Skip to main content
Kidney International Reports logoLink to Kidney International Reports
. 2022 Sep 30;8(1):17–29. doi: 10.1016/j.ekir.2022.09.026

Molecular Mechanisms of Rhabdomyolysis-Induced Kidney Injury: From Bench to Bedside

Jessica F Hebert 1,4,, Kevin G Burfeind 1,4, Darren Malinoski 2,3, Michael P Hutchens 1,3
PMCID: PMC9831947  PMID: 36644345

Abstract

Rhabdomyolysis-induced acute kidney injury (RIAKI) occurs following damage to the muscular sarcolemma sheath, resulting in the leakage of myoglobin and other metabolites that cause kidney damage. Currently, the sole recommended clinical treatment for RIAKI is aggressive fluid resuscitation, but other potential therapies, including pretreatments for those at risk for developing RIAKI, are under investigation. This review outlines the mechanisms and clinical significance of RIAKI, investigational treatments and their specific targets, and the status of ongoing research trials.

Keywords: acute kidney injury, rhabdomyolysis, RIAKI, treatment


Acute skeletal muscle destruction, termed rhabdomyolysis, can be caused by a variety of etiologies, often in austere environments. Examples include crush injury after an earthquake or blast injury, overexertion during intense athletic activity, and various forms of ischemia-reperfusion injury, including stasis and compression due to general anesthesia, obtundation by other drugs, or vascular occlusion from a tourniquet or thromboembolism.1,2 Once skeletal muscle contents are in circulation, a well-understood sequence of events occurs, involving the release and potentially lethal accumulation of toxins, including potassium, lactic acid, and myoglobin, leading to multiple organ failures, most notably acute kidney injury (AKI). This phenomenon, referred to as RIAKI, is a common complication affecting up to 46% of patients hospitalized and 80% of those requiring intensive care unit for rhabdomyolysis.3,4 Even with excellent care, mortality is greater than 15%.5 The incidence of RIAKI has increased 10-fold in the last decade,6 fueled in part by popular interest in studio fitness training, such as CrossFit.7, 8, 9, 10 RIAKI is a significant comorbidity for injured soldiers who are are 3 to 4 times more likely to develop rhabdomyolysis than civilians.11 It is also common in critically ill patients with COVID-19, because COVID-AKI shares many features with RIAKI even without specific markers of RIAKI.12, 13, 14, 15, 16, 17, 18, 19

RIAKI takes on greater importance as the leading cause of death in immediate survivors of earthquakes. Nearly 400 million people live in cities in earthquake-prone areas, a number that is projected to double by 2050, making RIAKI treatment strategies an essential part of any disaster relief plan. The Renal Disaster Relief Task Force has contributed substantially to saving lives during natural disasters by establishing permanent networks for support as well as response plans.20, 21, 22 However, despite these efforts and more than 50 years of mechanistic insight, significant challenges imposed by austere care environments and investigative limitations have prevented specific therapy for RIAKI from reaching the bedside. Treatment is limited to supportive care, emphasizing early intravenous fluid administration, reduction of serum potassium, and forced diuresis.2 Studies demonstrate benefits from these treatments, including reduction in need for dialysis,1,5 but this moderately effective supportive care for RIAKI requires highly trained personnel, extensive access to medical supplies, and advanced modalities such as dialysis.23 Specific, molecular mechanism-based treatment would be expected to reduce these barriers.

Recent advances in kidney physiology and interest in improving care in austere environments have advanced mechanistic knowledge and brought new hope for specific therapy. Herein, we review these challenges and respective advances, focusing on how knowledge gained from preclinical studies can be translated into therapies to improve clinical management in humans. We first present known molecular mechanisms of RIAKI and how these can be used to develop novel therapies. We then briefly summarize current treatment paradigms, emphasizing challenges and shortcomings, and identify potential future directions.

Mechanisms of Rhabdomyolysis-Induced Kidney Injury: Avenues for Novel Therapies

Animal Models of RIAKI

The importance of RIAKI in veterinary medicine is an indication of a critically conserved balance between kidney function and muscle integrity. Many domesticated species develop RIAKI, including racing greyhound dogs, cattle, and horses. Equine exertional rhabdomyolysis (“tying-up” or “azoturia”) develops after intense physical effort or resuming physical work following a long period of inactivity.24 Physical symptoms in horses are similar to those in humans and include stiff movements, dark reddish urine, and swollen, tender muscles. Exertional rhabdomyolysis is also observed in captured wild animals, including flamingos, baboons, and pronghorn, in response to stress and physical efforts to escape.25 The conserved nature of this critical muscle-kidney response supports the use of animal models for investigation. In rodents, RIAKI may be induced by direct infusion of myoglobin,26 administration of drugs that have rhabdomyolysis as an adverse effect (e.g., ciprofloxacin and atorvastatin),27 crush injury or vascular occlusion,28 or destruction of muscle by intramuscular glycerol injection.29, 30, 31, 32, 33, 34, 35 It is vital to contextualize mechanistic insights obtained from animal models as folllows: systemic inflammation may be altered, for example, in a glycerol-treated rodent compared with a human with a crush injury.

Primary Injury by Myoglobin

Damage to the sarcolemma sheath of muscle impairs cellular impermeability to sodium and calcium ions, affecting the sodium potassium pump and calcium transporter functionality.36,37 The resultant high intracellular calcium concentration activates calcium-dependent enzymes that further disrupt cellular structure and allow leakage of myoglobin and other proteins and metabolites into the bloodstream, which can increase vascular permeability leading to interstitial edema and systemic intravascular hypovolemia.38 Myoglobin is readily filtered into the proximal tubule of the kidney. The route of released myoglobin from muscle to AKI mechanism is illustrated in Figure 1.

Figure 1.

Figure 1

Rhabdomyolysis-induced acute kidney injury is caused by myoglobin. (a) Muscle takes damage and releases myoglobin and other metabolites into circulation. (b) Myoglobin is circulated to the kidney for filtration, causing capillary damage and hypovolemia en route. (c) Myoglobin reaches the kidney and is filtered by the glomerulus. (d) Heme oxygenase-1 degrades heme transported into the proximal tubule by Heme carrier protein 1 to release free ferrous iron. (e) Iron bound to substrates, including myoglobin, is transported into the proximal kidney tubule by megalin and cubilin, further increasing the concentration of free ferrous iron. (f) Ferritin, which oxidizes Fe(2+) to Fe(3+) and stores it, fails to keep up with incoming free ferrous. Fe(2+) reacts with hydrogen peroxide in the Fenton reaction, producing hydroxyl radicals, lipid peroxidation, and overwhelming superoxide dismutase activity, resulting in the formation of damaging reactive oxygen species (ROS). (g) Myoglobin combines with Tamm-Horsfall protein (THP), found in the thick segment of the ascending limb, forming a precipitate. (h) THP-Myoglobin precipitate forms obstructive tubular casts in the distal convoluted tubule. (i) Urine output decreases, resulting in reduced potassium excretion and perturbation of water, pH, and sodium balances, putting further pressure on the vascular system. ROS, reactive oxygen species.

As myoglobin transits the nephron, it causes injury at 2 primary points followed by cascading effects. Proximal tubule cells internalize myoglobin via their apical endocytic complex, composed of megalin, cubilin, and amnionless.39 Within tubular epithelial cells (TECs), heme oxygenase-1 catalyzes the breakdown of heme to iron, carbon monoxide, and biliverdin.40 Iron in excess of heme oxygenase capacity combines with hydrogen peroxide to create ferric (Fe(3+)) iron. When ferric iron storage capacity is reached, free ferric iron increases hydroxyl ion concentrations via the iron redox cycle; this results in increased formation of reactive oxygen species (ROS) and peroxidation of tubular cell membrane lipids. In addition to iron-mediated hydroxyl radical production, ROS are also generated via the redox cycle creation of lipid peroxidation from excessive myoglobin and other nephrotoxins released from the muscle.41

Beyond the proximal convoluted tubule, in the thick ascending limb of the loop of Henle, urinary myoglobin combines with Tamm-Horsfall protein, forming a precipitate. This pH-dependent precipitate forms tubular casts that occlude the distal tubule.26,42 Although the extent to which AKI depends on this mechanism is controversial, tubular cast obstruction is believed to increase intratubular pressure to above interstitial pressure, reducing vascular inflow and perfusion, promoting inflammation, and directly reducing glomerular filtration rate by altering Starling forces.43, 44, 45

Intravascular hypovolemia and hypotension are induced by post-trauma muscular edema, leading to sympathetic and renin-angiotensin system-mediated vasoconstriction due to increased angiotensin II, compounding kidney ischemia by impairing the release of vasodilating nitric oxide.2 Kidney micropuncture in glycerol-induced RIAKI rats demonstrated that low kidney intratubular hydrostatic pressure failed to clear tubular casts; therefore, tubular casts are a consequence of reduced intratubular pressure rather than a cause.46 Pressure is related to vascular tone; increased vasopressin, noradrenaline, and adrenaline lead to kidney arteriole vasoconstriction, potentially as a compensatory mechanism for tubular obstruction.47,48 With decreased glomerular filtration rate, urine output decreases, reducing myoglobin's excretion. Tubular myoglobin concentration rises, augmenting apoptosis and necrosis of TECs as ROS increases. The resultant kidney tubule pathology observed in RIAKI is associated with insufficient compensatory response as follows: inadequate levels of free-radical scavengers, excessive neutrophil adhesion and cytokine release, and impaired microvascular blood flow.49 Thus myoglobin directly causes injury to tubular epithelial cells and indirectly compounds the injury by direct and indirect vascular effects, including the release of local vasoconstrictors in the vasa recta and peritubular capillaries.50

Immune-Mediated Mechanisms

In addition to processes directly related to myoglobin toxicity, products released from skeletal muscle and TEC damage act as immunogenic damage-associated molecular patterns, activating resident macrophages and recruiting circulating immune cells into the kidney interstitium. These cells then produce inflammatory cytokines and additional cytotoxic molecules, resulting in a positive feedback loop, potentiating cell damage. Immune activation is thought to be an essential aspect of RIAKI; however, the immune system's role in RIAKI is still not well understood, and although specific therapies targeting immune cells have been tested in preclinical studies, no translation has occurred. Becuase the immune response represents a “second hit,” i.e., a dysfunctional, secondary response to injury, treatments do not necessarily need to be administered immediately after injury, offering a highly translational window for intervention. In this section, we discuss proposed and potential immune-mediated mechanisms of RIAKI and how these may be exploited to develop specific therapies.

Because adaptive immunity takes a week or more to respond to injury, The critical immune mediation to RIAKI is thought to be primarily innate immune-driven. AKI immunology suffers from limited clinical data because the field is highly dependent on tissue studies, and biopsies are rarely performed in AKI.51,52 Thorough immunophenotyping throughout the course of kidney injury in animal models has not been conducted. Case data available demonstrate macrophage (innate) infiltration53,54 but also T-cell infiltration, suggesting the possibility that there may be adaptive immune involvement.55 The involvement of immune cells in kidney inflammation during RIAKI is depicted in Figure 2.

Figure 2.

Figure 2

Kidney inflammation during RIAKI and associated molecular targets. Myoglobin from the tubular system infiltrates into the interstitial space, resulting in immune activation. Studies thus far demonstrate that primarily innate immune cells are involved in kidney inflammation during RIAKI. These cells include monocytes, macrophages, natural killer cells, and neutrophils. Resident macrophages express IL-1β receptor, activation of which promotes production of inflammatory cytokines and cytotoxic macrophage extracellular traps, similar to neutrophil extracellular traps. Depicted in this figure are several potential molecular targets that have not yet been investigated in RIAKI. IL-β, interleukin 1 beta; IL-R, interleukin receptor.

Macrophages

Studies performed in rodent models of RIAKI have focused primarily on macrophages, which expand shortly after rhabdomyolysis.54,56, 57, 58 Macrophages are highly heterogeneous innate immune cells with numerous functions,59 including inflammatory cytokine production, pathogen phagocytosis, ROS generation, tissue repair, and cellular debris scavenging. They can be derived from circulating monocytes that transmigrate into the tissue or from kidney-resident cells of the reticuloendothelial system.56,57,60

Macrophages are producers of the most potent proinflammatory cytokine, interleukin 1 beta (IL-1β). IL-1β is an apical inflammatory signaling mediator, because its production is required to generate a mature immune response.61 However, dysregulated IL-1β production is deleterious. Signaling through the IL-1β receptor results in the transcription of additional inflammatory cytokines.61 Macrophage-derived IL-1β induces pathology in diabetes,62 cholestasis,63 and sepsis.64 In the kidney, macrophages produce IL-1β during intravenous contrast-induced AKI, inhibition of which prevents kidney injury.65 In addition, myoglobin induces IL-1β production in macrophages in vitro.56 Lastly, IL-1β induces production of extracellular matrix components in kidney organoids, suggesting a role in the development of fibrosis.66 Though IL-1β inhibition led to decreased fibrosis and improved glomerular filtration rate in obesity-associated CKD,67 to our knowledge, no studies have deleted IL-1β specifically from macrophages, which would provide key insight into the mechanisms of macrophage-mediated kidney injury.

Neutrophils

Neutrophils are the most abundant leukocyte population in the circulation and are first responders to injury and infection. Though these cells are essential for an appropriate infection response, inappropriate recruitment can cause tissue damage and drive organ dysfunction during several diseases, such as cancer,68 atherosclerosis,69 Alzheimer's disease,70 and systemic lupus erythematosus.71 These cells are potent ROS generators, produce a myriad of inflammatory cytokines, phagocytose bacteria, and form cytotoxic neutrophil extracellular traps. Neutrophils accumulate in the kidney during ischemia-repurfusion injury,72 and neutrophil depletion attenuates kidney damage following ischemia-repurfusion injury.73 Our laboratory has recently observed a significant increase in neutrophils 24 hours after RIAKI following muscular glycerol injection (Figure 3). However, no studies have investigated the role of neutrophils in RIAKI.

Figure 3.

Figure 3

Neutrophils infiltrate the kidney in a mouse model of RIAKI. (a) Schematic of a RIAKI mouse model, generated by glycerol injection into the anterior thigh after 4 hours of water deprivation, resulting in rhabdomyolysis and acute kidney injury. The kidney immune landscape was then characterized by flow cytometry 24 hours after injection. (b) Representative flow cytometry plots depicting immune cells (live, single cell, CD45+), identifying kidney lymphocytes (CD11b-) and myeloid cells (CD11b+) from glycerol- or saline-treated mice. (c) Representative flow cytometry plots depicting different myeloid cell populations from glycerol-treated or saline-treated mice. By gating on CD11b+ myeloid cells, Ly6CloLy6G- myeloid cells (mainly resident macrophages), Ly6ChiLy6G- monocytes (inflammatory monocytes), and Ly6CmidLy6G+ neutrophils were identified. (d) Quantification of kidney immune cells 24 hrs after glycerol injection, demonstrating an increase in myeloid cells, particularly neutrophils, in glycerol-injected mice. n = 2/group. CD, cluster of differentiation; CD11b, integrin alpha M; CD45, Protein tyrosine phosphatase, receptor type, C.

RIAKI Diagnosis and Treatment

Diagnostic Criteria

The most used clinical test for rhabdomyolysis is the measurement of plasma creatine kinase. Typically, a level greater than 10,000 IU/l is considered diagnostic confirmation of severe rhabdomyolysis and an increased risk for RIAKI.74 However, measuring creatine kinase alone may give a partial and late view of the problem. Hidden compartment syndrome (i.e., compartment syndrome not clinically obvious initially) is an uncommon yet serious condition that can be overlooked in evaluation of a trauma patient and obscure the value of creatinine kinase evaluation. This is especially true for trauma or surgical patients, but can occur in young healthy athletes.75 Therefore, urine or plasma myoglobin may be useful in certain clinical scenarios. It should also be noted that serum creatinine should not be used to estimate glomerular filtration rate in the context of massive muscle breakdown, which results in a large amount phosphocreatinine (which is converted to creatinine) released into the circulation.

Current Treatment Paradigms

The standard of care for acute rhabdomyolysis is fluid resuscitation directed toward preventing TEC injury by promoting the excretion of myoglobin. Aggressive fluid resuscitation with balanced salt solutions such as normal saline or lactated Ringer's is the first step. Although protocols vary, we demonstrated success using a protocol with initial bolus infusion of 1 to 2 liters followed by additional fluid at a rate of up to 1l/hr, adjusted to urine output, pH, and other physiologic parameters.76 The goal is to restore intravascular volume and dilute intratubular myoglobin, preventing tubular cell injury. Though this may be challenging in austere trauma conditions (e.g., earthquakes, combat zones), experiences from previous natural disasters suggest that immediate hydration into a protruding limb before extraction may be life-saving, considering that extraction could take well over an hour.77,78 Up to 10 liters of intravenous fluid may be required in the first 48 hours of treatment, yet comorbid conditions such as respiratory distress syndrome or chronic heart failure may limit the amount of fluid an injured person can receive, emphasizing the importance of individualized treatment strategy and careful clinical monitoring.

Treatment protocols often include sodium bicarbonate and mannitol in addition to intravenous fluid, sometimes guided by plasma and urine acid-base balance to provide optimal conditions for myoglobin solubility and promotion of excretion in urine. Myoglobin precipitates in acidic urine, leading to the use of bicarbonate. Mannitol, an osmotic diuretic, increases urine flow and serves as a ROS scavenger,79 reducing TEC exposure. We conducted a 10-year retrospective study of surgical patients with creatinine kinase levels over 10,000 IU/L, which suggested that protocolized treatment with fluid and the latter adjunctive therapies reduces the development of RIAKI.76 However, there remains controversy regarding this strategy. Clinical evidence to support these claims is based on few and small patient cohorts. In addition, they are challenged by the most extensive retrospective study of trauma-induced rhabdomyolysis, which showed no change in kidney failure, dialysis, or mortality after treatment with bicarbonate and mannitol.80 The methodologic heterogeneity of these conflicting reports and lack of an adequately randomized controlled trial prevent definitive conclusions from being drawn. Lastly, the potential benefit of bicarbonate should be weighed against the risk of promoting calcium phosphate precipitation in muscle. Thus, although the standard of care is guided by mechanistic insight, a greater understanding of connections and disconnects between injury due to intratubular myoglobin and plasma volume, plasma pH, and osmotic diuretic use is necessary.

Extracorporeal technologies such as dialysis also play a role in RIAKI treatment. Dialysis is necessary once kidney failure is established or significant hyperkalemia occurs. Indeed, a newly liberated crushed extremity containing 150 mEq/l of intracellular potassium can produce hyperkalemia which can only be resolved with some form of dialysis. The challenge of providing dialysis in epidemics, disasters and military environments has driven innovation in use and design of dialysis equipment, including redesign or repurposing of fluidics and pumps,81, 82, 83 sorbent-based dialyzers,84,85 and extended use of critical care-based equipment.86,87 As with specific medical therapy, current evidence for these technologies is preliminary, but tantalizing.

Developing Mechanism-Based Treatments for RIAKI

The intervention window for preventing RIAKI is unknown but suspected to be within hours following injury as AKI develops quickly; therefore, treatments with rapid effects that can be readily administered are of utmost importance.88

Inhibition of Myoglobin Endocytosis

A well-characterized endocytic complex composed of megalin, cubilin, and amnionless transports many filtered proteins, including myoglobin, into TEC.39 Removing or blocking megalin activity prevents tubular myoglobin uptake.39 Mice with proximal tubule-specific megalin deletion are profoundly protected from myoglobin and demonstrate greater than 10-fold more myoglobin clearance than wild type mice. Our laboratory has recently identified a kidney dipeptidase inhibitor, cilastatin, with similar megalin-inhibitory characteristics.89 Administering cilastatin at the time of glycerol-induced rhabdomyolysis in mice results in a renoprotective effect, which recapitulates the effect seen in megalin-deleted mice.90 This efficacy of contemporaneous administration of cilastatin with RIAKI indicates a novel postinjury treatment for rhabdomyolysis to prevent kidney injury if given in a limited window following muscular trauma. Cilastatin is currently approved by the United States Food and Drug Administration for another indication and is nearly nontoxic; therefore, it is an promising drug for additional studies in related models and clinical trials.

Oxidative Damage Amelioration

ROS generated during RIAKI have been the target of several drug therapies, primarily antioxidants, designed to reduce ROS in vivo by inhibiting or scavenging, thereby protecting cells from further damage. Acetaminophen, an inhibitor of the formation of peroxide-generated radicals, was found to decrease ROS damage in the kidney and improve kidney function by counteracting lipid peroxidation and reducing global free radicals in a rat model of RIAKI. However, in high doses, it may generate n-acetyl-p-benzoquinone imine, a toxic byproduct.91,92 In some studies involving chronic kidney disease, n-acetyl cysteine has been shown to beneficially increase ROS scavenging and improve cardiovascular and kidney function.93 Other studies show no effect of n-acetyl cysteine on serum creatinine and other markers of kidney function.94,95 The use of n-acetyl cysteine to ameliorate RIAKI is, therefore, controversial.

Glutathione oxidation-reduction cycling is the object of other potential treatments for oxidative stress, including selenium. Glutathione peroxidase (and other reductases) are selenium-dependent enzymes that reduce superoxides.96 Selenium also increases peroxisome proliferator-activated receptor gamma coactivator 1-alpha, critical to many cellular metabolic pathways, including mitochondrial biogenesis.97 Providing pre-RIAKI dietary selenium to rats increased kidney superoxide dismutase and nitric oxide with reduced proteinuria, but other classical markers for AKI such as plasma urea and creatinine remained unchanged.98 Reduced proteinuria may result in less myoglobin clearance, and with unchanged creatinine levels, it is unlikely that this method of selenium administration is the most effective treatment.

Antioxidant vitamins C and E have been widely tested in AKI. Vitamin C reduced oxidative stress, kidney damage, tubular cast formation, and proteinuria in RIAKI, but vitamin E only inhibited lipid peroxidation with no subsequent improvement in kidney function or cellular condition.99 Curcumin delivered as an oral dietary supplement in rats following RIAKI has been found in one study to reduce kidney cell apoptosis and oxidative stress via regulation of 5' adenosine monophosphate-activated protein kinase, Nrf2/HO01, and PI3K/Akt pathways.35 Future renoprotective treatments may also come from mitochondria-targeted and global antioxidants that have successfully reduced ROS in other tissues, including mitochondrial-derived peptides like humanin and the superoxide dismutase stimulant metformin.100 At current, the most promising global antioxidants affecting AKI appear to be vitamin C and curcumin, both of which have low toxicity and are readily abundant, but targeting moieties discussed later in this paper may improve the effectiveness of these and other antioxidants in treating AKI.

Immune-Targeting Treatments

Despite the centrality of IL-1β to RIAKI and other forms of AKI, no studies have assessed whether targeting IL-1β can improve outcomes after RIAKI. However, translational investigation is suggested, because several inhibitors of IL-1β signaling are approved for the treatment of inflammatory disease in humans. These include canakinumab (an IL-1β-neutralizing antibody),101 anakinra (a recombinant IL-1β receptor antagonist), rilonacept (an IL-1β-neutralizing small molecule), and gevokizumab (an IL-1β-binding antibody). Several clinical trials investigating the efficacy of these treatments for non-AKI kidney disease are ongoing or were recently completed.102 For example, the Canakinumab Antiinflammatory Thrombosis Outcomes Study demonstrated decreased cardiovascular events in patients with CKD.103 In addition, canakinumab decreased proteinuria in a patient with kidney amyloidosis.104 CD44-overexpression stimulates IL-1β, but targeting these cells to counter increased IL-1β with elamipretide (SS-31) conjugated to nanopolyplexes reduced oxidative stress, inflammatory response, and apoptosis in tubular cells affected by AKI.105

Macrophages can also release cytotoxic molecules, which are essential for killing pathogens but can also damage surrounding tissue. A recent study in rodents demonstrated that kidney macrophages release their cellular contents during RIAKI, producing cytotoxic extracellular traps, called "macrophage extracellular traps" (METs).58 METs are similar to extracellular traps produced by neutrophils (termed neutrophil extracellular traps), which are potently bactericidal106 but can be aberrantly produced during conditions of sterile inflammation, such as cancer,68 Alzheimer's disease,70 and systemic lupus erythematosus,107 leading to tissue damage. During RIAKI, platelets and iron induce MET formation, which is dependent on integrin αMβ2 (also referred to as Mac-1).58 MET inhibition prevents TEC damage in RIAKI, presenting multiple therapeutic targets. A Mac-1 inhibitor was demonstrated to prevent cytokine storm and pathological damage in a mouse model of sepsis.108 In addition, lactoferrin, a naturally occurring glycoprotein with antibacterial properties, inhibited platelet-induced METs.58 Lastly, there are several inhibitors of peptidyl arginine deiminase, a key enzyme for neutrophil extracellular trap (and presumably MET) formation.109,110

Neutrophils present an unexplored cellular target for RIAKI. Several therapies targeting neutrophils are currently under investigation or already Food and Drug Administration approved.111 Brensocatib inhibits dipeptidyl peptidase-1, an enzyme key in activating neutrophil serine proteases. A recently published phase II clinical trial (NCT03218917) demonstrated that brensocatib administration was associated with increased time to first exacerbation in patients with bronchiectasis.112 In addition, secukinumab is a monoclonal antibody against IL-17A, which is important for neutrophil activation. Secukinumab is Food and Drug Administration-approved for psoriatic arthritis, psoriasis, and ankylosing spondylitis.113 To our knowledge, no studies have investigated the efficacy of these treatments in inflammatory kidney disease. This presents an exciting avenue for future RIAKI treatments.

Kidney-Targeted Delivery Systems

Kidney-targeted delivery systems, including nanoparticles (NPs) and microbubbles (MBs), are recent advances in medical technology to deliver drugs, proteins, and other pharmaceutical payloads to prevent and treat kidney dysfunction. Both NPs and MBs have benefits versus nonconjugated treatments as follows: they improve bioactivity and targeting and reduce toxicity and adverse side effects associated with other methods of treatment administration.114 NPs are generally 1 to 100 nm in diameter; though there is variation in how they are constructed, typically, a multifunctional “backbone” or envelope structure is bound to therapeutic and targeting moieties. Either the backbone alone or additional structural motifs may be added to target the molecule to the kidney specifically. The unique physiology of filtration and reabsorption in the kidney offers the potential for precise targeting, which may increase the efficacy of agents which have broad, systemic mechanisms, such as antioxidants.115,116 Although there are currently no clinically approved kidney-targeted NPs, NP treatment of rodent in vivo models of glycerol-induced rhabdomyolysis has produced promising results. AKI induced by ischemia-reperfusion injury has been the treatment focus of several NPs, including triptolide-encapsulated, resveratrol-conjugated, polyethylene glycol-treated ceria conjugated to atorvastatin, and oltipraz-loaded poly lactic-co-glycolic acid NPs.105,117, 118, 119, 120 Kidney clearable nanochelators designed to reduce excess iron have successfully targeted rat kidneys, with a 7-fold increase in urinary iron excretion, indicating a potential mechanism of reduced ROS creation and proximal tubule damage via the Fenton reaction.121 In addition, dextran, dendrimer, and poly lactic-co-glycolic acid-polyethylene glycol NPs, cationic quantum dots, and carbon nanotubes have been used to target kidney tubular cells in chronic kidney disease and may be options to treat RIAKI.115 Overall, the ability to specifically target the kidney and perhaps the proximal tubule renders NPs a promising emerging area. These targeting strategies may ameliorate many of the dose-limiting and treatment-limiting off-target effects of otherwise effective drugs.

Using selenium and rutin-coated gold NPs to pretreat before causing glycerol-induced rhabdomyolysis also showed favorable outcomes. Selenium NPs reduced elevated creatinine, serum creatine phosphokinase, and damage severity compared to injured but not uninjured rats, which was similar to what was observed using rutin-coated gold NP pretreatment.122 Curcumin-conjugated polyethylene glycol NPs were used to assess treatment efficacy in a human in vitro (HK-2 cells) and rodent in vivo model. Curcumin-NPs reduced oxidative stress and cellular apoptosis in vitro; creatinine, serum creatine phosphokinase, and the severity of kidney tubule damage were reduced to control levels in vivo.123 Quercetin-derived treatments such as rutin, ubiquinol (MitoQ), and plastoquinone (SkQ1) have also been used to reduce ROS via novel mechanisms that are still not fully understood but have produced promising results in other injury paradigms, suggesting that they may be worth considering for trials in AKI treatment.100,124

MBs have been used since the 1990s to facilitate ultrasound imaging. MBs are 1 to 10 μm in diameter, comprised of a gas core surrounded by a protein, lipid, or polymer envelope embedded with targeting molecules.125 MBs can be used to deliver treatment agents targeted to specific organs by applying ultrasound to the target, either destroying the bubble membrane or increasing local vascular permeability to facilitate uptake.126 Targeting moieties that have been employed include Smad 7, miR 433, NFκB, siTNFα, and VEGFR2.127 MBs have not yet been shown to ameliorate AKI, but modifications to the MB to improve its time in circulation have been proposed as potential carriers for treatments. This modality offers promise, especially with the widespread adoption of handheld ultrasound for clinical use; however, additional research into specific targets and protocols will be required.

Current Treatments for RIAKI, Clinical Trials, and Proposed Preclinical Therapies Conclusion

In Table 1 we provide a summary of current approved therapies for RIAKI as well as several novel therapies in various stages of investigation. Muscular trauma, particularly following crush injury, results in rhabdomyolysis and AKI primarily through proximal tubule endocytosis of myoglobin and subsequent immune-driven response. Current clinically approved treatments are limited, but recent research advances provide promising new avenues to improved treatment, including inhibition of myoglobin endocytosis, prevention and treatment of oxidative damage, and immune cell targets. Technology-related advances in NP and MB delivery may further enhance outcomes. Advancements in these areas would potentially prevent the long-term effects caused by the transition of AKI later to CKD.

Table 1.

RIAKI treatments (current and proposed) and their molecular targets

Current therapies
Treatment Molecular target Investigated in RIAKI? Investigation stage Reference(s)
Intravenous fluid Tubular flow Y Current recommended treatment 76
Sodium bicarbonate Tubular pH, myoglobin precipitation Y Current treatment at some centers 76,80
Mannitol Tubular flow, ROS Y Current treatment at some centers 76,80
Proposed therapies
Treatment Molecular target Investigated in RIAKI? Investigation stage Reference(s)
Cilastatin Megalin/tubular endocytosis Y Preclinical 89,90
High flux dialysis Myoglobin Y Phase I - NCT01467180 128
N Acetylcystine Reactive oxygen species Y Phase II - NCT00391911 93, 94, 95
CytoSorb device Myoglobin Y Phase II - NCT02111018 129,130
Peptidyl arginine deaminase NET/MET formation N-lupus Preclinical 131
Brensocatib Dipeptidyl peptidase-1 N- brochiectasis Phase II - NCT03218917 112
Secukinumab IL-17A N-rheumatoid diseases FDA approved for rheumatoid diseases 132
Lactoferrin MET formation Y Preclinical 58
Anti-Mac-1 antibody Mac-1 Y Preclinical 58
Canakinumab IL-1B N-CKD Phase III - NCT01327846 103
Anakinra IL-1B N-inflammation in CKD Phase II - NCT00420290, Phase II - NCT02278562 133
Rilonacept IL-1B N-inflammation in CKD Phase II - NCT00897715 134
Gevokizumab IL-1B N-Type 2 diabetic kidney disease Phase II - EudraCT2013–003610–41 135

CKD, chronic kidney disease; IL-B, interleukin beta; MET, macrophage extracellular traps; NET, neutrophil extracellular traps; RIAKI, rhabdomyolysis-induced acute kidney injury; ROS, reactive oxygen species; Y, yes.

Disclosure

The authors have no conflicting financial interests to disclose. JFH is supported by the Oregon Students Learn and Experience Research (OSLER) TL1 grant TL1TR002371. KGB received research support resources provided by the OHSU Department of Anesthesiology & Perioperative Medicine. This work was supported in part by the United States Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development, Biomedical Laboratory Research and Development (VA Merit Award #1I01BX004288 to MPH), and the United States Department of Defense (W81XWH2010196 to MPH). This material is also the result of work supported by resources and the use of facilities at the Portland Veterans Affairs Medical Center in Portland, Oregon. The contents do not represent the views of the US Department of Veterans Affairs or the US Government.

References

  • 1.Vanholder R., Sever M.S., Erek E., Lameire N. Rhabdomyolysis. J Am Soc Nephrol. 2000;11:1553–1561. doi: 10.1681/ASN.V1181553. [DOI] [PubMed] [Google Scholar]
  • 2.Bosch X., Poch E., Grau J.M. Rhabdomyolysis and acute kidney injury. N Engl J Med. 2009;361:62–72. doi: 10.1056/NEJMra0801327. [DOI] [PubMed] [Google Scholar]
  • 3.Candela N., Silva S., Georges B., et al. Short- and long-term renal outcomes following severe rhabdomyolysis: a French multicenter retrospective study of 387 patients. Ann Intensive Care. 2020;10:27. doi: 10.1186/s13613-020-0645-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Melli G., Chaudhry V., Cornblath D.R. Rhabdomyolysis: an evaluation of 475 hospitalized patients. Med (Baltim) 2005;84:377–385. doi: 10.1097/01.md.0000188565.48918.41. [DOI] [PubMed] [Google Scholar]
  • 5.Nielsen F.E., Cordtz J.J., Rasmussen T.B., Christiansen C.F. The association between rhabdomyolysis, acute kidney injury, renal replacement therapy, and mortality. Clin Epidemiol. 2020;12:989–995. doi: 10.2147/CLEP.S254516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Boden BP, Isaacs DJ, Ahmed AE, Anderson SA. Epidemiology of exertional rhabdomyolysis in the United States: analysis of NEISS database 2000 to 2019. Phys Sportsmed. 2021:1-8. doi:10.1080/00913847.2021.1956288 [DOI] [PubMed]
  • 7.Hopkins B.S., Li D., Svet M., et al. CrossFit and rhabdomyolysis: A case series of 11 patients presenting at a single academic institution. J Sci Med Sport. 2019;22:758–762. doi: 10.1016/j.jsams.2019.01.019. [DOI] [PubMed] [Google Scholar]
  • 8.Knapik J.J. Extreme conditioning programs: potential benefits and potential risks. J Spec Oper Med. 2015;15:108–113. doi: 10.55460/8J8E-2Q8D. [DOI] [PubMed] [Google Scholar]
  • 9.Meyer M., Sundaram S., Schafhalter-Zoppoth I. Exertional and crossfit-induced rhabdomyolysis. Clin J Sport Med. 2018;28:e92–e94. doi: 10.1097/JSM.0000000000000480. [DOI] [PubMed] [Google Scholar]
  • 10.Mitchell F., Henderson H.J., Gardner F. Cluster of exertional rhabdomyolysis in three young women. BMJ Case Rep. 2018;2018 doi: 10.1136/bcr-2017-223022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hill O.T., Wahi M.M., Carter R., 3rd, et al. Rhabdomyolysis in the US active duty army, 2004–2006. Med Sci Sports Exerc. 2012;44:442–449. doi: 10.1249/MSS.0b013e3182312745. [DOI] [PubMed] [Google Scholar]
  • 12.Albaba I., Chopra A., Al-Tarbsheh A.H., et al. Incidence, risk factors, and outcomes of rhabdomyolysis in hospitalized patients with COVID-19 infection. Cureus. 2021;13 doi: 10.7759/cureus.19802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Haroun M.W., Dieiev V., Kang J., et al. Rhabdomyolysis in COVID-19 patients: A retrospective observational study. Cureus. 2021;13 doi: 10.7759/cureus.12552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Smarz-Widelska I., Grywalska E., Morawska I., et al. Pathophysiology and clinical manifestations of COVID-19-related acute kidney injury—the current state of knowledge and future perspectives. Int J Mol Sci. 2021;22:7082. doi: 10.3390/ijms22137082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Werion A., Belkhir L., Perrot M., et al. SARS-CoV-2 causes a specific dysfunction of the kidney proximal tubule. Kidney Int. 2020;98:1296–1307. doi: 10.1016/j.kint.2020.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bouquegneau A., Erpicum P., Grosch S., et al. COVID-19-associated nephropathy includes tubular necrosis and capillary congestion, with evidence of SARS-CoV-2 in the nephron. Kidney360. 2021;2:639–652. doi: 10.34067/KID.0006992020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chan L., Chaudhary K., Saha A., et al. AKI in hospitalized patients with COVID-19. J Am Soc Nephrol. 2021;32:151–160. doi: 10.1681/ASN.2020050615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Papadimitriou J.C., Drachenberg C.B., Kleiner D., et al. Tubular epithelial and peritubular capillary endothelial injury in COVID-19 AKI. Kidney Int Rep. 2021;6:518525. doi: 10.1016/j.ekir.2020.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Mokhtari A.K., Maurer L.R., Christensen M.A., et al. Rhabdomyolysis in severe COVID-19: male sex, high body mass index, and prone positioning confer high risk. J Surg Res. 2021;266:35–43. doi: 10.1016/j.jss.2021.03.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Vanholder R., Sever M.S., Lameire N. The role of the renal disaster relief task force. Nat Clin Pract Nephrol. 2008;4:347. doi: 10.1038/ncpneph0862. [DOI] [PubMed] [Google Scholar]
  • 21.Sever M.S., Vanholder R., Lameire N. Management of crush-related injuries after disasters. N Engl J Med. 2006;354:1052–1063. doi: 10.1056/NEJMra054329. [DOI] [PubMed] [Google Scholar]
  • 22.Lameire N., Sever M.S., Van Biesen W., Vanholder R. Role of the international and national renal organizations in natural disasters: strategies for renal rescue. Semin Nephrol. 2020;40:393–407. doi: 10.1016/j.semnephrol.2020.06.007. [DOI] [PubMed] [Google Scholar]
  • 23.Sever M.S., Remuzzi G., Vanholder R. Disaster medicine and response: optimizing life-saving potential. Am J Disaster Med. 2018;13:253–264. doi: 10.5055/ajdm.2018.0305. [DOI] [PubMed] [Google Scholar]
  • 24.Valberg S.J., Mickelson J.R., Gallant E.M, MacLeay J.M., De La Corte L. Lentz. F. Exertional rhabdomyolysis in Quarter Horses and Thoroughbreds: one syndrome, multiple aetiologies. Equine Vet J Suppl. 1999;31:533–553. doi: 10.1111/j.2042-3306.1999.tb05279.x. [DOI] [PubMed] [Google Scholar]
  • 25.Bartsch R.C., Mcconnell E.E., Imes G.D., Schmidt J.M. A review of exertional rhabdomyolysis in wild and domestic animals and man. Vet Pathol. 1977;14:314–324. doi: 10.1177/030098587701400402. [DOI] [PubMed] [Google Scholar]
  • 26.Zager R.A. Rhabdomyolysis and myohemoglobinuric acute renal failure. Kidney Int. 1996;49:314–326. doi: 10.1038/ki.1996.48. [DOI] [PubMed] [Google Scholar]
  • 27.Matsubara A., Oda S., Akai S., et al. Establishment of a drug-induced rhabdomyolysis mouse model by co-administration of ciprofloxacin and atorvastatin. Toxicol Lett. 2018;291:184–193. doi: 10.1016/j.toxlet.2018.04.016. [DOI] [PubMed] [Google Scholar]
  • 28.Zhu H., Hou J., Roe J.L., et al. Amelioration of ischemia-reperfusion-induced muscle injury by the recombinant human MG53 protein. Muscle Nerve. 2015;52:852–858. doi: 10.1002/mus.24619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wilson D.R., Thiel G., Arce M.L., Oken D.E. Glycerol induced hemoglobinuric acute renal failure in the rat. 3. Micropuncture study of the effects of mannitol and isotonic saline on individual nephron function. Nephron. 1967;4:337–355. doi: 10.1159/000179594. [DOI] [PubMed] [Google Scholar]
  • 30.Wolfert A.I., Oken D.E. Glomerular hemodynamics in established glycerol-induced acute renal failure in the rat. J Clin Invest. 1989;84:1967–1973. doi: 10.1172/JCI114386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Nath K.A., Balla G., Vercellotti G.M., et al. Induction of heme oxygenase is a rapid, protective response in rhabdomyolysis in the rat. J Clin Invest. 1992;90:267–270. doi: 10.1172/JCI115847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wei Q., Hill W.D., Su Y., Huang S., Dong Z. Heme oxygenase-1 induction contributes to renoprotection by G-CSF during rhabdomyolysis-associated acute kidney injury. Am J Physiol Ren Physiol. 2011;301:F162–F170. doi: 10.1152/ajprenal.00438.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zager R.A., Burkhart K.M., Conrad D.S., Gmur D.J. Iron, heme oxygenase, and glutathione: effects on myohemoglobinuric proximal tubular injury. Kidney Int. 1995;48:1624–1634. doi: 10.1038/ki.1995.457. [DOI] [PubMed] [Google Scholar]
  • 34.Thomsen H.S., Larsen S., Hemmingsen L., Holm J., Skaarup P. Nephropathy induced by intramuscularly administered glycerol and contrast media in rats. A comparison between diatrizoate, iohexol and ioxilan. Acta Radiol. 1989;30:217–222. [PubMed] [Google Scholar]
  • 35.Wu J., Pan X., Fu H., et al. Effect of curcumin on glycerol-induced acute kidney injury in rats. Sci Rep. 2017;7:10114. doi: 10.1038/s41598-017-10693-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Jackson M.J., Jones D.A., Edwards R.H.T. Experimental skeletal muscle damage: the nature of the calcium-activated degenerative processes. Eur J Clin Investig. 1984;14:369–374. doi: 10.1111/j.1365-2362.1984.tb01197.x. [DOI] [PubMed] [Google Scholar]
  • 37.Blaustein M.P. Sodium ions, calcium ions, blood pressure regulation, and hypertension: a reassessment and a hypothesis. Am J Physiol Cell Physiol. 1977;232:C165–C173. doi: 10.1152/ajpcell.1977.232.5.C165. [DOI] [PubMed] [Google Scholar]
  • 38.Zhang M.H., Zhang M.-H. Rhabdomyolosis and its pathogenesis. World J Emerg Med. 2012;3:11–15. doi: 10.5847/wjem.j.issn.1920-8642.2012.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Gburek J., Birn H., Verroust P.J., et al. Renal uptake of myoglobin is mediated by the endocytic receptors megalin and cubilin. Am J Physiol Ren Physiol. 2003;285:F451–F458. doi: 10.1152/ajprenal.00062.2003. [DOI] [PubMed] [Google Scholar]
  • 40.Agarwal A., Bolisetty S. Adaptive responses to tissue injury: role of heme oxygenase-1. Trans Am Clin Climatol Assoc. 2013;124:111–122. [PMC free article] [PubMed] [Google Scholar]
  • 41.Petejova N., Martinek A. Acute kidney injury due to rhabdomyolysis and renal replacement therapy: a critical review. Crit Care (London, England) 2014;18:224. doi: 10.1186/cc13897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Gilbert S.J., Weiner D.E., Gipson D.S., et al. Elsevier/Saunders; 2014. National kidney foundation’s primer on kidney diseases. [Google Scholar]
  • 43.Sheerin N.S., Sacks S.H. Leaked protein and interstitial damage in the kidney: is complement the missing link? Clin Exp Immunol. 2002;130:1–3. doi: 10.1046/j.1365-2249.2002.01979.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hegarty N.J., Young L.S., Kirwan C.N., et al. Nitric oxide in unilateral ureteral obstruction: effect on regional renal blood flow. Kidney Int. 2001;59:1059–1065. doi: 10.1046/j.1523-1755.2001.0590031059.x. [DOI] [PubMed] [Google Scholar]
  • 45.Wei J., Song J., Jiang S., et al. Role of intratubular pressure during the ischemic phase in acute kidney injury. Am J Physiol Ren Physiol. 2017;312:F1158–F1165. doi: 10.1152/ajprenal.00527.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Oken D.E., Arce M.L., Wilson D.R. Glycerol-induced hemoglobinuric acute renal failure in the rat. I. Micropuncture study of the development of oliguria. J Clin Invest. 1966;45:724–735. doi: 10.1172/JCI105387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ito S., Abe K. Tubuloglomerular feedback. Jpn Heart J. 1996;37:153–163. doi: 10.1536/ihj.37.153. [DOI] [PubMed] [Google Scholar]
  • 48.Messerer D.A.C., Halbgebauer R., Nilsson B., et al. Immunopathophysiology of trauma-related acute kidney injury. Nat Rev Nephrol. 2021;17:91–111. doi: 10.1038/s41581-020-00344-9. [DOI] [PubMed] [Google Scholar]
  • 49.Huerta-Alardín A.L., Varon J., Marik P.E. Bench-to-bedside review: rhabdomyolysis -- an overview for clinicians. Crit Care (London, England) 2005;9:158–169. doi: 10.1186/cc2978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Malinoski D.J., Slater M.S., Mullins R.J. Crush injury and rhabdomyolysis. Crit Care Clin. 2004;20:171–192. doi: 10.1016/s0749-0704(03)00091-5. [DOI] [PubMed] [Google Scholar]
  • 51.Zuk A., Palevsky P.M., Fried L., et al. Overcoming translational barriers in acute kidney injury: A report from an NIDDK workshop. Clin J Am Soc Nephrol. 2018;13:1113–1123. doi: 10.2215/CJN.06820617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Waikar S.S., McMahon G.M. Expanding the role for kidney biopsies in acute kidney injury. Semin Nephrol. 2018;38:12–20. doi: 10.1016/j.semnephrol.2017.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sinniah R., Lye W. Acute renal failure from myoglobinuria secondary to myositis from severe falciparum malaria. Am J Nephrol. 2000;20:339–343. doi: 10.1159/000013611. [DOI] [PubMed] [Google Scholar]
  • 54.Rubio-Navarro A., Carril M., Padro D., et al. CD163-macrophages are involved in rhabdomyolysis-induced kidney injury and may be detected by MRI with targeted gold-coated iron oxide nanoparticles. Theranostics. 2016;6:896–914. doi: 10.7150/thno.14915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Grivei A., Giuliani K.T.K., Wang X., et al. Oxidative stress and inflammasome activation in human rhabdomyolysis-induced acute kidney injury. Free Radic Biol Med. 2020;160:690–695. doi: 10.1016/j.freeradbiomed.2020.09.011. [DOI] [PubMed] [Google Scholar]
  • 56.Belliere J., Casemayou A., Ducasse L., et al. Specific macrophage subtypes influence the progression of rhabdomyolysis-induced kidney injury. J Am Soc Nephrol. 2015;26:1363–1377. doi: 10.1681/ASN.2014040320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kim J.H., Lee D.W., Jung M.H., et al. Macrophage depletion ameliorates glycerol-induced acute kidney injury in mice. Nephron Exp Nephrol. 2014;128:21–29. doi: 10.1159/000365851. [DOI] [PubMed] [Google Scholar]
  • 58.Okubo K., Kurosawa M., Kamiya M., et al. Macrophage extracellular trap formation promoted by platelet activation is a key mediator of rhabdomyolysis-induced acute kidney injury. Nat Med. 2018;24:232–238. doi: 10.1038/nm.4462. [DOI] [PubMed] [Google Scholar]
  • 59.Murray P.J., Wynn T.A. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11:723–737. doi: 10.1038/nri3073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ide S., Yahara Y., Kobayashi Y., et al. Yolk-sac-derived macrophages progressively expand in the mouse kidney with age. eLife. 2020;9 doi: 10.7554/eLife.51756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Weber A., Wasiliew P., Kracht M. Interleukin-1 (IL-1) pathway. Sci Signal. 2010;3:cm1. doi: 10.1126/scisignal.3105cm1. [DOI] [PubMed] [Google Scholar]
  • 62.Monnerat G., Alarcón M.L., Vasconcellos L.R., et al. Macrophage-dependent IL-1β production induces cardiac arrhythmias in diabetic mice. Nat Commun. 2016;7:13344. doi: 10.1038/ncomms13344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.El Kasmi K.C., Vue P.M., Anderson A.L., et al. Macrophage-derived IL-1β/NF-κB signaling mediates parenteral nutrition-associated cholestasis. Nat Commun. 2018;9:1393. doi: 10.1038/s41467-018-03764-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Rodriguez A.E., Ducker G.S., Billingham L.K., et al. Serine metabolism supports macrophage IL-1β production. Cell Metab. 2019;29:1003–1011. doi: 10.1016/j.cmet.2019.01.014. e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Lau A., Chung H., Komada T., et al. Renal immune surveillance and dipeptidase-1 contribute to contrast-induced acute kidney injury. J Clin Invest. 2018;128:2894–2913. doi: 10.1172/JCI96640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lemos D.R., McMurdo M., Karaca G., et al. Interleukin-1β activates a MYC-dependent metabolic switch in kidney stromal cells necessary for progressive tubulointerstitial fibrosis. J Am Soc Nephrol. 2018;29:1690–1705. doi: 10.1681/ASN.2017121283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lei Y., Devarapu S.K., Motrapu M., et al. Interleukin-1β inhibition for chronic kidney disease in obese mice with Type 2 diabetes. Front Immunol. 2019;10:1223. doi: 10.3389/fimmu.2019.01223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Burfeind K.G., Zhu X., Norgard M.A., et al. Circulating myeloid cells invade the central nervous system to mediate cachexia during pancreatic cancer. eLife. 2020;9 doi: 10.7554/eLife.54095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Drechsler M., Megens R.T., van Zandvoort M., et al. Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis. Circulation. 2010;122:1837–1845. doi: 10.1161/CIRCULATIONAHA.110.961714. [DOI] [PubMed] [Google Scholar]
  • 70.Zenaro E., Pietronigro E., Bianca V.D., et al. Neutrophils promote Alzheimer’s disease–like pathology and cognitive decline via LFA-1 integrin. Nat Med. 2015;21:880–886. doi: 10.1038/nm.3913. [DOI] [PubMed] [Google Scholar]
  • 71.Kaplan M.J. Neutrophils in the pathogenesis and manifestations of SLE. Nat Rev Rheumatol. 2011;7:691–699. doi: 10.1038/nrrheum.2011.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Awad A.S., Rouse M., Huang L., et al. Compartmentalization of neutrophils in the kidney and lung following acute ischemic kidney injury. Kidney Int. 2009;75:689–698. doi: 10.1038/ki.2008.648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kim H.J., Lee J.S., Kim J.D., et al. Reverse signaling through the costimulatory ligand CD137L in epithelial cells is essential for natural killer cell-mediated acute tissue inflammation. Proc Natl Acad Sci U S A. 2012;109:E13–E22. doi: 10.1073/pnas.1112256109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.de Meijer A.R., Fikkers B.G., de Keijzer M.H., et al. Serum creatine kinase as predictor of clinical course in rhabdomyolysis: a 5-year intensive care survey. Intensive Care Med. 2003;29:1121–1125. doi: 10.1007/s00134-003-1800-5. [DOI] [PubMed] [Google Scholar]
  • 75.McQuerry J.L., Burnham J.M., Ireland M.L., Wright R.D. Delayed presentation of compartment syndrome of the thigh in a previously undiagnosed factor VII-deficient high school football athlete: A case report. JBJS Case Connect. 2018;8:e4. doi: 10.2106/JBJS.CC.17.00123. [DOI] [PubMed] [Google Scholar]
  • 76.Nielsen J.S., Sally M., Mullins R.J., et al. Bicarbonate and mannitol treatment for traumatic rhabdomyolysis revisited. Am J Surg. 2017;213:73–79. doi: 10.1016/j.amjsurg.2016.03.017. [DOI] [PubMed] [Google Scholar]
  • 77.Sever M.S., Erek E., Vanholder R., et al. Lessons learned from the catastrophic Marmara earthquake: factors influencing the final outcome of renal victims. Clin Nephrol. 2004;61:413–421. doi: 10.5414/cnp61413. [DOI] [PubMed] [Google Scholar]
  • 78.Better O.S., Stein J.H. Early management of shock and prophylaxis of acute renal failure in traumatic rhabdomyolysis. N Engl J Med. 1990;322:825–829. doi: 10.1056/NEJM199003223221207. [DOI] [PubMed] [Google Scholar]
  • 79.Conrozier T., Mathieu P., Rinaudo M. Mannitol preserves the viscoelastic properties of hyaluronic acid in an in vitro model of oxidative stress. Rheumatol Ther. 2014;1:45–54. doi: 10.1007/s40744-014-0001-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Brown C.V.R., Rhee P., Chan L., et al. Preventing renal failure in patients with rhabdomyolysis: do bicarbonate and mannitol make a difference? J Trauma Inj Infect Crit Care. 2004;56:1191–1196. doi: 10.1097/01.ta.0000130761.78627.10. [DOI] [PubMed] [Google Scholar]
  • 81.Hoareau G.L., Beyer C.A., Kashtan H.W., et al. Improvised field expedient method for renal replacement therapy in a porcine model of acute kidney injury. Disaster Med Public Health Prep. 2021;15:741–749. doi: 10.1017/dmp.2020.107. [DOI] [PubMed] [Google Scholar]
  • 82.Hoareau G.L., Kashtan H., Walker L.E., et al. A novel perfusion system for damage control of hyperkalemia in swine. Shock. 2018;50:677–683. doi: 10.1097/SHK.0000000000001079. [DOI] [PubMed] [Google Scholar]
  • 83.Chawla L.S. Single lumen alternating micro-batch hemodiafiltration (SLAMB-HDF): A device for minimally invasive renal replacement therapy. Kidney360. 2020;1:969–973. doi: 10.34067/KID.0001462020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Clemens M.S., Stull M.C., Rall J.M., et al. Extracorporeal filtration of potassium in a swine model of bilateral hindlimb ischemia-reperfusion injury with severe acute hyperkalemia. Mil Med. 2018;183:e335–e340. doi: 10.1093/milmed/usy189. [DOI] [PubMed] [Google Scholar]
  • 85.Hoareau G.L., Beyer C.A., Wilson C., et al. Extracorporeal potassium binding for the management of hyperkalemia in an anephric model of crush injury. J Trauma Acute Care Surg. 2019;86:694–701. doi: 10.1097/TA.0000000000002178. [DOI] [PubMed] [Google Scholar]
  • 86.Ronco C., Reis T. Continuous renal replacement therapy and extended indications. Semin Dial. 2021;34:550–560. doi: 10.1111/sdi.12963. [DOI] [PubMed] [Google Scholar]
  • 87.Hoareau G.L., Beyer C.A., Walker L.E., et al. Renal replacement therapy capability for the treatment of combat-associated acute kidney injury: A historical perspective to plan for future conflicts. Mil Med. 2019;184:81–83. doi: 10.1093/milmed/usy221. [DOI] [PubMed] [Google Scholar]
  • 88.Thadhani R., Pascual M., Bonventre J.V. Acute renal failure. N Engl J Med. 1996;334:1448–1460. doi: 10.1056/NEJM199605303342207. [DOI] [PubMed] [Google Scholar]
  • 89.Hori Y., Aoki N., Kuwahara S., et al. Megalin blockade with cilastatin suppresses drug-induced nephrotoxicity. J Am Soc Nephrol. 2017;28:1783–1791. doi: 10.1681/ASN.2016060606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Matsushita K., Mori K., Saritas T., et al. Cilastatin ameliorates rhabdomyolysis-induced acute kidney injury in mice. J Am Soc Nephrol: ASN. 2021;32:2579–2594. doi: 10.1681/ASN.2020030263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Boutaud O., Moore K.P., Reeder B.J., et al. Acetaminophen inhibits hemoprotein-catalyzed lipid peroxidation and attenuates rhabdomyolysis-induced renal failure. Proc Natl Acad Sci U S A. 2010;107:2699–2704. doi: 10.1073/pnas.0910174107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Heard K.J. Acetylcysteine for acetaminophen poisoning. N Engl J Med. 2008;359:285–292. doi: 10.1056/NEJMct0708278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Shimizu M.H., Coimbra T.M., de Araujo M., et al. N-acetylcysteine attenuates the progression of chronic renal failure. Kidney Int. 2005;68:2208–2217. doi: 10.1111/j.1523-1755.2005.00677.x. [DOI] [PubMed] [Google Scholar]
  • 94.Rehman T., Fought J., Solomon R. N-acetylcysteine effect on serum creatinine and cystatin C levels in CKD patients. Clin J Am Soc Nephrol CJASN. 2008;3:1610–1614. doi: 10.2215/CJN.01560408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Mainra R., Gallo K., Moist L. Effect of N-acetylcysteine on renal function in patients with chronic kidney disease. Nephrology (Carlton) 2007;12:510–513. doi: 10.1111/j.1440-1797.2007.00833.x. [DOI] [PubMed] [Google Scholar]
  • 96.Mehta S.L., Kumari S., Mendelev N., Li P.A. Selenium preserves mitochondrial function, stimulates mitochondrial biogenesis, and reduces infarct volume after focal cerebral ischemia. BMC Neurosci. 2012;13:79. doi: 10.1186/1471-2202-13-79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Mendelev N., Mehta S.L., Idris H., et al. Selenite stimulates mitochondrial biogenesis signaling and enhances mitochondrial functional performance in murine hippocampal neuronal cells. PLoS One. 2012;7 doi: 10.1371/journal.pone.0047910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Shanu A., Groebler L., Kim H.B., et al. Selenium inhibits renal oxidation and inflammation but not acute kidney injury in an animal model of rhabdomyolysis. Antioxid Redox Signal. 2013;18:756–769. doi: 10.1089/ars.2012.4591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Panizo N., Rubio-Navarro A., Amaro-Villalobos J.M., et al. Molecular mechanisms and novel therapeutic approaches to rhabdomyolysis-induced acute kidney injury. Kidney Blood Press Res. 2015;40:520–532. doi: 10.1159/000368528. [DOI] [PubMed] [Google Scholar]
  • 100.Hebert J.F., Myatt L. Placental mitochondrial dysfunction with metabolic diseases: therapeutic approaches. Biochim Biophys Acta Mol Basis Dis. 2021;1867:165967. doi: 10.1016/j.bbadis.2020.165967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Dhimolea E. Canakinumab. mAbs. 2010;2:3–13. doi: 10.4161/mabs.2.1.10328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Anders H.J. Of inflammasomes and alarmins: il-1β and IL-1α in kidney disease. J Am Soc Nephrol. 2016;27:2564–2575. doi: 10.1681/ASN.2016020177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ridker P.M., MacFadyen J.G., Glynn R.J., et al. Inhibition of interleukin-1β by canakinumab and cardiovascular outcomes in patients with chronic kidney disease. J Am Coll Cardiol. 2018;71:2405–2414. doi: 10.1016/j.jacc.2018.03.490. [DOI] [PubMed] [Google Scholar]
  • 104.Sozeri B., Gulez N., Ergin M., Serdaroglu E. The experience of canakinumab in renal amyloidosis secondary to Familial Mediterranean fever. Mol Cell Pediatr. 2016;3:33. doi: 10.1186/s40348-016-0058-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Liu D., Jin F., Shu G., et al. Enhanced efficiency of mitochondria-targeted peptide SS-31 for acute kidney injury by pH-responsive and AKI-kidney targeted nanopolyplexes. Biomaterials. 2019;211:57–67. doi: 10.1016/j.biomaterials.2019.04.034. [DOI] [PubMed] [Google Scholar]
  • 106.Brinkmann V., Reichard U., Goosmann C., et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–1535. doi: 10.1126/science.1092385. [DOI] [PubMed] [Google Scholar]
  • 107.Villanueva E., Yalavarthi S., Berthier C.C., et al. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol: Baltimore, Md. 2011;187:538–552. doi: 10.4049/jimmunol.1100450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wolf D., Anto-Michel N., Blankenbach H., et al. A ligand-specific blockade of the integrin Mac-1 selectively targets pathologic inflammation while maintaining protective host-defense. Nat Commun. 2018;9:525. doi: 10.1038/s41467-018-02896-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Aliko A., Kamińska M., Falkowski K., et al. Discovery of novel potential reversible peptidyl arginine deiminase inhibitor. Int J Mol Sci. 2019;20:2174. doi: 10.3390/ijms20092174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Luo Y., Arita K., Bhatia M., et al. Inhibitors and inactivators of protein arginine deiminase 4: functional and structural characterization. Biochemistry. 2006;45:11727–11736. doi: 10.1021/bi061180d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Chiang C.-C., Korinek M., Cheng W.-J., Hwang T.-L. Targeting neutrophils to treat acute respiratory distress syndrome in coronavirus disease. Front Pharmacol. 2020;11:572009. doi: 10.3389/fphar.2020.572009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chalmers J.D., Haworth C.S., Metersky M.L., et al. Phase 2 trial of the DPP-1 inhibitor brensocatib in bronchiectasis. N Engl J Med. 2020;383:2127–2137. doi: 10.1056/NEJMoa2021713. [DOI] [PubMed] [Google Scholar]
  • 113.Patel D.D., Lee D.M., Kolbinger F., Antoni C. Effect of IL-17A blockade with secukinumab in autoimmune diseases. Ann Rheum Dis. 2013;72(suppl 2):ii116–ii123. doi: 10.1136/annrheumdis-2012-202371. [DOI] [PubMed] [Google Scholar]
  • 114.Klibanov A.L., Hossack J.A. Ultrasound in radiology: from anatomic, functional, molecular imaging to drug delivery and image-guided therapy. Invest Radiol. 2015;50:657–670. doi: 10.1097/RLI.0000000000000188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Ma Y., Cai F., Li Y., et al. A review of the application of nanoparticles in the diagnosis and treatment of chronic kidney disease. Bioact Mater. 2020;5:732–743. doi: 10.1016/j.bioactmat.2020.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Mauricio M.D., Guerra-Ojeda S., Marchio P., et al. Nanoparticles in medicine: A focus on vascular oxidative stress. Oxid Med Cell Longev. 2018;2018 doi: 10.1155/2018/6231482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Yu H., Jin F., Liu D., et al. ROS-responsive nano-drug delivery system combining mitochondria-targeting ceria nanoparticles with atorvastatin for acute kidney injury. Theranostics. 2020;10:2342–2357. doi: 10.7150/thno.40395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Yu H., Lin T., Chen W., et al. Size and temporal-dependent efficacy of oltipraz-loaded PLGA nanoparticles for treatment of acute kidney injury and fibrosis. Biomaterials. 2019;219:119368. doi: 10.1016/j.biomaterials.2019.119368. [DOI] [PubMed] [Google Scholar]
  • 119.Li H.L., Yan Z., Ke Z.P., et al. IGFBP2 is a potential biomarker in acute kidney injury (AKI) and resveratrol-loaded nanoparticles prevent AKI. Oncotarget. 2018;9:36551–36560. doi: 10.18632/oncotarget.25663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Deng X., Zeng T., Li J., et al. Kidney-targeted triptolide-encapsulated mesoscale nanoparticles for high-efficiency treatment of kidney injury. Biomater Sci. 2019;7:5312–5323. doi: 10.1039/c9bm01290g. [DOI] [PubMed] [Google Scholar]
  • 121.Kang H., Han M., Xue J., et al. Renal clearable nanochelators for iron overload therapy. Nat Commun. 2019;10:5134. doi: 10.1038/s41467-019-13143-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.AlBasher G., Alfarraj S., Alarifi S., et al. Nephroprotective role of selenium nanoparticles against glycerol-induced acute kidney injury in rats. Biol Trace Elem Res. 2020;194:444–454. doi: 10.1007/s12011-019-01793-5. [DOI] [PubMed] [Google Scholar]
  • 123.Chen X., Sun J., Li H., et al. Curcumin-loaded nanoparticles protect against rhabdomyolysis-induced acute kidney injury. Cell Physiol Biochem. 2017;43:2143–2154. doi: 10.1159/000484233. [DOI] [PubMed] [Google Scholar]
  • 124.Khajevand-Khazaei M.-R., Mohseni-Moghaddam P., Hosseini M., et al. Rutin, a quercetin glycoside, alleviates acute endotoxemic kidney injury in C57BL/6 mice via suppression of inflammation and up-regulation of antioxidants and SIRT1. Eur J Pharmacol. 2018;833:307–313. doi: 10.1016/j.ejphar.2018.06.019. [DOI] [PubMed] [Google Scholar]
  • 125.Sirsi S., Borden M. Microbubble compositions, properties and biomedical applications. Bubble Sci Eng Technol. 2009;1:3–17. doi: 10.1179/175889709X446507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hull T.D., Agarwal A., Hoyt K. New ultrasound techniques promise further advances in AKI and CKD. J Am Soc Nephrol. 2017;28:3452–3460. doi: 10.1681/ASN.2017060647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Huang S., Ren Y., Wang X., et al. Application of ultrasound-targeted microbubble destruction–mediated exogenous gene transfer in treating various renal diseases. Hum Gene Ther. 2018;30:127–138. doi: 10.1089/hum.2018.070. [DOI] [PubMed] [Google Scholar]
  • 128.Balgobin S., Morena M., Brunot V., et al. Continuous veno-venous high cut-off hemodialysis compared to continuous veno-venous hemodiafiltration in Intensive Care Unit acute kidney injury patients. Blood Purif. 2018;46:248–256. doi: 10.1159/000489082. [DOI] [PubMed] [Google Scholar]
  • 129.Dilken O., Ince C., van der Hoven B., et al. Successful reduction of creatine kinase and myoglobin levels in severe rhabdomyolysis using extracorporeal blood purification (CytoSorb®) Blood Purif. 2020;49:743–747. doi: 10.1159/000505899. [DOI] [PubMed] [Google Scholar]
  • 130.Scharf C., Liebchen U., Paal M., et al. Blood purification with a cytokine adsorber for the elimination of myoglobin in critically ill patients with severe rhabdomyolysis. Crit Care. 2021;25:41. doi: 10.1186/s13054-021-03468-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Knight J.S., Subramanian V., O’Dell A.A., et al. Peptidylarginine deiminase inhibition disrupts NET formation and protects against kidney, skin and vascular disease in lupus-prone MRL/lpr mice. Ann Rheum Dis. 2015;74:2199–2206. doi: 10.1136/annrheumdis-2014-205365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Blanco F.J., Möricke R., Dokoupilova E., et al. Secukinumab in active rheumatoid arthritis: A Phase III randomized, double-blind, active comparator- and placebo-controlled study. Arthritis Rheumatol. 2017;69:1144–1153. doi: 10.1002/art.40070. [DOI] [PubMed] [Google Scholar]
  • 133.Hung A.M., Ellis C.D., Shintani A., et al. IL-1β receptor antagonist reduces inflammation in hemodialysis patients. J Am Soc Nephrol. 2011;22:437–442. doi: 10.1681/ASN.2010070760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Nowak K.L., Chonchol M., Ikizler T.A., et al. IL-1 inhibition and vascular function in CKD. J Am Soc Nephrol. 2017;28:971–980. doi: 10.1681/ASN.2016040453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Perez-Gomez M.V., Sanchez-Niño M.D., Sanz A.B., et al. Horizon 2020 in diabetic kidney disease: the clinical trial pipeline for add-on therapies on top of renin angiotensin system blockade. J Clin Med. 2015;4:1325–1347. doi: 10.3390/jcm4061325. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Kidney International Reports are provided here courtesy of Elsevier

RESOURCES