.53, 95% confidence interval [CI] 0.38 to 0.68; P = 0.70) and at 24 hours postoperatively (AUC-ROCC 0.55, 95% CI 0.40 to 0.71; P = 0.48), urinary IL-18 was not better than chance in predicting acute kidney injury. All findings were confirmed when urinary IL-18 was adjusted for urinary creatinine. Urinary IL-18 correlated with duration of cardiopulmonary bypass (P < 0.001). CONCLUSION In adults, early postoperative measurement of urinary IL-18 appears not to be valuable in identifying patients who develop acute kidney injury after cardiac surgery, but rather represents a nonspecific marker of cardiopulmonary bypass-associated systemic inflammation.
Collapse
Affiliation(s)
- Michael Haase
- Department of Intensive Care, Austin Health, Melbourne, 145 Studley Rd, Heidelberg 3084, Australia
| | | | | | | | | |
Collapse
|
413
|
Ichimura T, Asseldonk EJPV, Humphreys BD, Gunaratnam L, Duffield JS, Bonventre JV. Kidney injury molecule-1 is a phosphatidylserine receptor that confers a phagocytic phenotype on epithelial cells. J Clin Invest 2008; 118:1657-68. [PMID: 18414680 DOI: 10.1172/jci34487] [Citation(s) in RCA: 555] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Accepted: 02/29/2008] [Indexed: 01/15/2023] Open
Abstract
Following injury, the clearance of apoptotic and necrotic cells is necessary for mitigation and resolution of inflammation and tissue repair. In addition to macrophages, which are traditionally assigned to this task, neighboring epithelial cells in the affected tissue are postulated to contribute to this process. Kidney injury molecule-1 (KIM-1 or TIM-1) is an immunoglobulin superfamily cell-surface protein not expressed by cells of the myeloid lineage but highly upregulated on the surface of injured kidney epithelial cells. Here we demonstrate that injured kidney epithelial cells assumed attributes of endogenous phagocytes. Confocal images confirm internalization of apoptotic bodies within KIM-1-expressing epithelial cells after injury in rat kidney tubules in vivo. KIM-1 was directly responsible for phagocytosis in cultured primary rat tubule epithelial cells and also porcine and canine epithelial cell lines. KIM-1 was able to specifically recognize apoptotic cell surface-specific epitopes phosphatidylserine, and oxidized lipoproteins, expressed by apoptotic tubular epithelial cells. Thus, KIM-1 is the first nonmyeloid phosphatidylserine receptor identified to our knowledge that transforms epithelial cells into semiprofessional phagocytes.
Collapse
Affiliation(s)
- Takaharu Ichimura
- Renal Division, Brigham and Women's Hospital, Harvard Institutes of Medicine, Room 550, 4 Blackfan Circle, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | |
Collapse
|
414
|
Abstract
The diagnosis of acute kidney injury (AKI) is usually based on measurements of blood urea nitrogen (BUN) and serum creatinine. BUN and serum creatinine are not very sensitive or specific for the diagnosis of AKI because they are affected by many renal and nonrenal factors that are independent of kidney injury or kidney function. Biomarkers of AKI that are made predominantly by the injured kidney have been discovered in preclinical studies. In clinical studies of patients with AKI, some of these biomarkers (eg, interleukin-18, neutrophil gelatinase-associated lipocalin, and kidney injury molecule-1) have been shown to increase in the urine before the increase in serum creatinine. These early biomarkers of AKI are being tested in different types of AKI and in larger clinical studies. Biomarkers of AKI may also predict long-term kidney outcomes and mortality.
Collapse
|
415
|
Kidney injury molecule-1 as a biomarker of acute kidney injury in renal transplant recipients. ACTA ACUST UNITED AC 2008; 4:362-3. [DOI: 10.1038/ncpneph0828] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Accepted: 04/03/2008] [Indexed: 01/26/2023]
|
416
|
Immunolocalization of Kim-1, RPA-1, and RPA-2 in kidney of gentamicin-, mercury-, or chromium-treated rats: relationship to renal distributions of iNOS and nitrotyrosine. Toxicol Pathol 2008; 36:397-409. [PMID: 18441258 DOI: 10.1177/0192623308315832] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immunohistochemical studies for kidney injury molecule-1 (Kim-1), renal papillary antigen-1 (RPA-1), and renal papillary antigen-2 (RPA-2) were conducted to explore their relationship to inducible nitric oxide synthase (iNOS) and nitrotyrosine expression. Male Sprague-Dawley rats were exposed to gentamicin (100 mg/kg/day Gen, sc, for 3 days), mercury (0.25 mg Hg/kg, iv, single dose), or chromium (5 mg Cr/kg, sc, single dose) and kidney tissue was examined 24 hours or 72 hours after the last dose of the nephrotoxicant. Another group of kidneys was evaluated 24 hours after rats were administered 3 daily doses (50, 100, 150, 200, or 300 mg/kg/day) of Gen. Gen- and Cr-treated rats exhibited increased immunoreactivity of Kim-1, RPA-1, and RPA-2 largely in the S1/S2 segments and to a lesser extent in the S3 segments of the proximal tubule of the kidney, whereas Hg-treated rats showed increased immunoreactivity of Kim-1, RPA-1, and RPA-2 in the S3 segments. Up-regulation of Kim-1, RPA-1, and RPA-2 expression correlated with injured tubular epithelial cells and also correlated with immunoreactivity of iNOS and nitrotyrosine. It is possible that iNOS activation with nitrotyrosine production in injured nephron segments may be involved in the induction of Kim-1, RPA-1, and RPA-2 following exposure to nephrotoxicants.
Collapse
|
417
|
Devarajan P. Emerging urinary biomarkers in the diagnosis of acute kidney injury. ACTA ACUST UNITED AC 2008; 2:387-398. [PMID: 19079800 DOI: 10.1517/17530059.2.4.387] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND: Acute kidney injury (AKI) represents a common and devastating problem in clinical medicine. The lack of early biomarkers for AKI has led to a delay in initiating potentially effective therapies. OBJECTIVE: Identification of novel urinary biomarkers for AKI that have progressed to the clinical phase of the biomarker discovery process. METHODS: A literature review (PubMed, MedLine) from 2000 to the present. RESULTS/CONCLUSIONS;: The most promising AKI biomarkers include neutrophil gelatinase-associated lipocalin, IL-18, kidney injury molecule-1 and liver-type fatty acid binding protein. Studies to validate the sensitivity and specificity of these biomarkers in clinical samples from large cohorts and from multiple clinical situations are in progress, facilitated by the development of commercial tools for their measurement.
Collapse
Affiliation(s)
- Prasad Devarajan
- University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Nephrology and Hypertension, MLC 7022, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
418
|
Infiltrated macrophages contribute to recovery after ischemic injury but not to ischemic preconditioning in kidneys. Transplantation 2008; 85:447-55. [PMID: 18301336 DOI: 10.1097/tp.0b013e318160f0d1] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Macrophages are associated with ischemia/reperfusion (I/R) injury; however, the role of macrophages that have infiltrated into tissues remains unclear. Therefore, we investigated whether infiltrated macrophages influence recovery after kidney I/R injury and affect the phenomenon of ischemic preconditioning, in which previous ischemia affords the kidney resistance to subsequent ischemia. METHODS Mice were subjected to 30 min of bilateral renal ischemia on day 0, then intravenously administered either liposome-encapsulated dichloromethylene bisphosphonate (Cl2MBP; Lipo-clodronate, a remover of tissue macrophages) or PBS (Lipo-PBS) on day 6 and were then subjected to an additional 30 min of bilateral renal ischemia on day 8. RESULTS Administration of lipoclodronate removed the infiltrated macrophages after I/R. The number of apoptotic and necrotic cells, as well as superoxide and peroxynitrite levels in kidneys from mice that received Lipo-clodronate, was significantly greater than those in kidneys from mice that were administered Lipo-PBS. Proliferating cell nuclear antigen (PCNA) expression was greater in kidneys from mice that were treated with Lipo-clodronate than in those from mice treated with Lipo-PBS. Thirty min of ischemic preconditioning protected the kidneys from 30 min of ischemia induced 8 days later. There was no difference in the plasma creatinine levels of mice treated with Lipo-clodronate or Lipo-PBS. CONCLUSIONS Our results demonstrated that the infiltrated macrophages removed dead and dying cells and accelerated recovery after ischemia/reperfusion injury but did not make a critical contribution to ischemic preconditioning.
Collapse
|
419
|
Abstract
Acute kidney injury (AKI) is a common condition with a high risk of death. The standard metrics used to define and monitor the progression of AKI, such as serum creatinine and blood urea nitrogen levels, are insensitive, nonspecific, and change significantly only after significant kidney injury and then with a substantial time delay. This delay in diagnosis not only prevents timely patient management decisions, including administration of putative therapeutic agents, but also significantly affects the preclinical evaluation of toxicity thereby allowing potentially nephrotoxic drug candidates to pass the preclinical safety criteria only to be found to be clinically nephrotoxic with great human costs. Studies to establish effective therapies for AKI will be greatly facilitated by two factors: (a) development of sensitive, specific, and reliable biomarkers for early diagnosis/prognosis of AKI in preclinical and clinical studies, and (b) development and validation of high-throughput innovative technologies that allow rapid multiplexed detection of multiple markers at the bedside.
Collapse
Affiliation(s)
- Vishal S Vaidya
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
420
|
Ferguson MA, Vaidya V, Bonventre JV. Biomarkers of nephrotoxic acute kidney injury. Toxicology 2008; 245:182-93. [PMID: 18294749 PMCID: PMC4038970 DOI: 10.1016/j.tox.2007.12.024] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Accepted: 12/14/2007] [Indexed: 12/15/2022]
Abstract
Acute kidney injury (AKI) is a common condition with significant associated morbidity and mortality. Epidemiologic data suggest that a significant proportion of AKI cases is at least partially attributable to nephrotoxin exposure. This is not surprising given intrinsic renal susceptibility to toxicant-induced injury, a consequence of the unique physiologic and biochemical properties of the normally functioning kidney. A number of pathophysiologic mechanisms have been identified that mediate toxic effects on the kidney, resulting in a variety of clinical syndromes ranging from subtle changes in tubular function to fulminant renal failure. Unfortunately, standard metrics used to diagnose and monitor kidney injury, such as blood urea nitrogen and serum creatinine, are insensitive and nonspecific, resulting in delayed diagnosis and intervention. Considerable effort has been made to identify biomarkers that will allow the earlier diagnosis of AKI. Further characterization of these candidate biomarkers will clarify their utility in the setting of acute nephrotoxicity, define new diagnostic and prognostic paradigms for kidney injury, facilitate clinical trials, and lead to novel effective therapies.
Collapse
Affiliation(s)
- Michael A. Ferguson
- Fellow in Pediatric Nephrology, Division of Nephrology, Children’s Hospital Boston, Hunnewell 319, Boston, MA 02115, Tel: 617-355-6129, Fax: 617-730-0569,
| | - Vishal Vaidya
- Instructor, Harvard Medical School, Renal Division, Harvard Institutes of Medicine, Room 576, 4 Blackfan Circle, Boston, MA 02115, Tel: 617-525-5974, Fax: 617-525-5965,
| | - Joseph V. Bonventre
- Director, Renal Division, Brigham and Women’s Hospital, Robert H. Ebert Professor of Medicine, Harvard Medical School, Harvard Institutes of Medicine, Room 576, 4 Blackfan Circle, Boston, MA 02115, Tel: 617-525-5969, Fax: 617-525-5965,
| |
Collapse
|
421
|
Famulski KS, Sis B, Billesberger L, Halloran PF. Interferon-gamma and donor MHC class I control alternative macrophage activation and activin expression in rejecting kidney allografts: a shift in the Th1-Th2 paradigm. Am J Transplant 2008; 8:547-56. [PMID: 18294151 DOI: 10.1111/j.1600-6143.2007.02118.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Organ allografts deficient in interferon-gamma (Ifng) or major histocompatibility complex (MHC) class I products develop accelerated necrosis when rejection develops, depending on perforin and granzymes. Thus Ifng-induced donor class I products deliver inhibitory signals to host inflammatory cells. We used microarrays to investigate whether Ifng-induced donor class I products also control inflammation patterns in mouse kidney allografts. Compared to wild-type (WT) allografts, many transcripts were increased in both Ifng-deficient allografts (Ifng-suppressed transcripts [GSTs]) and class I-deficient allografts (class I-suppressed transcripts [CISTs]), with 73% overlap between GSTs and CISTs. Some GSTs and CISTs reflected increased necrosis, including known injury-induced transcripts. However, many GSTs and CISTs were independent of perforin, granzymes and necrosis, and were associated with alternative macrophage activation (AMA) (e.g. arginase I [Arg1], macrophage elastase [Mmp12] and macrophage mannose receptor 1 [Mrc1]). AMA transcripts were induced despite absence of host interleukin (IL)4 and IL13 receptors. The AMA inducer may be activins, whose genes (inhibin A [InhbA] and inhibin B [InhbB]) were increased in all allografts with AMA. We conclude that in allograft rejection, Ifng acts via donor Ifng receptors (Ifngr) to induce donor class Ia and Ib products, which engage host inflammatory cells to limit perforin-granzyme-mediated damage and prevent AMA associated with inhibition of activin expression. Thus, Ifng may control T helper type 2 (Th2) cell inflammation by induction of class I products.
Collapse
Affiliation(s)
- K S Famulski
- Department of Medicine, Division of Nephrology & Transplantation Immunology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
422
|
High urinary excretion of kidney injury molecule-1 is an independent predictor of graft loss in renal transplant recipients. Transplantation 2008; 84:1625-30. [PMID: 18165774 DOI: 10.1097/01.tp.0000295982.78039.ef] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Chronic transplant dysfunction is characterized by renal function decline and proteinuria. Kidney injury molecule (KIM)-1, a transmembrane tubular protein with unknown function, is undetectable in normal kidneys, but markedly induced after injury. Urinary KIM-1 excretion has been quantified as biomarker of renal damage. We prospectively studied whether urinary KIM-1 predicts graft loss, independent of renal function and proteinuria. METHODS Renal transplant recipients (n=145) visiting our outpatient clinic between August 2001 and July 2003 collected 24-hour urine samples for assessment of baseline urinary KIM-1 excretion (microsphere-based Luminex technology), and were followed for graft loss. RESULTS Recipients participated at a median (interquartile range) of 6.0 (2.5-12.0) years posttransplant in baseline measurements. Follow-up beyond baseline was 4.0 (3.2-4.5) years. Urinary KIM-1 excretion was 0.72 (0.42-1.37) ng per 24 hours. Occurrence of graft loss increased over tertiles of KIM-1 excretion: 3 (6.3%), 11 (22.4%), and 17 cases (35.4%; P=0.001), respectively. High KIM-1 excretion was associated with proteinuria, low creatinine clearance, and high donor age (all P<0.01). In multivariate Cox regression analyses, prediction of graft loss by KIM-1 appeared independent of creatinine clearance, proteinuria, and donor age. Hazard ratios (95% CI) for the second and third tertile of KIM-1 excretion were 3.6 (0.9-13.5) and 5.1 (1.5-17.8) in the final model. CONCLUSIONS Urinary excretion of KIM-1 is an independent predictor of long-term graft loss and therefore a promising new biomarker in early prediction of graft loss.
Collapse
|
423
|
Rached E, Hoffmann D, Blumbach K, Weber K, Dekant W, Mally A. Evaluation of putative biomarkers of nephrotoxicity after exposure to ochratoxin a in vivo and in vitro. Toxicol Sci 2008; 103:371-81. [PMID: 18308701 DOI: 10.1093/toxsci/kfn040] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The kidney is one of the main targets of xenobiotic-induced toxicity, but early detection of renal damage is difficult. Recently, several novel biomarkers of nephrotoxicity have been identified by transcription profiling, including kidney injury molecule-1 (Kim-1), lipocalin-2, tissue inhibitor of metalloproteinases-1 (Timp-1), clusterin, osteopontin (OPN), and vimentin, and suggested as sensitive endpoints for acute kidney injury in vivo. However, it is not known if these cellular marker molecules may also be useful to predict chronic nephrotoxicity or to detect nephrotoxic effects in vitro. In this study, a panel of new biomarkers of renal toxicity was assessed via quantitative real-time PCR, immunohistochemistry, and immunoblotting in rats treated with the nephrotoxin ochratoxin A (OTA) for up to 90 days and in rat proximal tubule cells (NRK-52E) treated with OTA in vitro. Repeated administration of OTA to male F344/N rats for 14, 28, or 90 days resulted in a dose- and time-dependent increase in the expression of Kim-1, Timp-1, lipocalin-2, OPN, clusterin, and vimentin. Changes in gene expression were found to correlate with the progressive histopathological alterations and preceded effects on traditional clinical parameters indicative of impaired kidney function. Induction of Kim-1 messenger RNA expression was the earliest and most prominent response observed, supporting the use of this marker as sensitive indicator of chronic kidney injury. In contrast, no significant increase in the expression of putative marker genes and proteins were evident in NRK-52E cells after exposure to OTA for up to 48 h, suggesting that they may not be suitable endpoints for sensitive detection of nephrotoxic effects in vitro.
Collapse
Affiliation(s)
- Eva Rached
- Department of Toxicology, University of Würzburg, Würzburg D-97078, Germany
| | | | | | | | | | | |
Collapse
|
424
|
|
425
|
Zhang Z, Humphreys BD, Bonventre JV. Shedding of the urinary biomarker kidney injury molecule-1 (KIM-1) is regulated by MAP kinases and juxtamembrane region. J Am Soc Nephrol 2008; 18:2704-14. [PMID: 17898236 DOI: 10.1681/asn.2007030325] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Kidney injury molecule-1 (KIM-1) is markedly upregulated in renal proximal tubule cells by stimuli that promote dedifferentiation, including ischemic or toxic injury, as well as in cases of tubulointerstitial disease, polycystic kidney disease, and renal cell carcinoma. Structurally, KIM-1 possesses a single transmembrane domain and undergoes membrane-proximal cleavage, which leads to the release of soluble KIM-1 ectodomain into the urine. Urinary KIM-1 ectodomain is a promising sensitive and specific biomarker for acute kidney injury in humans, and therefore it is important to determine what regulates KIM-1 shedding. We found that constitutive cleavage of KIM-1 is mediated by ERK activation, and that cleavage is accelerated by p38 MAP kinase activation. After cleavage, a 14-kD membrane-bound fragment of KIM-1, which contains two highly conserved tyrosine residues, was tyrosine-phosphorylated. Mutagenesis studies demonstrated that the juxtamembrane secondary structure, not the primary amino acid sequence, was critical to the cleavage of KIM-1.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Renal Division, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, and Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Boston, Massachusetts, USA
| | | | | |
Collapse
|
426
|
Abstract
Acute kidney injury (AKI), previously referred to as acute renal failure, represents a common and devastating problem in clinical medicine. Despite significant improvements in therapeutics, the mortality and morbidity associated with AKI remain high. A major reason for this is the lack of early markers for AKI, and hence an unacceptable delay in initiating therapy. Fortunately, the application of innovative technologies such as functional genomics and proteomics to human and animal models of AKI has uncovered several novel biomarkers and therapeutic targets. The most promising of these are chronicled in this review. These include the identification of biomarker panels in plasma (neutrophil gelatinase-associated lipocalin and cystatin C) and urine (neutrophil gelatinase-associated lipocalin, kidney injury molecule-1, interleukin-18, cystatin C, alpha1-microglobulin, Fetuin-A, Gro-alpha, and meprin). It is likely that the AKI panels will be useful for timing the initial insult, and assessing the duration and severity of AKI. It is also probable that the AKI panels will distinguish between the various etiologies of AKI and predict clinical outcomes. It will be important in future studies to validate the sensitivity and specificity of these biomarker panels in clinical samples from large cohorts and from multiple clinical situations. Such studies will be facilitated markedly by the development of commercial tools for the reproducible measurement of biomarkers across different laboratories.
Collapse
Affiliation(s)
- Prasad Devarajan
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
427
|
Abstract
Acute kidney injury is a common condition in hospitalized patients with significant associated morbidity and mortality. Although impressive progress has been made in the understanding of the molecular and biochemical mechanisms of kidney injury, the ability to effectively treat and thus impact on the outcomes of acute kidney injury has been disappointing. One of the major reasons for this has been the reliance on current measures of renal dysfunction, such as serum creatinine and blood urea nitrogen, which are insensitive to small changes in renal function and subtle signs of kidney injury. This insensitivity of commonly used biomarkers of renal dysfunction not only prevents timely diagnosis and estimation of injury severity, but also delays administration of putative therapeutic agents. A number of serum and urinary proteins have been identified that may herald acute kidney injury prior to a rise in serum creatinine. Further characterization of these candidate biomarkers will clarify their utility and define new diagnostic and prognostic paradigms for acute kidney injury, facilitate clinical trials and lead to novel effective therapies.
Collapse
Affiliation(s)
- Mitchell H Rosner
- University of Virginia Health System, Division of Nephrology, Box 800133, Charlottesville, VA 22908, USA
| |
Collapse
|
428
|
Wang EJ, Snyder RD, Fielden MR, Smith RJ, Gu YZ. Validation of putative genomic biomarkers of nephrotoxicity in rats. Toxicology 2008; 246:91-100. [PMID: 18289764 DOI: 10.1016/j.tox.2007.12.031] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 12/20/2007] [Accepted: 12/24/2007] [Indexed: 11/16/2022]
Abstract
Drug-induced renal injury is a common finding in the early preclinical phase of drug development. But the specific genes responding to renal injury remain poorly defined. Identification of drug-induced gene changes is critical to provide insights into molecular mechanisms and detection of renal damage. To identify genes associated with the development of drug-induced nephrotoxicity, a literature survey was conducted and a panel of 48 genes was selected based on gene expression changes in multiple published studies. Male Sprague-Dawley rats were dosed daily for 1, 3 or 5 days to the known nephrotoxicants gentamicin, bacitracin, vancomycin and cisplatin, or the known hepatotoxicants ketoconazole, 1-naphthyl isothiocyanate and 4,4-diaminodiphenylmethane. Histopathological evaluation and clinical chemistry revealed renal proximal tubular necrosis in rats treated with the nephrotoxicants, but not from those treated with the hepatotoxicants. RNA was extracted from the kidney, and RT-PCR was performed to evaluate expression profiles of the selected genes. Among the genes examined, 24 genes are confirmed to be highly induced or repressed in rats treated with nephrotoxicants; further investigation identified that 5 of the 24 genes were also altered by hepatotoxicants. These data led to the identification of a set of genomic biomarker candidates whose expression in kidney is selectively regulated only by nephrotoxicants. Among those genes displaying the highest expression changes specifically in nephrotoxicant-treated rats were kidney injury molecule 1 (Kim1), lipocalin 2 (Lcn2), and osteopontin (Spp1). The establishment of such a genomic marker set offers a new tool in our ongoing quest to monitor nephrotoxicity.
Collapse
Affiliation(s)
- Er-Jia Wang
- Department of Genetic and Molecular Toxicology, Drug Safety and Metabolism Division, Schering-Plough Corporation, Summit, NJ 07901, USA
| | | | | | | | | |
Collapse
|
429
|
Abstract
There is a need for new biomarkers to enable faster detection of adverse events due to drugs and disease processes. One would prefer biomarkers that are useful in multiple species (i.e., translational or bridging biomarkers) so that it would be possible to directly link responses between species and follow such injury in both preclinical and clinical settings. This chapter will explore some of the issues surrounding the use of pharmacogenomics to identify and qualify such biomarkers, and examples will be provided.
Collapse
Affiliation(s)
- Donna L Mendrick
- Department of Toxicogenomics, Gene Logic Inc., Gaithersburg, Maryland, USA
| |
Collapse
|
430
|
Nguyen MT, Devarajan P. Biomarkers for the early detection of acute kidney injury. Pediatr Nephrol 2008; 23:2151-7. [PMID: 17394022 PMCID: PMC6904376 DOI: 10.1007/s00467-007-0470-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Revised: 02/21/2007] [Accepted: 02/22/2007] [Indexed: 12/29/2022]
Abstract
Acute kidney injury (AKI), previously referred to as acute renal failure (ARF), represents a persistent problem in clinical medicine. Despite significant improvements in therapeutics, the mortality and morbidity associated with AKI remain high. A major reason for this is the lack of early markers for AKI, akin to troponins in acute myocardial disease, and hence an unacceptable delay in initiating therapy. Fortunately, the application of innovative technologies such as functional genomics and proteomics to human and animal models of AKI has uncovered several novel genes and gene products that are emerging as biomarkers. The most promising of these are chronicled in this article. These include a plasma panel [neutrophil gelatinase-associated lipocalin (NGAL) and cystatin C] and a urine panel [NGAL, interleukin 18 (IL-18), and kidney injury molecule 1 (KIM)-1]. As they represent sequentially expressed biomarkers, it is likely that the AKI panels will be useful for timing the initial insult and assessing the duration of AKI. Based on the differential expression of the biomarkers, it is also likely that the AKI panels will distinguish between the various types and etiologies of AKI. It will be important in future studies to validate the sensitivity and specificity of these biomarker panels in clinical samples from large cohorts and from multiple clinical situations.
Collapse
Affiliation(s)
- Mai T. Nguyen
- Nephrology and Hypertension, MLC 7022, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039 USA
| | - Prasad Devarajan
- Nephrology and Hypertension, MLC 7022, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039 USA
| |
Collapse
|
431
|
Zhang PL, Rothblum LI, Han WK, Blasick TM, Potdar S, Bonventre JV. Kidney injury molecule-1 expression in transplant biopsies is a sensitive measure of cell injury. Kidney Int 2007; 73:608-14. [PMID: 18160964 DOI: 10.1038/sj.ki.5002697] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Kidney injury molecule-1 (KIM-1) is a specific histological biomarker for diagnosing early tubular injury on renal biopsies. In this study, KIM-1 expression was quantitated in renal transplant biopsies by immunohistochemistry and correlated with renal function. None of the 25 protocol biopsies showed detectable tubular injury on histologic examination, yet 28% had focal positive KIM-1 expression. Proximal tubule KIM-1 expression was present in all biopsies from patients with histological changes showing acute tubular damage and deterioration of kidney function. In this group, higher KIM-1 staining predicted a better outcome with improved blood urea nitrogen (BUN), serum creatinine, and estimated glomerular filtration rate (eGFR) over an ensuing 18 months. KIM-1 was expressed focally in affected tubules in 92% of kidney biopsies from patients with acute cellular rejection. By contrast, there was little positive staining for Ki-67, a cell proliferation marker, in any of the groups. KIM-1 expression significantly correlated with serum creatinine and BUN, and inversely with the eGFR on the biopsy day. Our study shows that KIM-1 staining sensitively and specifically identified proximal tubular injury and correlated with the degree of renal dysfunction. KIM-1 expression is more sensitive than histology for detecting early tubular injury, and its level of expression in transplant biopsies may indicate the potential for recovery of kidney function.
Collapse
Affiliation(s)
- P L Zhang
- Division of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
432
|
Wong MG, Suzuki Y, Tanifuji C, Akiba H, Okumura K, Sugaya T, Yamamoto T, Horikoshi S, Tan SY, Pollock C, Tomino Y. Peritubular ischemia contributes more to tubular damage than proteinuria in immune-mediated glomerulonephritis. J Am Soc Nephrol 2007; 19:290-7. [PMID: 18094368 DOI: 10.1681/asn.2007020226] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Muh Geot Wong
- Division of Nephrology, Department of Internal Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 1138421, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
433
|
|
434
|
|
435
|
Chirino YI, Trujillo J, Sánchez-González DJ, Martínez-Martínez CM, Cruz C, Bobadilla NA, Pedraza-Chaverri J. Selective iNOS inhibition reduces renal damage induced by cisplatin. Toxicol Lett 2007; 176:48-57. [PMID: 18063323 DOI: 10.1016/j.toxlet.2007.10.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Revised: 10/17/2007] [Accepted: 10/17/2007] [Indexed: 12/13/2022]
Abstract
Cisplatin is a chemotherapeutic agent used in the treatment of several cancer tumors; however, nephrotoxicity has restricted its use. Reactive oxygen species and peroxynitrite, which is formed by the reaction between superoxide anion and nitric oxide (NO*), are implicated in cisplatin-induced nephrotoxicity. In contrast, both toxic and beneficial effects of NO* have been suggested in cisplatin-induced nephrotoxicity. Therefore, nowadays the role of NO* in this experimental model remains controversial. The aim of the present work was to elucidate the role of NO* in cisplatin-induced renal damage using N-[3-(aminomethyl)benzyl]acetamidine (1400W), a selective and irreversible inhibitor of iNOS. The mRNA levels of iNOS were increased in cisplatin-treated rats. The administration of 1400W reduced the cisplatin induced histological damage, renal dysfunction (increase in proteinuria and kidney injury molecule expression and decrease in creatinine clearance), tubulointerstitial infiltration, oxidative stress (increase in renal malondialdehyde and inmmunostaining for 4-hydroxy-2-nonenal) and nitrosative stress (immunostaining for 3-nitrotyrosine). In addition, the administration of 1400W was unable to modify systolic blood pressure in control rats. Our data demonstrate that selective iNOS inhibition reduces the cisplatin-induced nephrotoxicity and nitrosative stress which strongly suggest that in this experimental model (1) the NO* production is toxic and (2) iNOS is the main source of NO*.
Collapse
Affiliation(s)
- Yolanda I Chirino
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Mexico
| | | | | | | | | | | | | |
Collapse
|
436
|
Zhou Y, Vaidya VS, Brown RP, Zhang J, Rosenzweig BA, Thompson KL, Miller TJ, Bonventre JV, Goering PL. Comparison of kidney injury molecule-1 and other nephrotoxicity biomarkers in urine and kidney following acute exposure to gentamicin, mercury, and chromium. Toxicol Sci 2007; 101:159-70. [PMID: 17934191 DOI: 10.1093/toxsci/kfm260] [Citation(s) in RCA: 222] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Sensitive biomarkers are needed to detect kidney injury at the earliest stages. The objective of this study was to determine whether the appearance of kidney injury molecule-1 (Kim-1) protein ectodomain in urine and kidney injury molecule-1/hepatitis A viral cellular receptor-1 (Kim-1/Havcr1) gene expression in kidney tissue may be more predictive of renal injury after exposure to nephrotoxicants when compared to traditionally used biomarkers. Male Sprague-Dawley rats were injected with a range of doses of gentamicin, mercury (Hg; HgCl2), or chromium (Cr; K2Cr2O7). The results showed that increases in urinary Kim-1 and kidney Kim-1/Havcr1 gene expression paralleled the degree of severity of renal histopathology and were detected at lower doses of nephrotoxicants when compared to blood urea nitrogen (BUN), serum creatinine, and urinary N-acetyl-beta-D-glucosaminidase (NAG). In a time course study, urinary Kim-1 was elevated within 24 h after exposure to gentamicin (100 mg/kg), Hg (0.25 mg/kg), or Cr (5 mg/kg) and remained elevated through 72 h. NAG responses were nephrotoxicant dependent with elevations occurring early (gentamicin), late (Cr), or no change (Hg). At 72 h, after treatment with any of the three nephrotoxicants, there was increased Kim-1 immunoreactivity and necrosis involving approximately 50% of the proximal tubules; however, only urinary Kim-1 was significantly increased, while BUN, serum creatinine, and NAG were not different from controls. In rats treated with the hepatotoxicant galactosamine (1.1 mg/kg), serum alanine aminotransferase was increased, but no increase in urinary Kim-1 was observed. Urinary Kim-1 and kidney Kim-1/Havcr1 expression appear to be sensitive and tissue-specific biomarkers that will improve detection of early acute kidney injury following exposure to nephrotoxic chemicals and drugs.
Collapse
Affiliation(s)
- Yuzhao Zhou
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, White Oak Life Sciences Laboratory, Silver Spring, MD 20993, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
437
|
Chen G, Bridenbaugh EA, Akintola AD, Catania JM, Vaidya VS, Bonventre JV, Dearman AC, Sampson HW, Zawieja DC, Burghardt RC, Parrish AR. Increased susceptibility of aging kidney to ischemic injury: identification of candidate genes changed during aging, but corrected by caloric restriction. Am J Physiol Renal Physiol 2007; 293:F1272-81. [PMID: 17670906 PMCID: PMC2758575 DOI: 10.1152/ajprenal.00138.2007] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aging is associated with an increased incidence and severity of acute renal failure. However, the molecular mechanism underlying the increased susceptibility to injury remains undefined. These experiments were designed to investigate the influence of age on the response of the kidney to ischemic injury and to identify candidate genes that may mediate this response. Renal slices prepared from young (5 mo), aged ad libitum (aged-AL; 24 mo), and aged caloric-restricted (aged-CR; 24 mo) male Fischer 344 rats were subjected to ischemic stress (100% N(2)) for 0-60 min. As assessed by biochemical and histological evaluation, slices from aged-AL rats were more susceptible to injury than young counterparts. Importantly, caloric restriction attenuated the increased susceptibility to injury. In an attempt to identify the molecular pathway(s) underlying this response, microarray analysis was performed on tissue harvested from the same animals used for the viability experiments. RNA was isolated and the corresponding cDNA was hybridized to CodeLink Rat Whole Genome Bioarray slides. Subsequent gene expression analysis was performed using GeneSpring software. Using two-sample t-tests and a twofold cut-off, the expression of 92 genes was changed during aging and attenuated by caloric restriction, including claudin-7, kidney injury molecule-1 (Kim-1), and matrix metalloproteinase-7 (MMP-7). Claudin-7 gene expression peaked at 18 mo; however, increased protein expression in certain tubular epithelial cells was seen at 24 mo. Kim-1 gene expression was not elevated at 8 or 12 mo but was at 18 and 24 mo. However, changes in Kim-1 protein expression were only seen at 24 mo and corresponded to increased urinary levels. Importantly, these changes were attenuated by caloric restriction. MMP-7 gene expression was decreased at 8 mo, but an age-dependent increase was seen at 24 mo. Increased MMP-7 protein expression in tubular epithelial cells at 24 mo was correlated with the gene expression pattern. In summary, we identified genes changed by aging and changes attenuated by caloric restriction. This will facilitate investigation into the molecular mechanism mediating the age-related increase in susceptibility to injury.
Collapse
Affiliation(s)
- G. Chen
- Department of Systems Biology and Translational Medicine, College of Medicine, Texas A&M University System Health Science Center, College Station
| | - E. A. Bridenbaugh
- Department of Systems Biology and Translational Medicine, College of Medicine, Texas A&M University System Health Science Center, College Station
| | - A. D. Akintola
- Department of Systems Biology and Translational Medicine, College of Medicine, Texas A&M University System Health Science Center, College Station
| | - J. M. Catania
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - V. S. Vaidya
- Renal Division, Brigham and Women’s Hospital, Harvard Institutes of Medicine, Harvard Medical School, Boston, Massachusetts
| | - J. V. Bonventre
- Renal Division, Brigham and Women’s Hospital, Harvard Institutes of Medicine, Harvard Medical School, Boston, Massachusetts
| | - A. C. Dearman
- Department of Systems Biology and Translational Medicine, College of Medicine, Texas A&M University System Health Science Center, College Station
| | - H. W. Sampson
- Department of Systems Biology and Translational Medicine, College of Medicine, Texas A&M University System Health Science Center, College Station
| | - D. C. Zawieja
- Department of Systems Biology and Translational Medicine, College of Medicine, Texas A&M University System Health Science Center, College Station
| | - R. C. Burghardt
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas
| | - A. R. Parrish
- Department of Systems Biology and Translational Medicine, College of Medicine, Texas A&M University System Health Science Center, College Station
| |
Collapse
|
438
|
Prozialeck WC, Vaidya VS, Liu J, Waalkes MP, Edwards JR, Lamar PC, Bernard AM, Dumont X, Bonventre JV. Kidney injury molecule-1 is an early biomarker of cadmium nephrotoxicity. Kidney Int 2007; 72:985-93. [PMID: 17687258 PMCID: PMC2747605 DOI: 10.1038/sj.ki.5002467] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cadmium (Cd) exposure results in injury to the proximal tubule characterized by polyuria and proteinuria. Kidney injury molecule-1 (Kim-1) is a transmembrane glycoprotein not normally detected in the mature kidney, but is upregulated and shed into the urine following nephrotoxic injury. In this study, we determine if Kim-1 might be a useful early biomarker of Cd nephrotoxicity. Male Sprague-Dawley rats were given daily injections of Cd for up to 12 weeks. Weekly urine samples were analyzed for Kim-1, protein, creatinine, metallothionein, and Clara cell protein CC-16. Significant levels of Kim-1 were detected in the urine by 6 weeks and continued to increase throughout the treatment period. This appearance of Kim-1 occurred 4-5 weeks before the onset of proteinuria, and 1-3 weeks before the appearance of metallothionein and CC-16. Higher doses of Cd gave rise to higher Kim-1 excretion. Reverse transcriptase-polymerase chain reaction (RT-PCR) expression analysis showed that Kim-1 transcript levels were increased after 6 weeks at the low dose of Cd. Immunohistochemical analysis showed that Kim-1 was present in proximal tubule cells of the Cd-treated rats. Our results suggest that Kim-1 may be a useful biomarker of early stages of Cd-induced proximal tubule injury.
Collapse
Affiliation(s)
- W C Prozialeck
- Department of Pharmacology, Midwestern University, Downers Grove, Illinois 60515, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
439
|
Abstract
Renal dysfunction is common in patients with end-stage liver disease. Etiological factors include conditions as diverse as acute tubular necrosis, immunoglobulin A nephropathy and hepatorenal syndrome. Current standard tests of renal function, such as measurement of serum urea and creatinine levels, are inaccurate as the synthesis of these markers is affected by the native liver pathology. This article reviews novel markers of renal function and their potential use in patients with liver disease.
Collapse
Affiliation(s)
- Andrew J Portal
- Institute of Liver Studies, King's College Hospital, London, UK
| | | | | |
Collapse
|
440
|
Mendrick DL, Daniels KK. From the bench to the clinic and back again: translational biomarker discovery using in silico mining of pharmacogenomic data. Biomark Med 2007; 1:319-33. [PMID: 20477406 DOI: 10.2217/17520363.1.2.319] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
To improve drug efficacy and safety and advance medical intervention in diseases, new biomarkers are urgently needed. Pharmacogenomics can provide a tool to discover and begin to qualify biomarkers useful for these indications and is readily applicable to multiple species. One can begin and end with genes or focus on tissue-derived expression analysis of those genes that encode secreted proteins to discover potential biomarkers that can be monitored in body fluids. The paper will discuss issues surrounding such investigations, show examples of translational biomarkers and end with a summary of the US FDA’s work in this field over the last 6 years.
Collapse
Affiliation(s)
- Donna L Mendrick
- Gene Logic Inc., 50 West Watkins Mill Road, Gaithersburg, MA 20878, USA
| | - Kellye K Daniels
- Gene Logic Inc., 50 West Watkins Mill Road, Gaithersburg, MA 20878, USA
| |
Collapse
|
441
|
Smith KD. Iron metabolism at the host pathogen interface: lipocalin 2 and the pathogen-associated iroA gene cluster. Int J Biochem Cell Biol 2007; 39:1776-80. [PMID: 17714976 PMCID: PMC2695446 DOI: 10.1016/j.biocel.2007.07.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 07/07/2007] [Accepted: 07/09/2007] [Indexed: 01/10/2023]
Abstract
The host innate immune defense protein lipocalin 2 binds bacterial enterobactin siderophores to limit bacterial iron acquisition. To counteract this host defense mechanism bacteria have acquired the iroA gene cluster, which encodes enzymatic machinery and transporters that revitalize enterobactin in the form of salmochelin. The iroB enzyme introduces glucosyl residues at the C5 site on 2,3-dihydroxybenzoylserine moieties of enterobactin and thereby prevents lipocalin 2 binding. Additional strategies to evade lipocalin 2 have evolved in other bacteria, such as Mycobacteria tuberculosis and Bacillus anthracis. Targeting these specialized bacterial evasion strategy may provide a mechanism to reinvigorate lipocalin 2 in defense against specific pathogens.
Collapse
Affiliation(s)
- Kelly D Smith
- Department of Pathology, University of Washington, 1959 NE Pacific St. HSB E504, Box 357470, Seattle, WA 98195, United States.
| |
Collapse
|
442
|
Espandiari P, Zhang J, Rosenzweig BA, Vaidya VS, Sun J, Schnackenberg L, Herman EH, Knapton A, Bonventre JV, Beger RD, Thompson KL, Hanig J. The utility of a rodent model in detecting pediatric drug-induced nephrotoxicity. Toxicol Sci 2007; 99:637-48. [PMID: 17636248 PMCID: PMC2729403 DOI: 10.1093/toxsci/kfm184] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
A multi-age rat model was used to identify potential age-related differences in renal injury following exposure to gentamicin (GM). In this study, 10-, 25-, 40-, and 80-day-old Sprague-Dawley rats were dosed with GM at 0, 50, or 100 mg kg(-1) body weight per day (mkd) sc for 6 or 14 days. Urine samples were collected up to 72 h after initial dosing. The maximum tolerated dose was lower in 10-day-old rats than for other ages (none survived 11 days of treatment). Eighty-day-old rats given the highest dose showed a diminished rate of growth and an increase in serum creatinine, blood urea nitrogen (BUN), urinary kidney injury molecule-1 (Kim-1), and renal pathology. Ten- and 40-day-old rats given 100 mkd of GM for 6- or 14 days also had increased levels of serum BUN and Cr and renal pathology, whereas only mild renal alterations were found in 25-day-old rats. After 6 days of treatment with 100 mkd GM, significant increases in Havcr-1 (Kim-1) gene expression were detected only in 10- and 80-day-old rats. In urine samples, nuclear magnetic resonance and ultra performance liquid chromatography/mass spectrometry analysis detected changes related to GM efficacy (e.g., hippurate) and increases in metabolites related to antioxidant activity, which was greatest in the 80-day-old rats. The magnitude of the genomic, metabonomic, and serum chemistry changes appeared to correlate with the degree of nephropathy. These findings indicate that an experimental animal model that includes several developmental stages can detect age-related differences in drug-induced organ toxicities and may be a useful predictor of pediatric drug safety in preclinical studies.
Collapse
Affiliation(s)
- Parvaneh Espandiari
- Center for Drug Evaluation and Research, Silver Spring, Maryland 20993, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
443
|
van Timmeren MM, van den Heuvel MC, Bailly V, Bakker SJL, van Goor H, Stegeman CA. Tubular kidney injury molecule-1 (KIM-1) in human renal disease. J Pathol 2007; 212:209-17. [PMID: 17471468 DOI: 10.1002/path.2175] [Citation(s) in RCA: 350] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
KIM-1, a transmembrane tubular protein with unknown function, is undetectable in normal kidneys, but is markedly induced in experimental renal injury. The KIM-1 ectodomain is cleaved, detectable in urine, and reflects renal damage. KIM-1 expression in human renal biopsies and its correlation with urinary KIM-1 (uKIM-1) is unknown. In biopsies from various renal diseases (n = 102) and controls (n = 7), the fraction of KIM-1 positive tubules and different renal damage parameters were scored. Double labelling was performed for KIM-1 with macrophages (MØ), alpha-smooth muscle actin (alpha-SMA), proximal (aquaporin-1) and distal (E-cadherin) tubular markers and a dedifferentiation marker (vimentin). uKIM-1 at the time of biopsy (n = 53) was measured by ELISA. Renal KIM-1 was significantly increased in all diseases versus controls (p < 0.05), except minimal change. KIM-1 was primarily expressed at the luminal side of dedifferentiated proximal tubules, in areas with fibrosis (alpha-SMA) and inflammation (MØ). Independent of the disease, renal KIM-1 correlated positively with renal damage, negatively with renal function, but not with proteinuria. uKIM-1 was increased in renal patients versus controls (p < 0.001), including minimal change, and correlated positively with tissue KIM-1 and MØ, negatively with renal function, but not with proteinuria. In conclusion, KIM-1 is upregulated in renal disease and is associated with renal fibrosis and inflammation. uKIM-1 is also associated with inflammation and renal function, and reflects tissue KIM-1, indicating that it can be used as a non-invasive biomarker in renal disease.
Collapse
Affiliation(s)
- M M van Timmeren
- Department of Pathology and Laboratory Medicine, University Medical Centre Groningen and University of Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
444
|
Bibliography: current world literature. Curr Opin Anaesthesiol 2007; 20:157-63. [PMID: 17413401 DOI: 10.1097/aco.0b013e3280dd8cd1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
445
|
Abstract
PURPOSE OF REVIEW Acute renal failure causes considerable morbidity and mortality in critically ill patients. To date, there are few therapies available to clinicians that alter outcome. This review will focus on clinical and basic science research efforts related to the diagnosis, pathophysiology, prevention, and treatment of acute renal failure. RECENT FINDINGS The incidence of acute renal failure may be increasing and the mortality rate continues to be significant. The development of sensitive, predictive biomarkers of acute renal failure may help to diagnose the syndrome earlier and allow for meaningful therapeutic intervention. A number of new therapies are in development for acute renal failure. Many show promise in animal models of acute renal failure but prospective studies in humans are lacking. SUMMARY Despite the present lack of therapies for the treatment and prevention of acute renal failure, there are reasons to be optimistic. Recent research has led to the development of several different strategies that may provide a breakthrough in the treatment of acute renal failure.
Collapse
Affiliation(s)
- Dean R Jones
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, NY 10032-3784, USA
| | | |
Collapse
|
446
|
Bagshaw SM, Langenberg C, Haase M, Wan L, May CN, Bellomo R. Urinary biomarkers in septic acute kidney injury. Intensive Care Med 2007; 33:1285-1296. [PMID: 17487471 DOI: 10.1007/s00134-007-0656-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2007] [Accepted: 04/06/2007] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To appraise the literature on the value of urinary biomarkers in septic acute kidney injury (AKI). DESIGN Systematic review. SETTING Academic medical centre. PATIENTS AND PARTICIPANTS Human studies of urinary biomarkers. INTERVENTIONS None. MEASUREMENTS AND RESULTS Fourteen articles fulfilled inclusion criteria. Most studies were small, single-centre, and included mixed medical/surgical adult populations. Few focused solely on septic AKI and all had notable limitations. Retrieved articles included data on low-molecular-weight proteins (beta2-microglobulin, alpha1-microglobulin, adenosine deaminase binding protein, retinol binding protein, cystatin C, renal tubular epithelial antigen-1), enzymes (N-acetyl-beta-glucosaminidase, alanine-aminopeptidase, alkaline phosphatase; lactate dehydrogenase, alpha/pi-glutathione-S-transferase, gamma-glutamyl transpeptidase), cytokines [platelet activating factor (PAF), interleukin-18 (IL-18)] and other biomarkers [kidney injury molecule-1, Na/H exchanger isoform-3 (NHE3)]. Increased PAF, IL-18, and NHE3 were detected early in septic AKI and preceded overt kidney failure. Several additional biomarkers were evident early in AKI; however, their diagnostic value in sepsis remains unknown. In one study, IL-18 excretion was higher in septic than in non-septic AKI. IL-18 also predicted deterioration in kidney function, with increased values preceding clinically significant kidney failure by 24-48 h. Detection of cystatin C, alpha1-microglobulin, and IL-18 predicted need for renal replacement therapy (RRT). CONCLUSIONS Few clinical studies of urinary biomarkers in AKI have included septic patients. However, there is promising evidence that selected biomarkers may aid in the early detection of AKI in sepsis and may have value for predicting subsequent deterioration in kidney function. Additional prospective studies are needed to accurately describe their diagnostic and prognostic value in septic AKI.
Collapse
Affiliation(s)
- Sean M Bagshaw
- Division of Critical Care Medicine, University of Alberta Hospital, 3C1.16 Walter C. Mackenzie Centre, 8440-112 Street, T6G 2B7, Edmonton, Alberta, Canada.
- Department of Intensive Care Medicine, Austin Hospital, Studley Road, 3084, Heidelberg, VIC, Australia.
| | - Christoph Langenberg
- Department of Intensive Care Medicine, Austin Hospital, Studley Road, 3084, Heidelberg, VIC, Australia
- Howard Florey Institute, University of Melbourne, 161 Barry Street, 3053, South Carlton, VIC, Australia
| | - Michael Haase
- Department of Intensive Care Medicine, Austin Hospital, Studley Road, 3084, Heidelberg, VIC, Australia
| | - Li Wan
- Department of Intensive Care Medicine, Austin Hospital, Studley Road, 3084, Heidelberg, VIC, Australia
- Howard Florey Institute, University of Melbourne, 161 Barry Street, 3053, South Carlton, VIC, Australia
| | - Clive N May
- Howard Florey Institute, University of Melbourne, 161 Barry Street, 3053, South Carlton, VIC, Australia
| | - Rinaldo Bellomo
- Department of Intensive Care Medicine, Austin Hospital, Studley Road, 3084, Heidelberg, VIC, Australia
- Department of Medicine, Melbourne University, Melbourne, Australia
| |
Collapse
|
447
|
Herzog C, Seth R, Shah SV, Kaushal GP. Role of meprin A in renal tubular epithelial cell injury. Kidney Int 2007; 71:1009-18. [PMID: 17377510 DOI: 10.1038/sj.ki.5002189] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Meprins are zinc-dependent metalloproteinases that are highly expressed in the brush-border membranes of both the kidney and the intestines. Meprins are capable of proteolytically degrading extracellular matrix proteins, proteolytically processing bioactive proteins, and play a role in inflammatory processes. In this study, the function of meprin A in the acute kidney injury (AKI) model of cisplatin nephrotoxicity was examined. Normal linear localization of meprin A in the brush border membranes of proximal tubules was altered in AKI. The meprin A alpha-subunit was detected in the urine of both control and cisplatin-treated mice. A cleaved product of the meprin A beta-subunit, undetected in the urine of control mice, was found to be significantly increased in the urine during the progression of cisplatin nephrotoxicity. The excretion of this beta-fragment was found to be before the rise in serum creatinine and blood urea nitrogen (BUN) suggesting usefulness as a biomarker for AKI. Pretreatment of mice with a meprin A inhibitor afforded protection from cisplatin nephrotoxicity as reflected by significant decreases in serum creatinine, BUN, and the excretion of kidney injury molecule-1. These decreases in serum and urine biomarkers were accompanied by significant decreases in histologic markers such as leukocyte infiltration and apoptosis. Meprin A appears to be an important therapeutic target and urinary excretion appears to be a potential biomarker of AKI.
Collapse
Affiliation(s)
- C Herzog
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | | | | |
Collapse
|
448
|
Prozialeck WC, Edwards JR. Cell adhesion molecules in chemically-induced renal injury. Pharmacol Ther 2007; 114:74-93. [PMID: 17316817 PMCID: PMC1913814 DOI: 10.1016/j.pharmthera.2007.01.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Accepted: 01/05/2007] [Indexed: 01/15/2023]
Abstract
Cell adhesion molecules are integral cell-membrane proteins that maintain cell-cell and cell-substrate adhesion and in some cases act as regulators of intracellular signaling cascades. In the kidney, cell adhesion molecules, such as the cadherins, the catenins, the zonula occludens protein-1 (ZO-1), occludin and the claudins are essential for maintaining the epithelial polarity and barrier integrity that are necessary for the normal absorption/excretion of fluid and solutes. A growing volume of evidence indicates that these cell adhesion molecules are important early targets for a variety of nephrotoxic substances including metals, drugs, and venom components. In addition, it is now widely appreciated that molecules, such as intracellular adhesion molecule-1 (ICAM-1), integrins, and selectins play important roles in the recruitment of leukocytes and inflammatory responses that are associated with nephrotoxic injury. This review summarizes the results of recent in vitro and in vivo studies indicating that these cell adhesion molecules may be primary molecular targets in many types of chemically-induced renal injury. Some of the specific agents that are discussed include cadmium (Cd), mercury (Hg), bismuth (Bi), cisplatin, aminoglycoside antibiotics, S-(1,2-dichlorovinyl)-l-cysteine (DCVC), and various venom toxins. This review also includes a discussion of the various mechanisms, by which these substances can affect cell adhesion molecules in the kidney.
Collapse
Affiliation(s)
- Walter C Prozialeck
- Department of Pharmacology, Midwestern University, 555 31st Street, Downers Grove, IL 60515, United States.
| | | |
Collapse
|
449
|
Heemskerk S, van Koppen A, van den Broek L, Poelen GJM, Wouterse AC, Dijkman HBPM, Russel FGM, Masereeuw R. Nitric oxide differentially regulates renal ATP-binding cassette transporters during endotoxemia. Pflugers Arch 2007; 454:321-34. [PMID: 17285300 PMCID: PMC1915652 DOI: 10.1007/s00424-007-0210-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2006] [Revised: 12/22/2006] [Accepted: 01/08/2007] [Indexed: 01/28/2023]
Abstract
Nitric oxide (NO) is an important regulator of renal transport processes. In the present study, we investigated the role of NO, produced by inducible NO synthase (iNOS), in the regulation of renal ATP-binding cassette (ABC) transporters in vivo during endotoxemia. Wistar–Hannover rats were injected with lipopolysaccharide (LPS+) alone or in combination with the iNOS inhibitor, aminoguanidine. Controls received detoxified LPS (LPS−). After LPS+, proximal tubular damage and a reduction in renal function were observed. Furthermore, iNOS mRNA and protein, and the amount of NO metabolites in plasma and urine, increased compared to the LPS− group. Coadministration with aminoguanidine resulted in an attenuation of iNOS induction and reduction of renal damage. Gene expression of 20 ABC transporters was determined. After LPS+, a clear up-regulation in Abca1, Abcb1/P-glycoprotein (P-gp), Abcb11/bile salt export pump (Bsep), and Abcc2/multidrug resistance protein (Mrp2) was found, whereas Abcc8 was down-regulated. Up-regulation of Abcc2/Mrp2 was accompanied by enhanced calcein excretion. Aminoguanidine attenuated the effects on transporter expression. Our data indicate that NO, produced locally by renal iNOS, regulates the expression of ABC transporters in vivo. Furthermore, we showed, for the first time, expression and subcellular localization of Abcb11/Bsep in rat kidney.
Collapse
Affiliation(s)
- Suzanne Heemskerk
- Department of Pharmacology and Toxicology (149), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Arianne van Koppen
- Department of Pharmacology and Toxicology (149), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Luc van den Broek
- Department of Pharmacology and Toxicology (149), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Geert J. M. Poelen
- Central Animal Laboratory, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Alfons C. Wouterse
- Department of Pharmacology and Toxicology (149), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Henry B. P. M. Dijkman
- Department of Pathology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Frans G. M. Russel
- Department of Pharmacology and Toxicology (149), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Rosalinde Masereeuw
- Department of Pharmacology and Toxicology (149), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
450
|
Liangos O, Perianayagam MC, Vaidya VS, Han WK, Wald R, Tighiouart H, MacKinnon RW, Li L, Balakrishnan VS, Pereira BJG, Bonventre JV, Jaber BL. Urinary N-acetyl-beta-(D)-glucosaminidase activity and kidney injury molecule-1 level are associated with adverse outcomes in acute renal failure. J Am Soc Nephrol 2007; 18:904-12. [PMID: 17267747 DOI: 10.1681/asn.2006030221] [Citation(s) in RCA: 348] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The role of urinary biomarkers of kidney injury in the prediction of adverse clinical outcomes in acute renal failure (ARF) has not been well described. The relationship between urinary N-acetyl-beta-(D)-glucosaminidase activity (NAG) and kidney injury molecule-1 (KIM-1) level and adverse clinical outcomes was evaluated prospectively in a cohort of 201 hospitalized patients with ARF. NAG was measured by spectrophotometry, and KIM-1 was measured by a microsphere-based Luminex technology. Mean Acute Physiology, Age, Chronic Health Evaluation II (APACHE II) score was 16, 43% had sepsis, 39% required dialysis, and hospital mortality was 24%. Urinary NAG and KIM-1 increased in tandem with APACHE II and Multiple Organ Failure scores. Compared with patients in the lowest quartile of NAG, the second, third, and fourth quartile groups had 3.0-fold (95% confidence interval [CI] 1.3 to 7.2), 3.7-fold (95% CI 1.6 to 8.8), and 9.1-fold (95% CI 3.7 to 22.7) higher odds, respectively, for dialysis requirement or hospital death (P < 0.001). This association persisted after adjustment for APACHE II, Multiple Organ Failure score, or the combined covariates cirrhosis, sepsis, oliguria, and mechanical ventilation. Compared with patients in the lowest quartile of KIM-1, the second, third, and fourth quartile groups had 1.4-fold (95% CI 0.6 to 3.0), 1.4-fold (95% CI 0.6 to 3.0), and 3.2-fold (95% CI 1.4 to 7.4) higher odds, respectively, for dialysis requirement or hospital death (P = 0.034). NAG or KIM-1 in combination with the covariates cirrhosis, sepsis, oliguria, and mechanical ventilation yielded an area under the receiver operator characteristic curve of 0.78 (95% CI 0.71 to 0.84) in predicting the composite outcome. Urinary markers of kidney injury such as NAG and KIM-1 can predict adverse clinical outcomes in patients with ARF.
Collapse
Affiliation(s)
- Orfeas Liangos
- Division of Nephrology, Caritas St. Elizabeth's Medical Center, 736 Cambridge Street, Boston, MA 02135, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|