Skip to main content
Clinical and Translational Science logoLink to Clinical and Translational Science
. 2011 Dec 7;5(1):93–101. doi: 10.1111/j.1752-8062.2011.00302.x

Enabling Innovative Translational Research in Acute Kidney Injury

Abolfazl Zarjou 1, Paul W Sanders 1,2, Ravindra L Mehta 3, Anupam Agarwal 1,2,
PMCID: PMC3292183  NIHMSID: NIHMS309472  PMID: 22376265

Abstract

Acute kidney injury (AKI) is a common, heterogeneous, and detrimental clinical condition that has significant attributable morbidity and mortality. Despite major advances in understanding the epidemiology, pathogenesis, and outcomes of AKI, preventive measures remain inadequate and therapeutic approaches (except for renal replacement therapy) have largely proven futile so far. Critical to the process of designing rational therapies is translational research, which involves the transition between the basic research discoveries and everyday clinical applications to prevent, diagnose, and treat human diseases. Progress in innovative approaches has been hampered due in part to the reliance on functional markers (serum creatinine and blood urea nitrogen) that are neither sensitive nor specific to diagnose AKI. This limitation has created a great deal of interest and intense investigation to identify a “troponin‐like marker” that would facilitate recognition of AKI and allow for timely implementation of the precise therapeutic agent. The other major obstacle in this field is the diverse and complex nature of AKI that involves multiple independent and overlapping pathways, making it difficult to cure AKI with a single approach. In this review, we will summarize the advances, ongoing studies, and future perspectives in the field of translational research of AKI. Clin Trans Sci 2012; Volume 5: 93–101

Keywords: kidney, nephrology, translational research

Introduction

The kidney is a complex organ that has evolved to serve a number of important functions: excretion of the waste products of the metabolism, regulation of body water and salt, maintenance of appropriate acidbalance, and secretion of a variety of hormones and autocoids. To accomplish these intricate functions, the kidney is elaborately composed of several cell types that function as a unit (the nephron) and not as individual cells. This exceptional characteristic also implies an intricate process of regeneration and hence requires special attention for early diagnosis and appropriate preventive and therapeutic approaches. This statement has unfortunately proven far more challenging, highlighting the vital need for relevant investigations to enhance the “bench to bedside” transition. 1 , 2 Even the exact definition of acute kidney injury (AKI) has been the subject of considerable debate and incongruity has long surrounded the term making cross‐comparison among studies difficult. 3 , 4 Recently, a new definition has been proposed for AKI (previously known as acute renal failure) by the AKI network (AKIN) that has been validated and gained broad consensus among clinicians. 5 This proposal defines AKI based on a >0.3 mg/dT rise or a >50% (1.5‐fold from baseline) increase in serum creatinine or the development of oliguria as defined by urine output <0.5 mL/kg/h for more than 6 hours. 5 In contrast, the RIFTE classification initially defined several stages of AKI including Risk, Injury, Failure, Toss of function, and End Stage Renal Disease, which have more recently been modified to three main stages of severity of AKI (Risk, Injury, and Failure). 6 The “Risk” criteria is defined by an increase in serum creatinine ≥1.5× baseline or decrease in glomerular filtration rate (GFR) ≥25% or a urine output of <0.5 mT/kg/h for ≥6 hours. The “Injury” is defined by an increase in serum creatinine ≥2.0× baseline or decrease in GFR ≥50% or a urine output of <0.5 mT/kg/h for ≥12 hours. Finally, “Failure” is defined by increase in serum creatinine ≥3.0× baseline or decrease in GFR ≥ 75% or an absolute serum creatinine ≥354 μmol/L with an acute rise of at least 44 μmol/L or a urine output of <0.3 mL/kg/h for ≥24 hours or anuria for ≥12 hours.

AKI is a common clinical condition encountered both in the hospital and outpatient settings and is an important cause of morbidity and mortality regardless of etiology and setting. 7 , 8 , 9 , 10 The incidence of AKI is increasing at a disturbing pace 10 , 11 and is attributed to several factors including shifts in patient demographics (older, more comorbid illness), severity of illness (multiple organ dysfunction syndrome), and AKI associated with expansion of invasive and complex medical and surgical procedures (organ transplantation, cardiac surgery). 7 , 8 Additionally, as the population continues to age, it is anticipated that there will be an even more substantial rise in the incidence of AKI. 12 , 13 It is estimated that AKI accounts for 1% of hospital admissions in the United States. 14 In addition, approximately 25% of patients in the intensive care unit (ICU) develop AKI and 5% of the patients in the ICU require renal replacement therapy among whom mortality rates range between 40% and 60%. 15 , 16 , 17 , 18 , 19 Moreover, AKI is associated with an increased length of hospital stay, which is an obvious and significant burden on the health care system. 20 , 21 For instance, total hospital cost of patients with creatinine levels of 2.0 mg/dT is increased by approximately $34,000. 21 Furthermore, because of the potential for involvement of multiple organ systems, AKI should be regarded as a systemic disease. The distant organ effects of AKI are increasingly being recognized, 22 and deterioration of renal function has a detrimental consequence on virtually all organ systems and body homeostasis. In addition, numerous primary systemic conditions affect the kidney and can present with AKI. In conjunction with the increasing recognition of clinical AKI, several advances have been made in identifying the mechanisms and pathways of disease and a number of targets have been identified in preclinical models. Despite these advances, trials in humans have not proven successful. These data underscore the urgent need of translational studies in the field of AKI. The purpose of this review is to briefly discuss the current status of translational research in AKI and provide a framework for future studies.

Translational research and AKI

Translational research is an integral part of most, if not all, fields of medicine. 23 Although translational research has been generally considered as “bench to bedside transition,” it is now considered to have a much wider scope and encompasses additional domains representing the continuum of preclinical discovery and application to humans. 24 , 25 , 26 These domains overlap in a number of aspects with an ultimate goal of improving global health. The first domain (T1) represents research findings in “basic” research 24 , 25 , 26 encompassing discovery in preclinical models, human physiology, biomarker discovery, proof of concept, and Phase 1 studies in humans. The (T2) domain provides knowledge of the performance of specific interventions in carefully selected populations to test for safety and efficacy in Phase 2 and Phase 3 clinical trials. The application of findings to general practice is encompassed in the (T3) domain and includes Phase 4 trials and health services and outcomes research. The (T4) domain comprises the study of population health and outcomes including factors such as social determinants, for example, economic conditions limiting access to care. The applicability of the spectrum of T1–T4 research to the field of AKI is schematized in Figure 1 . Although these four domains are somewhat distinct, it is implicit that the findings from each area of translational research can and should inform the design and conduct of future studies. For instance, knowledge from Phase 2 and Phase 3 studies of a new compound may result in a reassessment of the molecule in preclinical studies based on the efficacy of the compound in specific populations. Similarly, T3 research could identify areas of clinical need where new diagnostic and therapeutic molecules would improve management of patients. In this sense, translational research is not just “bench to bedside” but an integrative “bench to bedside and back” approach. In this review, we utilize this framework to delineate specific elements in AKI and provide information on resources that are currently available in this field.

Figure 1.

Figure 1

Spectrum of translational research (T1‐T4) opportunities in acute kidney injury.

Diagnostic modalities—the biomarker revolution

It is clear that implementation of any appropriate therapeutic agent first requires accurate diagnosis of the clinical condition. This is why so much effort and investment has been directed at finding a suitable biomarker to diagnose AKI. An ideal biomarker: (1) should be noninvasive and easily performed; (2) should be highly sensitive and specific to ensure timely diagnosis; and (3) must exhibit strong biomarker performance on statistical analysis. 27 , 28 , 29 , 30 Remarkable advances in other diagnostic fields of medicine have made it possible for immediate interventions with specific agents to manage life‐threatening conditions. One such example is the field of cardiology where translational research has introduced the utilization of cardiac enzymes to facilitate recognition of myocardial cell injury and development of newer therapeutic approaches that has significantly decreased the mortality and morbidity in this group of patients. 31 In contrast, the field of nephrology relies on evidence of renal dysfunction—elevation of serum creatinine and oliguria—to diagnose AKI. 27 , 30 Serum creatinine is a relatively late indicator of renal injury. This is due in part to multiple factors that can modulate serum levels, including race, gender, protein intake, certain drugs, muscle mass, and total body volume. 32 , 33 In addition, significant AKI can occur without major increases in serum creatinine levels that could be explained by increased tubular secretion or renal reserve. 34 This implies that significant changes in serum creatinine will only be detected once the kidneys have experienced at least 50% reduction in function. Moreover, the rise in serum creatinine does not by itself distinguish among pre‐, intra‐ and, postrenal causes of AKI. Another dilemma is the determination of baseline renal function in hospitalized patients. Ideally, a baseline creatinine should reflect the level of renal function at steady state prior to the hospitalization. In most studies, a baseline creatinine is not available for most of the patients and, therefore, the first creatinine measured in the hospital is considered as “baseline.” The major limitation of this concept is that creatinine level at hospital admission likely reflects the effects of the disease process that occurred before hospital admission, or during the initial phase of hospital care. 35 , 36 Hence, it is evident that there is an urgent need to introduce sensitive and specific biomarkers that indicate cellular damage and enable physicians to intervene early on and potentially limit the extent of renal injury. In fact, the American Society of Nephrology has designated the development of such biomarkers for early detection of AKI as a top research priority. 37

Over the past few years, several groups of investigators have identified novel biomarkers in experimental animals as well as humans with AKI ( Table 1 ). The utility of such biomarkers could serve multiple imperative functions. These include identification of the exact site of injury within the nephron, recognition of the specific etiologies causing AKI, and could also be a valuable tool to evaluate prognosis and response to treatment. One extensively studied example is the neutrophil gelatinase‐associated lipocalin (NGAL), which is a 25‐kDa protein that is expressed only at very low levels under physiological condition in several human tissues but its levels are markedly increased following injury in epithelial cells, including the kidney, colon, liver, and lung. 38 , 39 , 40 A growing body of evidence suggests that NGAL is one of the most upregulated genes in the kidney very early after acute injury in animal models of sepsis, ischemia, and nephrotoxic AKI. 38 , 39 , 40 Recently, an NGAL reporter mouse model was generated by inserting a double‐fusion reporter gene encoding luciferase‐2 and mCherry into the NGAL locus. 41 It was elegantly shown that the NGAL‐Luc2‐mC reporter accurately illuminates injury in vivo in real time. Strikingly, specific cells of the distal nephron were found to be the source of urinary NGAL. 41 These studies were followed with human studies in various settings including sepsis, cardiac surgery, transplant, and nephrotoxicity. 42 , 43 , 44 , 45 Another biomarker that has gained increasing interest and provided promising results is kidney injury molecule‐1 (KIM‐1). KIM‐1 is a transmembrane glycoprotein that is released into urine with proximal tubular injury. 46 This feature is intriguing because the bulk of all ischemic and toxic AKI is exerted at the proximal tubules. 47 More importantly, KIM‐1 expression is not detectable under normal circumstances, while expression is significantly upregulated in various models of AKI. 48 , 49 These include cisplatin, cyclosporine, radio contrast, ischemia/reperfusion, folic acid, and even experimental models of autosomal dominant polycystic kidney disease. 49 On the other hand, several functions have been attributed to KIM‐1 (such as a role in autophagy) that are under extensive investigation. 50 Nonetheless, KIM‐1 has vital diagnostic and predictive properties in the context of AKI. This has been confirmed not only in animal models of AKI but also in human studies. 47 , 48 , 49 , 50 Although there is a strong body of evidence that supports the potential of NGAL or KIM‐1 to be used as a standard diagnostic tool, it maybe more feasible to use a panel of multiple biomarkers for the precise and well‐timed diagnosis of AKI.

Table 1.

A partial list of emerging biomarkers for early detection of AKI.

Biomarker Source of sample Main method of detection Time of elevation in AKI References
Tubular enzymes (AP, γ‐GT, and others) Urine ELISA Intermediate 94, 95
Cystatin C Plasma Nephlometric immunoassay Intermediate 28, 96
IL‐18 Urine ELISA Intermediate 28, 95
NGAL Plasma Western blot Early 38, 39, 97
L‐FABP Urine ELISA Early 98, 99
KIM‐1 Urine ELISA Intermediate 49, 50
Netrin‐1 Urine ELISA Early 100, 101

AP: alkaline phosphatase; γ‐GT: γ‐glutamyl transferase; NGAL: neutrophil gelatinase‐associated lipocalin; L‐FABP: L‐type fatty acid‐binding protein; KIM‐1: kidney injury molecule‐1.

Recent studies by Endre et al. 51 have utilized a novel approach by using urinary biomarkers to triage intervention in the EARLYARF trial to evaluate the efficacy of erythropoietin versus placebo in incipient AKI. While the results of the intervention with erythropoietin were negative, the experimental design in this study offers a prototype for using biomarkers to trigger randomization.

Current therapeutic approaches and future targets

There has been outstanding progress in the T1 and T2 category related to AKI. Numerous studies have identified different aspects of AKI and complications related to it. This has resulted in an evolution in understanding the pathophysiological nature of AKI. 52 Despite such substantial advances the successful “bench‐bedside” transition has remained elusive and almost no meaningful progress has been made to cure or reverse AKI. Hence clinicians rely on conservative measures and renal replacement therapy for the management of AKI. Remarkable technological advances have been made in the field of hemodialysis. However, the optimal time of intervention with dialysis in AKI still remains to be clarified. The current practice prescribes early intervention based on clinical criteria and volume status rather than just biochemical parameters. With regards to the dose of dialysis, two recent large, multicenter, randomized, controlled trials examined the role of intensifying the dosage of dialysis (25–40 mL/kg/h) versus the standard prescribed dose (20 mL/kg/h). 53 , 54 Both studies concluded that increasing the dosage and frequency of dialysis did not translate to a decrement in mortality rates in critically ill patients with AKI. Nevertheless, it is important to note that the results of these studies indicate that a minimal dosing threshold must be achieved to optimize clinical outcomes. 55 Refinements in dialysis modalities including the advantage of high‐flux membranes with higher filtration rates, which are more permissive to “middle molecules,” and enhanced cytokine removal compared to low‐flux membranes and convective over pure diffusive strategies are yet to be conclusively validated. 56 , 57 , 58 , 59

The feasibility of a “bioartificial kidney” has been examined in a number of animal studies. Humes and colleagues demonstrated that a “renal assist device” composed of renal proximal tubule progenitor cells can effectively replace all the maj or functions of the kidney in acutely uremic dogs following bilateral nephrectomy 60 , 61 as well as animal models of sepsis. 62 These exciting reports led to Phase 1 and 2 clinical trials that tested the applicability, safety, and efficacy of this novel approach in humans. 63 Unfortunately, the pivotal Phase 3 trial was terminated early due to concerns of higher mortality in the treatment group potentially related to some changes in the protocol. These studies highlight the difficulties in transitioning from small Phase 2 to larger Phase 3 studies. Although this approach could be potentially beneficial, larger, randomized, multicenter trials are required to unequivocally confirm its role as a novel therapeutic modality in AKI.

Because of the vast nature of kidney‐offending conditions and agents, numerous pathways have been proposed to prevent, minimize, or reverse AKI. For instance, extensive studies targeted different deranged pathways during sepsis‐induced AKI. These targets include inhibition of inflammatory mediators, enhancement of renal perfusion by blocking vasoconstrictor mechanisms and intensifying vasodilator mechanisms, attenuation of leukocyte infiltration, inhibition of the coagulation cascade, and administration of growth factors to accelerate renal recovery. 64 , 65 , 66 Most of the mentioned attempts provided solid evidence in animal models of sepsis‐induced AKI but unfortunately none could provide beneficial effects in this group of patients. In fact, only activated protein C 67 and steroid replacement therapy 68 have been shown to reduce mortality in patients with sepsis and are now accepted adjunctive treatment options for sepsis in general. 64 , 66

Another example is the pharmacological interventions that were designed to prevent renal ischemia or modulate the ensuing inflammatory or hormonal milieu. Low‐dose dopamine, long thought to prevent AKI by improving renal perfusion, was shown to have no effect on mortality and renal replacement requirement. 69 , 70 Another vasoactive hormone, atrial natriuretic peptide that enhances glomerular filtration rate by dilating the afferent and constricting the efferent arteriole (thereby increasing the intraglomerular hydrostatic pressure and subsequently promoting filtration), 71 seemed to be an appropriate therapeutic option. Although there appeared to be a trend toward improvement of AKI, this peptide also promoted hypotension and arrhythmias in high doses and the beneficial potential is still a subject of ongoing debate. 72 , 73 On the other hand, recombinant human insulin‐like growth factor‐1 that showed encouraging results at decreasing renal tubular apoptosis and inflammation in mice when administered immediately after renal ischemia 74 failed to show any benefit in a small randomized control trial of 72 critically ill patients with AKI. 75

In spite of challenges and diverse failures, some exciting results and achievements have also emerged in the recent years. Knowledge gained from preclinical models has led to the initiation of several clinical studies targeting specific pathways. Phase 1 studies ( Table 2 ) are most commonly pursued in cardiac surgery patients where the timing and nature of insult can be well defined. In contrast, Phase 2 and 3 studies ( Table 3 ) target a broader segment of AKI but the preponderance is in settings where the nature of the insult can be defined, for example, contrast nephropathy.

Table 2.

Tl (Phase 1) Drug studies currently underway for AKI*.

Clinical trials identifier Study sponsor Drug/biologic intervention Setting Target
NCT00683553 Quark Pharmaceuticals 15NP (small interfering RNA) Cardiac surgery Apoptosis
NCT00733876 AlloCure Inc. Mesenchymal stem cells On‐pump cardiac surgery Repair
NCT00821522 Maine Medical Center Research Institute Remote ischemic preconditioning Cardiac surgery Primary prevention in ischemia reperfusion injury
NCT01275612 Mario Negri Institute for Pharmacological Research Ex vivo expanded mesenchymal stem cells Cisplatin‐induced AKI Repair
NCT00861822 University Health Network, Toronto RBC transfusion presurgery Cardiac surgery Oxygen delivery
NCT00878956 Austin Health Bicarbonate Cardiac surgery Oxidative stress

Table 3.

T2 studies (Phase 2 and 3) in AKI*.

Clinical trials identifier Study sponsor Drug/biologic intervention Setting Target
NCT00878956 Austin Health Bicarbonate Cardiac surgery Oxidative stress
NCT01251029 Assaf‐Harofeh Medical Center N‐acetylcysteine Major surgery under general anesthesia Oxidative stress
NCT01225094 Lawson Health Research Institute Curcumin Elective Abdominal Aortic Aneurysm Repair Antiinflammatory
NCT01066351 Thammasat University Beta‐erythropoietin CKD patients cardiac surgery Antiapoptotic and anti‐inflammatory
NCT00982527 Bambino Gesù Hospital and Research Institute Fenoldopam Pediatric cardiac surgery Dopamine D‐l receptor
NCT00557219 Medical University of Gdansk Fenoldopam and Ketanserin Cardiac surgery Dopamine D‐1 receptor agonist Adrenergic blocker and platelet aggregation
NCT01072682 CytoPherx, Inc Selective cytopheretic device (SCD) ICU AKI Immunomodulation
NCT01245595 Stanford University Aminophylline Pediatrie cardiac surgery Phosphodiesterase inhibitors
NCT01146925 CorMedix CRMD‐001‐Deferiprone Cardiac angio contrast exposure Oxidative stress
NCT00791648 Vanderbilt University Atorvastatin Cardiac surgery Oxidative stress
NCT00978354 University of Alberta Furosemide ICU AKI Diuresis
NCT01256372 Action Pharma A/S AP214 Cardiac surgery Inflammation

One promising therapy is the utilization of mesenchymal stem cells (MSC) as a novel therapeutic modality in AKI. 76 , 77 MSC are unique and versatile multipotent cells that possess antiinflammatory and immunomdulatory properties. 78 , 79 These characteristics have ignited great interest and extensive study of MSC in different clinical conditions. MSC have proven beneficial in numerous animal models of injury including myocardial ischemia, 80 inflammatory lung diseases, 81 sepsis, 82 and AKI. 76 , 77 It is now evident that MSC mainly exert their beneficial effects via endocrine and paracrine and differentiation‐independent mechanisms. 83 , 84 These and other findings have led to exciting clinical trials that are evaluating the efficacy and safety of administration of MSC in cardiac surgery‐related AKI (http://clinicaltrials.gov,NCT00733876).

Another potential approach is the utilization of endogenous antioxidative enzymes. One such enzyme is HO‐1 that is a stress inducible enzyme. 85 HO‐1 cleaves the porphyrin ring at the alpha‐methene bridge to form equimolar amounts of iron, biliverdin, and carbon monoxide. 86 Biliverdin is then converted to bilirubin by biliverdin reductase. 87 HO‐1 has important antiapoptotic and antiinflammatory functions that have been attributed to one (or more) of its byproducts. 88 Induction of HO‐1 has been shown to be protective in several forms ofinjury including AKI. 89 Furthermore, evidence suggests that polymorphism of HO‐1 (short GT repeats) in humans is associated with better clinical outcome in various conditions including renal transplantation 90 and arteriovenous fistula patency in hemodialysis patients. 91 There are now ongoing clinical trials that are examining the beneficial effects of HO‐1 byproducts including carbon monoxide in kidney transplantation (http://clinicaltrials.gov,NCT00531856) and bilirubin in endotoxemia (http://clinicaltrials.gov,NCT00916448).

As mentioned, many therapeutic agents that showed great potential in animals failed to complete the translational steps. This may have various explanations most likely of which is the fact that these agents were utilized only after AKI was established. This further highlights the urgent need for novel biomarkers to diagnose AKI so that proper treatment can be implemented in a timely and accurate manner.

What is needed to enhance translational research in AKI?

It is evident from the discussion above that progress in the field requires sharing of information across the domains of basic discovery and clinical care. Future clinical and translational research in AKI will require the development of collaborative networks of investigators drawn from various disciplines who are willing to share their expertise and resources. Since AKI is a heterogeneous disorder, like sepsis, it is helpful to consider the scope of activities that are currently required to enhance fundamental knowledge for the diagnosis and treatment of this disorder. Based upon this information, we can develop strategies for facilitating clinical and translational research activities and define the infrastructure that will be needed. Table 4 describes some of the main elements that need to be considered. An approach is to build a network of centers that will be responsible for developing a databank of AKI patients that are carefully characterized with respect to their clinical course, alterations in renal function, and various markers of renal injury and renal pathology. Given the wide variation in the natural history and management of this disorder globally, it is essential that we develop mechanisms for sharing information and for collaboration among centers. Several key issues will need to be addressed including the infrastructure needed (database, protocols for web‐based information transfer, etc.) the requirements for sharing information with regulatory issues, training needs for developing researchers and the resources that are required, and what will be the hurdles to be overcome in establishing such networks. A key aspect of these networks is that the number of centers involved varies for different activities, for example, all centers could participate in the registry function, but only a few in longitudinal data collection and some centers may only participate in clinical trials activity.

Table 4.

Elements for clinical investigation in AKI and requirements for translational research.

Component Reason Proposed tools Infrastructure required Examples from other areas
Observational studies
Cross‐sectional Prevalence of disease in various settings is not well known. Do not have knowledge of key elements, e.g., renal pathology Develop a registry of patients with AKI Web‐based techniques for minimal data collection, HIPAA and IRB compliant, consent issues. Coordination center for data analysis and reports. PICARD network, USRDS (registry model), sepsis, and critical care Registries. Pediatric CRRT Registry, Latin America AKI Registry
Longitudinal Incidence of disease. Establish natural history of ARF; characterize the phenotype and genotype of patients with AKI, establish relationships of various patient and process factors with outcomes. Collect biological samples Develop a longitudinal dataset that includes biological samples (including renal biopsy material) collected at specific time points in the course of AKI Web‐based techniques for data acquisition, HIPAA compliance, informed consent, Data coordination center, biological sample repository. Data analysis Diabetes GoKIND study, AIDS collaborative networks, PICARD network, O'Brien AKI Registry*
Intervention studies
Single/multi‐center clinical research Establish mechanisms of disease Develop translational research networks linking basic scientists to clinicians to evaluate specific diagnostic interventions and therapeutic targets in limited numbers of patients, e.g., Physiological studies Link basic scientists working on AKI with clinical centers with patients PICARD network, Glomerulo‐nephritis network
Single‐center clinical trials Phase 1 and 2 clinical trials Develop mechanisms to support safety and efficacy and proof of concept trials in well‐characterized patients with AKI CTSA resources, Biological sample processing. Industry‐sponsored studies, AKIN network
Multicenter clinical trials Phase 2–4 trials Develop networks of centers with access to large numbers of patients with AKI for efficacy and effectiveness trials. Clinical trial protocols, resources for trial implementation DCC for CRF design and development data acquisition, and analysis. VA‐NIH clinical trial for dialysis in AKI; Hemo trial ARDS network, AKIN
Health services research
Evidence‐based guidelines Best practices for management of AKI Develop evidence‐based guidelines Compilation and analysis and grading of available evidence, recommendations for nondialytic and dialytic management of AKI, treatment algorithms ADQI, KDIGO
Outcomes research Health economics and relationship of AKI to development of CKD Establish benefit‐cost and cost effectiveness of various strategies for AKI. Establish links of AKI to CKD Mechanisms to collect data prospectively on costs and outcomes from AKI USRDS for CKD, sepsis trials, large database analysis, e.g., Kaiser, Veterans Medical Centers
Comparative effectiveness research Generation and synthesis of evidence that compares the benefits and harms of alternate methods to prevent, diagnose, treat, and monitor a clinical condition or to improve the delivery of care eg Dialysis timing, dose modality in AKI Systematic reviews AKI Registry for longitudinal observational data ADHERE heart failure Registry, O'Brien AKI Registry*
Experimental studies including RCTs Data coordination for support of multicenter RCTs
Observation studies including Registries Statistical support
Retrospective analysis including administrative data Decision modeling
Decision support

*UAB‐UCSD O'Brien Center (http://www.ObrienAKI.org).

One major barrier for clinical trials in AKI is related to the selection of appropriate endpoints for outcome measures. Most clinical trials rely on one or more criteria that relate to (1) changes in serum creatinine (doubling or a 50% increase in creatinine from baseline levels), (2) dialysis initiation, (3) impact on length of stay in the ICU, and (3) overall mortality. Each of these criteria has inherent problems. The criteria involving change from baseline creatinine would vary depending on the selection of actual “baseline” value, which have not been consistent. Studies have used the first admission creatinine, inpatient nadir, or an outpatient creatinine level to define baseline values and depending on which value is used, the end point of doubling or 50% increase in serum creatinine may not be an accurate reflection of the status of AKI in a given patient. 35 The timing of dialysis initiation in AKI is highly variable and influenced by not only comorbidities but also variations in decision‐making processes of physicians taking care of these patients. 92 The length of stay in the hospital or ICU and overall mortality are dependent on several factors and not solely on the status of AKI. For example, renal function following a given therapeutic intervention may improve, but the patient could succumb to nonrenal causes (e.g., respiratory or cardiac failure). These barriers need to be carefully considered and better endpoints (possibly through the use of novel biomarkers) would certainly improve clinical trial designs.

Recognizing the need to accelerate research in this field, several resources have been developed to assist with the complexities associated with the pathogenesis and treatment of AKI. The University of Alabama, Birmingham and the University of California, San Diego NIH‐NIDDK funded O'Brien Center for AKI research (http://www.obrienaki.org) offers some of the infrastructure described for this purpose and more details are provided in Table 4 . The center provides multiple interdisciplinary cores that cover all areas of translational research. The prospects provided for the T1 category include development and training in the use of rodent models of AKI, multimodality small animal imaging core for state‐of‐the‐art molecular imaging and a physiology core. The physiology core provides expertise including measurements of GFR, tubular reabsorption, renal hemodynamics, and isolation of primary renal and vascular cells in culture from rodents.

The objective of the Clinical Research Core (O'Brien Core A) is to provide resources to enable interdisciplinary clinical investigation in AKI that will advance our understanding of the natural history and pathophysiology of human AKI, ascertain genetic contributions for susceptibility and prognosis of AKI, enhance our diagnostic specificity, and expand our preventive and therapeutic approaches for this disorder. The core provides a Web‐based database for the conduct of a wide range of observational and interventional clinical studies in AKI and is flexible in accommodating the research objectives of individual investigators and collaborating centers. This database has been used to establish a multicenter international registry of AKI that currently has 13 centers participating. Additionally, the core provides a genomics resource to facilitate the investigation of genetic determinants (both risk and outcome) of AKI, and enable investigators to perform systematic genomic analyses to allow correlation with clinical phenotypic information. Furthermore, the core provides access to various biomarker assays and a biological sample repository linked to the clinical database to enable further characterization of patients. Moreover, the center is actively involved in education and training as well as multiple registry projects that cover different areas of T2‐T4 categories. Finally, recognizing the need not only for continued bench‐type research but also the call for an integrative “bench to bedside and back” approach to AKI, the O'Brien Center also provides a core for Preclinical Studies in AKI (Core B).

Conclusion

AKI remains a serious clinical problem, with an alarming rate of morbidity, mortality, and rising incidence. Although investigators at all levels of translational research related to AKI have made important contributions, most of these have failed to translate into clinical care for patients with AKI. Diverse factors have thwarted progress in developing effective therapeutics for AKI. 93 These include (1) a reliance on traditional diagnostic tools that have major limitations for precise and judicious implementation of therapeutic agents, (2) the complex pathophysiology of AKI with involvement of multiple overlapping and temporal pathways, (3) heterogeneous patient population, and (4) lack of standardized clinical trial designs. The availability of novel biomarkers and improved knowledge of the natural history of AKI will continue to enhance our understanding of this complex disease. We believe that improving outcomes from AKI will require a concerted effort to enhance collaborative research and facilitate translation of knowledge not only from the “bench to bedside” but from the “bedside to the bench and back” (B2B). Several resources are now available through the NIDDK‐funded UAB‐UCSD O'Brien Center for AKI research to facilitate this process. We hope that this review will stimulate additional interest in the field and spark investigators to leverage these resources.

Acknowledgments

The work was supported by NIH grants R01 DK059600, R01 DK075332 and O'Brien Center P30 DK079337 (to AA). The authors have no conflicting financial interests.

References

  • 1. Rondon‐Berrios H, Palevsky PM. Treatment of acute kidney injury: an update on the management of renal replacement therapy. Curr Opin Nephrol Hypertens. 2007; 16(2): 64–70. [DOI] [PubMed] [Google Scholar]
  • 2. Himmelfarb J, Ikizler TA. Acute kidney injury: changing lexicography, definitions, and epidemiology. Kidney Int. 2007; 71 (10): 971–976. [DOI] [PubMed] [Google Scholar]
  • 3. Srisawat N, Hoste EE, Kellum JA. Modern classification of acute kidney injury. Blood Purif. 2010; 29(3): 300–307. [DOI] [PubMed] [Google Scholar]
  • 4. Kellum JA, Bellomo R, Ronco C. Definition and classification of acute kidney injury. Nephron Clin Pract. 2008; 109(4): c182–c187. [DOI] [PubMed] [Google Scholar]
  • 5. Mehta RL, Kellum JA, Shah SV, Molitoris BA, Ronco C, Warnock DG, Levin A. Acute Kidney Injury Network: report of an initiative to improve outcomes in acute kidney injury. Crit Care. 2007; 11(2): R31 Available at: http://ccforum.com/content/11/2/R31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Ricci Z, Cruz D, Ronco C. The RIFLE criteria and mortality in acute kidney injury: a systematic review. Kidney Int. 2008; 73(5): 538–546. [DOI] [PubMed] [Google Scholar]
  • 7. Hoste EA, Schurgers M. Epidemiology of acute kidney injury: how big is the problem? Crit Care Med. 2008; 36(Suppl 4): S146–151. [DOI] [PubMed] [Google Scholar]
  • 8. Zappitelli M. Epidemiology and diagnosis of acute kidney injury. Semin Nephrol. 2008; 28(5) 436–446. [DOI] [PubMed] [Google Scholar]
  • 9. Uchino S. The epidemiology of acute renal failure in the world. Curr Opin Crit Care. 2006; 12(6): 538–543. [DOI] [PubMed] [Google Scholar]
  • 10. Xue JL, Daniels F, Star RA, Kimmel PL, Eggers PW, Molitoris BA, Himmelfarb J, Collins AJ. Incidence and mortality of acute renal failure in Medicare beneficiaries, 1992 to 2001. J Am Soc Nephrol. 2006; 17(4): 1135–1142. [DOI] [PubMed] [Google Scholar]
  • 11. Goldberg R, Dennen P. Long‐term outcomes of acute kidney injury. Adv Chronic Kidney Dis. 2008; 15(3): 297–307. [DOI] [PubMed] [Google Scholar]
  • 12. Rosner MH. The pathogenesis of susceptibility to acute kidney injury in the elderly. Curr Aging Sci. 2009; 2(2): 158–164. [DOI] [PubMed] [Google Scholar]
  • 13. Weinstein JR, Anderson S. The aging kidney: physiological changes. Adv Chronic Kidney Dis. 2010; 17(4): 302–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Kaufman J, Dhakal M, Patel B, Hamburger R. Community‐acquired acute renal failure. Am J Kidney Dis. 1991; 17(2): 191–198. [DOI] [PubMed] [Google Scholar]
  • 15. Schiffl H. Renal recovery from acute tubular necrosis requiring renal replacement therapy: a prospective study in critically ill patients. Nephrol Dial Transplant. 2006; 21 (5): 1248–1252. [DOI] [PubMed] [Google Scholar]
  • 16. Uchino S, Kellum JA, Bellomo R, Doig GS, Morimatsu H, Morgera S, Schetz M, Tan I, Bouman C, E Macedo, al et. Acute renal failure in critically ill patients: a multinational, multicenter study. JAMA. 2005; 294(7): 813–818. [DOI] [PubMed] [Google Scholar]
  • 17. McCarthy JT. Prognosis of patients with acute renal failure in the intensive‐care unit: a tale of two eras. Mayo Clin Proc. 1996; 71(2): 117–126. [DOI] [PubMed] [Google Scholar]
  • 18. de Mendonca A, Vincent JL, Suter PM, Moreno R, Dearden NM, Antonelli M, Takala J, Sprung C, Cantraine F. Acute renal failure in the ICU: risk factors and outcome evaluated by the SOFA score. Intensive Care Med. 2000; 26(7): 915–921. [DOI] [PubMed] [Google Scholar]
  • 19. Metnitz PG, Krenn CG, Steltzer H, Lang T, Ploder J, Lenz K, Le Gall JR, Druml W. Effect of acute renal failure requiring renal replacement therapy on outcome in critically ill patients. Crit Care Med. 2002; 30(9): 2051–2058. [DOI] [PubMed] [Google Scholar]
  • 20. Coca SG, Peixoto AJ, Garg AX, Krumholz HM, Parikh CR. The prognostic importance of a small acute decrement in kidney function in hospitalized patients: a systematic review and meta‐analysis. Am J Kidney Dis. 2007; 50(5): 712–720. [DOI] [PubMed] [Google Scholar]
  • 21. Chertow GM, Burdick E, Honour M, Bonventre JV, Bates DW. Acute kidney injury, mortality, length of stay, and costs in hospitalized patients. J Am Soc Nephrol. 2005; 16(11): 3365–3370. [DOI] [PubMed] [Google Scholar]
  • 22. Kelly KJ. Distant effects of experimental renal ischemia/reperfusion injury. J Am Soc Nephrol. 2003; 14(6): 1549–1558. [DOI] [PubMed] [Google Scholar]
  • 23. Zerhouni EA. Translational research: moving discovery to practice. Clin Pharmacol Ther. 2007; 81(1): 126–128. [DOI] [PubMed] [Google Scholar]
  • 24. Woolf SH. The meaning of translational research and why it matters. JAMA. 2008; 299(2): 211–213. [DOI] [PubMed] [Google Scholar]
  • 25. Westfall JM, Mold J, Fagnan L. Practice‐based research‐“Blue Highways” on the NIH roadmap. JAMA. 2007; 297(4): 403–406. [DOI] [PubMed] [Google Scholar]
  • 26. Feldman AM. The clinical and translational science “tent”. Clin Transl Sci. 2010; 3(1): 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Edelstein CL. Biomarkers of acute kidney injury. Adv Chronic Kidney Dis. 2008; 15(3): 222–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Coca SG, Yalavarthy R, Concato J, Parikh CR. Biomarkers for the diagnosis and risk stratification of acute kidney injury: a systematic review. Kidney Int. 2008; 73(9): 1008–1016. [DOI] [PubMed] [Google Scholar]
  • 29. Lisowska‐Myjak B. Serum and urinary biomarkers of acute kidney injury. Blood Purif. 2010; 29(4): 357–365. [DOI] [PubMed] [Google Scholar]
  • 30. Parikh CR, Devarajan P. New biomarkers of acute kidney injury. Crit Care Med. 2008; 36(Sup‐pl4):S159–S165. [DOI] [PubMed] [Google Scholar]
  • 31. Chan D, Ng LL. Biomarkers in acute myocardial infarction. BMC Med. 2010; 8: 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Tomlanovich S, Golbetz H, Perlroth M, Stinson E, Myers BD. Limitations of creatinine in quantifying the severity of cyclosporine‐induced chronic nephropathy. Am J Kidney Dis. 1986; 8(5): 332–337. [DOI] [PubMed] [Google Scholar]
  • 33. Bjornsson TD. Use of serum creatinine concentrations to determine renal function. Clin Phar-macokinet. 1979; 4(3): 200–222. [DOI] [PubMed] [Google Scholar]
  • 34. Herrera J, Rodriguez‐lturbe B. Stimulation of tubular secretion of creatinine in health and in conditions associated with reduced nephron mass. Evidence for a tubular functional reserve. Nephrol Dial Transplant. 1998; 13(3): 623–629. [DOI] [PubMed] [Google Scholar]
  • 35. Siew ED, Matheny ME, Ikizler TA, Lewis JB, Miller RA, Waitman LR, Go AS, Parikh CR, Peterson JF. Commonly used surrogates for baseline renal function affect the classification and prognosis of acute kidney injury. Kidney Int. 2010; 77(6): 536–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Bagshaw SM, Gibney RT. Conventional markers of kidney function. Crit Care Med. 2008; 36(Suppl 4): S152–S158. [DOI] [PubMed] [Google Scholar]
  • 37. American Society of Nephrology Renal Research Report . J Am Soc Nephrol. 2005; 16(7): 1886–1903. [DOI] [PubMed] [Google Scholar]
  • 38. Devarajan P. Review: neutrophil gelatinase‐associated lipocalin: a troponin‐like biomarker for human acute kidney injury. Nephrology (Carlton). 2010; 15(4): 419–428. [DOI] [PubMed] [Google Scholar]
  • 39. Di Grande A, Giuffrida C, Carpinteri G, Narbone G, Pirrone G, Di Mauro A, Calandra S, Noto P, Le Moli C, B Alongi, al et. Neutrophil gelatinase‐associated lipocalin: a novel biomarker for the early diagnosis of acute kidney injury in the emergency department. Eur Rev Med Pharmacol Sci. 2009; 13(3): 197–200. [PubMed] [Google Scholar]
  • 40. Ronco C. N‐GAL: diagnosing AKI as soon as possible. Crit Care. 2007; 11 (6): 173 Available at: http://ccforum.com/content/11/6/173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Paragas N, Qiu A, Zhang Q, Samstein B, Deng SX, Schmidt‐Ott KM, Viltard M, Yu W, Forster CS, G Gong, al et. The Ngal reporter mouse detects the response of the kidney to injury in real time. Nat Med. 2011; 17(2): 216–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Przybylowski P, Malyszko J, Malyszko JS. Serum neutrophil gelatinase‐associated lipocalin correlates with kidney function in heart allograft recipients. Transplant Proc. 2010; 42(5): 1797–1802. [DOI] [PubMed] [Google Scholar]
  • 43. Moore E, Bellomo R, Nichol A. Biomarkers of acute kidney injury in anesthesia, intensive care and major surgery: from the bench to clinical research to clinical practice. Minerva Anestesiol. 2010; 76(6): 425–440. [PubMed] [Google Scholar]
  • 44. Portal AJ, McPhail MJ, Bruce M, Coltart I, Slack A, Sherwood R, Heaton ND, Shawcross D, Wendon JA, Heneghan MA. Neutrophil gelatinase‐associated lipocalin predicts acute kidney injury in patients undergoing liver transplantation. Liver Transpl. 2010; 16(11): 1257–1266. [DOI] [PubMed] [Google Scholar]
  • 45. Hollmen ME, Kyllonen LE, Inkinen KA, Lalla ML, Salmela KT. Urine neutrophil gelatinase‐associated lipocalin is a marker of graft recovery after kidney transplantation. Kidney Int. 2010; 79: 89–98. [DOI] [PubMed] [Google Scholar]
  • 46. Bailly V, Zhang Z, Meier W, Cate R, Sanicola M, Bonventre JV. Shedding of kidney injury mole‐cule‐1, a putative adhesion protein involved in renal regeneration. J Biol Chem. 2002; 277(42): 39739–39748. [DOI] [PubMed] [Google Scholar]
  • 47. van Timmeren MM, van den Heuvel MC, Bailly V, Bakker SJ, van Goor H, Stegeman CA. Tubular kidney injury molecule‐1 (KIM‐1) in human renal disease. J Pathol. 2007; 212(2): 209–217. [DOI] [PubMed] [Google Scholar]
  • 48. Bonventre JV. Kidney Injury Molecule‐1 (KIM‐1): a specific and sensitive biomarker of kidney injury. Scand J Clin Lab Invest Suppl. 2008; 241: 78–83. [DOI] [PubMed] [Google Scholar]
  • 49. Waanders F, van Timmeren MM, Stegeman CA, Bakker SJ, van Goor H. Kidney injury molecule‐1 in renal disease. J Pathol. 2010; 220(1): 7–16. [DOI] [PubMed] [Google Scholar]
  • 50. Huo W, Zhang K, Nie Z, Li Q, Jin F. Kidney injury molecule‐1 (KIM‐1): a novel kidney‐specific injury molecule playing potential double‐edged functions in kidney injury. Transplant Rev (Orlando). 2010; 24(3): 143–146. [DOI] [PubMed] [Google Scholar]
  • 51. Endre ZH, Walker RJ, Pickering JW, Shaw GM, Frampton CM, Henderson SJ, Hutchison R, Mehrtens JE, Robinson JM, JB Schollum, al et. Early intervention with erythropoietin does not affect the outcome of acute kidney injury (the EARLYARF trial). Kidney Int. 2010; 77(11): 1020–1030. [DOI] [PubMed] [Google Scholar]
  • 52. Lattanzio MR, Kopyt NP. Acute kidney injury: new concepts in definition, diagnosis, pathophy‐siology, and treatment. J Am Osteopath Assoc. 2009; 109(1): 13–19. [PubMed] [Google Scholar]
  • 53. Palevsky PM, Zhang JH, O'Connor TZ, Chertow GM, Crowley ST, Choudhury D, Finkel K, Kellum JA, Paganini E, RM Schein, al et. Intensity of renal support in critically ill patients with acute kidney injury. N Engl J Med. 2008; 359(1): 7–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Tolwani AJ, Campbell RC, Stofan BS, Lai KR, Oster RA, Wille KM. Standard versus high‐dose CVVHDF for ICU‐related acute renal failure. J Am Soc Nephrol. 2008; 19(6): 1233–1238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Palevsky PM. Renal support in acute kidney injury‐how much is enough? N Engl J Med. 2009; 361(17): 1699–1701. [DOI] [PubMed] [Google Scholar]
  • 56. de Francisco AL, Pinera C. Challenges and future of renal replacement therapy. Hemodial Int. 2006; 10(Suppl 1):S19–S23. [DOI] [PubMed] [Google Scholar]
  • 57. Jacobs C. Renal replacement therapy by hemodialysis: an overview. Nephrol Ther. 2009; 5(4): 306–312. [DOI] [PubMed] [Google Scholar]
  • 58. Lameire N, Van Biesen W, Vanholder R. Did 20 years of technological innovations in hemodialysis contribute to better patient outcomes? Clin J Am Soc Nephrol. 2009; 4(Suppl 1): S30–S40. [DOI] [PubMed] [Google Scholar]
  • 59. Schiffl H. Daily haemodialysis in acute renal failure. Old wine in a new bottle? Minerva Urol Nefrol. 2004; 56(3): 265–277. [PubMed] [Google Scholar]
  • 60. Humes HD, Buffington DA, MacKay SM, Funke AJ, Weitzel WF. Replacement of renal function in uremic animals with a tissue‐engineered kidney. Nat Biotechnol. 1999; 17(5): 451–455. [DOI] [PubMed] [Google Scholar]
  • 61. Humes HD, MacKay SM, Funke AJ, Buffington DA. Tissue engineering of a bioartificial renal tubule assist device: in vitro transport and metabolic characteristics. Kidney Int. 1999; 55(6): 2502–2514. [DOI] [PubMed] [Google Scholar]
  • 62. Fissell WH, Dyke DB, Weitzel WF, Buffington DA, Westover AJ, MacKay SM, Gutierrez JM, Humes HD. Bioartificial kidney alters cytokine response and hemodynamics in endotoxin‐challenged uremic animals. Blood Purif. 2002; 20(1): 55–60. [DOI] [PubMed] [Google Scholar]
  • 63. Ding F, Humes HD. The bioartificial kidney and bioengineered membranes in acute kidney injury. Nephron Exp Nephrol. 2008; 109(4): el18–el22. [DOI] [PubMed] [Google Scholar]
  • 64. De Vriese AS, Bourgeois M. Pharmacologic treatment of acute renal failure in sepsis. Curr Opin Crit Care. 2003; 9(6): 474–480. [DOI] [PubMed] [Google Scholar]
  • 65. Pelte CH, Chawla LS. Novel therapeutic targets for prevention and therapy of sepsis associated acute kidney injury. Curr Drug Targets. 2009; 10(12): 1205–1211. [DOI] [PubMed] [Google Scholar]
  • 66. Polderman KH, Girbes AR. Drug intervention trials in sepsis: divergent results. Lancet. 2004; 363(9422): 1721–1723. [DOI] [PubMed] [Google Scholar]
  • 67. Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF, Lopez‐Rodriguez A, Steingrub JS, Garber GE, Helterbrand JD, EW Ely, al et. Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med. 2001; 344(10): 699–709. [DOI] [PubMed] [Google Scholar]
  • 68. Annane D, Sebille V, Charpentier C, Bollaert PE, Francois B, Korach JM, Capellier G, Cohen Y, Azoulay E, G Troche, al et. Effect of treatment with low doses of hydrocortisone and fludrocortisone on mortality in patients with septic shock. JAMA. 2002; 288(7): 862–871. [DOI] [PubMed] [Google Scholar]
  • 69. Bellomo R, Chapman M, Finfer S, Hickling K, Myburgh J. Low‐dose dopamine in patients with early renal dysfunction: a placebo‐controlled randomised trial. Australian and New Zealand Intensive Care Society (ANZICS) Clinical Trials Group. Lancet. 2000; 356(9248): 2139–2143. [DOI] [PubMed] [Google Scholar]
  • 70. Friedrich JO, Adhikari N, Herridge MS, Beyene J. Meta‐analysis: low‐dose dopamine increases urine output but does not prevent renal dysfunction or death. Ann Intern Med. 2005; 142(7): 510–524. [DOI] [PubMed] [Google Scholar]
  • 71. Knepper MA, Lankford SP, Terada Y. Renal tubular actions of ANF. Can J Physiol Pharmacol. 1991; 69(10): 1537–1545. [DOI] [PubMed] [Google Scholar]
  • 72. Nigwekar SU, Navaneethan SD, Parikh CR, Hix JK. Atrial natriuretic peptide for preventing and treating acute kidney injury. Cochrane Database Syst Rev 2009(4):CD006028. [DOI] [PubMed]
  • 73. Nigwekar SU, Navaneethan SD, Parikh CR, Hix JK. Atrial natriuretic peptide for management of acute kidney injury: a systematic review and meta‐analysis. Clin J Am Soc Nephrol. 2009; 4(2): 261–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Goes N, Urmson J, Vincent D, Ramassar V, Halloran PF. Effect of recombinant human insulinlike growth factor‐1 on the inflammatory response to acute renal injury. J Am Soc Nephrol. 1996; 7(5): 710–720. [DOI] [PubMed] [Google Scholar]
  • 75. Hirschberg R, Kopple J, Lipsett P, Benjamin E, Minei J, Albertson T, Munger M, Metzler M, Zaloga G, M Murray, al et. Multicenter clinical trial of recombinant human insulin‐like growth factor I in patients with acute renal failure. Kidney Int. 1999; 55(6): 2423–2432. [DOI] [PubMed] [Google Scholar]
  • 76. Imai N, Kaur T, Rosenberg ME, Gupta S. Cellular therapy of kidney diseases. Semin Dial. 2009; 22(6): 629–635. [DOI] [PubMed] [Google Scholar]
  • 77. Humphreys BD, Bonventre JV. Mesenchymal stem cells in acute kidney injury. Annu Rev Med. 2008; 59:311–325. [DOI] [PubMed] [Google Scholar]
  • 78. Jones BJ, McTaggart SJ. Immunosuppression by mesenchymal stromal cells: from culture to clinic. Exp Hematol. 2008; 36(6): 733–741. [DOI] [PubMed] [Google Scholar]
  • 79. Bunnell BA, Betancourt AM, Sullivan DE. New concepts on the immune modulation mediated by mesenchymal stem cells. Stem Cell Res Ther. 2010; 1 (5): 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Schafer R, Northoff H. Cardioprotection and cardiac regeneration by mesenchymal stem cells. Panminerva Med. 2008; 50(1): 31–39. [PubMed] [Google Scholar]
  • 81. Iyer SS, Co C, Rojas M. Mesenchymal stem cells and inflammatory lung diseases. Panminerva Med. 2009; 51(1): 5–16. [PubMed] [Google Scholar]
  • 82. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, Robey PG, Leelahavanichkul K, Koller BH, JM Brown, al et. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)‐dependent reprogramming of host macrophages to increase their interleukin‐10 production. Nat Med. 2009; 15(1): 42–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Togel F, Hu Z, Weiss K, Isaac J, Lange C, Westenfelder C. Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation‐independent mechanisms. Am J Physiol Renal Physiol. 2005; 289(1): F31–F42. [DOI] [PubMed] [Google Scholar]
  • 84. Zarjou A, Kim J, Traylor AM, Sanders PW, Balla J, Agarwal A, Curtis LM. Paracrine effects of mesenchymal stem cells in cisplatin‐induced renal injury require heme oxygenase‐1. Am J Physiol Renal Physiol. 2010; 300: 254–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Hill‐Kapturczak N, Chang SH, Agarwal A. Heme oxygenase and the kidney. DNA Cell Biol. 2002; 21 (4): 307–321. [DOI] [PubMed] [Google Scholar]
  • 86. Nath KA. Heme oxygenase‐1: a provenance for cytoprotective pathways in the kidney and other tissues. Kidney Int. 2006; 70(3): 432–443. [DOI] [PubMed] [Google Scholar]
  • 87. Maines MD. New insights into biliverdin reductase functions: linking heme metabolism to cell signaling. Physiology (Bethesda). 2005; 20: 382–389. [DOI] [PubMed] [Google Scholar]
  • 88. Gozzelino R, Jeney V, Soares MP. Mechanisms of cell protection by heme oxygenase‐1. Annu Rev Pharmacol Toxicol. 2010; 50: 323–354. [DOI] [PubMed] [Google Scholar]
  • 89. Jarmi T, Agarwal A. Heme oxygenase and renal disease. Curr Hypertens Rep. 2009; 11(1): 56–62. [DOI] [PubMed] [Google Scholar]
  • 90. Ozaki KS, Marques GM, Nogueira E, Feitoza RQ, Cenedeze MA, Franco MF, Mazzali M, Soares MP, Pacheco‐Silva A, Camara NO. Improved renal function after kidney transplantation is associated with heme oxygenase‐1 polymorphism. Clin Transplant. 2008; 22(5): 609–616. [DOI] [PubMed] [Google Scholar]
  • 91. Lin CC, Yang WC, Lin SJ, Chen TW, Lee WS, Chang CF, Lee PC, Lee SD, Su TS, CS Fann, al et. Length polymorphism in heme oxygenase‐1 is associated with arteriovenous fistula patency in hemodialysis patients. Kidney Int. 2006; 69(1): 165–172. [DOI] [PubMed] [Google Scholar]
  • 92. Liu KD, Glidden DV. Clinical trials for acute kidney injury: design challenges and possible solutions. Curr Drug Targets. 2009; 10(12): 1190–1195. [DOI] [PubMed] [Google Scholar]
  • 93. Jo SK, Rosner MH, Okusa MD. Pharmacologic treatment of acute kidney injury: why drugs haven't worked and what is on the horizon. Clin J Am Soc Nephrol. 2007; 2(2): 356–365. [DOI] [PubMed] [Google Scholar]
  • 94. Marchewka Z, Kuzniar J, Dlugosz A. Enzymuria and beta2‐mikroglobulinuria in the assessment of the influence of proteinuria on the progression of glomerulopathies. Int Urol Nephrol. 2001; 33(4): 673–676. [DOI] [PubMed] [Google Scholar]
  • 95. Trof RJ, Di Maggio F, Leemreis J, Groeneveld AB. Biomarkers of acute renal injury and renal failure. Shock. 2006; 26(3): 245–253. [DOI] [PubMed] [Google Scholar]
  • 96. Devarajan P. Proteomics for the investigation of acute kidney injury. Contrib Nephrol. 2008; 160: 1–16. [DOI] [PubMed] [Google Scholar]
  • 97. Bagshaw SM, Bennett M, Haase M, Haase‐Fielitz A, Egi M, Morimatsu H, D'Amico G, Goldsmith D, Devarajan P, Bellomo R. Plasma and urine neutrophil gelatinase‐associated lipocalin in septic versus non‐septic acute kidney injury in critical illness. Intensive Care Med. 2010; 36(3): 452–461. [DOI] [PubMed] [Google Scholar]
  • 98. Nakamura T, Sugaya T, Koide H. Urinary liver‐type fatty acid‐binding protein in septic shock: effect of polymyxin B‐immobilized fiber hemoperfusion. Shock. 2009; 31 (5): 454–459. [DOI] [PubMed] [Google Scholar]
  • 99. Kamijo‐Ikemori A, Sugaya T, Kimura K. Urinary fatty acid binding protein in renal disease. Clin Chim Acta. 2006; 374(1‐2): 1–7. [DOI] [PubMed] [Google Scholar]
  • 100. Basnakian AG. Netrin‐1: a potential universal biomarker for acute kidney injury. Am J Physiol Renal Physiol. 2008; 294(4): F729–F730. [DOI] [PubMed] [Google Scholar]
  • 101. Reeves WB, Kwon O, Ramesh G. Netrin‐1 and kidney injury. II. Netrin‐1 is an early biomarker of acute kidney injury. Am J Physiol Renal Physiol. 2008; 294(4): F731–F738. [DOI] [PubMed] [Google Scholar]

Articles from Clinical and Translational Science are provided here courtesy of Wiley

RESOURCES