Background: Perioperative acute kidney injury (AKI) is common in surgical patients and is associated with high morbidity and mortality. There are currently few options for AKI prevention and treatment. Due to its complex pathophysiology, there is no efficient medication therapy to stop the onset of the injury or repair the damage already done. Certain anesthetics, however, have been demonstrated to affect the risk of perioperative AKI in some studies. The impact of anesthetics on renal function is particularly important as it is closely related to the prognosis of patients. Some anesthetics can induce anti-inflammatory, anti-necrotic, and anti-apoptotic effects. Propofol, sevoflurane, and dexmedetomidine are a few examples of anesthetics that have protective association with AKI in the perioperative period. Summary: In this study, we reviewed the clinical characteristics, risk factors, and pathogenesis of AKI. Subsequently, the protective effects of various anesthetic agents against perioperative AKI and the latest research are introduced. Key Message: This work demonstrates that a thorough understanding of the reciprocal effects of anesthetic drugs and AKI is crucial for safe perioperative care and prognosis of patients. However, more complete mechanisms and pathophysiological processes still need to be further studied.

Perioperative acute kidney injury (AKI) is one of the most frequent complications in surgery, which increases the incidence of postoperative complications, mortality, and the risk of post-discharge hemodialysis and has become a major burden on medical cost resources. Despite advances in anesthesia technology, perioperative AKI remains unpredictable and the current diagnostic criteria for perioperative AKI still have limitations [1]. Besides, there is no effective targeted drug therapy for AKI, and its treatment is limited to supportive care and continuous renal replacement therapy when necessary [2]. Hence, the prevention of high-risk patients and the improvement of AKI are major goals. Perioperative AKI is usually caused by a combination of factors, mainly including renal hypoperfusion, oxidative damage, and inflammation. Among them, renal hypoperfusion is the most important mechanism.

Nowadays, more and more research has found that intraoperative anesthesia can affect renal function in different ways, and several anesthetic drugs can improve perioperative AKI through antioxidant, anti-inflammatory, and other mechanisms, like propofol, sevoflurane, isoflurane, and articaine [3]. For example, an animal experiment has shown that sevoflurane can alleviate kidney injury in liver transplantation rats by greatly decreasing the 24-h serum creatinine (Scr) after reperfusion and neutrophil gelatinase-associated lipocalin (NGAL) concentrations after 2 h of reperfusion [4]. Another study has shown that propofol can protect renal function in mice after cecum ligation and puncture surgery to improve survival outcomes and alleviation of AKI [5]. The mechanisms underlying the association between these perioperative anesthetics and AKI have also been extensively studied in recent years. For instance, a study by Wang et al. found that dexmedetomidine (DEX) can improve renal function and ameliorate AKI by regulating p75 neurotrophin receptor/p38 mitogen-activated protein kinase/Jun N-terminal kinase signaling pathways [6]. However, the specific mechanisms of these anesthetic drugs for AKI still have many ambiguities. Considering the current lack of summary about the protection and mechanisms of tranquilizers and anesthetic drugs to perioperative AKI, we reviewed the pathogenesis and mechanism of perioperative AKI and analyzed the risk factors involved in it. Meanwhile, we summarized the latest research and mechanisms of perioperative anesthesia on AKI and concluded the clinical data of related drugs in Table 1. With this knowledge, this article may indicate the directions of the safe use of tranquilizers and DEX drugs, and make people better understand perioperative management for preventing perioperative AKI.

Table 1.

Overview of clinical studies on the effects of anesthetic agents on AKI

Anesthetic agentsClinical studyNumber of patients and incidence of AKIClinical effectsLimitationsReference
Comparison of propofol and sevoflurane Effect of both propofol and sevoflurane on AKI after pediatric liver transplantation 120; propofol – 47% (28/60), sevoflurane – 8% (17/60) Sevoflurane anesthesia modestly reduces the incidence of AKI compared to isoproterenol Small sample size; the serum examination might be a poor specificity test [7
 Effect of propofol (total intravenous anesthesia) versus sevoflurane (inhalation anesthesia) on the occurrence of AKI after pneumonectomy 1,322; propofol – 3.5% (23/661), sevoflurane – 3.6% (24/661) There was no significant difference in the incidence of AKI after pneumonectomy between the two groups Selection bias; a single-center study; underdiagnosed AKI within the study cohort; limitation of accuracy of the preoperative eGFR data [8
 Effect of propofol (total intravenous anesthesia) versus sevoflurane (inhaled anesthesia) on the occurrence of AKI after bilateral pulmonary oxygenation 60; propofol – 13% (4/30), sevoflurane – 38% (11/30) Significantly fewer patients in the isoproterenol group developed AKI compared to the sevoflurane group Whether blood concentrations are sufficient for renal protection remains controversial; many factors may affect the protective effect of narcotic drugs. At present, it is difficult to draw the conclusion that propofol is not related to the attenuation of oxidative stress. The anti-inflammatory effect of midazolam may have an effect on cytokine levels [9
 Effect of both on the occurrence of AKI after cardiac surgery with extracorporeal circulation 112; propofol – 10.7% (6/56), sevoflurane – 37.5% (21/56) Compared with sevoflurane anesthesia, isoproterenol anesthesia significantly reduces the incidence and severity of AKI in CPB patients undergoing heart valve surgery Objective defects; small sample size; the relative risk of AKI between the propofol group and sevoflurane group probably overestimated the true effect [10
 Effect of both on the occurrence of AKI after colorectal surgery 4,320; propofol – 8.9% (192/2,160), sevoflurane – 11.2% (242/2,160) Sevoflurane anesthesia may be associated with a modest increase in the incidence of AKI compared to isoproterenol anesthesia Confounding parameters; the results were applied only to patients undergoing colorectal surgery [11
DEX Effect of DEX on the occurrence of AKI after cardiac valve replacement with extracorporeal circulation 72; placebo – 25% (9/36), DEX – 8.3% (3/36) DEX attenuates renal injury and reduces the incidence of AKI in patients undergoing extracorporeal heart valve replacement The effect of DEX on renal blood flow and the effect of renal blood flow on renal function during CPB have not been explored; a long-time follow-up of patients has not been performed; the diagnosis of AKI was mainly based on urine volume and serum Cr which may affect the accuracy [12
 Effect of DEX on the occurrence of AKI after endovascular aortic repair in Stanford type B aortic coarctation 98; placebo – 22% (11/50), DEX – 6.3% (3/48) DEX reduces the incidence of AKI after endovascular aortic repair in patients with aortic coarctation Restricted patient selection; the current sample size precludes any firm statistical or clinical inferences for the secondary and long-term outcomes; hydroxyethyl starch 6% 130/0.4 was used; a single-center study [13
 Effect of DEX on the occurrence of AKI after aortic surgery requiring extracorporeal circulation 108; placebo – 31% (17/54), DEX – 13% (7/54) DEX reduces the incidence of AKI after active surgery requiring extracorporeal circulation A lower-than-predicted incidence of AKI; results should not be extrapolated to patients with decreased cardiac function, renal function, or both; did not simultaneously assess the levels of plasma catecholamines or inflammatory markers [14
Midazolam Effect of isoproterenol versus midazolam treatment on the incidence of AKI in patients in the ICU during the first 7 days 1,396; propofol – 55.0%, midazolam – 67.3% Patients treated with isoproterenol have a lower risk of AKI, fluid-related complications A single-center data source; low number of patients receiving midazolam; the retrospective nature [15
Setrons Effect of ondansetron on the occurrence of AKI in ICU patients 149; ondansetron – 12.3% (6/49), prochloraz – 0% (0/49), metoclopramide – 5.48% (5/91) The use of antiemetics did not change the risk of AKI. Ondansetron was associated with a significant reduction in 5-day mortality The difficulty in clearly defining the temporality of antiemetic administration and the development of AKI [16
Anesthetic agentsClinical studyNumber of patients and incidence of AKIClinical effectsLimitationsReference
Comparison of propofol and sevoflurane Effect of both propofol and sevoflurane on AKI after pediatric liver transplantation 120; propofol – 47% (28/60), sevoflurane – 8% (17/60) Sevoflurane anesthesia modestly reduces the incidence of AKI compared to isoproterenol Small sample size; the serum examination might be a poor specificity test [7
 Effect of propofol (total intravenous anesthesia) versus sevoflurane (inhalation anesthesia) on the occurrence of AKI after pneumonectomy 1,322; propofol – 3.5% (23/661), sevoflurane – 3.6% (24/661) There was no significant difference in the incidence of AKI after pneumonectomy between the two groups Selection bias; a single-center study; underdiagnosed AKI within the study cohort; limitation of accuracy of the preoperative eGFR data [8
 Effect of propofol (total intravenous anesthesia) versus sevoflurane (inhaled anesthesia) on the occurrence of AKI after bilateral pulmonary oxygenation 60; propofol – 13% (4/30), sevoflurane – 38% (11/30) Significantly fewer patients in the isoproterenol group developed AKI compared to the sevoflurane group Whether blood concentrations are sufficient for renal protection remains controversial; many factors may affect the protective effect of narcotic drugs. At present, it is difficult to draw the conclusion that propofol is not related to the attenuation of oxidative stress. The anti-inflammatory effect of midazolam may have an effect on cytokine levels [9
 Effect of both on the occurrence of AKI after cardiac surgery with extracorporeal circulation 112; propofol – 10.7% (6/56), sevoflurane – 37.5% (21/56) Compared with sevoflurane anesthesia, isoproterenol anesthesia significantly reduces the incidence and severity of AKI in CPB patients undergoing heart valve surgery Objective defects; small sample size; the relative risk of AKI between the propofol group and sevoflurane group probably overestimated the true effect [10
 Effect of both on the occurrence of AKI after colorectal surgery 4,320; propofol – 8.9% (192/2,160), sevoflurane – 11.2% (242/2,160) Sevoflurane anesthesia may be associated with a modest increase in the incidence of AKI compared to isoproterenol anesthesia Confounding parameters; the results were applied only to patients undergoing colorectal surgery [11
DEX Effect of DEX on the occurrence of AKI after cardiac valve replacement with extracorporeal circulation 72; placebo – 25% (9/36), DEX – 8.3% (3/36) DEX attenuates renal injury and reduces the incidence of AKI in patients undergoing extracorporeal heart valve replacement The effect of DEX on renal blood flow and the effect of renal blood flow on renal function during CPB have not been explored; a long-time follow-up of patients has not been performed; the diagnosis of AKI was mainly based on urine volume and serum Cr which may affect the accuracy [12
 Effect of DEX on the occurrence of AKI after endovascular aortic repair in Stanford type B aortic coarctation 98; placebo – 22% (11/50), DEX – 6.3% (3/48) DEX reduces the incidence of AKI after endovascular aortic repair in patients with aortic coarctation Restricted patient selection; the current sample size precludes any firm statistical or clinical inferences for the secondary and long-term outcomes; hydroxyethyl starch 6% 130/0.4 was used; a single-center study [13
 Effect of DEX on the occurrence of AKI after aortic surgery requiring extracorporeal circulation 108; placebo – 31% (17/54), DEX – 13% (7/54) DEX reduces the incidence of AKI after active surgery requiring extracorporeal circulation A lower-than-predicted incidence of AKI; results should not be extrapolated to patients with decreased cardiac function, renal function, or both; did not simultaneously assess the levels of plasma catecholamines or inflammatory markers [14
Midazolam Effect of isoproterenol versus midazolam treatment on the incidence of AKI in patients in the ICU during the first 7 days 1,396; propofol – 55.0%, midazolam – 67.3% Patients treated with isoproterenol have a lower risk of AKI, fluid-related complications A single-center data source; low number of patients receiving midazolam; the retrospective nature [15
Setrons Effect of ondansetron on the occurrence of AKI in ICU patients 149; ondansetron – 12.3% (6/49), prochloraz – 0% (0/49), metoclopramide – 5.48% (5/91) The use of antiemetics did not change the risk of AKI. Ondansetron was associated with a significant reduction in 5-day mortality The difficulty in clearly defining the temporality of antiemetic administration and the development of AKI [16

AKI, acute kidney injury; CPB, cardiopulmonary bypass; DEX, dexmedetomidine; ICU, intensive care unit.

Clinical Features and Risk Factors

Perioperative AKI is defined as an acute decrease in renal function that occurs from preoperative days 5–7 to postoperative days 7–12 [17]. However, AKI has not had a unified definition for many years. The Kidney Disease: Improving Global Outcomes (KDIGO) guidelines published in 2012 have now become widely accepted diagnostic criteria for AKI: (1) a 0.3 mg/dL increase in Scr within 48 h, (2) a 50% rise in Scr within 7 days, or (3) a urine output of <0.5 mL/kg/h for >6 h. The guidelines also classify AKI into three stages by different Scr and/or urine output [18]. The reported incidence of perioperative AKI is 2–18% in patients and 22–57% in intensive care patients [19]. In the USA, it is estimated that 30–40% of AKI cases occur postoperatively [20]. Cardiac surgery, especially using cardiopulmonary bypass (CPB), has a perioperative incidence of more than 20% and is the surgery with the highest incidence of AKI. Vascular surgery also carries a higher risk of AKI, with postoperative AKI rates ranging from 20% to 70% [21]. Furthermore, the incidence of AKI after orthotopic liver transplantation ranges from 17% to 95% depending on the definition, which is typically due to a combination of factors [22]. Notably, AKI occurs in the first week after liver transplantation surgery due possibly to prerenal azotemia and acute tubular necrosis, while in 2–4 weeks the majority of causes are sepsis and calcineurin inhibitor toxicity [23].

There are many types of risk factors related to perioperative AKI. The more risk factors, the greater the likelihood of developing AKI. These risk factors can be classified into six groups: adverse clinical status, underlying disease, use of nephrotoxic drugs, contrast injection, some surgical interventions, and anesthetic factors, and are summarized in Table 2 according to preoperative, intraoperative, and postoperative factors. First, patients with poor clinical status are more prone to AKI, including age ≥56 years, preoperative creatinine >106 mol/L, hypoalbuminemia, smoking, coagulopathy, obesity, etc. [24, 25]. Patients with underlying diseases are also more likely to develop perioperative AKI, such as chronic kidney diseases (CKD), hypertension, peripheral vascular disease, diabetes, and chronic obstructive pulmonary disease [18, 26]. Among them, preoperative CKD history is a major risk factor for AKI, conferring as much as a 10-fold risk. At the same time, patients with AKI are also more likely to develop CKD [27]. Besides, up to 25% of severe AKI cases are thought to be caused by nephrotoxic drugs, including nonsteroidal anti-inflammatory drugs, angiotensin-converting enzyme inhibitors/angiotensin receptor blockers, and some antibiotics [28]. Intravenous or arterial injection of contrast media also increases the risk of perioperative AKI, and arterial injection of contrast media is more nephrotoxic [29]. Some special operations may also increase the risk of AKI, such as CPB, intraoperative blood transfusion, increased intra-abdominal pressure in abdominal surgery, and so on. CPB lasting for more than 2 h has been verified to induce a significant increase in the incidence of AKI after cardiac surgery [30]. Finally, anesthesia factors have also been found to have a significant impact on perioperative AKI. A study showed that epidural analgesia during subtotal hepatectomy may also induce AKI with sympatholysis, peripheral vasodilation, and hypotension [31]. Rodgers et al. [32] have demonstrated that patients treated with spinal anesthesia are less likely to develop AKI than those treated with general anesthesia. Nevertheless, the relationship between perioperative anesthetics and AKI is still unclear at present, and more experimental evidence is needed to explore its underlying mechanism.

Table 2.

Risk factors for perioperative AKI

Risk factorsDetailsReferences
Preoperative risk factors Clinical status Gender Women appear to be associated with an increased risk of developing AKI during cardiac surgery, while men in general surgery have a two-fold higher risk of developing postoperative AKI [33
  Advanced age (>56 years)  [34
  Obesity (BMI >25.0) Increased oxidative stress, intra-abdominal hypertension, inflammation, and underestimated fluid requirements [35, 36
  Creatinine >106 mol/L  [24
 Underlying diseases CKD Decreased kidney function [24
  Peripheral vascular disease  [37
  Anemia Renal hypoxia [24, 38
  Diabetes Glucose metabolism disorder could generate many glycosylation products, which are then deposited in glomerular endothelial cells, thereby causing inflammation [39
  Congestive heart failure Sudden decrease in renal blood flow and GFR [40
  COPD Airflow limitation, which may lead to chronic renal hypoxia [41
  Malnutrition Significant changes in renal hemodynamics and renal concentration capacity [42
  Sepsis, crush injury, liver failure Acute preoperative factors [18
 Drugs ACEI/ARB Renal arterial vasoconstriction leading to renal hypoperfusion; low blood pressure [43, 44
  NSAIDs Directly reduces renal blood flow and inhibits vasodilatory prostaglandins, thus leading to renal vasoconstriction; may cause interstitial nephritis [24, 26
  Diuretics Low blood volume [39
  Several antibiotics (like β-lactam, neoquinolones, and aminoglycosides) Dose-dependent tubular necrosis; allergic tubulointerstitial nephritis; tubular crystal formation [45, 46
 Intravenous/arterial contrast agents  Direct toxic effects; renal medullary hypoxia caused by changes in renal hemodynamics; renal microcirculation disturbances [24
Intraoperative risk factors Surgical factors CPB Systemic inflammatory response, coagulopathy, and embolism [18, 26
  Direct urinary tract injury  [47
  Transfusion Progressive hematological changes that occur during blood storage damage the kidneys [4
  Increase of intra-abdominal pressure Venous congestion, increased intrarenal pressure, and decreased renal perfusion [48
 Anesthetic factors Hypotension (MAP <60 mm Hg 20 min, or <55 mm Hg 10 min) Vasodilation, decreased renal perfusion [49
  Epidural analgesia Sympatholysis, peripheral vasodilation, and hypotension [31
  Anesthetic drugs (sevoflurane, propofol, etc.)  [4, 5
Postoperative factors  Decreased cardiac output  [50
  Preoperative comorbid illnesses Diabetes and any chronic disease of the heart, lung, or liver [51
  Elevated American Society of Anesthesiologists physical status classification score Patient has severe systemic diseases, and the organ function of the body has been in a decompensated state [51, 52
  24-h intracardiac intubation  [26
  Mechanical ventilation Oliguria and sodium and water retention [53
Risk factorsDetailsReferences
Preoperative risk factors Clinical status Gender Women appear to be associated with an increased risk of developing AKI during cardiac surgery, while men in general surgery have a two-fold higher risk of developing postoperative AKI [33
  Advanced age (>56 years)  [34
  Obesity (BMI >25.0) Increased oxidative stress, intra-abdominal hypertension, inflammation, and underestimated fluid requirements [35, 36
  Creatinine >106 mol/L  [24
 Underlying diseases CKD Decreased kidney function [24
  Peripheral vascular disease  [37
  Anemia Renal hypoxia [24, 38
  Diabetes Glucose metabolism disorder could generate many glycosylation products, which are then deposited in glomerular endothelial cells, thereby causing inflammation [39
  Congestive heart failure Sudden decrease in renal blood flow and GFR [40
  COPD Airflow limitation, which may lead to chronic renal hypoxia [41
  Malnutrition Significant changes in renal hemodynamics and renal concentration capacity [42
  Sepsis, crush injury, liver failure Acute preoperative factors [18
 Drugs ACEI/ARB Renal arterial vasoconstriction leading to renal hypoperfusion; low blood pressure [43, 44
  NSAIDs Directly reduces renal blood flow and inhibits vasodilatory prostaglandins, thus leading to renal vasoconstriction; may cause interstitial nephritis [24, 26
  Diuretics Low blood volume [39
  Several antibiotics (like β-lactam, neoquinolones, and aminoglycosides) Dose-dependent tubular necrosis; allergic tubulointerstitial nephritis; tubular crystal formation [45, 46
 Intravenous/arterial contrast agents  Direct toxic effects; renal medullary hypoxia caused by changes in renal hemodynamics; renal microcirculation disturbances [24
Intraoperative risk factors Surgical factors CPB Systemic inflammatory response, coagulopathy, and embolism [18, 26
  Direct urinary tract injury  [47
  Transfusion Progressive hematological changes that occur during blood storage damage the kidneys [4
  Increase of intra-abdominal pressure Venous congestion, increased intrarenal pressure, and decreased renal perfusion [48
 Anesthetic factors Hypotension (MAP <60 mm Hg 20 min, or <55 mm Hg 10 min) Vasodilation, decreased renal perfusion [49
  Epidural analgesia Sympatholysis, peripheral vasodilation, and hypotension [31
  Anesthetic drugs (sevoflurane, propofol, etc.)  [4, 5
Postoperative factors  Decreased cardiac output  [50
  Preoperative comorbid illnesses Diabetes and any chronic disease of the heart, lung, or liver [51
  Elevated American Society of Anesthesiologists physical status classification score Patient has severe systemic diseases, and the organ function of the body has been in a decompensated state [51, 52
  24-h intracardiac intubation  [26
  Mechanical ventilation Oliguria and sodium and water retention [53

AKI, acute kidney injury; BMI, body mass index; CKD, chronic kidney disease; GFR, glomerular filtration rate; COPD, chronic obstructive pulmonary disease; ACEI/ARB, angiotensin-converting enzyme inhibitor/angiotensin receptor blocker; NSAIDs, nonsteroidal anti-inflammatory drugs; CPB, cardiopulmonary bypass; MAP, mean artery pressure.

Pathogenesis

Perioperative AKI is usually caused by multiple injuries from complex causes, mainly due to the combined effect of renal hypoperfusion, oxidative damage, and inflammation (Fig. 1). Among them, renal hypoperfusion is the main factor. During surgery and critical illness, systemic changes in cardiac output, systemic vascular resistance, and renal venous pressure can alter renal cortical and medullary perfusion. Symons et al. [54] classified the causes of renal hypoperfusion into four categories: volume depletion, decreased cardiac output, decreased systemic vascular resistance, and increased renal artery resistance. First, the kidneys can release vasopressin (ADH) and angiotensin II by activating the sympathetic nervous system, and activate the renin-angiotensin-aldosterone system to make water-sodium retent to maintain glomerular filtration rate (GFR) [24]. However, when renal hypoperfusion lasts too long, angiopoietin II can promote afferent arteriole contraction, which instead leads to renal tubule ischemia and decreased GFR. However, compared to norepinephrine alone, Meersch proved that angiotensin II could reduce renin plasma concentrations significantly in patients at risk for AKI (high postoperative Δ-renin) with hypotonia after cardiac surgery, which acts as a hormone substitution therapy [55]. Moreover, ischemia-reperfusion (IR) induces renal mitochondria to produce reactive oxygen species, which impairs endothelial function and perfusion homeostasis by decreasing nitric oxide production. Furthermore, it will activate pro-inflammatory transcription factors such as nuclear factor-kappa B (NF-кB), and destroy surrounding tissue by oxidizing lipids, DNA, and other proteins [37]. Systemic inflammation induced by surgical stress and cytokine release can induce irreversible microcirculatory disturbances in renal tubules, further increasing systemic inflammation [56]. Neutrophils and macrophages are also recruited to the renal interstitium to regulate inflammation. Damaged endothelial cells will increase the production of intercellular adhesion molecule-1, which can stimulate white blood cells and cause blockage of small blood vessels, thereby increasing local inflammation within kidney tissue [18]. In addition, Paneth cells located in the small intestine can be activated by AKI-induced inflammation, which can induce great release of inflammatory mediators such as interleukin-17A (IL-17A) and may lead to sepsis and multiple organ failure [57]. Although there are a lot of studies have been performed on perioperative AKI, most of them are retrospective studies. In the future, we still need more prospective work to explore the pathogenesis of perioperative AKI.

Fig. 1.

Pathogenesis of perioperative AKI. AKI, acute kidney injury; ANGⅡ, angiopoietin-2; GFR, glomerular filtration rate; ROS, reactive oxygen species; NO, nitric oxide; NF‐κB, nuclear factor-кB; ICAM-1, intercellular adhesion molecule; WBC, white blood cell; IL-17A, interleukin-17A; MOF, multiple organ failure.

Fig. 1.

Pathogenesis of perioperative AKI. AKI, acute kidney injury; ANGⅡ, angiopoietin-2; GFR, glomerular filtration rate; ROS, reactive oxygen species; NO, nitric oxide; NF‐κB, nuclear factor-кB; ICAM-1, intercellular adhesion molecule; WBC, white blood cell; IL-17A, interleukin-17A; MOF, multiple organ failure.

Close modal

Anesthesia is an important factor affecting renal function in the perioperative period. Proper anesthetic management can reduce the risk of postoperative renal complications, and the choice of anesthetic may also affect renal outcome [58]. Anesthetics we list below appear to exhibit the potential to attenuate AKI through different mechanisms (Table 3, Fig. 2).

Table 3.

Overview of common anesthetic agents and their protective effects on perioperative AKI

ActionDrugResearch objectResearch findingsReference
Intravenous anesthetic Propofol Human and animals Anti-inflammatory, antioxidant, and cytoprotective effects [59‒61
  Human Cystatin C, serum IL-6, C-reactive protein, and fractional neutrophil count↓ [10
Animals Cx32 protein expression ↓; ROS and oxidative stress ↓ [62
Animals BMP-7 expression ↑; inflammatory cytokines (TNF-α, MCP-1) expression ↓; free radical generation and cell death ↓; oxidative stress ↓ [63
Animals miR-290-5p expression ↑; CCL-2 and inflammatory cytokines levels ↓ [5
Volatile anesthetic Sevoflurane Animals Necrosis and inflammatory renal cell death ↓ [64
 Isoflurane Human and animals Expression of NGAL, TNF-α, IL-6, and NF-кB ↓ [4, 65‒67
Human and animals Release of TGF-β1 ↑ [68‒70
Animals Level of PTEN ↓, pAkt ↑ [71
Animals Release of TGF-β1 ↑; IL-11 mRNA and protein synthesis↑ [72
Animals CD73 and adenosine generation ↑ [73
Local anesthetic Articaine Animals NF‐ĸB activation and TLR4 expression ↓; NLRP3 inflammasome pathway ↓ [74
ActionDrugResearch objectResearch findingsReference
Intravenous anesthetic Propofol Human and animals Anti-inflammatory, antioxidant, and cytoprotective effects [59‒61
  Human Cystatin C, serum IL-6, C-reactive protein, and fractional neutrophil count↓ [10
Animals Cx32 protein expression ↓; ROS and oxidative stress ↓ [62
Animals BMP-7 expression ↑; inflammatory cytokines (TNF-α, MCP-1) expression ↓; free radical generation and cell death ↓; oxidative stress ↓ [63
Animals miR-290-5p expression ↑; CCL-2 and inflammatory cytokines levels ↓ [5
Volatile anesthetic Sevoflurane Animals Necrosis and inflammatory renal cell death ↓ [64
 Isoflurane Human and animals Expression of NGAL, TNF-α, IL-6, and NF-кB ↓ [4, 65‒67
Human and animals Release of TGF-β1 ↑ [68‒70
Animals Level of PTEN ↓, pAkt ↑ [71
Animals Release of TGF-β1 ↑; IL-11 mRNA and protein synthesis↑ [72
Animals CD73 and adenosine generation ↑ [73
Local anesthetic Articaine Animals NF‐ĸB activation and TLR4 expression ↓; NLRP3 inflammasome pathway ↓ [74

AKI, acute kidney injury; Cx32, connexin32; ROS, reactive oxygen species; BMP-7, morphogenetic protein-7; TNF-α, tumor necrosis factor; MCP-1, monocyte chemotactic protein; miR, microRNAs; CCL-2, chemokine C-C motif ligand 2; IL-6, interleukin-6; NF-кB, nuclear factor-кB; TGF-β1, transforming growth factor-β1; PTEN, chromosome 10; pAkt, phosphorylated protein kinase B; IL-11, interleukin-11; CD73, ecto-5′-nucleotidase; TLR4, toll‐like receptor 4; NLRP3, nucleotide-binding oligomerization domain-like receptor protein-3.

Fig. 2.

Overview of common anesthetic agents and their protective effects on perioperative AKI. AKI, acute kidney injury; Cx32, connexin32; BMP-7, morphogenetic protein-7; TNF-α, tumor necrosis factor; MCP-1, monocyte chemotactic protein; miR, microRNAs; CCL-2, chemokine C-C motif ligand 2; IRI, ischemia/reperfusion injury; IL-6, interleukin-6; NF-кB, nuclear factor-кB; TGF-β1, transforming growth factor-β1; PTEN, chromosome 10; PI3K/Akt, phosphoinositide 3-kinase/protein kinase B; pAkt, phosphorylated protein kinase B; IL-1, interleukin-1; IL-8, interleukin-8; IL-11, interleukin-11; CD73, ecto-5′-nucleotidase.

Fig. 2.

Overview of common anesthetic agents and their protective effects on perioperative AKI. AKI, acute kidney injury; Cx32, connexin32; BMP-7, morphogenetic protein-7; TNF-α, tumor necrosis factor; MCP-1, monocyte chemotactic protein; miR, microRNAs; CCL-2, chemokine C-C motif ligand 2; IRI, ischemia/reperfusion injury; IL-6, interleukin-6; NF-кB, nuclear factor-кB; TGF-β1, transforming growth factor-β1; PTEN, chromosome 10; PI3K/Akt, phosphoinositide 3-kinase/protein kinase B; pAkt, phosphorylated protein kinase B; IL-1, interleukin-1; IL-8, interleukin-8; IL-11, interleukin-11; CD73, ecto-5′-nucleotidase.

Close modal

Intravenous Anesthetic

Propofol, an ultra-fast-acting intravenous anesthetic with anti-inflammatory and antioxidant properties [59, 60] and few adverse effects [75], has been widely used in clinical applications. Among the widely used anesthetics, accumulated data suggest that it has a protective effect against renal ischemia/reperfusion injury (IRI) [76]. With the continuous in-depth study of its renal protective mechanism, propofol is regarded as a promising renal protectant and various clinical studies have shown that it can reduce the risk of perioperative AKI. A study comparing the first 48 h of outcomes in intensive care unit patients receiving propofol or midazolam showed that receiving propofol was associated with a lower risk of AKI, fluid-related complications, and the need for RRT [15]. In another clinical study, Yoo et al. demonstrated a significant reduction in the incidence and severity of AKI in CPB patients undergoing valvular heart surgery using propofol anesthesia compared to sevoflurane. It was also reported that postoperative cystatin C, serum IL-6, C-reactive protein, and fractional neutrophil count were significantly decreased [10], which may be related to the anti-inflammatory property of propofol [60]. However, due to the limitations of the experiment such as the small sample size, the effect of propofol on AKI has always been controversial. In a retrospective observational study involving 5,663 individuals (blood creatinine not tested 1 month before surgery was not included in the study), Oh et al. failed to observe an association between propofol use in the perioperative period and the occurrence of AKI after total hip or knee arthroplasty. Therefore, more clinical trials need to be conducted.

However, the research progress of propofol in mice with AKI has made remarkable achievements. Propofol exhibited the ability to lessen oxidative stress and decrease AKI after autologous orthotopic liver transplantation by reducing connexin32 (Cx32) activity, according to a study on male rats which were given the selective Cx32 inhibitor, 2-aminoethoxydiphenyl borate or propofol or not [62]. Another study showed that propofol treatment could increase bone morphogenetic protein-7 expression, decrease inflammatory cytokines (tumor necrosis factor [TNF-α] and monocyte chemotactic protein [MCP-1]), and inhibit oxidative stress (free radical production, H2O2-induced oxidative stress damage) to reduce the risk of kidney exposure to AKI caused by sepsis [63]. Small noncoding RNAs known as microRNAs (miRNAs) have the potential to serve as biomarkers for the early detection of AKI [77]. Chemokine C-C motif ligand 2 (CCL-2) may be central to the pathological process of renal injury and has potential as a predictor of renal function in patients with kidney disease [78, 79]. Zheng et al. found that miR-290-5p could inversely regulate the expression of CCL-2. In cecal ligation and puncture-treated mice, propofol treatment was shown to inhibit inflammation and reduce renal injury in vivo by increasing miR-290-5p levels and decreasing CCL-2 expression [5]. Furthermore, propofol is a potent membranous antioxidant and cytoprotective agent, which can alleviate hypothermia and ischemic AKI in renal transplantation [61].

Volatile Anesthetic

Historically, several volatile anesthetics used clinically in the past are nephrotoxic (e.g., methoxyflurane) and were no longer routinely administered, but recent studies have shown that modern halogenated volatile anesthetics induce potent anti-inflammatory and anti-necrotic effects and protect the organism from ischemic AKI (e.g., isoflurane, desflurane, and sevoflurane) [80]. Sevoflurane is a widely used volatile anesthetic with antioxidant stress and anti-inflammatory activities, which has a powerful multi-organ protective effect in the perioperative period. Sevoflurane could improve renal function and effectively regulate IR injury and prevent ischemic AKI in vivo and in vitro [80, 81]. In a pediatric liver transplantation trial, patients in the sevoflurane group had more stable hemodynamics and significantly lower levels of TNF-α, IL-18, and serum NGAL during hepatic reperfusion than those in the propofol group. NGAL is a marker of renal injury which is significantly elevated in urine after AKI [65, 66]. Compared to propofol, sevoflurane anesthesia may be associated with a small decrease in the incidence of AKI after pediatric liver transplantation [7]. Anyway, due to the different evaluation criteria and the existence of experimental limitations, the effect of propofol and sevoflurane has always been controversial. Sevoflurane performed worse than propofol during anesthesia in a single-center parallel randomized controlled study in terms of decreased urine production, salt excretion, and elevated plasma renin. Sevoflurane-induced renal hypoperfusion could raise the risk of renal impairment in patients with additional risk factors, such as chronic kidney disease, even if there is not a clear link between intraoperative oliguria and postoperative AKI [82]. In another Volatile Anesthetic Protection of Renal Transplants-1 (VAPOR-1) randomized controlled trial, sevoflurane anesthesia increased the risk of kidney damage compared to propofol anesthesia [83]. However, multiple studies demonstrate the protective effect of volatile anesthetics against renal IR injury [76], and more experiments are needed before selecting anesthetics.

Decades ago, sevoflurane was found to degrade into compound A, which is nephrotoxic in rats. However, it has not been discovered that compound A affects human renal function [84]. IRI is one of the major contributors of AKI [85]. If tissue damage caused by IRI can be moderately alleviated, AKI could be controlled. Early in 2004, volatile anesthetics were reported to protect the kidney from IRI by reducing necrosis and inflammatory renal cell death. In addition, this study suggests that the reduction of cytokine and chemokine expression by volatile anesthetics may contribute to the decrease of renal injury after IR [64]. A subsequent study conducted on liver transplantation mice anesthetized by sevoflurane showed significant reductions in plasma TNF-α and IL-6 concentrations and a significantly lower NGAL (which means improved renal function) after 2 h of reperfusion, which is consistent with previous findings [4]. Similar experimental results were revealed in another research [67]. In the following years, Lee also proposed that sevoflurane prevents renal IRI via transforming growth factor-β1 (TGF-β1). Through TGF-β1-mediated anti-inflammatory and anti-necrotic effects, volatile anesthesia presents protective effects on the kidney [68‒70]. Recently, it was reported that miRNAs play a key role in renal diseases and the regulation of IRI [86]. Data from Yamamoto et al. [71] suggested that pretreatment anesthesia and ischemic preconditioning with sevoflurane could affect different miRNAs that can similarly regulate the chromosome 10 (PTEN)/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway (suppressed PTEN and increased phosphorylated Akt), resulting in renal protection. PTEN blocks the downstream activity of the PI3K/Akt pathway, while Akt activation can promote cell survival through anti-apoptosis [87]. This means sevoflurane can reduce renal IRI by inhibiting miRNAs, which would be a good target to prevent AKI. However, these animal studies have not yet been translated into human studies, so further human studies are needed.

The use of sedative and antiemetic drugs is often necessary for the perioperative care of patients. Many studies have reported the different effects and protective mechanisms of these drugs against AKI (Table 4, Fig. 3). The risk of AKI is further reduced by providing a tailored dosing regimen that ensures optimal comfort.

Table 4.

Overview of common sedatives and antiemetics and their protective effects on perioperative AKI

ActionDrugResearch objectResearch findingsReference
Sedative Midazolam  Need further studies  
 DEX Human and animals Anti-inflammatory, antioxidant, and anti-apoptosis effects [6
 Human and animals Sympathy-adrenal hyperactivity ↓; maintaining hemodynamic variables; diuresis [13, 88
Human SOD activity ↑ [12
Animals Inflammation and premature cellular senescence ↓ [89
Animals Inactivation of GSK-3β ↑; level of Nrf2 ↑ [90
Animals iNOS activity ↓; expression of iNOS mRNA↓; NO production ↓ [91
Animals Oxidative stress damage and NLRP3 inflammasome activation ↓; mRNA and protein expressions of TLR4, NOX4, NF‐κB, and NLRP3 ↓ [92
Antiemetic Ondansetron Human NF-кB pathway and JAK-STAT pathway ↓ [93
  Human Anti-inflammatory, antiplatelet aggregation, and anti-apoptosis effects [94
Tropisetron Animals Expression of inflammatory molecules including TNF-α, IL-1β, and iNOS ↓ [95
Diphenhydramine Animals Inflammatory cytokines, apoptosis, and oxidative stress ↓; H1R’s preventive effect ↑; OCT 2 ↓ [96
ActionDrugResearch objectResearch findingsReference
Sedative Midazolam  Need further studies  
 DEX Human and animals Anti-inflammatory, antioxidant, and anti-apoptosis effects [6
 Human and animals Sympathy-adrenal hyperactivity ↓; maintaining hemodynamic variables; diuresis [13, 88
Human SOD activity ↑ [12
Animals Inflammation and premature cellular senescence ↓ [89
Animals Inactivation of GSK-3β ↑; level of Nrf2 ↑ [90
Animals iNOS activity ↓; expression of iNOS mRNA↓; NO production ↓ [91
Animals Oxidative stress damage and NLRP3 inflammasome activation ↓; mRNA and protein expressions of TLR4, NOX4, NF‐κB, and NLRP3 ↓ [92
Antiemetic Ondansetron Human NF-кB pathway and JAK-STAT pathway ↓ [93
  Human Anti-inflammatory, antiplatelet aggregation, and anti-apoptosis effects [94
Tropisetron Animals Expression of inflammatory molecules including TNF-α, IL-1β, and iNOS ↓ [95
Diphenhydramine Animals Inflammatory cytokines, apoptosis, and oxidative stress ↓; H1R’s preventive effect ↑; OCT 2 ↓ [96

SOD, superoxide dismutase; GSK-3β, glycogen synthase kinase‐3β; Nrf2, nuclear factor erythroid 2‐related factor 2; iNOS, inducible nitric oxide synthase; NO, nitric oxide; NLRP3, nucleotide-binding oligomerization domain-like receptor protein-3; TLR4, toll‐like receptor 4; NOX4, NADPH oxidase 4; NF‐κB, nuclear factor-кB; JAK-STAT, Janus kinases-signal transducer and activator of transcription; TNF-α, tumor necrosis factor; IL-1β, interleukin-1β; H1R, histamine H1 receptor; OCT2, organ cation transport 2.

Fig. 3.

Overview of common sedatives and antiemetics and their protective effects on perioperative AKI. AKI, acute kidney injury; GFR, glomerular filtration rate; iNOS, inducible nitric oxide synthase; NO, nitric oxide; DEX, dexmedetomidine; GSK-3β, glycogen synthase kinase‐3β; Nrf2, nuclear factor erythroid 2‐related factor 2; α2-AR, α2 adrenergic receptor; CKD, chronic kidney disease; NF‐κB, nuclear factor-кB; JAK-STAT, Janus kinases-signal transducer and activator of transcription; TNF-α, tumor necrosis factor; IL-1β, interleukin-1β; H1R, histamine H1 receptor; OCT2, organ cation transport 2.

Fig. 3.

Overview of common sedatives and antiemetics and their protective effects on perioperative AKI. AKI, acute kidney injury; GFR, glomerular filtration rate; iNOS, inducible nitric oxide synthase; NO, nitric oxide; DEX, dexmedetomidine; GSK-3β, glycogen synthase kinase‐3β; Nrf2, nuclear factor erythroid 2‐related factor 2; α2-AR, α2 adrenergic receptor; CKD, chronic kidney disease; NF‐κB, nuclear factor-кB; JAK-STAT, Janus kinases-signal transducer and activator of transcription; TNF-α, tumor necrosis factor; IL-1β, interleukin-1β; H1R, histamine H1 receptor; OCT2, organ cation transport 2.

Close modal

Sedative

Midazolam

Midazolam is a benzodiazepine that can be used for the management of palliative sedation. It is mainly metabolized in the liver by cytochrome P450 (CYP) and excreted by the kidneys. In the case of inflammation and organ failure, midazolam clearance is reduced which could cause drug accumulation in the kidneys [97, 98]. Kirwan et al. suggested that increased severity and duration of AKI were associated with reduced elimination of midazolam. This may be caused by impaired CYP3A activity secondary to AKI [99]. The structures of propofol and midazolam both have phenol, but the experimental data did not show that midazolam had any antioxidant effect similar to propofol on the kidneys of rats after IR [100]. In patients with renal impairment, excretion of midazolam-active metabolites and parent drugs is prolonged. Midazolam should be used with caution in patients with acute renal failure [101]. Currently, studies on midazolam and AKI are scarce, thus further elaborations are also needed.

Dexmedetomidine

DEX, a class of fast-onset sedative drugs with proven neuroprotective and nephroprotective effects, has been widely introduced in the perioperative period due to its beneficial physiological effects and limited adverse effects [102]. Previous experiments have shown that DEX causes diuresis by inhibiting vasopressin secretion and increasing urine output [88], and protects against AKI by inhibiting inflammation, oxidation, and apoptosis [6]. Currently, an increasing number of studies confirm its protective effects against perioperative AKI. Shan et al. [13] found that DEX treatment attenuated renal insufficiency and reduced the incidence of AKI after endovascular aortic repair, which may be due to the reduction of sympathy-adrenal hyperactivity and stable maintenance of hemodynamic variables caused by DEX, resulting in enhanced GFR and increased urine output. Superoxide dismutase, which can reduce the risk of free radical overproduction, plays an important role in reducing oxidative stress [103]. In another cardiac valve replacement under CPB study, DEX was found to alleviate renal injury by increasing superoxide dismutase activity [12]. Besides, early intervention of DEX before ischemic injury appears to be essential for its organ-protective effect against IRI. In Cho’s research, preemptive DEX administration during an initial period lasting 24 h after surgery is effective in preventing AKI after cardiac surgery [104]. Similarly, data from Soh et al. [14] presented a lower incidence of AKI in an aortic operation under CPB after 24 h of preemptive DEX administration starting after anesthetic induction.

DEX is a selective α2 adrenergic receptor (α2-AR) agonist, and its protection mechanism of tissue damage through antioxidant, anti-apoptotic, and anti-inflammatory pathways [6] has been demonstrated by several animal studies. In a surgical bilateral renal IR model, DEX attenuated renal fibrosis after renal IR by reducing inflammation and premature cellular senescence mainly through α2-AR, which effectively prevented the renal IR-induced AKI-CKD transition [89]. One study on lipopolysaccharide‐induced AKI noted that the nephroprotective effect of DEX is to reduce inflammation and oxidative stress through glycogen synthase kinase‐3β (GSK-3β)/nuclear factor erythroid 2‐related factor 2 (Nrf2) signaling mediated by α2-AR pathway [90]. Nrf2 is thought to be crucial in the prevention of oxidative stress diseases [105‒107]. In pathological state, Nrf2 binds to antioxidant response elements in gene promoters, initiates Nrf2-mediated transcription process, regulates downstream gene expression such as heme oxygenase-, and promotes cytoprotective mechanisms such as anti-oxidative stress, anti-inflammatory, and anti-apoptosis [108]. GSK-3β is widely found in the kidney [109], which regulates Nrf2 activity by inhibiting GSK-3β [110]. GSK-3β/Nrf2 pathway may be an emerging therapeutic target for AKI. Furthermore, DEX can also ameliorate AKI through various pathways such as inducible nitric oxide synthase/nitric oxide pathway and toll‐like receptor 4 (TLR4)/NADPH oxidase 4/NF-κB pathway [91, 92]; however, they were not listed here due to few relevant studies.

Antiemetic

Setrons are competitive antagonists of serotonin receptors (5-HT3R), which are effective in preventing nausea and vomiting induced by chemotherapy and radiotherapy [111]. Cisplatin is a highly emetogenic anticancer drug with nephrotoxicity, which has been recommended for use with 5-HT3R antagonists [112, 113]. Therefore, protecting the renal function during administration is needed. However, a study indicated that the first-generation 5-HT3R antagonists (ondansetron, granisetron, tropisetron) exacerbated cisplatin-induced renal injury, whereas the second-generation 5-HT3R antagonists (palonosetron) showed greater anti-cisplatin-induced nephrotoxicity than other 5-HT3R antagonists [114, 115]. A study reported that ondansetron is associated with an increasing risk of AKI [116]. But interestingly, in a recent study, ondansetron was found to be correlated with lower mortality among AKI patients in intensive care unit. In addition, by comparing the gene expression characteristics, this effect may be induced by nuclear factor-kappa B pathway and Janus kinases-signal transducer and activator of transcription pathway [93]. In another retrospective cohort study, ondansetron use was also found to be associated with lower risk-adjusted mortality among patients with AKI, possibly due to the anti-inflammatory, antiplatelet aggregation, and anti-apoptosis effects [94]. However, it is worth noting that differences in the number of subjects and age groups as well as experimental indicators may contribute to different experimental results. Some animal studies have shown that the combination of ondansetron and cisplatin increases nephrotoxicity [117], while some other studies demonstrated tropisetron treatment ameliorated the cisplatin-induced nephrotoxicity [95]. These results are consistent with Kou’s clinical trial [118]. At present, few studies have been conducted to investigate the protective effect of setron drugs against AKI. Different types and doses of therapeutic drugs can cause different effects of setrons on renal function, which needs further clinical studies.

The renoprotective effects of the abovementioned anesthetic agents have been studied in several studies, but there is no single anesthetic agent that is widely accepted and included in standard perioperative anesthetic regimens. Therefore, whether or not an anesthetic agent of choice is used in the perioperative period or whether it is included as an add-on therapy for patients at high risk for AKI depends on a combination of factors. For example, when selecting an anesthetic agent during surgery, factors such as the type of surgery, the patient’s underlying health status, drug effects, and metabolic pathways are taken into account, in addition to other anesthesia-related factors such as blood pressure control and analgesic needs. Overall, more research is needed to confirm these findings and to find the best way to use them, based on a comprehensive assessment of the patient's overall condition and other clinical conditions, in order to fully protect renal function.

A single anesthetic (or treatment for that matter) may not change AKI outcomes, and multiple small interventions are needed to improve outcomes. AKI's “care bundle” concept has been proposed as a structured approach aimed at improving care processes and outcomes [119]. Care bundle is not a single intervention but a number of independent elements (usually between three and six) delivered together as a complex intervention designed to change clinician behavior [120]. The biomarker-guided KDIGO bundle has been shown in previous Prev-AKI trials to be effective in reducing AKI in cardiac surgery patients [121, 122]. Moreover, multiple BIGPAC trials have shown that early biomarker-based prediction of AKI after implementation of the KDIGO care package reduces the risk of postoperative AKI [123, 124]. Anesthetics can improve the level of traditional biomarkers, while the new biomarkers applied by care bundle can be diagnosed earlier. The study of pairing certain anesthetics with care bundles may provide a new direction for AKI prevention.

In fact, the application of perioperative medication of anesthesia on perioperative AKI in clinics still has many challenges. On the one hand, many studies are still at the animal level and the results have not been translated to humans, such as few clinical studies have explored the renal protection effects of DEX by modulating Nrf2 activity. On the other hand, regarding animal models (or humans), there may be sex differences in response to drugs. Due to the higher incidence of postoperative AKI in female gender, the gender ratio of patients needs to be carefully considered in clinical studies. In addition, there are a few basic studies using male rodents, however, gender is an under-researched factor in this population. Moreover, the interactions between the various anesthetics and other cells, tissues, and organs of the body have not been very clearly studied. Therefore, the selection of clinical anesthetics needs a comprehensive evaluation of various factors and more experimental support.

AKI is very common in surgical patients, and the influence of anesthetic drugs on renal function is particularly important during the perioperative period. In recent years, some anesthetic drugs seem to have therapeutic value for AKI through anti-inflammatory, antioxidant, anti-apoptotic, and other mechanisms, which is a hot topic of intense research. However, there are also many researchers studying the clinical effect of anesthetic drugs, with mixed results. The underlying mechanism of renal protection is still unclear; further studies are needed to clarify the molecular mechanisms of anesthetics and to resolve discrepant findings.

In this study, we reviewed the protective mechanism of various anesthetic drugs on AKI and the latest clinical research, in order to better understand anesthetic administration for perioperative AKI. Intravenous anesthetics (e.g., propofol), volatile anesthetics (e.g., sevoflurane), and sedatives (e.g., DEX) seem to be outstanding in the prevention or treatment of AKI. These drugs achieve anti-inflammatory, antioxidant, and cell protection through expression regulation and signal transduction, such as propofol and sevoflurane can both reduce IL-6 level. However, the effect of other drugs such as setrons and midazolam on AKI is still controversial. Nowadays, there are still many challenges to the clinical application of perioperative medication of anesthesia on perioperative AKI. Therefore, we also present relevant basic studies to further elucidate the renal protective mechanisms of anesthetic drugs.

In any case, there is no doubt that studies on the renal protection of anesthetic agents will be continued, and these studies will help further evaluate and improve the administration of anesthetic agents in perioperative AKI. This paper not only provides a theoretical basis for perioperative anesthesia but also provides a reference for the precise treatment of AKI. More importantly, we hope that the above summary can provide a better choice of anesthesia drugs during the perioperative period and provide future direction for the treatment of AKI.

The graphical abstracts were created with BioRender software (BioRender.com).

The authors have no conflicts of interest to declare.

This work was supported by the Natural Science Foundation of Jiangxi Province (No. 20224ACB216009); the National Natural Science Foundation of China (Grant No. 82160371, No. 82100869, No. 82360162, No. 81960669); the Jiangxi Province Thousands of Plans (No. jxsq2023201105); and the Hengrui Diabetes Metabolism Research Fund (No. Z-2017-26-2202-4).

Bin Zeng: Conceptualization, Writing - Original Draft, Writing - Review & Editing Preparation, Supervision, Funding acquisition.

Yi-Nuo Liu: Conceptualization, Writing - Original Draft, Writing - Review & Editing Preparation.

Jia-Wei Xua: Writing - Original Draft, Writing - Review & Editing Preparation.

Li-Yan Niu: Visualization, Writing - Review & Editing Preparation.

Yuting Wu: Writing - Review & Editing Preparation.

De-Ju Zhang: Writing - Review & Editing Preparation.

Xiao-Yi Tang: Writing - Review & Editing Preparation.

Zi-Cheng Zhu: Visualization.

Yi-Xuan Chen: Visualization.

Lei-Lei Hu: Visualization.

Shu-Chun Yu: Visualization.

Peng Yu: Conceptualization, Writing - Review & Editing Preparation, Supervision, Project administration, Funding acquisition.

Wen-Ting Wang: Conceptualization, Supervision.

Jing Zhang: Conceptualization, Supervision, Funding acquisition.

Additional Information

Bin Zeng, Yinuo Liu, and Jiawei Xu are co-first authors.

1.
Ostermann
M
,
Joannidis
M
.
Acute kidney injury 2016: diagnosis and diagnostic workup
.
Crit Care
.
2016
;
20
(
1
):
299
.
2.
Wang
HE
,
Muntner
P
,
Chertow
GM
,
Warnock
DG
.
Acute kidney injury and mortality in hospitalized patients
.
Am J Nephrol
.
2012
;
35
(
4
):
349
55
.
3.
Sun
LY
,
Wijeysundera
DN
,
Tait
GA
,
Beattie
WS
.
Association of intraoperative hypotension with acute kidney injury after elective noncardiac surgery
.
Anesthesiology
.
2015
;
123
(
3
):
515
23
.
4.
Kong
HY
,
Zhu
SM
,
Wang
LQ
,
He
Y
,
Xie
HY
,
Zheng
SS
.
Sevoflurane protects against acute kidney injury in a small-size liver transplantation model
.
Am J Nephrol
.
2010
;
32
(
4
):
347
55
.
5.
Zheng
G
,
Qu
H
,
Li
F
,
Ma
W
,
Yang
H
.
Propofol attenuates sepsis-induced acute kidney injury by regulating miR-290-5p/CCL-2 signaling pathway
.
Braz J Med Biol Res
.
2018
;
51
(
11
):
e7655
.
6.
Wang
Z
,
Wu
J
,
Hu
Z
,
Luo
C
,
Wang
P
,
Zhang
Y
.
Dexmedetomidine alleviates lipopolysaccharide-induced acute kidney injury by inhibiting p75NTR-mediated oxidative stress and apoptosis
.
Oxid Med Cell Longev
.
2020
;
2020
:
5454210
.
7.
Li
H
,
Weng
Y
,
Yuan
S
,
Liu
W
,
Yu
H
,
Yu
W
.
Effect of sevoflurane and propofol on acute kidney injury in pediatric living donor liver transplantation
.
Ann Transl Med
.
2019
;
7
(
14
):
340
.
8.
Oh
T
,
Kim
J
,
Han
S
,
Kim
K
,
Jheon
S
,
Ji
E
.
Effect of sevoflurane-based or propofol-based anaesthesia on the incidence of postoperative acute kidney injury: a retrospective propensity score-matched analysis
.
Eur J Anaesthesiol
.
2019
;
36
(
9
):
649
55
.
9.
Song
Y
,
Paik
HC
,
Kim
N
,
Jung
H
,
Lee
JG
,
Yoo
YC
.
Effect of propofol versus sevoflurane anesthesia on acute kidney injury after lung transplantation surgery: a prospective randomized controlled trial
.
J Clin Med
.
2022
;
11
(
22
):
6862
.
10.
Yoo
Y
,
Shim
J
,
Song
Y
,
Yang
S
,
Kwak
Y
.
Anesthetics influence the incidence of acute kidney injury following valvular heart surgery
.
Kidney Int
.
2014
;
86
(
2
):
414
22
.
11.
Bang
JY
,
Lee
J
,
Oh
J
,
Song
JG
,
Hwang
GS
.
The influence of propofol and sevoflurane on acute kidney injury after colorectal surgery: a retrospective cohort study
.
Anesth Analg
.
2016
;
123
(
2
):
363
70
.
12.
Zhai
M
,
Kang
F
,
Han
M
,
Huang
X
,
Li
J
.
The effect of dexmedetomidine on renal function in patients undergoing cardiac valve replacement under cardiopulmonary bypass: a double-blind randomized controlled trial
.
J Clin Anesth
.
2017
;
40
:
33
8
.
13.
Shan
XS
,
Dai
HR
,
Zhao
D
,
Yang
BW
,
Feng
XM
,
Liu
H
.
Dexmedetomidine reduces acute kidney injury after endovascular aortic repair of Stanford type B aortic dissection: a randomized, double-blind, placebo-controlled pilot study
.
J Clin Anesth
.
2021
;
75
:
110498
.
14.
Soh
S
,
Shim
JK
,
Song
JW
,
Bae
JC
,
Kwak
YL
.
Effect of dexmedetomidine on acute kidney injury after aortic surgery: a single-centre, placebo-controlled, randomised controlled trial
.
Br J Anaesth
.
2020
;
124
(
4
):
386
94
.
15.
Leite
T
,
Macedo
E
,
Martins
Id S
,
Neves
F
,
Libório
A
.
Renal outcomes in critically ill patients receiving propofol or midazolam
.
Clin J Am Soc Nephrol
.
2015
;
10
(
11
):
1937
45
.
16.
Gray
M
,
Priyanka
P
,
Kane-Gill
S
,
Wang
L
,
Kellum
JA
.
Kidney and mortality outcomes associated with ondansetron in critically ill patients
.
J Intensive Care Med
.
2022
;
37
(
10
):
1403
10
.
17.
Kellum
JA
,
Levin
N
,
Bouman
C
,
Lameire
N
.
Developing a consensus classification system for acute renal failure
.
Curr Opin Crit Care
.
2002
;
8
(
6
):
509
14
.
18.
Gomelsky
A
,
Abreo
K
,
Khater
N
,
Abreo
A
,
Amin
B
,
Craig
MK
.
Perioperative acute kidney injury: stratification and risk reduction strategies
.
Best Pract Res Clin Anaesthesiol
.
2020
;
34
(
2
):
167
82
.
19.
Lewington
AJ
,
Cerda
J
,
Mehta
RL
.
Raising awareness of acute kidney injury: a global perspective of a silent killer
.
Kidney Int
.
2013
;
84
(
3
):
457
67
.
20.
Meersch
M
,
Schmidt
C
,
Zarbock
A
.
Perioperative acute kidney injury: an under-recognized problem
.
Anesth Analg
.
2017
;
125
(
4
):
1223
32
.
21.
Ojo
AO
,
Held
PJ
,
Port
FK
,
Wolfe
RA
,
Leichtman
AB
,
Young
EW
.
Chronic renal failure after transplantation of a nonrenal organ
.
N Engl J Med
.
2003
;
349
(
10
):
931
40
.
22.
Barri
YM
,
Sanchez
EQ
,
Jennings
LW
,
Melton
LB
,
Hays
S
,
Levy
MF
.
Acute kidney injury following liver transplantation: definition and outcome
.
Liver Transpl
.
2009
;
15
(
5
):
475
83
.
23.
Hobson
C
,
Lysak
N
,
Huber
M
,
Scali
S
,
Bihorac
A
.
Epidemiology, outcomes, and management of acute kidney injury in the vascular surgery patient
.
J Vasc Surg
.
2018
;
68
(
3
):
916
28
.
24.
Yu
X
,
Feng
Z
.
Analysis of risk factors for perioperative acute kidney injury and management strategies
.
Front Med
.
2021
;
8
:
751793
.
25.
Roche
M
,
Rondeau
P
,
Singh
NR
,
Tarnus
E
,
Bourdon
E
.
The antioxidant properties of serum albumin
.
FEBS Lett
.
2008
;
582
(
13
):
1783
7
.
26.
Boyer
N
,
Eldridge
J
,
Prowle
JR
,
Forni
LG
.
Postoperative acute kidney injury
.
Clin J Am Soc Nephrol
.
2022
;
17
(
10
):
1535
45
.
27.
Grams
ME
,
Astor
BC
,
Bash
LD
,
Matsushita
K
,
Wang
Y
,
Coresh
J
.
Albuminuria and estimated glomerular filtration rate independently associate with acute kidney injury
.
J Am Soc Nephrol
.
2010
;
21
(
10
):
1757
64
.
28.
Dong
M
,
Jiao
Z
,
Liu
T
,
Guo
F
,
Li
G
.
Effect of administration route on the renal safety of contrast agents: a meta-analysis of randomized controlled trials
.
J Nephrol
.
2012
;
25
(
3
):
290
301
.
29.
Kheterpal
S
,
Tremper
KK
,
Heung
M
,
Rosenberg
AL
,
Englesbe
M
,
Shanks
AM
.
Development and validation of an acute kidney injury risk index for patients undergoing general surgery: results from a national data set
.
Anesthesiology
.
2009
;
110
(
3
):
505
15
.
30.
Ichai
C
,
Vinsonneau
C
,
Souweine
B
,
Armando
F
,
Canet
E
,
Clec’h
C
.
Acute kidney injury in the perioperative period and in intensive care units (excluding renal replacement therapies)
.
Ann Intensive Care
.
2016
;
6
(
1
):
48
.
31.
Kambakamba
P
,
Slankamenac
K
,
Tschuor
C
,
Kron
P
,
Wirsching
A
,
Maurer
K
.
Epidural analgesia and perioperative kidney function after major liver resection
.
Br J Surg
.
2015
;
102
(
7
):
805
12
.
32.
Rodgers
A
,
Walker
N
,
Schug
S
,
McKee
A
,
Kehlet
H
,
van Zundert
A
.
Reduction of postoperative mortality and morbidity with epidural or spinal anaesthesia: results from overview of randomised trials
.
BMJ
.
2000
;
321
(
7275
):
1493
.
33.
Neugarten
J
,
Sandilya
S
,
Singh
B
,
Golestaneh
L
.
Sex and the risk of AKI following cardio-thoracic surgery: a meta-analysis
.
Clin J Am Soc Nephrol
.
2016
;
11
(
12
):
2113
22
.
34.
Grams
ME
,
Sang
Y
,
Coresh
J
,
Ballew
S
,
Matsushita
K
,
Molnar
MZ
.
Acute kidney injury after major surgery: a retrospective analysis of veterans health administration data
.
Am J Kidney Dis
.
2016
;
67
(
6
):
872
80
.
35.
Fidalgo
P
,
Ahmed
M
,
Meyer
SR
,
Lien
D
,
Weinkauf
J
,
Cardoso
FS
.
Incidence and outcomes of acute kidney injury following orthotopic lung transplantation: a population-based cohort study
.
Nephrol Dial Transplant
.
2014
;
29
(
9
):
1702
9
.
36.
Shashaty
MGS
,
Forker
CM
,
Miano
TA
,
Wu
Q
,
Yang
W
,
Oyster
ML
.
The association of post-lung transplant acute kidney injury with mortality is independent of primary graft dysfunction: a cohort study
.
Clin Transplant
.
2019
;
33
(
10
):
e13678
.
37.
Billings
FT
4th
,
Lopez
MG
,
Shaw
AD
.
The incidence, risk, presentation, pathophysiology, treatment, and effects of perioperative acute kidney injury
.
Can J Anaesth
.
2021
;
68
(
3
):
409
22
.
38.
Walsh
M
,
Garg
AX
,
Devereaux
PJ
,
Argalious
M
,
Honar
H
,
Sessler
DI
.
The association between perioperative hemoglobin and acute kidney injury in patients having noncardiac surgery
.
Anesth Analg
.
2013
;
117
(
4
):
924
31
.
39.
Hobson
C
,
Ruchi
R
,
Bihorac
A
.
Perioperative acute kidney injury: risk factors and predictive strategies
.
Crit Care Clin
.
2017
;
33
(
2
):
379
96
.
40.
Kalamas
AG
,
Niemann
CU
.
Patients with chronic kidney disease
.
Med Clin North Am
.
2013
;
97
(
6
):
1109
22
.
41.
Haase-Fielitz
A
,
Haase
M
,
Bellomo
R
,
Calzavacca
P
,
Spura
A
,
Baraki
H
.
Perioperative hemodynamic instability and fluid overload are associated with increasing acute kidney injury severity and worse outcome after cardiac surgery
.
Blood Purif
.
2017
;
43
(
4
):
298
308
.
42.
Li
C
,
Xu
L
,
Guan
C
,
Zhao
L
,
Luo
C
,
Zhou
B
.
Malnutrition screening and acute kidney injury in hospitalised patients: a retrospective study over a 5-year period from China
.
Br J Nutr
.
2020
;
123
(
3
):
337
46
.
43.
Arora
P
,
Rajagopalam
S
,
Ranjan
R
,
Kolli
H
,
Singh
M
,
Venuto
R
.
Preoperative use of angiotensin-converting enzyme inhibitors/angiotensin receptor blockers is associated with increased risk for acute kidney injury after cardiovascular surgery
.
Clin J Am Soc Nephrol
.
2008
;
3
(
5
):
1266
73
.
44.
Roshanov
PS
,
Rochwerg
B
,
Patel
A
,
Salehian
O
,
Duceppe
E
,
Belley-Cote
EP
.
Withholding versus continuing angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers before noncardiac surgery: an analysis of the vascular events in noncardiac surgery patIents cOhort evaluatioN prospective cohort
.
Anesthesiology
.
2017
;
126
(
1
):
16
27
.
45.
Bell
S
,
Davey
P
,
Nathwani
D
,
Marwick
C
,
Vadiveloo
T
,
Sneddon
J
.
Risk of AKI with gentamicin as surgical prophylaxis
.
J Am Soc Nephrol
.
2014
;
25
(
11
):
2625
32
.
46.
Elyasi
S
,
Khalili
H
,
Dashti-Khavidaki
S
,
Mohammadpour
A
.
Vancomycin-induced nephrotoxicity: mechanism, incidence, risk factors and special populations. A literature review
.
Eur J Clin Pharmacol
.
2012
;
68
(
9
):
1243
55
.
47.
Wong
JMK
,
Bortoletto
P
,
Tolentino
J
,
Jung
MJ
,
Milad
MP
.
Urinary tract injury in gynecologic laparoscopy for benign indication: a systematic review
.
Obstet Gynecol
.
2018
;
131
(
1
):
100
8
.
48.
Dalfino
L
,
Tullo
L
,
Donadio
I
,
Malcangi
V
,
Brienza
N
.
Intra-abdominal hypertension and acute renal failure in critically ill patients
.
Intensive Care Med
.
2008
;
34
(
4
):
707
13
.
49.
Nash
DM
,
Mustafa
RA
,
McArthur
E
,
Wijeysundera
DN
,
Paterson
JM
,
Sharan
S
.
Combined general and neuraxial anesthesia versus general anesthesia: a population-based cohort study
.
Can J Anaesth
.
2015
;
62
(
4
):
356
68
.
50.
Goren
O
,
Matot
I
.
Perioperative acute kidney injury
.
Br J Anaesth
.
2015
115
Suppl 2
ii3
14
.
51.
Prowle
JR
,
Forni
LG
,
Bell
M
,
Chew
MS
,
Edwards
M
,
Grams
ME
.
Postoperative acute kidney injury in adult non-cardiac surgery: joint consensus report of the Acute Disease Quality Initiative and PeriOperative Quality Initiative
.
Nat Rev Nephrol
.
2021
;
17
(
9
):
605
18
.
52.
Daabiss
M
.
American Society of Anaesthesiologists physical status classification
.
Indian J Anaesth
.
2011
;
55
(
2
):
111
5
.
53.
Lassnigg
A
,
Schmid
ER
,
Hiesmayr
M
,
Falk
C
,
Druml
W
,
Bauer
P
.
Impact of minimal increases in serum creatinine on outcome in patients after cardiothoracic surgery: do we have to revise current definitions of acute renal failure
.
Crit Care Med
.
2008
;
36
(
4
):
1129
37
.
54.
Symons
JM
.
Moving beyond supportive care: current status of specific therapies in pediatric acute kidney injury
.
Pediatr Nephrol
.
2014
;
29
(
2
):
173
81
.
55.
Meersch
M
,
Weiss
R
,
Massoth
C
,
Küllmar
M
,
Saadat-Gilani
K
,
Busen
M
.
The association between angiotensin II and renin kinetics in patients after cardiac surgery
.
Anesth Analg
.
2022
;
134
(
5
):
1002
9
.
56.
Riegel
AK
,
Faigle
M
,
Zug
S
,
Rosenberger
P
,
Robaye
B
,
Boeynaems
JM
.
Selective induction of endothelial P2Y6 nucleotide receptor promotes vascular inflammation
.
Blood
.
2011
;
117
(
8
):
2548
55
.
57.
Idzko
M
,
Ferrari
D
,
Riegel
AK
,
Eltzschig
HK
.
Extracellular nucleotide and nucleoside signaling in vascular and blood disease
.
Blood
.
2014
;
124
(
7
):
1029
37
.
58.
McKinlay
J
,
Tyson
E
,
Forni
L
.
Renal complications of anaesthesia
.
Anaesthesia
.
2018
73
Suppl 1
85
94
.
59.
Han
J
,
Tao
W
,
Cui
W
,
Chen
J
.
Propofol via antioxidant property attenuated hypoxia-mediated mitochondrial dynamic imbalance and malfunction in primary rat hippocampal neurons
.
Oxid Med Cell Longev
.
2022
;
2022
:
6298786
.
60.
Hsiao
H
,
Liu
Y
,
Wang
J
,
Lin
Y
,
Liu
Y
.
The analgesic effect of propofol associated with the inhibition of hypoxia inducible factor and inflammasome in complex regional pain syndrome
.
J Biomed Sci
.
2019
;
26
(
1
):
74
.
61.
Snoeijs
MG
,
Vaahtera
L
,
de Vries
EE
,
Schurink
GW
,
Haenen
GR
,
Peutz-Kootstra
CJ
.
Addition of a water-soluble propofol formulation to preservation solution in experimental kidney transplantation
.
Transplantation
.
2011
;
92
(
3
):
296
302
.
62.
Luo
C
,
Yuan
D
,
Li
X
,
Yao
W
,
Luo
G
,
Chi
X
.
Propofol attenuated acute kidney injury after orthotopic liver transplantation via inhibiting gap junction composed of connexin 32
.
Anesthesiology
.
2015
;
122
(
1
):
72
86
.
63.
Hsing
C
,
Chou
W
,
Wang
J
,
Chen
H
,
Yeh
C
.
Propofol increases bone morphogenetic protein-7 and decreases oxidative stress in sepsis-induced acute kidney injury
.
Nephrol Dial Transplant
.
2011
;
26
(
4
):
1162
72
.
64.
Lee
HT
,
Ota-Setlik
A
,
Fu
Y
,
Nasr
SH
,
Emala
CW
.
Differential protective effects of volatile anesthetics against renal ischemia-reperfusion injury in vivo
.
Anesthesiology
.
2004
;
101
(
6
):
1313
24
.
65.
Mårtensson
J
,
Martling
CR
,
Bell
M
.
Novel biomarkers of acute kidney injury and failure: clinical applicability
.
Br J Anaesth
.
2012
;
109
(
6
):
843
50
.
66.
Zou
C
,
Wang
C
,
Lu
L
.
Advances in the study of subclinical AKI biomarkers
.
Front Physiol
.
2022
;
13
:
960059
.
67.
Zhang
Y
,
Liu
L
,
Guo
S
,
Song
J
,
Zhu
C
,
Yue
Z
.
Deciphering TAL effectors for 5-methylcytosine and 5-hydroxymethylcytosine recognition
.
Nat Commun
.
2017
;
8
(
1
):
901
10
.
68.
Lee
HT
,
Kim
M
,
Kim
J
,
Kim
N
,
Emala
CW
.
TGF-beta1 release by volatile anesthetics mediates protection against renal proximal tubule cell necrosis
.
Am J Nephrol
.
2007
;
27
(
4
):
416
24
.
69.
Lee
HT
,
Kim
M
,
Song
JH
,
Chen
SW
,
Gubitosa
G
,
Emala
CW
.
Sevoflurane-mediated TGF-beta1 signaling in renal proximal tubule cells
.
Am J Physiol Renal Physiol
.
2008
294
2
F371
8
.
70.
Lee
HT
,
Chen
SW
,
Doetschman
TC
,
Deng
C
,
D'Agati
VD
,
Kim
M
.
Sevoflurane protects against renal ischemia and reperfusion injury in mice via the transforming growth factor-beta1 pathway
.
Am J Physiol Renal Physiol
.
2008
295
1
F128
36
.
71.
Yamamoto
M
,
Morita
T
,
Ishikawa
M
,
Sakamoto
A
.
Specific microRNAs are involved in the reno-protective effects of sevoflurane preconditioning and ischemic preconditioning against ischemia reperfusion injury in rats
.
Int J Mol Med
.
2020
;
45
(
4
):
1141
9
.
72.
Ham
A
,
Kim
M
,
Kim
JY
,
Brown
KM
,
Yeh
J
,
D'Agati
VD
.
Critical role of interleukin-11 in isoflurane-mediated protection against ischemic acute kidney injury in mice
.
Anesthesiology
.
2013
;
119
(
6
):
1389
401
.
73.
Kim
M
,
Ham
A
,
Kim
JY
,
Brown
KM
,
D’Agati
VD
,
Lee
HT
.
The volatile anesthetic isoflurane induces ecto-5’-nucleotidase (CD73) to protect against renal ischemia and reperfusion injury
.
Kidney Int
.
2013
;
84
(
1
):
90
103
.
74.
Zhao
G
,
Lu
S
,
Li
L
,
Fan
X
.
Local anesthetic articaine ameliorates LPS-induced acute kidney injury via inhibition of NF-ĸB activation and the NLRP3 inflammasome pathway
.
J Biochem Mol Toxicol
.
2020
;
34
(
10
):
e22554
.
75.
Lundström
S
,
Twycross
R
,
Mihalyo
M
,
Wilcock
A
.
Propofol
.
J Pain Symptom Manage
.
2010
;
40
(
3
):
466
70
.
76.
Motayagheni
N
,
Phan
S
,
Eshraghi
C
,
Nozari
A
,
Atala
A
.
A review of anesthetic effects on renal function: potential organ protection
.
Am J Nephrol
.
2017
;
46
(
5
):
380
9
.
77.
Da-Silva
C
,
Anauate
A
,
Guirao
T
,
Novaes
A
,
Maquigussa
E
,
Boim
M
.
Analysis of exosome-derived microRNAs as early biomarkers of lipopolysaccharide-induced acute kidney injury in rats
.
Front Physiol
.
2022
;
13
:
944864
.
78.
Neves
J
,
Ibiapina
H
,
Magalhães-Gama
F
,
Sachett
J
,
Silva
I
,
Coelho
K
.
CCL-2 and CXCL-8: potential prognostic biomarkers of acute kidney injury after a Bothrops atrox snakebite
.
Mediators Inflamm
.
2022
;
2022
:
8285084
.
79.
Jung
YJ
,
Lee
AS
,
Nguyen-Thanh
T
,
Kim
D
,
Kang
KP
,
Lee
S
.
SIRT2 regulates LPS-induced renal tubular CXCL2 and CCL2 expression
.
J Am Soc Nephrol
.
2015
;
26
(
7
):
1549
60
.
80.
Fukazawa
K
,
Lee
HT
.
Volatile anesthetics and AKI: risks, mechanisms, and a potential therapeutic window
.
J Am Soc Nephrol
.
2014
;
25
(
5
):
884
92
.
81.
Liang
T
,
Peng
S
,
Ma
M
,
Li
H
,
Wang
Z
,
Chen
G
.
Protective effects of sevoflurane in cerebral ischemia reperfusion injury: a narrative review
.
Med Gas Res
.
2021
;
11
(
4
):
152
4
.
82.
Franzén
S
,
Semenas
E
,
Taavo
M
,
Mårtensson
J
,
Larsson
A
,
Frithiof
R
.
Renal function during sevoflurane or total intravenous propofol anaesthesia: a single-centre parallel randomised controlled study
.
Br J Anaesth
.
2022
;
128
(
5
):
838
48
.
83.
Nieuwenhuijs-Moeke
GJ
,
Nieuwenhuijs
VB
,
Seelen
MAJ
,
Berger
SP
,
van den Heuvel
MC
,
Burgerhof
JGM
.
Propofol-based anaesthesia versus sevoflurane-based anaesthesia for living donor kidney transplantation: results of the VAPOR-1 randomized controlled trial
.
Br J Anaesth
.
2017
;
118
(
5
):
720
32
.
84.
Anders
MW
.
Formation and toxicity of anesthetic degradation products
.
Annu Rev Pharmacol Toxicol
.
2005
;
45
:
147
76
.
85.
Pefanis
A
,
Ierino
FL
,
Murphy
JM
,
Cowan
PJ
.
Regulated necrosis in kidney ischemia-reperfusion injury
.
Kidney Int
.
2019
;
96
(
2
):
291
301
.
86.
Godwin
JG
,
Ge
X
,
Stephan
K
,
Jurisch
A
,
Tullius
SG
,
Iacomini
J
.
Identification of a microRNA signature of renal ischemia reperfusion injury
.
Proc Natl Acad Sci U S A
.
2010
;
107
(
32
):
14339
44
.
87.
Sun
G
,
Zhou
Y
,
Li
H
,
Guo
Y
,
Shan
J
,
Xia
M
.
Over-expression of microRNA-494 up-regulates hypoxia-inducible factor-1 alpha expression via PI3K/Akt pathway and protects against hypoxia-induced apoptosis
.
J Biomed Sci
.
2013
;
20
(
1
):
100
.
88.
Villela
NR
,
do Nascimento Júnior
P
,
de Carvalho
LR
,
Teixeira
A
.
Effects of dexmedetomidine on renal system and on vasopressin plasma levels. Experimental study in dogs
.
Rev Bras Anestesiol
.
2005
;
55
(
4
):
429
40
.
89.
Li
Q
,
Chen
C
,
Chen
X
,
Han
M
,
Li
J
.
Dexmedetomidine attenuates renal fibrosis via α2-adrenergic receptor-dependent inhibition of cellular senescence after renal ischemia/reperfusion
.
Life Sci
.
2018
;
207
:
1
8
.
90.
Feng
X
,
Guan
W
,
Zhao
Y
,
Wang
C
,
Song
M
,
Yao
Y
.
Dexmedetomidine ameliorates lipopolysaccharide-induced acute kidney injury in rats by inhibiting inflammation and oxidative stress via the GSK-3β/Nrf2 signaling pathway
.
J Cell Physiol
.
2019
;
234
(
10
):
18994
9009
.
91.
Chen
Y
,
Luan
L
,
Wang
C
,
Song
M
,
Zhao
Y
,
Yao
Y
.
Dexmedetomidine protects against lipopolysaccharide-induced early acute kidney injury by inhibiting the iNOS/NO signaling pathway in rats
.
Nitric Oxide
.
2019
;
85
:
1
9
.
92.
Yao
Y
,
Hu
X
,
Feng
X
,
Zhao
Y
,
Song
M
,
Wang
C
.
Dexmedetomidine alleviates lipopolysaccharide-induced acute kidney injury by inhibiting the NLRP3 inflammasome activation via regulating the TLR4/NOX4/NF-κB pathway
.
J Cell Biochem
.
2019
;
120
(
10
):
18509
23
.
93.
Guo
X
,
Qi
X
,
Fan
P
,
Gilbert
M
,
La
AD
,
Liu
Z
.
Effect of ondansetron on reducing ICU mortality in patients with acute kidney injury
.
Sci Rep
.
2021
;
11
(
1
):
19409
.
94.
Tao
L
,
Zhou
S
,
Chang
P
,
An
S
.
Effects of ondansetron use on outcomes of acute kidney injury in critically ill patients: an analysis based on the MIMIC-IV database
.
J Crit Care
.
2021
;
66
:
117
22
.
95.
Zirak
MR
,
Rahimian
R
,
Ghazi-Khansari
M
,
Abbasi
A
,
Razmi
A
,
Mehr
SE
.
Tropisetron attenuates cisplatin-induced nephrotoxicity in mice
.
Eur J Pharmacol
.
2014
;
738
:
222
9
.
96.
Hamano
H
,
Ikeda
Y
,
Goda
M
,
Fukushima
K
,
Kishi
S
,
Chuma
M
.
Diphenhydramine may be a preventive medicine against cisplatin-induced kidney toxicity
.
Kidney Int
.
2021
;
99
(
4
):
885
99
.
97.
Prommer
E
.
Midazolam: an essential palliative care drug
.
Palliat Care Soc Pract
.
2020
;
14
:
2632352419895527
.
98.
Vet
NJ
,
Brussee
JM
,
de Hoog
M
,
Mooij
MG
,
Verlaat
CW
,
Jerchel
IS
.
Inflammation and organ failure severely affect midazolam clearance in critically ill children
.
Am J Respir Crit Care Med
.
2016
;
194
(
1
):
58
66
.
99.
Kirwan
CJ
,
MacPhee
IA
,
Lee
T
,
Holt
DW
,
Philips
BJ
.
Acute kidney injury reduces the hepatic metabolism of midazolam in critically ill patients
.
Intensive Care Med
.
2012
;
38
(
1
):
76
84
.
100.
Erol
U
,
Gurdal
M
,
Erol
A
,
Aslan
R
,
Konukoğlu
D
,
Onmus
H
.
Is midazolam effective as an antioxidant in preventing reperfusion injury in rat kidney
.
Int Urol Nephrol
.
2002
;
34
(
1
):
121
7
.
101.
Zaporowska-Stachowiak
I
,
Szymański
K
,
Oduah
MT
,
Stachowiak-Szymczak
K
,
Łuczak
J
,
Sopata
M
.
Midazolam: safety of use in palliative care: a systematic critical review
.
Biomed Pharmacother
.
2019
;
114
:
108838
.
102.
Mahmoud
M
,
Mason
KP
.
Dexmedetomidine: review, update, and future considerations of paediatric perioperative and periprocedural applications and limitations
.
Br J Anaesth
.
2015
;
115
(
2
):
171
82
.
103.
Rosa
AC
,
Bruni
N
,
Meineri
G
,
Corsi
D
,
Cavi
N
,
Gastaldi
D
.
Strategies to expand the therapeutic potential of superoxide dismutase by exploiting delivery approaches
.
Int J Biol Macromol
.
2021
;
168
:
846
65
.
104.
Cho
JS
,
Shim
JK
,
Soh
S
,
Kim
MK
,
Kwak
YL
.
Perioperative dexmedetomidine reduces the incidence and severity of acute kidney injury following valvular heart surgery
.
Kidney Int
.
2016
;
89
(
3
):
693
700
.
105.
Chen
QM
.
Nrf2 for cardiac protection: pharmacological options against oxidative stress
.
Trends Pharmacol Sci
.
2021
;
42
(
9
):
729
44
.
106.
Liu
XF
,
Hao
JL
,
Xie
T
,
Malik
TH
,
Lu
CB
,
Liu
C
.
Nrf2 as a target for prevention of age-related and diabetic cataracts by against oxidative stress
.
Aging Cell
.
2017
;
16
(
5
):
934
42
.
107.
Lee
C
.
Therapeutic modulation of virus-induced oxidative stress via the nrf2-dependent antioxidative pathway
.
Oxid Med Cell Longev
.
2018
;
2018
:
6208067
.
108.
Zhang
Q
,
Liu
J
,
Duan
H
,
Li
R
,
Peng
W
,
Wu
C
.
Activation of Nrf2/HO-1 signaling: an important molecular mechanism of herbal medicine in the treatment of atherosclerosis via the protection of vascular endothelial cells from oxidative stress
.
J Adv Res
.
2021
;
34
:
43
63
.
109.
Ge
Y
,
Si
J
,
Tian
L
,
Zhuang
S
,
Dworkin
LD
,
Gong
R
.
Conditional ablation of glycogen synthase kinase 3β in postnatal mouse kidney
.
Lab Invest
.
2011
;
91
(
1
):
85
96
.
110.
Liu
T
,
Fang
Y
,
Liu
S
,
Yu
X
,
Zhang
H
,
Liang
M
.
Limb ischemic preconditioning protects against contrast-induced acute kidney injury in rats via phosphorylation of GSK-3β
.
Free Radic Biol Med
.
2015
;
81
:
170
82
.
111.
Basak
S
,
Kumar
A
,
Ramsey
S
,
Gibbs
E
,
Kapoor
A
,
Filizola
M
.
High-resolution structures of multiple 5-HT(3A)R-setron complexes reveal a novel mechanism of competitive inhibition
.
Elife
.
2020
;
9
:
e57870
.
112.
Hesketh
PJ
,
Kris
MG
,
Basch
E
,
Bohlke
K
,
Barbour
SY
,
Clark-Snow
RA
.
Antiemetics: ASCO guideline update
.
J Clin Oncol
.
2020
;
38
(
24
):
2782
97
.
113.
Fang
CY
,
Lou
DY
,
Zhou
LQ
,
Wang
JC
,
Yang
B
,
He
QJ
.
Natural products: potential treatments for cisplatin-induced nephrotoxicity
.
Acta Pharmacol Sin
.
2021
;
42
(
12
):
1951
69
.
114.
Goda
M
,
Kanda
M
,
Yoshioka
T
,
Yoshida
A
,
Murai
Y
,
Zamami
Y
.
Effects of 5-HT(₃) receptor antagonists on cisplatin-induced kidney injury
.
Clin Transl Sci
.
2021
;
14
(
5
):
1906
16
.
115.
Takemura
M
,
Ikemura
K
,
Kondo
M
,
Yamane
F
,
Ueda
M
,
Okuda
M
.
Concomitant palonosetron ameliorates cisplatin-induced nephrotoxicity, nausea, and vomiting: a retrospective cohort study and pharmacovigilance analysis
.
J Pharm Health Care Sci
.
2022
;
8
(
1
):
21
.
116.
Nishtala
PS
,
Chyou
TY
.
Identifying drug combinations associated with acute kidney injury using association rules method
.
Pharmacoepidemiol Drug Saf
.
2020
;
29
(
4
):
467
73
.
117.
Li
Q
,
Guo
D
,
Dong
Z
,
Zhang
W
,
Zhang
L
,
Huang
SM
.
Ondansetron can enhance cisplatin-induced nephrotoxicity via inhibition of multiple toxin and extrusion proteins (MATEs)
.
Toxicol Appl Pharmacol
.
2013
;
273
(
1
):
100
9
.
118.
Kou
W
,
Qin
H
,
Hanif
S
,
Wu
X
.
Nephrotoxicity evaluation on cisplatin combined with 5-HT(3) receptor antagonists: a retrospective study
.
BioMed Res Int
.
2018
;
2018
:
1024324
.
119.
Bagshaw
SM
.
Acute kidney injury care bundles
.
Nephron
.
2015
;
131
(
4
):
247
51
.
120.
Selby
NM
,
Kolhe
NV
.
Care bundles for acute kidney injury: do they work
.
Nephron
.
2016
;
134
(
3
):
195
9
.
121.
Zarbock
A
,
Küllmar
M
,
Ostermann
M
,
Lucchese
G
,
Baig
K
,
Cennamo
A
.
Prevention of cardiac surgery-associated acute kidney injury by implementing the KDIGO guidelines in high-risk patients identified by biomarkers: the PrevAKI-multicenter randomized controlled trial
.
Anesth Analg
.
2021
;
133
(
2
):
292
302
.
122.
Meersch
M
,
Schmidt
C
,
Hoffmeier
A
,
Van Aken
H
,
Wempe
C
,
Gerss
J
.
Prevention of cardiac surgery-associated AKI by implementing the KDIGO guidelines in high risk patients identified by biomarkers: the PrevAKI randomized controlled trial
.
Intensive Care Med
.
2017
;
43
(
11
):
1551
61
.
123.
Göcze
I
,
Jauch
D
,
Götz
M
,
Kennedy
P
,
Jung
B
,
Zeman
F
.
Biomarker-guided intervention to prevent acute kidney injury after major surgery: the prospective randomized BigpAK study
.
Ann Surg
.
2018
;
267
(
6
):
1013
20
.
124.
von Groote
T
,
Meersch
M
,
Romagnoli
S
,
Ostermann
M
,
Ripollés-Melchor
J
,
Schneider
AG
.
Biomarker-guided intervention to prevent acute kidney injury after major surgery (BigpAK-2 trial): study protocol for an international, prospective, randomised controlled multicentre trial
.
BMJ Open
.
2023
;
13
(
3
):
e070240
.