Abstract
The current lack of effective therapeutics for patients with acute kidney injury (AKI) represents an important and unmet medical need. Given the importance of the clinical problem, it is time for us to take a few steps back and reexamine current practices. The focus of this review is to explore the extent to which failure of therapeutic translation from animal studies to human studies stems from deficiencies in the preclinical models of AKI. We will evaluate whether the preclinical models of AKI that are commonly used recapitulate the known pathophysiologies of AKI that are being modeled in humans, focusing on four common scenarios that are studied in clinical therapeutic intervention trials: cardiac surgery-induced AKI; contrast-induced AKI; cisplatin-induced AKI; and sepsis associated AKI. Based on our observations, we have identified a number of common limitations in current preclinical modeling of AKI that could be addressed. In the long term, we suggest that progress in developing better preclinical models of AKI will depend on developing a better understanding of human AKI. To this this end, we suggest that there is a need to develop greater in-depth molecular analyses of kidney biopsy tissues coupled with improved clinical and molecular classification of patients with AKI.
Keywords: acute kidney injury, animal models, human disease, preclinical research, drug discovery
acute kidney injury (AKI) is characterized by an abrupt deterioration in kidney function that disrupts metabolic, fluid and electrolyte homeostasis over a period of hours or days. An analysis of 305 AKI cohort studies found that AKI occurred in 21% of adults hospitalized in developed countries with an acute illness (153). The risk of AKI is increased in the elderly, in patients with preexisting chronic kidney disease (CKD), and in patients with diabetes mellitus (130). An episode of AKI is also a strong predictor of mortality: in patients with severe AKI requiring dialysis mortality approaches 60% in some series (130).
US and European guidelines for the management of patients with AKI outline a series of diagnostic, renal support strategies and approaches to reduce further damage to the kidney (8a, 70). However, other than optimization of hemodynamics and avoidance of nephrotoxins, no therapeutic interventions have been shown to improve clinical outcomes in patients with AKI (31, 104, 117). Given the high mortality and morbidity associated with AKI, there is an important unmet medical need to develop effective therapies for these patients. This failure cannot be attributed to a lack of preclinical research or from a failure to conduct clinical trials (72). Indeed, numerous potential therapies for AKI have been identified from preclinical research (17, 95, 127, 154). Unfortunately, most of the phase 2 proof-of-concept trials in AKI have yielded conflicting results, and none of the therapeutic interventions tested in definitive phase 3 trials have shown improvements in clinically meaningful end points (25, 103, 112, 173).
This translational gap between preclinical efficacy studies in animal models and clinical trials in patients with AKI may be occurring for a number of different reasons. These include 1) deficiencies in clinical study design and power (68, 118, 173); 2) failure to identify and distinguish between patients with different pathophysiological subtypes of AKI (34); 3) lack of definitive AKI biomarkers that are predictive of clinically meaningful outcomes and/or responses to therapeutic interventions (41); and 4) analysis of surrogate measures that do not predict improvements in long-term clinically meaningful renal outcomes after AKI (25). However, there is also concern that the animal models may not be predictive of therapeutic responses in humans with AKI (39, 45, 57, 114, 137). In this review, we discuss some of the reasons current preclinical models of AKI may not be translated into clinical practice and will outline strategies that could enhance the translatability of preclinical AKI research.
Preclinical Models of Human AKI
There has been an effort to develop preclinical models that more closely mimic injury in patients with AKI (113). Ideally, preclinical AKI models should be tailored to model specific clinical scenarios that are being planned for future clinical intervention trials. However, in practice this is rarely done. To illustrate this, we have surveyed the preclinical studies that have been used to support completed phase 2 and phase 3 AKI studies registered with www.clinicaltrials.gov (Tables 1 and 2). All clinical trials for drugs that might be marketed in the United States have been registered on this website since 2007. We have focused on clinical studies involving therapeutic interventions and have only included those studies that were annotated as “completed” in the database. Therefore, while our search does not include all of the clinical trials in AKI, it provides a snapshot of clinical studies that have been performed along with the preclinical data that have been used to support these therapeutic intervention studies. Studies meeting these criteria are restricted to four clinical scenarios: cardiac surgery-associated AKI (CSA-AKI), contrast-induced AKI (CI-AKI), cisplatin-induced AKI (CP-AKI), and sepsis-associated AKI (SA-AKI).
Table 1.
Clinical trials for AKI
| Drug | Indication | Risk Factors | Timing of Therapy | Result | Reference or Study Identifier |
|---|---|---|---|---|---|
| N-acetylcysteine+fenoldopam | CSA-AKI | CKD | Prevention | Positive | 15 |
| Sodium bicarbonate | CSA-AKI | CKD | Prevention | No effect | 101 |
| Erythropoietin (or erythropoietin analogs) | CSA-AKI | CKD | Prevention | Positive | 159 |
| None | Prevention | Positive | 111 | ||
| Includes CKD | Postoperative | No effect | 39 | ||
| Rasburicase | CSA-AKI | CKD | Prevention | No effect | 47 |
| Neseritide (brain natriuretic peptide) | CSA-AKI | CKD | Prevention | Positive | 35 |
| Insulin | CSA-AKI | None | Prevention | No effect | 164 |
| Sodium bicarbonate | CI-AKI | CKD | Prevention +12 h | Worse vs. NaCl | 76 |
| N-acetylcysteine | CI-AKI | CKD/DM | Prevention | No effect | 7 |
| Defiprone (iron chelator) | CI-AKI | CKD/DM | Prevention | Unknown | NCT01146925 |
| Vitamin E+N-acetylcysteine | CI-AKI | CKD | Prevention | No effect | 74 |
| Vitamin E | CI-AKI | CKD/DM | Prevention | Positive | 158 |
| CKD | Prevention | Unknown | NCT02070679 | ||
| Wine/beer | CI-AKI | CKD/DM | Prevention | Unknown | NCT01562925 |
| Silymarin | CP-AKI | CKD | Prevention | Unknown | 142 |
| Sodium bicarbonate | SA-AKI | None | Presence of SIRS and elevated N-Gal | No effect (toxicity noted) | 138 |
| Erythropoietin | SA-AKI | Includes CKD | Prevention | Unknown | NCT00676234 Part reported in Ref. 39 |
| Hydroxyethyl starch (HES) | SA-AKI | None | For volume resuscitation | Worse | 124 |
| Insulin | SA-AKI | None | Prevention | No effect | 11 |
| Insulin+HES | SA-AKI | None | For volume resuscitation | Worse | 28 |
AKI, acute kidney injury. See text for additional definitions. Clinical studies were identified from www.clinicaltrials.gov using search terms “acute kidney injury” and “phase 2” or “phase 3” studies. Only completed studies that evaluated drug interventions and in which efficacy was evaluated with renal function, dialysis, or death as primary end points appear. Renal transplantation and cell-based therapies were excluded. Clinical study identification number from the www.clinicaltrials.gov site is provided when published references are not available. Timing of therapy refers to timing in relation to the initiating event. Prevention studies include interventions that were delivered at any time before or at the same time as the initiating event.
Table 2.
Preclinical studies supporting clinical trials identified in Table 1
| Drug | Model | Species | Timing of Therapy | Dose-Response | Target Analysis | Results | Reference(s) |
|---|---|---|---|---|---|---|---|
| Fenoldopam | IR-AKI | Rats | Prevention | No | No | Positive | 9 |
| Community-acquired AKI | Cats+dogs | Established AKI | No | No | No effect | 109 | |
| N-acetylcysteine | IR-AKI | Rats | Prevention | Yes | No | Positive | 43 |
| CPB-AKI | Rats | Prevention | Yes | Yes | Positive | 180 | |
| CI-AKI | Rats | Prevention | No | No | Positive | 178 | |
| CLP-AKI | Rats | Prevention | No | Yes | Positive | 29 | |
| Glycerol | Rats | Prevention | No | No | Positive | 74 | |
| Sodium bicarbonate | IR-AKI | Rats | Prevention | No | Yes | No effect | 152 |
| Mercuric chloride | Rats | Prevention | No | No | Worse vs. NaCl | 19 | |
| CI-AKI | Rats | Prevention | No | No | No effect | 80 | |
| Rats | Prevention | No | No | Positive | 166 | ||
| Erythropoietin (or erythropoietin analogs) | IR-AKI | Rats | Prevention | No | No | Positive | 146 |
| Glycerol | Rats | Prevention | No | No | Positive | 176 | |
| IR-AKI | Macaque | Prevention | No | No | Positive | 67 | |
| CP-AKI | Rats | Prevention | No | No | Positive | 13, 79 | |
| CLP-AKI | Rats | Prevention | No | No | Positive | 131 | |
| IR-AKI | Rats | 6 h post | No | No | Positive | 69 | |
| IR-AKI | Rats | Prevention | No | No | Positive | 151 | |
| CPB-AKI | Rats | Prevention | No | No | Positive | 168 | |
| Rasburicase | CP-AKI | Rats | Prevention | No | No | Positive | 132 |
| Brain natriuretic peptide (BNP) | Lipopolysaccharide | Dog | Prevention | No | No | Positive | 84 |
| Insulin | IR-AKI | Rats | Prevention | No | No | Positive | 102 |
| Deferoxamine (iron chelator) | Glycerol | Rats | Prevention | Yes | Yes | Positive | 33 |
| UUO | Mice | Prevention | No | Yes | Positive | 63 | |
| Deferoxamine+N-acetylcysteine | Gentamicin | Rats | Prevention | No | Yes | Positive | 125 |
| IR-AKI | Rats | Prevention | No | Yes | Positive | 20 | |
| Vitamin E+N-acetylcysteine | Gentamicin | Rats | Prevention +8 days post | No | Yes | Positive | 92 |
| Vitamin E+pentoxifylin | Glycerol | Rats | Prevention | No | No | No effect | 6 |
| Vitamin E | Mercuric chloride | Rats | Prevention +2 h post | No | Yes | Positive | 5 |
| IR-AKI | Rats | Prevention | No | No | Positive | 78 | |
| Resveratrol | Hypovolemic Shock | Rats | Resuscitate | No | Yes | No effect | 169 |
| IR-AKI | Rats | Prevention | No | Yes | Positive | 21, 73 | |
| Glycerol | Rats | Prevention | Yes | Yes | Positive | 32 | |
| Lipopolysaccharide | Mice | Prevention | No | No | Positive | 36 | |
| CLP-AKI | Mice | 6 h post | No | Yes | Positive | 61 | |
| Silymarin | CP-AKI | Rats | Prevention +2 h post | No | No | Positive | 71 |
| HES | Lipopolysaccharide | Rats | 1 h post | No | No | Positive | 167 |
UUO, unilateral ureteral obstruction. See the text for additional definitions. Preclinical studies were identified from PubMed and Google searches using generic, chemical, and/or brand names of drugs and “acute kidney injury” or “kidney injury” search terms. If multiple examples of the same model were found, only 1–2 references were included. Timing of therapy refers to timing in relation to the initiating event. Prevention refers to interventions that were delivered before or at the same time as the initiating event. Target analysis refers to analysis of known molecular targets of drug action.
CSA-AKI
Cardiac surgery has the highest postoperative risk of AKI compared with other major surgical procedures (55). Up to 30% of patients undergoing cardiac surgery develop AKI, and ∼1% of these patients require dialysis (134). Despite declining numbers of cardiac surgical procedures, those that are being performed are often more complex procedures that take longer to perform and are associated with an increased incidence of procedure-associated AKI (83, 161). CSA-AKI is an independent risk factor for mortality after cardiac surgery (126), and patients with CSA-AKI are at increased risk of CKD, particularly those patients with AKI requiring dialysis (86, 175). Common risk factors for CSA-AKI include preexisting CKD, old age, diabetes, and the female gender (134). Other risk factors include hemodynamic instability, cardiac failure, the type and length of the procedure, and use of cardiopulmonary bypass (CPB). In addition, patients who develop contrast-induced AKI following coronary angiography, which is often performed before surgery, are at increased risk of a second episode of AKI after cardiac surgery (51). A large number of therapeutic intervention studies have been performed, mostly prevention studies in which test drugs were administered before or at the time of cardiac surgery, but none have shown consistent beneficial effects on CSA-AKI incidence or outcomes (120, 179). In part, this can be attributed to weaknesses in study design since many of these studies only evaluated short-term effects on serum creatinine levels and were underpowered to detect clinically significant end points such as death, dialysis, and CKD (145).
Pathophysiology of CSA-AKI.
The pathogenesis of CSA-AKI is almost certainly multifactorial. A variety of mechanisms have been implicated. These include 1) reduced renal perfusion; 2) decreased autoregulation of renal blood flow; 3) hemodilution, resulting in reduced oxygen-carrying capacity of blood perfusing the kidney; 4) systemic inflammation caused by passage of blood through the bypass circuits and from leakage of bacterial endotoxins from the intestine; 5) renal ischemia or infarctions caused by macroscopic and microscopic renal embolization; 6) shear stress-induced intravascular hemolysis giving rise to pathogenic oxidative stress from excess free hemoglobin; 7) perioperative and postoperative hemodynamic instability; and 8) drug toxicities (56, 93, 134). Histological analyses of the kidneys from patients with CSA-AKI are limited to early autopsy series and indicate that while a proportion of patients have acute tubular necrosis or renal infarction consistent with macroscopic renal embolization, the majority of patients have “normal” renal histology (177). While this study was conducted more than 40 years ago and many of the surgical procedures and interventions associated with CSA-AKI have changed substantially, these data suggest that there maybe profound effects on cellular and physiological functions in the kidney without inducing histological evidence of cellular injury in many patients with CSA-AKI.
Clinical trials and preclinical models for CSA-AKI.
All of the clinical trials in our review of patients with CSA-AKI were trials to prevent AKI rather than to treat patients with established AKI (Table 1). Cardiac surgery provides a convenient model for studying the effects of interventions developed to prevent AKI, since the timing of injury is usually well defined and risk factors for AKI are well known. In addition, patients are admitted to the hospital before the procedure and kept in the hospital for a number of days after the procedure, facilitating follow-up and selection of patients at risk for developing CSA-AKI. Of the clinical studies that we identified, five of eight were performed in patients with preexisting CKD, and all of the studies evaluated short-term effects of the interventions on postoperative changes in serum creatinine. One study evaluated long-term effects on all-cause mortality but only as a secondary end point (111).
Of the animal studies used to support the use of the therapeutic interventions for patients with CSA-AKI summarized in Table 1 [fenoldopam, N-acetyl-cysteine (NAC), sodium bicarbonate, erythropoietin analogs, rasburicase, neseritide (BNP), and insulin], 8 of 23 of the studies used ischemia-reperfusion models of AKI (IR-AKI) and the majority evaluated the effects of treatment regimens designed to prevent rather than treat established AKI (Table 2). The majority of IR-AKI studies were performed in rats, but one IR-AKI study was performed in macaques (66). The rationale for using IR-AKI models is based on the observation that there is reduced renal blood flow during CPB and that this, along with reduced oxygen-carrying capacity of the diluted blood, may result in critical ischemia to metabolically active cells in the kidney. Other AKI models that were used to support use of these therapeutic interventions in patients with CSA-AKI included the cecal ligation and perforation (CLP) and endotoxin (LPS) models of sepsis in rats and dogs, respectively, as well as glycerol-induced rhabdomyolysis in rats. Each of these models mimics some but not all of the pathophysiological features of CSA-AKI. For example, there is activation of systemic inflammatory responses in the LPS, CLP, glycerol-induced AKI, and IR-AKI models, each of which may mimic some aspects of the systemic inflammatory response seen in patients after CPB (56, 60, 87, 134). Some drugs were shown to be effective in multiple models of AKI. Examples of this include the use erythropoietin analogs and NAC, both of which are effective in IR-AKI, CLP, and glycerol models of AKI. In addition, both NAC and erythropoietin analogs were evaluated in a rat model of CPB (168, 180). However, despite the demonstrable efficacy of these therapeutic interventions across multiple preclinical models, mixed results of clinical trials using the same interventions suggest that efficacy in multiple models, including the rat model of CPB, is not a guarantee that preclinical AKI studies will translate into effective therapeutics for patients with CSA-AKI. In part, failure of these preclinical therapeutic efficacy studies to translate into clinical practice may be because none of the preclinical studies incorporated known CSA-AKI risk factors, notably old age, CKD, and diabetes mellitus. These could potentially influence therapeutic responses to therapy. Other models have been developed that may better mimic clinical scenarios, including pig models of CPB (91, 105). However, these pig models are only available in a small number of specialized laboratories, and none of the published data that we were able to identify using pig models of CPB have extended beyond early time points after the surgical intervention (52, 53, 91, 96, 108, 121–123, 148, 150). Furthermore, it is unknown whether these large-animal models will better recapitulate the pathophysiology of human disease in which comorbidities contribute to AKI susceptibility. For example, a recent study suggests that obesity, which is one of the risk factors for CSA-AKI in patients (110), may actually protect against AKI after CPB in pigs (150).
CI-AKI
Iodinated contrast agents are used to optimize imaging for different X-ray imaging techniques. CI-AKI may have a varied course but is typically characterized by a rise in serum creatinine 2–3 days after contrast administration (99). While only a small number of patients develop dialysis-dependent AKI, a significant proportion of these patients do not recover baseline kidney function (54, 99). Moreover, patients who develop CI-AKI are at increased risk of serious cardiac events even if their serum creatinine values return to baseline (54, 99). Risk factors for CI-AKI include old age, female sex, CKD, and diabetes mellitus (16). In addition, the site of contrast administration can play a role. For example, AKI is more common after percutaneous coronary interventions in which contrast is given via intra-arterial injection than after intravenous contrast injections commonly used for CT imaging studies (10, 100, 107). The risk of CI-AKI is also affected by the volume of contrast agent given as well as the presence of hemodynamic instability (16). Rates of CI-AKI are reduced by ensuring patients are well hydrated, avoiding patients at high risk of CI-AKI, avoiding the use of concomitant nephrotoxins, and avoiding hyperosmolar contrast agents (12). Despite this, CI-AKI is still one of the commonest causes of AKI in hospitalized patients (135), and to date no therapeutic interventions have been shown to reduce the incidence of CI-AKI (173).
Pathophysiology of CI-AKI.
Contrast agents are freely filtered and concentrated in the collecting ducts and distal tubules as water and low-molecular weight solutes are preferentially reabsorbed (139). Increased concentrations of contrast agents in the glomerular filtrate may account for the higher rate of CI-AKI with intra-arterial vs. intravenous contrast agents. Animal studies indicate that there is an early reduction in renal medullary blood flow giving rise to tubular ischemia (140). It is unclear whether this is a direct effect on the renal vasculature or an indirect effect resulting from tubuloglomerular feedback (89). In addition to these vascular effects, in vitro studies using cultured tubular epithelial cells and ex vivo studies using isolated tubules indicate that contrast agents may also cause direct renal tubular toxicity associated with increased production of pathogenic reactive oxygen species (12). Histological studies in patients with CI-AKI are limited to isolated case reports, so it is unclear whether the pathophysiological mechanisms of renal injury reported in animal and in vitro models actually occur in humans. However, cases in which renal pathology has been reported do show evidence of tubular injury (85). Additional confounding factors may contribute to the pathology of renal injury in patients with CI-AKI, including the possibility of atheroembolization to the kidney from intra-arterial manipulations (106).
Clinical trials and preclinical models for CI-AKI.
All of the clinical trials we identified evaluated pretreatment interventions for CI-AKI (Table 1). The incidence of CI-AKI in these studies was relatively low (1–15%) despite selecting for patients with diabetes mellitus and/or mild to moderate CKD (7, 75, 76, 160). Most of the published studies were performed in patients treated with intra-arterial contrast agents at the time of percutaneous coronary intervention (7, 46, 76, 160). One study was conducted exclusively in patients treated with intravenous contrast agents at the time of computed tomography imaging (75). This study failed to show an effect because none of the patients developed AKI despite selecting for patients with preexisting CKD. Primary end points for all of these studies were based on short-term changes in serum creatinine. Benefits for long-term outcomes such as CKD and mortality were not assessed. Since clinical trials require large patient cohorts to identify changes in long-term CKD and mortality outcomes, there is an even greater need that the preclinical models of CI-AKI accurately mimic human CI-AKI. Of the animal studies used to evaluate the therapeutic interventions in patients with CI-AKI summarized in Table 1 [sodium bicarbonate, NAC, iron chelators (deferoxamine), and vitamin E], only 3 of 23 were performed using animal models of CI-AKI (Table 2) (80, 166, 178). The majority of animal studies were performed using IR-AKI, sepsis, or toxin models of AKI. This may reflect the paucity of animal models of CI-AKI that model the clinical scenarios commonly seen in patients. For example, of the preclinical models of CI-AKI used, one study protocol involved water deprivation before injection of the contrast agents (178), while another utilized high-osmolality contrast media (166), neither of which reflect current clinical practice in patients treated with contrast agents (12). Furthermore, only one-third of the preclinical CI-AKI studies that were performed used intra-arterial contrast injections (80), and none were performed in animals with preexisting CKD or diabetes. Despite these limitations, it is likely that these models of contrast-induced renal injury more closely mimic the pathophysiology of CI-AKI than currently utilized IR-AKI, sepsis, and toxin models.
CP-AKI
Platinum-based chemotherapies, of which cisplatin is the most commonly used and effective agent, have been in use for nearly 40 years and are important agents in the therapeutic arsenal for a variety of solid organ malignancies in adults, including head and neck, lung, testis, and ovarian and bladder cancer, as well as childhood osteosarcomas and hepatoblastomas (65, 171). The principal dose-limiting side effect of cisplatin is AKI (115, 136). Many patients who develop AKI are managed by their oncologist and do not see a nephrologist unless renal replacement therapy is required or severe metabolic complications, such as severe magnesium wasting, occur (136). Despite the use of standard precautions to ensure patients are well hydrated, do not have preexisting CKD, and avoiding the use of concomitant nephrotoxins, up to 30% of patients still develop AKI (136). The repeated dosing requirements for the majority of cisplatin-containing chemotherapy regimens carries a significant risk of CKD (136). Development of CKD is of particular concern for long-term follow-up in children treated and cured of their malignancy using cisplatin chemotherapy regimens (77). In patients who develop significant renal impairment after cisplatin treatment, increased mortality is observed due to markedly increased rates of tumor recurrence (24). Since clinical guidelines require reduced or missed doses of cisplatin if serum creatinine does not return to baseline (81), increased rates of tumor recurrence in patients with CP-AKI may result from a failure to complete the chemotherapy regimens. Therefore, the effectiveness of chemotherapy is an important consideration for outcomes of therapeutic trials of CP-AKI since, unlike other indications, therapeutic success of an AKI intervention study might be judged by its ability to ensure completion of chemotherapy.
Pathophysiology of CP-AKI.
The renal toxicity of cisplatin has been studied in vitro and in rodents and results from the concentration of cisplatin in the proximal tubular epithelium as a result of the selective uptake of cisplatin by organic cation transporters (50). Once in the cells, cisplatin induces DNA damage and activates a variety of signaling pathways that promote apoptosis as well as spontaneous necrosis and programmed necroptosis (115, 136). Limited data on human tissues obtained from autopsies show dominant proximal tubular degeneration, necrosis, and repair, but also damage to distal tubules and the collecting duct epithelium (158).
Clinical trials and preclinical models for CP-AKI.
We were only able to identify a single, small, completed clinical trial of a therapeutic intervention for prevention of CP-AKI after cyclical treatment with cisplatin (listed in www.clinicaltrials.gov) (Table 1). No reduction in AKI was observed in patients pretreated with the antioxidant silymarin (142). This clinical study was supported by one preclinical study that used a single dose of cisplatin in rats pretreated with silymarin (Table 2). Treated rats showed less tubular injury at 2 wk, although functional markers such as creatinine or blood urea nitrogen were not assessed (3). While this study is not representative of the breadth of preclinical studies that have been used to study CP-AKI (115), it does reflect a number of common weaknesses. Nearly all of the preclinical studies to date have been performed in mice or rats after a single high dose of cisplatin (from 5 to 30 mg/kg) (49, 114, 149). These models contrast with standard cisplatin treatment regimens in patients in whom the drug is usually administered at multiple lower doses (60–100 mg/m2) at 3- to 4-wk intervals for several months (136). To model this, the Siskind laboratory (144) recently developed a repeated dosing model in mice with administration of 7 mg/kg cisplatin once a week for 4 wk. An advantage of this repeated dosing model is that mice are able to survive the course of cisplatin treatment and can be aged out to examine the long-term effects on kidney pathology and function. In addition to differences in dosing regimens, while the majority of adult patients with head and neck and lung cancer are elderly, we are unaware of preclinical studies that have been performed in older rodents or in rodents with CP-AKI-associated risk factors, including CKD and diabetes.
The dearth of clinical trials for CP-AKI likely reflects the competing end points of preventing AKI and reducing tumor growth. A number of the drugs tested to prevent CP-AKI might also promote tumor growth directly or might inhibit chemotherapeutic efficacy. For example, while numerous studies have tested the efficacy of erythropoietin in preventing CP-AKI (39), erythropoietin use in patients with cancer is controversial since there is concern that erythropoietin may increase tumor growth (38). Other treatments that prevent CP-AKI in animal models, including inhibition of caspases and anti-TNF therapy (49, 128), might also limit the therapeutic efficacy of cisplatin against tumors. Despite this, we are aware of only two studies that have examined the dual effect of therapy on both CP-AKI and tumor growth (116, 129). The study by Ravichandran et al. (129) is a sobering reminder that the model of injury studied can result in very different results. In that study, the role of CD4 T cell depletion in the prevention of CP-AKI was examined in a CP-AKI model that was developed in tumor-bearing mice. Cisplatin administered weekly for 4 wk increased serum creatinine and caused tumor shrinkage by 2 wk. Although depletion of CD4 T cells protects against CP-AKI in the high dose, single injection model, in the 4-wk tumor model CD4 T cell depletion did not protect against CP-AKI and also reduced the effectiveness of cisplatin to shrink tumors: tumors were larger in the CD4-depleted mice treated with cisplatin. The results of this report highlight two critical issues in future cisplatin-induced AKI work: 1) results in the more clinically relevant low dose 4 wk model may not be consistent with results in the high-dose short-term models; and 2) measures that may protect against AKI may adversely affect tumor growth.
SA-AKI
SA-AKI is probably the most challenging and complex clinical scenario for studying therapeutic efficacy in AKI. AKI is a frequent complication of sepsis, occurring in >50% of patients with sepsis in intensive care units (62). SA-AKI is an independent predictor of mortality and progressive CKD, particularly in patients who require dialysis (14, 62). The incidence of sepsis has been progressively increasing over the last 20 years. This has been attributed to increased population age, survivable chronic diseases, therapeutic and human immunodeficiency virus-associated immunosuppression, and increasingly complex surgical interventions (97). Patients with SA-AKI usually present with a rapid decline in renal function after the onset of sepsis, but the clinical course of patients with sepsis is highly variable. For example, sepsis is associated with activation of a systemic inflammatory response during the acute illness, yet over time, a compensatory anti-inflammatory response may develop which predisposes patients to secondary infections (26). Patients may therefore present with a primary infection such as postoperative sepsis after bowel surgery, followed by more severe sepsis after they develop a secondary infection such as a hospital-acquired pneumonia (30). Other factors may complicate the diagnosis since sepsis may be treated with nephrotoxic antibiotics, and patients with AKI from other causes, such as CI-AKI and CSA-AKI, are also at increased risk of sepsis (94). AKI-specific risk factors in sepsis are shared with other causes of AKI and include preexisting CKD, hypertension, and diabetes mellitus (62).
Pathophysiology of SA-AKI.
SA-AKI is part of a complex clinical syndrome in which a profound systemic inflammatory response syndrome (SIRS) is activated in response to an infection (8). Organ dysfunction results from reduced oxygen delivery to and/or usage by tissues. Reduced oxygen delivery may result from reduced blood pressure in septic shock, as well as abnormalities in microcirculatory blood flow resulting from reduced red blood cell deformability, coagulopathy associated with disseminated intravascular coagulation, and endothelial dysfunction (8). In addition to reduced tissue oxygenation, hyperactivation of the innate immune response by activation of pathogen associated molecular pathways (PAMPS) gives rise to pathogenic oxidative stress and further tissue injury in sepsis. Activation of death-associated molecular pathways (DAMPS) in neighboring cells further enhances inflammation (8, 174). Despite these effects on tissue oxygenation, oxidative stress, and inflammation, autopsy series from patients with SA-AKI dying of severe sepsis fail to detect widespread cellular necrosis in the kidney (157). One explanation for this is that sepsis promotes mitochondrial damage in renal tubular epithelial cells that in turn impairs oxygen usage (163). This may make the tubular epithelium resistant to the effects of reduced oxygen delivery in sepsis, and would explain why there is evidence of widespread structural abnormalities in mitochondria and increased mitophagy in the kidneys of patients dying with SA-AKI (157).
Clinical trials and preclinical models for SA-AKI.
There have been a large number of completed and ongoing therapeutic trials in patients with SA-AKI (Table 1) (8, 154). None of these therapeutic interventions have been translated into clinical practice (37). A common weakness in clinical study designs for SA-AKI has been the failure to identify and select phenotypic subgroups of patients that are predictive of adverse outcomes (34). Given the heterogeneous pathophysiology of SA-AKI, in the absence of subgroup selection clinical studies may either show negative results or have to recruit prohibitively large numbers of patients to see significant treatment effects. In addition, when clinical studies are planned there has often been a failure to carefully consider how timing of the intervention in patients relates to timing of intervention in animal studies. For example, a commonly cited example of the failure of animal studies to translate to patients with sepsis is the failure to effectively translate anti-TNF therapy, which is beneficial in animals with sepsis, to patients (4). However, the clinical studies used anti-TNF therapies in patients with established sepsis whereas the beneficial effects of anti-TNF in animal models of sepsis required administration before the onset of the sepsis (58, 162). In a recent study from the Faubel laboratory (23) using both the cecal ligation and perforation (CLP) and endotoxin models of sepsis, it was determined that the onset of AKI occurs rapidly in the course of sepsis and that prophylactic administration of TNF antibodies protected against AKI. However, anti-TNF therapy administered 2 h after sepsis did not protect against AKI. Thus the failure in the clinical trials that administered anti-TNF therapy in established sepsis is consistent with these animal data, indicating that anti-TNF therapy is ineffective in established disease.
A number of different preclinical models of severe sepsis and SA-AKI have been developed in rodents and larger mammals (45, 165). Of the animal studies used to evaluate therapeutic interventions for patients with SA-AKI summarized in Table 1 [sodium bicarbonate, erythropoietin analogs, hydroxyethyl starch (HES), and insulin], the majority were based on nonsepsis models of AKI (Table 2). It can be argued that models such as IR-AKI, glycerol-induced rhabdomyolysis, and cardiopulmonary bypass-associated-AKI (CPB-AKI) model some of the alterations in renal hemodynamics and inflammatory responses seen in SA-AKI (8, 56, 60, 64). However, it is unlikely that these preclinical models share all of the pathophysiological features of SA-AKI. Only 1 of 15 preclinical studies identified in our search used the rat cecal ligation and puncture (CLP) model of SA-AKI to evaluate efficacy of therapeutic interventions for the prevention of SA-AKI (131). Failure to utilize sepsis models of SA-AKI is probably a practical issue reflecting difficulties establishing some of the more clinically relevant models of SA-AKI, such as the rat CLP model, in which fluid resuscitation and antibiotic therapy are used to mimic SA-AKI in patients (45). However, there are limitations even to these more clinically relevant models of SA-AKI. For example, CLP AKI in rodents is rarely studied for >24 h and yet patients with SA-AKI are often septic for several days. In addition, SA-AKI is associated not only with polymicrobial, bowel-associated sepsis but also with lung, intravascular, and urogenital sepsis (14). Apart from intravenous or intraperitoneal inoculation with bacteria (45), we are unaware of SA-AKI models that have used alternate sites of primary infection. In addition, the bacterial species may affect therapeutic responses to intervention. For example, pretreatment with TNF antibodies confers a marked improvement in survival after intravenous injection of Escherichia coli but has limited survival benefit after intravenous injections of two other gram negative bacteria, Pseudomonas aeruginosa and Klebsiella pneumoniae (147). While these studies did not evaluate effects on SA-AKI, they illustrate the importance of evaluating therapeutic efficacy for SA-AKI induced by different pathogens. Finally, there is evidence that preexisting CKD increases the severity of AKI and worsens survival in the rat CLP model (44). Importantly, preexisting CKD can also alter therapeutic responses in sepsis. For example, rats that have undergone ⅚ nephrectomies show reduced survival benefit of anti-vascular endothelial growth factor (VEGF) therapy after CLP but improved survival after anti-high-mobility box group protein 1 (HMGB1) treatment (82).
Additional Considerations
Pharmacokinetics and target engagement studies.
Differences in therapeutic responses between mammalian species may result from differences in drug metabolism and dosing, and it is often unclear whether doses that are administered achieve effective tissue concentrations of the drugs. For example, of 39 preclinical studies we identified, none included pharmacokinetic studies and only 4 included dose-response analyses (Table 2). An alternative approach is to evaluate whether the molecular pathways being targeted are efficiently engaged by the therapeutic intervention. Fourteen of thirty-nine studies evaluated target engagement in the kidney by the therapeutic intervention: 13 of these evaluated the effects of antioxidants on markers of oxidative stress in the kidney (Table 2). Since 20 of 39 of the studies evaluated effects of antioxidants in AKI, this suggests that while target engagement analysis is commonly performed in preclinical antioxidant studies, it is rarely performed for other therapeutic interventions. This is likely because oxidative stress markers are relatively easy to measure while the therapeutic targets engaged by other therapeutic interventions, such as erythropoietin, insulin, and brain natriuretic peptide, are poorly defined and difficult to assess.
Sex and genetic heterogeneity.
The majority of preclinical studies in AKI are performed in males because renal injury is generally more severe and models are less affected by hormonal fluctuations that occur in females during the estrus cycle (39, 119). However, while female mice are protected from IR-AKI, they have increased susceptibility to CP-AKI (172), and importantly may have different responses to therapeutic interventions after AKI. For example, while male rats are protected from CP-AKI after treatment with erythropoietin, erythropoietin has no protective effects on CP-AKI in females (48). Despite this and consistent with the published literature (39), only 4 of 39 of the preclinical studies that we identified performed studies in females (36, 84, 109, 178), and only 2 of these studies compared effects in males and females (84, 109). Humans are also genetically heterogeneous, and the genetic heterogeneity has been shown to affect the severity of AKI in both mice and rat models of IR-AKI (18, 90). Interestingly, the majority of the preclinical AKI drug efficacy studies we identified were performed using outbred, genetically heterogeneous rats (mostly Sprague-Dawley and Wistar). However, all four of the mouse preclinical AKI studies that we identified were performed using inbred, genetically identical mouse strains (Table 2).
Recommendations
Our inability to translate preclinical discovery into effective therapeutic interventions that improve clinical outcomes in patients with AKI suggests that our current approaches are flawed. While this is clearly a complex problem with deficiencies in clinical and preclinical study design, models, and approaches, we suggest it is an opportune time to take a few steps back and reexamine current practices. Our analysis of the preclinical models that have been used to support recent clinical efficacy studies for AKI have identified a number of common limitations that we feel could now be addressed.
Better modeling of comorbidities and risk factors.
The presence of comorbidities that affect the severity of AKI may affect responses to therapy. In addition to the effects of CKD on therapeutic responses in sepsis (82), another example of a comorbidity that may reduce therapeutic responsiveness in AKI is the reduced regenerative capacity of the aging kidney (2). Additional comorbidities, including CKD and diabetes, may affect therapeutic responses. For example, clinical studies of remote ischemic preconditioning (RIPC) may prove to be ineffective in clinical trials because many of the risk factors used to select patients for intervention also reduce the protective effects of RIPC after AKI (98). Other variables include the effect of gender on severity of injury and therapeutic responses after AKI (48, 119, 172), as well as genetic (18, 88) and environmental heterogeneity, including the effects of polypharmacy, which are particularly common in elderly patients with AKI (133). Future studies should focus on developing more complex models of AKI in older animals, both males and females, as well as studies in animals with preexisting CKD, diabetes, and should also evaluate the effects of commonly used polypharmacies on these therapeutic responses. In addition, while many of the preclinical studies in rats are performed using outbred, genetically heterogeneous strains, efforts should be made to study outbred strains in mouse studies as well.
Incorporating dose-response, timing, and target engagement studies.
Some attempt should be made to determine the optimal dosing and timing for preclinical therapeutic intervention studies. While many laboratories may not have the ability to perform pharmacokinetic studies, analysis of dose-response effects are likely to capture effects of inadequate dosing on drug efficacy. If the molecular target of the therapeutic intervention is known, efforts should be made to evaluate activity and/or expression of the target in treated mice to determine whether effective concentrations of the drug are reaching the target organ. Finally, while many preclinical studies have focused on prevention as a therapeutic goal, attempts should be made to compare effects of preventive treatment with delayed treatment regimens on AKI outcomes. By clearly defining both effective dosing and timing of therapeutic interventions, preclinical therapeutic efficacy studies can be better used to direct clinical study design in the future.
Better modeling of human disease.
As a guiding principal, we recommend that attempts be made to test therapeutic interventions in preclinical models that reflect the nature of renal injury that occurs in patients with AKI. For example, a drug that is being evaluated in patients with CI-AKI should also be tested in animal models of CI-AKI, while drugs being evaluated in patients with SA-AKI should be tested in valid models of SA-AKI. Our review indicates that this has not always been the case. It remains to be seen whether additional cost and time spent modeling more complex and varied clinical scenarios, such as the use of CPB in rodents and pigs to model CSA-AKI, or the use of intra-arterial contrast agents to model CI-AKI, will allow for the first convincing translation of a preclinical AKI drug discovery to patients. Finally, where effects of AKI interventions might adversely influence a concurrent disease process, such as the use of erythropoietin in patients with cancer undergoing cisplatin therapy, we recommend that attempts are made to evaluate effects of these therapeutic intervention on the progression of the associated disease in animal models before these interventions are tested in humans.
Improved understanding of the cellular and molecular pathways in human AKI.
There is a scarcity of renal biopsy material available to interrogate molecular pathways that are involved in the pathogenesis of human AKI. As a result, it is unclear whether the pathways being targeted in preclinical studies are relevant to human disease. This problem has received scant attention in the AKI literature, but a number of sobering examples of this can be found in the non-AKI literature. For example, while some mouse models of diabetic nephropathy recapitulate the pathological findings and progression of the human disease, only a minor subset of pathways that are activated in these preclinical models are activated in renal biopsies from patients with diabetic nephropathy (22, 59). There is also a divergence of the molecular pathways regulating inflammatory responses in mice and humans. This has precipitated a vigorous debate about the relevance of mouse models to the study inflammatory pathologies, including sepsis, in humans (141, 155, 156, 170). These concerns might be mitigated with the development of “humanized” mouse models, in which human genomic loci are transferred into the mouse genome (27, 42). However, for these models to be valid there is still a need to understand which pathways are involved in the pathogenesis of human disease. These questions underscore the need to develop a better understanding of the similarities and differences between the molecular pathogenesis of AKI in animal models and humans. In the long term, our ability to assess this is dependent on developing a deeper understanding of the comparative molecular pathogenesis of human and experimental AKI. This will only be possible if we are able to develop renal biopsy tissue repositories in patients with AKI. While waiting for this, we suggest preclinical efficacy studies are performed in multiple models of AKI: drugs that are effective in multiple models of AKI, particularly if they are effective in males and females and in different species, are more likely to be effective patients with AKI in which the pathophysiologies are often complex and multifactorial.
Conclusions
The lack of effective therapies for patients with AKI represents an important and unmet medical need. While there are many explanations for this, ranging from deficiencies in clinical study design, patient selection, and outcomes measures, the development of preclinical models that are predictive of therapeutic responses in patients with AKI is a challenge that can and should be addressed. This will take time and effort, and in all likelihood will increase the cost of preclinical AKI research. However, the cost of developing preclinical models which are predictive of therapeutic efficacy in patients with AKI pale compared with the costs of funding futile clinical trials. While recommendations in this review reflect the authors' opinions, we hope that these recommendations will provide a springboard for further discussion that will lead to the development of consensus guidelines for the nephrology research community. In the long term, this would serve as the foundation for collaborations and commitment from interested stakeholders in industry, academia, and the research funding agencies that will be required to support the development and application of these models more widely by the nephrology research community.
GRANTS
This work was supported by National Institutes of Health (NIH) Grants 1RC4DK090770, VUMC44339-R/P30DK079307-07, VUMC43321/U24 DK076169, and 2R01 DK069403, and the Vanderbilt Center for Kidney Disease (VCKD). A research grant from NephroGenex, Inc. supported M. P. de Caestecker; NIH Grant R01DK093462 supported L. J. Siskind; and S. Faubel was supported by Veterans Afairs Merit Review Grants I01 BX001498 and R56 HL095363.
DISCLOSURES
M. de Caestecker was paid as a consultant for NephroGenex.
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
Author contributions: N.I.S. and M.P.d.C. prepared figures; N.I.S. drafted manuscript; N.I.S., L.J.S., S.F., and M.P.d.C. approved final version of manuscript; L.J.S., S.F., and M.P.d.C. edited and revised manuscript.
REFERENCES
- 2.Abdel-Rahman EM, Okusa MD. Effects of aging on renal function and regenerative capacity. Nephron Clin Pract 127: 15–20, 2014. [DOI] [PubMed] [Google Scholar]
- 3.Abdelmeguid NE, Chmaisse HN, Abou Zeinab NS. Silymarin ameliorates cisplatin-induced hepatotoxicity in rats: histopathological and ultrastructural studies. Pak J Biol Sci 13: 463–479, 2010. [DOI] [PubMed] [Google Scholar]
- 4.Abraham E, Anzueto A, Gutierrez G, Tessler S, San Pedro G, Wunderink R, Dal Nogare A, Nasraway S, Berman S, Cooney R, Levy H, Baughman R, Rumbak M, Light RB, Poole L, Allred R, Constant J, Pennington J, Porter S. Double-blind randomised controlled trial of monoclonal antibody to human tumour necrosis factor in treatment of septic shock. NORASEPT II Study Group. Lancet 351: 929–933, 1998. [PubMed] [Google Scholar]
- 5.Agarwal R, Goel SK, Chandra R, Behari JR. Role of vitamin E in preventing acute mercury toxicity in rat. Environ Toxicol Phar 29: 70–78, 2010. [DOI] [PubMed] [Google Scholar]
- 6.Akpolat T, Akpolat I, Ozturk H, Sarikaya S, Cosar AM, Bedir A, Kandemir B. Effect of vitamin E and pentoxifylline on glycerol-induced acute renal failure. Nephron 84: 243–247, 2000. [DOI] [PubMed] [Google Scholar]
- 7.Amini M, Salarifar M, Amirbaigloo A, Masoudkabir F, Esfahani F. N-acetylcysteine does not prevent contrast-induced nephropathy after cardiac catheterization in patients with diabetes mellitus and chronic kidney disease: a randomized clinical trial. Trials 10: 45, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med 369: 840–851, 2013. [DOI] [PubMed] [Google Scholar]
- 8a.Anonymous. Kidney Disease: Improving Global Outcomes. Clinical Practice Guideline for Acute Kidney Injury. Kidney Int Suppl 2: 1–138, 2012. [Google Scholar]
- 9.Aravindan N, Samuels J, Riedel B, Shaw A. Fenoldopam improves corticomedullary oxygen delivery and attenuates angiogenesis gene expression in acute ischemic renal injury. Kidney Blood Press Res 29: 165–174, 2006. [DOI] [PubMed] [Google Scholar]
- 10.Aurelio A, Durante A. Contrast-induced nephropathy in percutaneous coronary interventions: pathogenesis, risk factors, outcome, prevention and treatment. Cardiology 128: 62–72, 2014. [DOI] [PubMed] [Google Scholar]
- 11.Azevedo JR, Azevedo RP, Lucena LC, Costa Nde N, Silva WS. Impact of glycemic control on the incidence of acute kidney injury in critically ill patients: a comparison of two strategies using the RIFLE criteria. Clinics 65: 769–773, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Azzalini L, Spagnoli V, Ly HQ. Contrast-induced nephropathy: from pathophysiology to preventive strategies. Can J Cardiol 32: 247–255, 2016. [DOI] [PubMed] [Google Scholar]
- 13.Bagnis C, Beaufils H, Jacquiaud C, Adabra Y, Jouanneau C, Le Nahour G, Jaudon MC, Bourbouze R, Jacobs C, Deray G. Erythropoietin enhances recovery after cisplatin-induced acute renal failure in the rat. Nephrol Dial Transplant 16: 932–938, 2001. [DOI] [PubMed] [Google Scholar]
- 14.Bagshaw SM, Uchino S, Bellomo R, Morimatsu H, Morgera S, Schetz M, Tan I, Bouman C, Macedo E, Gibney N, Tolwani A, Oudemans-van Straaten HM, Ronco C, Kellum JA. Beginning, and ending supportive therapy for the kidney. I. Septic acute kidney injury in critically ill patients: clinical characteristics and outcomes. Clin J Am Soc Nephrol 2: 431–439, 2007. [DOI] [PubMed] [Google Scholar]
- 15.Barr LF, Kolodner K. N-acetylcysteine and fenoldopam protect the renal function of patients with chronic renal insufficiency undergoing cardiac surgery. Crit Care Med 36: 1427–1435, 2008. [DOI] [PubMed] [Google Scholar]
- 16.Bartholomew BA, Harjai KJ, Dukkipati S, Boura JA, Yerkey MW, Glazier S, Grines CL, O'Neill WW. Impact of nephropathy after percutaneous coronary intervention and a method for risk stratification. Am J Cardiol 93: 1515–1519, 2004. [DOI] [PubMed] [Google Scholar]
- 17.Basile DP, Bonventre JV, Mehta R, Nangaku M, Unwin R, Rosner MH, Kellum JA, Ronco C; ADQI XIII Work Group. Progression after AKI: understanding maladaptive repair processes to predict and identify therapeutic treatments. J Am Soc Nephrol 27: 687–697, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Basile DP, Dwinell MR, Wang SJ, Shames BD, Donohoe DL, Chen S, Sreedharan R, Van Why SK. Chromosome substitution modulates resistance to ischemia reperfusion injury in Brown Norway rats. Kidney Int 83: 242–250, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Beaumont JE, Kotchen TA, Galla JH, Luke RG. Failure of loading with sodium bicarbonate to protect against acute renal failure induced by mercuric chloride in the rat. Clin Sci Mol Med 53: 149–154, 1977. [DOI] [PubMed] [Google Scholar]
- 20.Bernardi RM, Constantino L, Machado RA, Vuolo F, Budni P, Ritter C, Dal-Pizzol F. N-acetylcysteine and deferrioxamine protects against acute renal failure induced by ischemia/reperfusion in rats. Rev Bras Ter Intensiva 24: 219–223, 2012. [PubMed] [Google Scholar]
- 21.Bertelli AA, Migliori M, Panichi V, Origlia N, Filippi C, Das DK, Giovannini L. Resveratrol, a component of wine and grapes, in the prevention of kidney disease. Ann NY Acad Sci 957: 230–238, 2002. [DOI] [PubMed] [Google Scholar]
- 22.Bhalla V, Velez MG, Chertow GM. A transcriptional blueprint for human and murine diabetic kidney disease. Diabetes 62: 31–33, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bhargava R, Altmann CJ, Andres-Hernando A, Webb RG, Okamura K, Yang Y, Falk S, Schmidt EP, Faubel S. Acute lung injury and acute kidney injury are established by four hours in experimental sepsis and are improved with pre, but not post, sepsis administration of TNF-alpha antibodies. PLoS One 8: e79037, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bhat ZY, Cadnapaphornchai P, Ginsburg K, Sivagnanam M, Chopra S, Treadway CK, Lin HS, Yoo G, Sukari A, Doshi MD. Understanding the risk factors and long-term consequences of cisplatin-associated acute kidney injury: an observational cohort study. PLoS One 10: e0142225, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Billings FTt, Shaw AD. Clinical trial endpoints in acute kidney injury. Nephron Clin Pract 127: 89–93, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Boomer JS, To K, Chang KC, Takasu O, Osborne DF, Walton AH, Bricker TL, Jarman SD 2nd, Kreisel D, Krupnick AS, Srivastava A, Swanson PE, Green JM, Hotchkiss RS. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 306: 2594–2605, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Breyer MD. Drug discovery for diabetic nephropathy: trying the leap from mouse to man. Semin Nephrol 32: 445–451, 2012. [DOI] [PubMed] [Google Scholar]
- 28.Brunkhorst FM, Engel C, Bloos F, Meier-Hellmann A, Ragaller M, Weiler N, Moerer O, Gruendling M, Oppert M, Grond S, Olthoff D, Jaschinski U, John S, Rossaint R, Welte T, Schaefer M, Kern P, Kuhnt E, Kiehntopf M, Hartog C, Natanson C, Loeffler M, Reinhart K; German Competence Network Sepsis (SepNeet). Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med 358: 125–139, 2008. [DOI] [PubMed] [Google Scholar]
- 29.Campos R, Shimizu MHM, Volpini RA, de Braganca AC, Andrade L, Lopes FD, Olivo C, Canale D, Seguro AC. N-acetylcysteine prevents pulmonary edema and acute kidney injury in rats with sepsis submitted to mechanical ventilation. Am J Physiol Lung Cell Mol Physiol 302: L1640–L1650, 2012. [DOI] [PubMed] [Google Scholar]
- 30.Cawcutt KA, Peters SG. Severe sepsis and septic shock: clinical overview and update on management. Mayo Clin Proc 89: 1572–1578, 2014. [DOI] [PubMed] [Google Scholar]
- 31.Cerda J, Liu KD, Cruz DN, Jaber BL, Koyner JL, Heung M, Okusa MD, Faubel S; Nephrology AKI Advisory Group of the American Society of Nephrology. Promoting kidney function recovery in patients with AKI requiring RRT. Clin J Am Soc Nephrol 10: 1859–1867, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chander V, Chopra K. Protective effect of resveratrol, a polyphenolic phytoalexin on glycerol-induced acute renal failure in rat kidney. Ren Fail 28: 161–169, 2006. [DOI] [PubMed] [Google Scholar]
- 33.Chander V, Singh D, Chopra K. Attenuation of glycerol-induced acute renal failure in rats by trimetazidine and deferoxamine. Pharmacology 67: 41–48, 2003. [DOI] [PubMed] [Google Scholar]
- 34.Chawla LS. Disentanglement of the acute kidney injury syndrome. Curr Opin Crit Care 18: 579–584, 2012. [DOI] [PubMed] [Google Scholar]
- 35.Chen HH, Sundt TM, Cook DJ, Heublein DM, Burnett JC Jr. Low dose nesiritide and the preservation of renal function in patients with renal dysfunction undergoing cardiopulmonary-bypass surgery: a double-blind placebo-controlled pilot study. Circulation 116: I134–I138, 2007. [DOI] [PubMed] [Google Scholar]
- 36.Chen L, Yang S, Zumbrun EE, Guan H, Nagarkatti PS, Nagarkatti M. Resveratrol attenuates lipopolysaccharide-induced acute kidney injury by suppressing inflammation driven by macrophages. Mol Nutr Food Res 59: 853–864, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Cohen J, Vincent JL, Adhikari NK, Machado FR, Angus DC, Calandra T, Jaton K, Giulieri S, Delaloye J, Opal S, Tracey K, van der Poll T, Pelfrene E. Sepsis: a roadmap for future research. Lancet Infect Dis 15: 581–614, 2015. [DOI] [PubMed] [Google Scholar]
- 38.Debeljak N, Solar P, Sytkowski AJ. Erythropoietin and cancer: the unintended consequences of anemia correction. Front Immunol 5: 563, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.de Caestecker M, Humphreys BD, Liu KD, Fissell WH, Cerda J, Nolin TD, Askenazi D, Mour G, Harrell FE Jr, Pullen N, Okusa MD, Faubel S; ASN AKI Advisory Group. Bridging translation by improving preclinical study design in AKI. J Am Soc Nephrol 26: 2905–2916, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.de Seigneux S, Ponte B, Weiss L, Pugin J, Romand JA, Martin PY, Saudan P. Epoetin administrated after cardiac surgery: effects on renal function and inflammation in a randomized controlled study. BMC Nephrol 13: 132, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Devarajan P, Murray P. Biomarkers in acute kidney injury: are we ready for prime time? Nephron Clin Pract 127: 176–179, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Devoy A, Bunton-Stasyshyn RK, Tybulewicz VL, Smith AJ, Fisher EM. Genomically humanized mice: technologies and promises. Nat Rev Genet 13: 14–20, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Di Giorno C, Pinheiro HS, Heinke T, Franco MF, Galante NZ, Pacheco-Silva A, Camara NOS. Beneficial effect of N-acetyl-cysteine on renal injury triggered by ischemia and reperfusion. Transplant Proc 38: 2774–2776, 2006. [DOI] [PubMed] [Google Scholar]
- 44.Doi K, Leelahavanichkul A, Hu X, Sidransky KL, Zhou H, Qin Y, Eisner C, Schnermann J, Yuen PS, Star RA. Pre-existing renal disease promotes sepsis-induced acute kidney injury and worsens outcome. Kidney Int 74: 1017–1025, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Doi K, Leelahavanichkul A, Yuen PS, Star RA. Animal models of sepsis and sepsis-induced kidney injury. J Clin Invest 119: 2868–2878, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Duran PA, Concepcion LA. Survival after acute kidney injury requiring dialysis: long-term follow up. Hemodial Int 18, Suppl 1: S1–S6, 2014. [DOI] [PubMed] [Google Scholar]
- 47.Ejaz AA, Dass B, Lingegowda V, Shimada M, Beaver TM, Ejaz NI, Abouhamze AS, Johnson RJ. Effect of uric acid lowering therapy on the prevention of acute kidney injury in cardiovascular surgery. Int Urol Nephrol 45: 449–458, 2013. [DOI] [PubMed] [Google Scholar]
- 48.Eshraghi-Jazi F, Nematbakhsh M, Pezeshki Z, Nasri H, Talebi A, Safari T, Mansouri A, Mazaheri S, Ashrafi F. Sex differences in protective effect of recombinant human erythropoietin against cisplatin-induced nephrotoxicity in rats. Iran J Kidney Dis 7: 383–389, 2013. [PubMed] [Google Scholar]
- 49.Faubel S, Ljubanovic D, Reznikov L, Somerset H, Dinarello CA, Edelstein CL. Caspase-1-deficient mice are protected against cisplatin-induced apoptosis and acute tubular necrosis. Kidney Int 66: 2202–2213, 2004. [DOI] [PubMed] [Google Scholar]
- 50.Filipski KK, Mathijssen RH, Mikkelsen TS, Schinkel AH, Sparreboom A. Contribution of organic cation transporter 2 (OCT2) to cisplatin-induced nephrotoxicity. Clin Pharmacol Ther 86: 396–402, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Garcia S, Ko B, Adabag S. Contrast-induced nephropathy and risk of acute kidney injury and mortality after cardiac operations. Ann Thorac Surg 94: 772–776, 2012. [DOI] [PubMed] [Google Scholar]
- 52.Ghorbel MT, Patel NN, Sheikh M, Angelini GD, Caputo M, Murphy GJ. Changes in renal medulla gene expression in a pre-clinical model of post cardiopulmonary bypass acute kidney injury. BMC Genomics 15: 916, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Goebel U, Siepe M, Schwer CI, Schibilsky D, Foerster K, Neumann J, Wiech T, Priebe HJ, Schlensak C, Loop T. Inhaled carbon monoxide prevents acute kidney injury in pigs after cardiopulmonary bypass by inducing a heat shock response. Anesth Analg 111: 29–37, 2010. [DOI] [PubMed] [Google Scholar]
- 54.Goldenberg I, Chonchol M, Guetta V. Reversible acute kidney injury following contrast exposure and the risk of long-term mortality. Am J Nephrol 29: 136–144, 2009. [DOI] [PubMed] [Google Scholar]
- 55.Grams ME, Sang Y, Coresh J, Ballew S, Matsushita K, Molnar MZ, Szabo Z, Kalantar-Zadeh K, Kovesdy CP. Acute kidney injury after major surgery: a retrospective analysis of Veterans Health Administration data. Am J Kidney Dis [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Haase M, Haase-Fielitz A, Bellomo R. Cardiopulmonary bypass, hemolysis, free iron, acute kidney injury and the impact of bicarbonate. Contrib Nephrol 165: 28–32, 2010. [DOI] [PubMed] [Google Scholar]
- 57.Heyman SN, Rosen S, Rosenberger C. Animal models of renal dysfunction: acute kidney injury. Expert Opin Drug Discov 4: 629–641, 2009. [DOI] [PubMed] [Google Scholar]
- 58.Hinshaw LB, Tekamp-Olson P, Chang AC, Lee PA, Taylor FB Jr, Murray CK, Peer GT, Emerson TE Jr, Passey RB, Kuo GC. Survival of primates in LD100 septic shock following therapy with antibody to tumor necrosis factor (TNF alpha). Circ Shock 30: 279–292, 1990. [PubMed] [Google Scholar]
- 59.Hodgin JB, Nair V, Zhang H, Randolph A, Harris RC, Nelson RG, Weil EJ, Cavalcoli JD, Patel JM, Brosius FC 3rd, Kretzler M. Identification of cross-species shared transcriptional networks of diabetic nephropathy in human and mouse glomeruli. Diabetes 62: 299–308, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hoke TS, Douglas IS, Klein CL, He Z, Fang W, Thurman JM, Tao Y, Dursun B, Voelkel NF, Edelstein CL, Faubel S. Acute renal failure after bilateral nephrectomy is associated with cytokine-mediated pulmonary injury. J Am Soc Nephrol 18: 155–164, 2007. [DOI] [PubMed] [Google Scholar]
- 61.Holthoff JH, Wang Z, Seely KA, Gokden N, Mayeux PR. Resveratrol improves renal microcirculation, protects the tubular epithelium, and prolongs survival in a mouse model of sepsis-induced acute kidney injury. Kidney Int 81: 370–378, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hoste EA, Bagshaw SM, Bellomo R, Cely CM, Colman R, Cruz DN, Edipidis K, Forni LG, Gomersall CD, Govil D, Honore PM, Joannes-Boyau O, Joannidis M, Korhonen AM, Lavrentieva A, Mehta RL, Palevsky P, Roessler E, Ronco C, Uchino S, Vazquez JA, Vidal Andrade E, Webb S, Kellum JA. Epidemiology of acute kidney injury in critically ill patients: the multinational AKI-EPI study. Intensive Care Med 41: 1411–1423, 2015. [DOI] [PubMed] [Google Scholar]
- 63.Ikeda Y, Ozono I, Tajima S, Imao M, Horinouchi Y, Izawa-Ishizawa Y, Kihira Y, Miyamoto L, Ishizawa K, Tsuchiya K, Tamaki T. Iron chelation by deferoxamine prevents renal interstitial fibrosis in mice with unilateral ureteral obstruction. PLoS One 9: e89355, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ince C. The central role of renal microcirculatory dysfunction in the pathogenesis of acute kidney injury. Nephron Clin Pract 127: 124–128, 2014. [DOI] [PubMed] [Google Scholar]
- 65.Isakoff MS, Bielack SS, Meltzer P, Gorlick R. Osteosarcoma: current treatment and a collaborative pathway to success. J Clin Oncol 33: 3029–3035, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ishii Y, Sawada T, Murakami T, Sakuraoka Y, Shiraki T, Shimizu A, Kubota K, Fuchinoue S, Teraoka S. Renoprotective effect of erythropoietin against ischaemia-reperfusion injury in a non-human primate model. Nephrol Dial Transplant 26: 1157–1162, 2011. [DOI] [PubMed] [Google Scholar]
- 68.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 2: 356–365, 2007. [DOI] [PubMed] [Google Scholar]
- 69.Johnson DW, Pat B, Vesey DA, Guan Z, Endre Z, Gobe GC. Delayed administration of darbepoetin or erythropoietin protects against ischemic acute renal injury and failure. Kidney Int 69: 1806–1813, 2006. [DOI] [PubMed] [Google Scholar]
- 70.Jorres A, John S, Lewington A, ter Wee PM, Vanholder R, Van Biesen W, Tattersall J; ad-hoc working group of ERBP . European Renal Best Practice (ERBP) position statement on the Kidney Disease Improving Global Outcomes (KDIGO) Clinical Practice Guidelines on Acute Kidney Injury: part 2: renal replacement therapy. Nephrol Dial Transplant 28: 2940–2945, 2013. [DOI] [PubMed] [Google Scholar]
- 71.Karimi G, Ramezani M, Tahoonian Z. Cisplatin nephrotoxicity and protection by milk thistle extract in rats. Evidence-based complementary and alternative medicine. eCAM 2: 383–386, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Kaushal GP, Shah SV. Challenges and advances in the treatment of AKI. J Am Soc Nephrol 25: 877–883, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Khader A, Yang WL, Kuncewitch M, Prince JM, Marambaud P, Nicastro J, Coppa GF, Wang P. Novel resveratrol analogues attenuate renal ischemic injury in rats. J Surg Res 193: 807–815, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kim JH, Lee SS, Jung MH, Yeo HD, Kim HJ, Yang JI, Roh GS, Chang SH, Park DJ. N-acetylcysteine attenuates glycerol-induced acute kidney injury by regulating MAPKs and Bcl-2 family proteins. Nephrol Dial Transplant 25: 1435–1443, 2010. [DOI] [PubMed] [Google Scholar]
- 75.Kitzler TM, Jaberi A, Sendlhofer G, Rehak P, Binder C, Petnehazy E, Stacher R, Kotanko P. Efficacy of vitamin E and N-acetylcysteine in the prevention of contrast induced kidney injury in patients with chronic kidney disease: a double blind, randomized controlled trial. Wien Klin Wochenschr 124: 312–319, 2012. [DOI] [PubMed] [Google Scholar]
- 76.Klima T, Christ A, Marana I, Kalbermatter S, Uthoff H, Burri E, Hartwiger S, Schindler C, Breidthardt T, Marenzi G, Mueller C. Sodium chloride vs. sodium bicarbonate for the prevention of contrast medium-induced nephropathy: a randomized controlled trial. Eur Heart J 33: 2071–2079, 2012. [DOI] [PubMed] [Google Scholar]
- 77.Knijnenburg SL, Mulder RL, Schouten-Van Meeteren AY, Bokenkamp A, Blufpand H, van Dulmen-den Broeder E, Veening MA, Kremer LC, Jaspers MW. Early and late renal adverse effects after potentially nephrotoxic treatment for childhood cancer. Cochrane Database Syst Rev 10: CD008944, 2013. [DOI] [PubMed] [Google Scholar]
- 78.Koga H, Hagiwara S, Mei H, Hiraoka N, Kusaka J, Goto K, Kashima K, Noguchi T. The vitamin E derivative, ESeroS-GS, attenuates renal ischemia-reperfusion injury in rats. J Surg Res 176: 220–225, 2012. [DOI] [PubMed] [Google Scholar]
- 79.Kong DY, Zhuo L, Gao CL, Shi SZ, Wang N, Huang ZY, Li WG, Hao LR. Erythropoietin protects against cisplatin-induced nephrotoxicity by attenuating endoplasmic reticulum stress-induced apoptosis. J Nephrol 26: 219–227, 2013. [DOI] [PubMed] [Google Scholar]
- 80.Ladwig M, Flemming B, Seeliger E, Sargsyan L, Persson PB. Renal effects of bicarbonate versus saline infusion for iso- and lowosmolar contrast media in rats. Invest Radiol 46: 672–677, 2011. [DOI] [PubMed] [Google Scholar]
- 81.Launay-Vacher V, Rey JB, Isnard-Bagnis C, Deray G, Daouphars M; European Society of Clinical Pharmacy Special Interest Group on Cancer Care. Prevention of cisplatin nephrotoxicity: state of the art and recommendations from the European Society of Clinical Pharmacy Special Interest Group on Cancer Care. Cancer Chemother Pharmacol 61: 903–909, 2008. [DOI] [PubMed] [Google Scholar]
- 82.Leelahavanichkul A, Huang Y, Hu X, Zhou H, Tsuji T, Chen R, Kopp JB, Schnermann J, Yuen PS, Star RA. Chronic kidney disease worsens sepsis and sepsis-induced acute kidney injury by releasing high mobility group box protein-1. Kidney Int 80: 1198–1211, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lenihan CR, Montez-Rath ME, Mora Mangano CT, Chertow GM, Winkelmayer WC. Trends in acute kidney injury, associated use of dialysis, and mortality after cardiac surgery, 1999 to 2008. Ann Thorac Surg 95: 20–28, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Li N, Jin HX, Song Z, Bai CZ, Cui Y, Gao Y. Protective effect of recombinant human brain natriuretic peptide on acute renal injury induced by endotoxin in canines. Cell Biochem Biophys 70: 1317–1324, 2014. [DOI] [PubMed] [Google Scholar]
- 85.Light JA, Hill GS. Acute tubular necrosis in a renal transplant recipient. Complication from drip-infusion excretory urography. JAMA 232: 1267–1268, 1975. [PubMed] [Google Scholar]
- 86.Lim CC, Tan CS, Chia CM, Tan AK, Choo JC, Kaushik M, Tan HK. Long-term risk of progressive chronic kidney disease in patients with severe acute kidney injury requiring dialysis after coronary artery bypass surgery. Cardiorenal Med 5: 157–163, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liu KD, Altmann C, Smits G, Krawczeski CD, Edelstein CL, Devarajan P, Faubel S. Serum interleukin-6 and interleukin-8 are early biomarkers of acute kidney injury and predict prolonged mechanical ventilation in children undergoing cardiac surgery: a case-control study. Crit Care 13: R104, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Liu X, Zhang T, Xia W, Wang Y, Ma K. Recombinant human erythropoietin pretreatment alleviates renal glomerular injury induced by cardiopulmonary bypass by reducing transient receptor potential channel 6-nuclear factor of activated T-cells pathway activation. J Thorac Cardiovasc Surg 146: 681–687, 2013. [DOI] [PubMed] [Google Scholar]
- 89.Liu ZZ, Schmerbach K, Lu Y, Perlewitz A, Nikitina T, Cantow K, Seeliger E, Persson PB, Patzak A, Liu R, Sendeski MM. Iodinated contrast media cause direct tubular cell damage, leading to oxidative stress, low nitric oxide, and impairment of tubuloglomerular feedback. Am J Physiol Renal Physiol 306: F864–F872, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Lu X, Li N, Shushakova N, Schmitt R, Menne J, Susnik N, Meier M, Leitges M, Haller H, Gueler F, Rong S. C57BL/6 and 129/Sv mice: genetic difference to renal ischemia-reperfusion. J Nephrol 25: 738–743, 2012. [DOI] [PubMed] [Google Scholar]
- 91.Lundemoen S, Kvalheim VL, Svendsen OS, Mongstad A, Andersen KS, Grong K, Husby P. Intraaortic counterpulsation during cardiopulmonary bypass impairs distal organ perfusion. Ann Thorac Surg 99: 619–625, 2015. [DOI] [PubMed] [Google Scholar]
- 92.Manali BP, Deshpande S, Shah G. Evaluation of efficacy of vitamin E and N-acetyl cysteine in gentamicin-induced nephrotoxicity in rats. Ren Fail 33: 341–347, 2011. [DOI] [PubMed] [Google Scholar]
- 93.Mao H, Katz N, Ariyanon W, Blanca-Martos L, Adybelli Z, Giuliani A, Danesi TH, Kim JC, Nayak A, Neri M, Virzi GM, Brocca A, Scalzotto E, Salvador L, Ronco C. Cardiac surgery-associated acute kidney injury. Blood Purif 37, Suppl 2: 34–50, 2014. [DOI] [PubMed] [Google Scholar]
- 94.Matejovic M, Chvojka J, Radej J, Ledvinova L, Karvunidis T, Krouzecky A, Novak I. Sepsis and acute kidney injury are bidirectional. Contrib Nephrol 174: 78–88, 2011. [DOI] [PubMed] [Google Scholar]
- 95.Matejovic M, Ince C, Chawla LS, Blantz R, Molitoris BA, Rosner MH, Okusa MD, Kellum JA, Ronco C; ADQI XIII Work Group. Renal hemodynamics in AKI: in search of new treatment targets. J Am Soc Nephrol 27: 49–58, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Maulik N, Engelman RM, Wei Z, Liu X, Rousou JA, Flack JE, Deaton DW, Das DK. Drug-induced heat-shock preconditioning improves postischemic ventricular recovery after cardiopulmonary bypass. Circulation 92: II381–388, 1995. [DOI] [PubMed] [Google Scholar]
- 97.Mayr FB, Yende S, Angus DC. Epidemiology of severe sepsis. Virulence 5: 4–11, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.McCafferty K, Byrne C, Yaqoob MM. Ischaemic conditioning strategies for the nephrologist: a promise lost in translation? Nephrol Dial Transplant 29: 1827–1840, 2014. [DOI] [PubMed] [Google Scholar]
- 99.McCullough PA. Contrast-induced acute kidney injury. J Am Coll Cardiol 51: 1419–1428, 2008. [DOI] [PubMed] [Google Scholar]
- 100.McDonald JS, McDonald RJ, Comin J, Williamson EE, Katzberg RW, Murad MH, Kallmes DF. Frequency of acute kidney injury following intravenous contrast medium administration: a systematic review and meta-analysis. Radiology 267: 119–128, 2013. [DOI] [PubMed] [Google Scholar]
- 101.McGuinness SP, Parke RL, Bellomo R, Van Haren FM, Bailey M. Sodium bicarbonate infusion to reduce cardiac surgery-associated acute kidney injury: a phase II multicenter double-blind randomized controlled trial. Crit Care Med 41: 1599–1607, 2013. [DOI] [PubMed] [Google Scholar]
- 102.Melo RS, Visona I, Almeida WS, Campos AH. Glucose-insulin infusion reduces kidney injury in an experimental model of ischemic nephropathy. Am J Nephrol 32: 603–609, 2010. [DOI] [PubMed] [Google Scholar]
- 103.Molitoris BA, Okusa MD, Palevsky PM, Chawla LS, Kaufman JS, Devarajan P, Toto RM, Hsu CY, Greene TH, Faubel SG, Kellum JA, Wald R, Chertow GM, Levin A, Waikar SS, Murray PT, Parikh CR, Shaw AD, Go AS, Chinchilli VM, Liu KD, Cheung AK, Weisbord SD, Mehta RL, Stokes JB, Thompson AM, Thompson BT, Westenfelder CS, Tumlin JA, Warnock DG, Shah SV, Xie Y, Duggan EG, Kimmel PL, Star RA. Design of clinical trials in AKI: a report from an NIDDK workshop. Trials of patients with sepsis and in selected hospital settings. Clin J Am Soc Nephrol 7: 856–860, 2012. [DOI] [PubMed] [Google Scholar]
- 104.Molitoris BA, Okusa MD, Palevsky PM, Kimmel PL, Star RA. Designing clinical trials in acute kidney injury. Clin J Am Soc Nephrol 7: 842–843, 2012. [DOI] [PubMed] [Google Scholar]
- 105.Murphy GJ, Lin H, Coward RJ, Toth T, Holmes R, Hall D, Angelini GD. An initial evaluation of post-cardiopulmonary bypass acute kidney injury in swine. Eur J Cardiothorac Surg 36: 849–855, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Nasri H, Mubarak M. Contrast induced nephropathy has to be differentiated from kidney injury due to atheroembolic disease. J Renal Inj Prev 2: 107–108, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Newhouse JH, RoyChoudhury A. Quantitating contrast medium-induced nephropathy: controlling the controls. Radiology 267: 4–8, 2013. [DOI] [PubMed] [Google Scholar]
- 108.Nguyen B, Luong L, Naase H, Vives M, Jakaj G, Finch J, Boyle J, Mulholland JW, Kwak JH, Pyo S, de Luca A, Athanasiou T, Angelini G, Anderson J, Haskard DO, Evans PC. Sulforaphane pretreatment prevents systemic inflammation and renal injury in response to cardiopulmonary bypass. J Thorac Cardiovasc Surg 148: 690–697, 2014. [DOI] [PubMed] [Google Scholar]
- 109.Nielsen LK, Bracker K, Price LL. Administration of fenoldopam in critically ill small animal patients with acute kidney injury: 28 dogs and 34 cats (2008–2012). J Vet Emerg Crit Car 25: 396–404, 2015. [DOI] [PubMed] [Google Scholar]
- 110.O'Sullivan KE, Byrne JS, Hudson A, Murphy AM, Sadlier DM, Hurley JP. The effect of obesity on acute kidney injury after cardiac surgery. J Thorac Cardiovasc Surg 150: 1622–1628, 2015. [DOI] [PubMed] [Google Scholar]
- 111.Oh SW, Chin HJ, Chae DW, Na KY. Erythropoietin improves long-term outcomes in patients with acute kidney injury after coronary artery bypass grafting. J Korean Med Sci 27: 506–511, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Okusa MD, Molitoris BA, Palevsky PM, Chinchilli VM, Liu KD, Cheung AK, Weisbord SD, Faubel S, Kellum JA, Wald R, Chertow GM, Levin A, Waikar SS, Murray PT, Parikh CR, Shaw AD, Go AS, Chawla LS, Kaufman JS, Devarajan P, Toto RM, Hsu CY, Greene TH, Mehta RL, Stokes JB, Thompson AM, Thompson BT, Westenfelder CS, Tumlin JA, Warnock DG, Shah SV, Xie Y, Duggan EG, Kimmel PL, Star RA. Design of clinical trials in acute kidney injury: a report from an NIDDK workshop—prevention trials. Clin J Am Soc Nephrol 7: 851–855, 2012. [DOI] [PubMed] [Google Scholar]
- 113.Okusa MD, Rosner MH, Kellum JA, Ronco C; Acute Dialysis Quality Initiative XIII Workshop. Therapeutic targets of human AKI: harmonizing human and animal AKI. J Am Soc Nephrol 27: 44–48, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Ortiz A, Sanchez-Nino MD, Izquierdo MC, Martin-Cleary C, Garcia-Bermejo L, Moreno JA, Ruiz-Ortega M, Draibe J, Cruzado JM, Garcia-Gonzalez MA, Lopez-Novoa JM, Soler MJ, Sanz AB; Red de Investigacion Renal and Consorcio Madrileno para investigacion del fracaso renal aguda. Translational value of animal models of kidney failure. Eur J Pharmacol 759: 205–220, 2015. [DOI] [PubMed] [Google Scholar]
- 115.Ozkok A, Edelstein CL. Pathophysiology of cisplatin-induced acute kidney injury. Biomed Res Int 2014: 967826, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Pabla N, Dong G, Jiang M, Huang S, Kumar MV, Messing RO, Dong Z. Inhibition of PKCdelta reduces cisplatin-induced nephrotoxicity without blocking chemotherapeutic efficacy in mouse models of cancer. J Clin Invest 121: 2709–2722, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Palevsky PM, Liu KD, Brophy PD, Chawla LS, Parikh CR, Thakar CV, Tolwani AJ, Waikar SS, Weisbord SD. KDOQI US commentary on the 2012 KDIGO clinical practice guideline for acute kidney injury. Am J Kidney Dis 61: 649–672, 2013. [DOI] [PubMed] [Google Scholar]
- 118.Palevsky PM, Molitoris BA, Okusa MD, Levin A, Waikar SS, Wald R, Chertow GM, Murray PT, Parikh CR, Shaw AD, Go AS, Faubel SG, Kellum JA, Chinchilli VM, Liu KD, Cheung AK, Weisbord SD, Chawla LS, Kaufman JS, Devarajan P, Toto RM, Hsu CY, Greene T, Mehta RL, Stokes JB, Thompson AM, Thompson BT, Westenfelder CS, Tumlin JA, Warnock DG, Shah SV, Xie Y, Duggan EG, Kimmel PL, Star RA. Design of clinical trials in acute kidney injury: report from an NIDDK workshop on trial methodology. Clin J Am Soc Nephrol 7: 844–850, 2012. [DOI] [PubMed] [Google Scholar]
- 119.Park KM, Kim JI, Ahn Y, Bonventre AJ, Bonventre JV. Testosterone is responsible for enhanced susceptibility of males to ischemic renal injury. J Biol Chem 279: 52282–52292, 2004. [DOI] [PubMed] [Google Scholar]
- 120.Patel NN, Angelini GD. Pharmacological strategies for the prevention of acute kidney injury following cardiac surgery: an overview of systematic reviews. Curr Pharm Des 20: 5484–5488, 2014. [DOI] [PubMed] [Google Scholar]
- 121.Patel NN, Lin H, Toth T, Jones C, Ray P, Welsh GI, Satchell SC, Sleeman P, Angelini GD, Murphy GJ. Phosphodiesterase-5 inhibition prevents postcardiopulmonary bypass acute kidney injury in swine. Ann Thorac Surg 92: 2168–2176, 2011. [DOI] [PubMed] [Google Scholar]
- 122.Patel NN, Lin H, Toth T, Welsh GI, Jones C, Ray P, Satchell SC, Sleeman P, Angelini GD, Murphy GJ. Reversal of anemia with allogenic RBC transfusion prevents post-cardiopulmonary bypass acute kidney injury in swine. Am J Physiol Renal Physiol 301: F605–F614, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Patel NN, Toth T, Jones C, Lin H, Ray P, George SJ, Welsh G, Satchell SC, Sleeman P, Angelini GD, Murphy GJ. Prevention of post-cardiopulmonary bypass acute kidney injury by endothelin A receptor blockade. Crit Care Med 39: 793–802, 2011. [DOI] [PubMed] [Google Scholar]
- 124.Perner A, Haase N, Guttormsen AB, Tenhunen J, Klemenzson G, Aneman A, Madsen KR, Moller MH, Elkjaer JM, Poulsen LM, Bendtsen A, Winding R, Steensen M, Berezowicz P, Soe-Jensen P, Bestle M, Strand K, Wiis J, White JO, Thornberg KJ, Quist L, Nielsen J, Andersen LH, Holst LB, Thormar K, Kjaeldgaard AL, Fabritius ML, Mondrup F, Pott FC, Moller TP, Winkel P, Wetterslev J, Group ST; Scandinavian Critical Care Trials Group. Hydroxyethyl starch 130/042 versus Ringer's acetate in severe sepsis. N Engl J Med 367: 124–134, 2012. [DOI] [PubMed] [Google Scholar]
- 125.Petronilho F, Constantino L, de Souza B, Reinke A, Martins MR, Fraga CM, Ritter C, Dal-Pizzol F. Efficacy of the combination of N-acetylcysteine and desferrioxamine in the prevention and treatment of gentamicin-induced acute renal failure in male Wistar rats. Nephrol Dial Transplant 24: 2077–2082, 2009. [DOI] [PubMed] [Google Scholar]
- 126.Pickering JW, James MT, Palmer SC. Acute kidney injury and prognosis after cardiopulmonary bypass: a meta-analysis of cohort studies. Am J Kidney Dis 65: 283–293, 2015. [DOI] [PubMed] [Google Scholar]
- 127.Rabb H, Griffin MD, McKay DB, Swaminathan S, Pickkers P, Rosner MH, Kellum JA, Ronco C; Acute Dialysis Quality Initiative Consensus XIII Work Group. Inflammation in AKI: current understanding, key questions, and knowledge gaps. J Am Soc Nephrol 27: 371–379, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ramesh G, Reeves WB. TNF-alpha mediates chemokine and cytokine expression and renal injury in cisplatin nephrotoxicity. J Clin Invest 110: 835–842, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Ravichandran K, Wang Q, Ozkok A, Jani A, Li H, He Z, Ljubanovic D, Weiser-Evans MC, Nemenoff RA, Edelstein CL. CD4 T cell knockout does not protect against kidney injury and worsens cancer. J Mol Med (Berl) [Epub before print]. [DOI] [PubMed] [Google Scholar]
- 130.Rewa O, Bagshaw SM. Acute kidney injury-epidemiology, outcomes and economics. Nat Rev Nephrol 10: 193–207, 2014. [DOI] [PubMed] [Google Scholar]
- 131.Rodrigues CE, Sanches TR, Volpini RA, Shimizu MH, Kuriki PS, Camara NO, Seguro AC, Andrade L. Effects of continuous erythropoietin receptor activator in sepsis-induced acute kidney injury and multi-organ dysfunction. PLoS One 7: e29893, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Roncal CA, Mu W, Croker B, Reungjui S, Ouyang X, Tabah-Fisch I, Johnson RJ, Ejaz AA. Effect of elevated serum uric acid on cisplatin-induced acute renal failure. Am J Physiol Renal Physiol 292: F116–F122, 2007. [DOI] [PubMed] [Google Scholar]
- 133.Rosner MH. Acute kidney injury in the elderly. Clin Geriatr Med 29: 565–578, 2013. [DOI] [PubMed] [Google Scholar]
- 134.Rosner MH, Portilla D, Okusa MD. Cardiac surgery as a cause of acute kidney injury: pathogenesis and potential therapies. J Intensive Care Med 23: 3–18, 2008. [DOI] [PubMed] [Google Scholar]
- 135.Sadat U, Usman A, Boyle JR, Hayes PD, Solomon RJ. Contrast medium-induced acute kidney injury. Cardiorenal Med 5: 219–228, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Sanchez-Gonzalez PD, Lopez-Hernandez FJ, Lopez-Novoa JM, Morales AI. An integrative view of the pathophysiological events leading to cisplatin nephrotoxicity. Crit Rev Toxicol 41: 803–821, 2011. [DOI] [PubMed] [Google Scholar]
- 137.Sanz AB, Sanchez-Nino MD, Martin-Cleary C, Ortiz A, Ramos AM. Progress in the development of animal models of acute kidney injury and its impact on drug discovery. Expert Opin Drug Discov 8: 879–895, 2013. [DOI] [PubMed] [Google Scholar]
- 138.Schneider AG, Bellomo R, Reade M, Peck L, Young H, Eastwood GM, Garcia M, Moore E, Harley N. Safety evaluation of a trial of lipocalin-directed sodium bicarbonate infusion for renal protection in at-risk critically ill patients. Crit Care Resusc 15: 126–133, 2013. [PubMed] [Google Scholar]
- 139.Seeliger E, Lenhard DC, Persson PB. Contrast media viscosity versus osmolality in kidney injury: lessons from animal studies. Biomed Res Int 2014: 358136, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Seeliger E, Sendeski M, Rihal CS, Persson PB. Contrast-induced kidney injury: mechanisms, risk factors, and prevention. Eur Heart J 33: 2007–2015, 2012. [DOI] [PubMed] [Google Scholar]
- 141.Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, Richards DR, McDonald-Smith GP, Gao H, Hennessy L, Finnerty CC, Lopez CM, Honari S, Moore EE, Minei JP, Cuschieri J, Bankey PE, Johnson JL, Sperry J, Nathens AB, Billiar TR, West MA, Jeschke MG, Klein MB, Gamelli RL, Gibran NS, Brownstein BH, Miller-Graziano C, Calvano SE, Mason PH, Cobb JP, Rahme LG, Lowry SF, Maier RV, Moldawer LL, Herndon DN, Davis RW, Xiao W, Tompkins RG; Inflammation, and Host Response to Injury Large Scale Collaborative Research Program. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci USA 110: 3507–3512, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Shahbazi F, Sadighi S, Dashti-Khavidaki S, Shahi F, Mirzania M, Abdollahi A, Ghahremani MH. Effect of silymarin administration on cisplatin nephrotoxicity: report from a pilot, randomized, double-blinded, placebo-controlled clinical trial. Phytother Res 29: 1046–1053, 2015. [DOI] [PubMed] [Google Scholar]
- 144.Sharp CN, Doll MA, Dupre TV, Shah PP, Subathra M, Siow D, Arteel GE, Megyesi J, Beverly LJ, Siskind LJ. Repeated administration of low-dose cisplatin in mice induces fibrosis. Am J Physiol Renal Physiol 310: F560–F568, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Shaw A. Models of preventable disease: contrast-induced nephropathy and cardiac surgery-associated acute kidney injury. Contrib Nephrol 174: 156–162, 2011. [DOI] [PubMed] [Google Scholar]
- 146.Shen S, Jin Y, Li W, Liu X, Zhang T, Xia W, Wang Y, Ma K. Recombinant human erythropoietin pretreatment attenuates acute renal tubular injury against ischemia-reperfusion by restoring transient receptor potential channel-6 expression and function in collecting ducts. Crit Care Med 42: e663–e672, 2014. [DOI] [PubMed] [Google Scholar]
- 147.Silva AT, Bayston KF, Cohen J. Prophylactic and therapeutic effects of a monoclonal antibody to tumor necrosis factor-alpha in experimental gram-negative shock. J Infect Dis 162: 421–427, 1990. [DOI] [PubMed] [Google Scholar]
- 148.Singal RK, Docking LM, Girling LG, Graham MR, Nickerson PW, McManus BM, Magil AB, Walker EK, Warrian RK, Cheang MS, Mutch WA. Biologically variable bypass reduces enzymuria after deep hypothermic circulatory arrest. Ann Thorac Surg 82: 1480–1488, 2006. [DOI] [PubMed] [Google Scholar]
- 149.Singh AP, Junemann A, Muthuraman A, Jaggi AS, Singh N, Grover K, Dhawan R. Animal models of acute renal failure. Pharmacol Rep 64: 31–44, 2012. [DOI] [PubMed] [Google Scholar]
- 150.Sleeman P, Patel NN, Lin H, Walkden GJ, Ray P, Welsh GI, Satchell SC, Murphy GJ. High fat feeding promotes obesity and renal inflammation and protects against post cardiopulmonary bypass acute kidney injury in swine. Crit Care 17: R262, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Spandou E, Tsouchnikas I, Karkavelas G, Dounousi E, Simeonidou C, Guiba-Tziampiri O, Tsakiris D. Erythropoietin attenuates renal injury in experimental acute renal failure ischaemic/reperfusion model. Nephrol Dial Transplant 21: 330–336, 2006. [DOI] [PubMed] [Google Scholar]
- 152.Sporer H, Lang F, Oberleithner H, Greger R, Deetjen P. Inefficacy of bicarbonate infusions on the course of postischaemic acute renal failure in the rat. Eur J Clin Invest 11: 311–315, 1981. [DOI] [PubMed] [Google Scholar]
- 153.Susantitaphong P, Cruz DN, Cerda J, Abulfaraj M, Alqahtani F, Koulouridis I, Jaber B; Acute Kidney Injury Advisory Group of the American Society of Nephrology. World incidence of AKI: a meta-analysis. Clin J Am Soc Nephrol 8: 1482–1493, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Swaminathan S, Rosner MH, Okusa MD. Emerging therapeutic targets of sepsis-associated acute kidney injury. Semin Nephrol 35: 38–54, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Takao K, Hagihara H, Miyakawa T. Commonalities across species do exist and are potentially important. Proc Natl Acad Sci USA 112: E347–E348, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Takao K, Miyakawa T. Genomic responses in mouse models greatly mimic human inflammatory diseases. Proc Natl Acad Sci USA 112: 1167–1172, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Takasu O, Gaut JP, Watanabe E, To K, Fagley RE, Sato B, Jarman S, Efimov IR, Janks DL, Srivastava A, Bhayani SB, Drewry A, Swanson PE, Hotchkiss RS. Mechanisms of cardiac and renal dysfunction in patients dying of sepsis. Am J Respir Crit Care Med 187: 509–517, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Tanaka H, Ishikawa E, Teshima S, Shimizu E. Histopathological study of human cisplatin nephropathy. Toxicol Pathol 14: 247–257, 1986. [DOI] [PubMed] [Google Scholar]
- 159.Tasanarong A, Duangchana S, Sumransurp S, Homvises B, Satdhabudha O. Prophylaxis with erythropoietin versus placebo reduces acute kidney injury and neutrophil gelatinase-associated lipocalin in patients undergoing cardiac surgery: a randomized, double-blind controlled trial. BMC Nephrol 14: 136, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Tasanarong A, Vohakiat A, Hutayanon P, Piyayotai D. New strategy of alpha- and gamma-tocopherol to prevent contrast-induced acute kidney injury in chronic kidney disease patients undergoing elective coronary procedures. Nephrol Dial Transplant 28: 337–344, 2013. [DOI] [PubMed] [Google Scholar]
- 161.Thakar CV, Worley S, Arrigain S, Yared JP, Paganini EP. Improved survival in acute kidney injury after cardiac surgery. Am J Kidney Dis 50: 703–711, 2007. [DOI] [PubMed] [Google Scholar]
- 162.Tracey KJ, Fong Y, Hesse DG, Manogue KR, Lee AT, Kuo GC, Lowry SF, Cerami A. Anti-cachectin/TNF monoclonal antibodies prevent septic shock during lethal bacteraemia. Nature 330: 662–664, 1987. [DOI] [PubMed] [Google Scholar]
- 163.Tran M, Tam D, Bardia A, Bhasin M, Rowe GC, Kher A, Zsengeller ZK, Akhavan-Sharif MR, Khankin EV, Saintgeniez M, David S, Burstein D, Karumanchi SA, Stillman IE, Arany Z, Parikh SM. PGC-1alpha promotes recovery after acute kidney injury during systemic inflammation in mice. J Clin Invest 121: 4003–4014, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Umpierrez G, Cardona S, Pasquel F, Jacobs S, Peng L, Unigwe M, Newton CA, Smiley-Byrd D, Vellanki P, Halkos M, Puskas JD, Guyton RA, Thourani VH. Randomized Controlled Trial of Intensive Versus Conservative Glucose Control in Patients Undergoing Coronary Artery Bypass Graft Surgery: GLUCO-CABG Trial. Diabetes Care 38: 1665–1672, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.van der Poll T. Preclinical sepsis models. Surg Infect 13: 287–292, 2012. [DOI] [PubMed] [Google Scholar]
- 166.Vattimo M, dos Santos JG. [Protective effect of sodium bicarbonate on radiological contrast medium-induced nephropathy in rats]. Rev Esc Enferm USP 47: 722–727, 2013. [DOI] [PubMed] [Google Scholar]
- 167.Voigtsberger S, Urner M, Hasler M, Z'Graggen BR, Booy C, Spahn DR, Beck-Schimmer B. Modulation of early inflammatory response by different balanced and non-balanced colloids and crystalloids in a rodent model of endotoxemia. PLoS One 9: e93863, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Wang GM, Huang HR, Wu HW, Wu C, Xu YH, Wang L, Liu XL, Wang CT, Shen Y, Li DM, Jing H. Erythropoietin attenuates cardiopulmonary bypass-induced renal inflammatory injury by inhibiting nuclear factor-kappa B P65 expression. Eur J Pharmacol 689: 154–159, 2012. [DOI] [PubMed] [Google Scholar]
- 169.Wang H, Guan Y, Karamercan MA, Ye L, Bhatti T, Becker LB, Baur JA, Sims CA. Resveratrol rescues kidney mitochondrial function following hemorrhagic shock. Shock 44: 173–180, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Warren HS, Tompkins RG, Moldawer LL, Seok J, Xu W, Mindrinos MN, Maier RV, Xiao W, Davis RW. Mice are not men. Proc Natl Acad Sci USA 112: E345, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Watanabe K. Current chemotherapeutic approaches for hepatoblastoma. Int J Clin Oncol 18: 955–961, 2013. [DOI] [PubMed] [Google Scholar]
- 172.Wei Q, Wang MH, Dong Z. Differential gender differences in ischemic and nephrotoxic acute renal failure. Am J Nephrol 25: 491–499, 2005. [DOI] [PubMed] [Google Scholar]
- 173.Weisbord SD, Gallagher M, Kaufman J, Cass A, Parikh CR, Chertow GM, Shunk KA, McCullough PA, Fine MJ, Mor MK, Lew RA, Huang GD, Conner TA, Brophy MT, Lee J, Soliva S, Palevsky PM. Prevention of contrast-induced AKI: a review of published trials and the design of the prevention of serious adverse events following angiography (PRESERVE) trial. Clin J Am Soc Nephrol 8: 1618–1631, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Wiersinga WJ, Leopold SJ, Cranendonk DR, van der Poll T. Host innate immune responses to sepsis. Virulence 5: 36–44, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Xu JR, Zhu JM, Jiang J, Ding XQ, Fang Y, Shen B, Liu ZH, Zou JZ, Liu L, Wang CS, Ronco C, Liu H, Teng J. Risk factors for long-term mortality and progressive chronic kidney disease associated with acute kidney injury after cardiac surgery. Medicine 94: e2025, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Yang FL, Subeq YM, Chiu YH, Lee RP, Lee CJ, Hsu BG. Recombinant human erythropoietin reduces rhabdomyolysis-induced acute renal failure in rats. Injury 43: 367–373, 2012. [DOI] [PubMed] [Google Scholar]
- 177.Yeboah ED, Petrie A, Pead JL. Acute renal failure and open heart surgery. Br Med J 1: 415–418, 1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Yenicerioglu Y, Yilmaz O, Sarioglu S, Ormen M, Akan P, Celik A, Camsari T. Effects of N-acetylcysteine on radiocontrast nephropathy in rats. Scand J Urol Nephrol 40: 63–69, 2006. [DOI] [PubMed] [Google Scholar]
- 179.Zacharias M, Mugawar M, Herbison GP, Walker RJ, Hovhannisyan K, Sivalingam P, Conlon NP. Interventions for protecting renal function in the perioperative period. Cochrane Database Syst Rev 9: CD003590, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Zhu J, Yin R, Shao H, Dong G, Luo L, Jing H. N-acetylcysteine to ameliorate acute renal injury in a rat cardiopulmonary bypass model. J Thorac Cardiovasc Surg 133: 696–703, 2007. [DOI] [PubMed] [Google Scholar]
