Abstract
The histological substrate of many forms of intrinsic acute kidney injury (AKI) has been classically attributed to tubular necrosis. However, more recent studies indicate that necrosis is not the main form of cell death in AKI and that other forms such as apoptosis, regulated necrosis (i.e. necroptosis and parthanatos), autophagic cell death and mitotic catastrophe, also participate in AKI and that their contribution depends on the cause and stage of AKI. Herein, we briefly summarize the main characteristics of the major types of cell death and we also critically review the existing evidence on the occurrence of different types of cell death reported in the most common experimental models of AKI and human specimens. We also discuss the pathophysiological mechanisms linking tubule epithelial cell death with reduced glomerular filtration, azotaemia and hydroelectrolytic imbalance. For instance, special relevance is given to the analysis of the inflammatory component of some forms of cell death over that of others, as an important and differential pathophysiological determinant. Finally, known molecular mechanisms and signalling pathways involved in each cell death type pose appropriate targets to specifically prevent or reverse AKI, provided that further knowledge of their participation and repercussion in each AKI syndrome is progressively increased in the near future.
Keywords: apoptosis, autophagy, ferroptosis, necroptosis, pathophysiology
Acute kidney injury: an overview
Acute kidney injury (AKI) refers to a number of aetiologically different conditions suddenly resulting in decreased glomerular filtration, increased plasma creatinine and variably diminished urinary output [1]. AKI is a very serious condition from the sanitary and economic points of view. It is especially relevant in determined clinical circumstances, such as those related to patients in intensive care units, critically ill patients and patients with multiorgan failure. In these circumstances, mortality remains at 50–80% of cases [2]. Overall incidence of AKI is estimated at 1–2% of hospital admissions and 2–7% during hospital stay [2, 3]. Beyond acute consequences, AKI increases medium and long-term cardiovascular morbimortality, favours progression to chronic kidney disease, and is a cause of permanent dialysis dependence. AKI-associated cost poses 5% of hospital expenditure [4] and 1% of overall health expenditure [3] in developed countries. From the pathogenic point of view, AKI is often classified in three types: (i) pre-renal AKI; (ii) renal, parenchymal or intrinsic AKI; and (iii) post-renal or obstructive AKI. Insults may produce one of these three AKI types, although most frequently combined pathophysiological patterns result, which depend on the intensity, dose or exposure level of the insult and exposure time. Often, though, the situation is even more complex, as several insults may act simultaneously on the patient.
Pre-renal AKI accounts for 60–70% of all AKI cases [5]. Pre-renal syndromes develop with a transient increase of plasma creatinine that resolves by withdrawal of the cause and hydration. In pre-renal AKI, glomerular filtration decreases as a consequence of deregulated renal haemodynamics. This involves strong reduction in whole renal blood flow, reduction of intraglomerular pressure by altered (or overwhelmed) regulation of afferent and efferent arteriole synchronized contractility, or both. At least theoretically, pure pre-renal AKI courses with no injury to renal tissues, as it results merely from deregulated haemodynamics. Common causes of pre-renal AKI are severe hypotension (caused e.g. by surgical or traumatic blood loss, burns and mild sepsis), dehydration (as from vomiting, diarrhoea, bleeding or hypovolaemia), heart failure, liver failure, narrowing of renal arteries, renal microangiopathy, exposure to vasoactive drugs and toxins, and others [6]. It is uncertain whether purely pre-renal AKI is rife in the overall clinical AKI casuistry, the initial cause or simply a part of the aetiology of more complex syndromes also involving intrinsic damage, first because prolonged or severe pre-renal AKI may give rise to ischaemic scenarios leading to parenchymal damage, and second, because certain pre-renal AKI causes, such as drugs and toxins, may also cause parenchymal damage directly. Animal models of pre-renal AKI (Table 1) include (i) severe hypovolaemia (by exsanguination); (ii) drug-induced renal haemodynamic deregulation; (iii) hepatorenal syndrome (liver cirrhosis of any origin).
Table 1.
Major types of acute kidney injury in humans, including their major characteristics
| Human syndrome | Pre-renal/renal | Causes | Characteristics | Animal model |
|---|---|---|---|---|
| Pre-renal azotaemia | Pre-renal |
|
|
|
| Drug nephrotoxicity | Renal |
|
|
|
| Metal toxicity | Renal |
|
|
|
| CIN | Renal |
|
|
|
| Ischaemic AKI | Renal |
|
|
|
| Septic AKI | Renal |
|
|
|
| Rhabdomyolytic AKI | Renal |
|
|
|
Nephritis
|
Renal |
|
|
|
ATN, acute tubular necrosis; CIN, contrast-induced nephropathy; GMN, glomerulonephritis; ICN, iodinated contrast media; PN, pyelonephritis; TIN, tubulo-interstitial nephritis.
Unlike in pre-renal AKI, in renal and post-renal AKI, deadly or subdeadly alterations in renal structures, mostly involving renal tubuli, are the key pathophysiological element [5, 7]. The commonest form of renal AKI is acute tubular necrosis (ATN), characterized by tubular epithelial cell death and dysfunction in one or several tubular segments [8]. Tubular cell dysfunction, resulting from death or sublethal alterations, is the initiating event in ATN leading to renal dysfunction (i.e. reduced glomerular filtration rate, GFR) and renal failure. Different mechanisms link tubular damage with reduced glomerular filtration [9]: (i) tubular cell injury impedes appropriate tubular reuptake, which activates the tubuloglomerular feedback mechanism to reduce filtration and minimize hydroelectrolytic loss; (ii) tissue debris, dead cells and cell residues occlude renal tubules, which hampers filtration in obstructed nephrons and reduces overall glomerular filtration rate; (iii) tubular damage and cell death lead to activation of extant cells, which produce pro-inflammatory mediators and vasoactive cytokines. These, in turn, contribute to keep glomerular filtration low and to further amplify damage to different renal structures. Several rodent models of renal AKI exist (Table 1) that variably recreate the homologous human disease.
Post-renal AKI occurs upon occlusion of the ureters mostly from stones, cancer, trauma and congenital alterations. Ureteral occlusion leads to rapid degeneration and fibrosis of the occluded kidney. Unilateral occlusion develops with maintained renal function, whereas bilateral occlusion leads to dysfunction. In occluded kidneys, tubular cell death, tissue derangement and inflammation are the most important pathophysiological events. The popular animal model of post-renal AKI is recreated by temporary or irreversible, unilateral ureteral ligation [10].
The degree to which renal models reproduce human diseases is heterogeneous and variable. Ischaemic and toxic models often show strong parenchymal damage, which is not consistently observed to the same extent in the limited histopathological information obtained from humans, even for a similar degree of renal dysfunction [11]. It has been suggested that milder damage resulting from accumulation of sublethal comorbidities may more accurately model human AKI in laboratory animals [11]. Yet, knowledge from animal models has, with limitations, been useful for progressively increasing our understanding of simple and complex human conditions. Because the stronger the parenchymal damage the worse the prognosis, better knowledge of tubule cell death is required. In the last four decades, different forms of cell death have been identified with individual phenotypic and biochemical characteristics, and with yet uncertain or not fully unveiled biological and pathophysiological meaning. It is also uncertain whether some or many of these cell modes are epiphenomenons of the same process yielding to different appearances depending on the circumstances and type of insult or intracellular injury site, or whether they constitute truly differentiated processes. In this article we critically review the evidence existing on the occurrence of different cell modes in different experimental models and clinical studies of homologous or similar circumstances, and their pathophysiological importance and mechanisms.
Summary of cell death modes: differential mechanisms, signalling and characteristics
Since the first descriptions of programmed cell death mechanisms in the mid-1960s, many attempts have been made to classify cell death forms and their physiological and pathological consequences. The first classifications of cell death were based on the morphological characteristics of the dying cells. When biochemical pathways and genes involved in cell death started to be described, the classification of cell death types was based also on biochemical and molecular criteria (Table 2 and Figure 1). Because there are many classifications of cell death relying on different criteria, we have chosen to use in the present review the last published recommendations of The Nomenclature Committee on Cell Death (NCCD) [12]. This classification applies to both in vitro and in vivo settings and includes apoptosis, regulated necrosis, autophagic cell death and mitotic catastrophe, as well as some other types of cell death such as anoikis, entosis, NETosis, parthanatos, ferroptosis, and pyroptosis (Table 2 and Figure 1).
Table 2.
Types of cell death and criteria to define these types
| Criteria | Types of cell death |
|---|---|
| Morphological |
|
| Digestion |
|
| Enzymatic dependency |
|
| Functional meaning |
|
| Immunological |
|
| Caspase-independent intrinsic apoptosis |
|
| Anoikis |
|
| Pyroptosis |
|
| Regulated necrosis |
|
Fig. 1.
Schematic representation of the main signalling pathways of different cell death modes, and the pathophysiological mechanisms activated by cell death modes in acute kidney injury.
Apoptosis
Apoptosis is the collapse of a cell through an active, highly regulated process requiring metabolic activity by the dying cell, and characterized by membrane blebbing, cell shrinkage, chromatin condensation and DNA fragmentation, followed by rapid engulfment of the corpse by neighbouring cells, without rupture of the cell membrane [13]. Activation of executioner caspases, a family of cysteine proteases, is necessary to complete this process. The term apoptosis is often used interchangeably with programmed cell death. However, in the strictest sense, programmed cell death may be applied to other forms of cell death that require gene expression without fulfilling some of the morphological criteria of apoptosis. [24]. The signalling mechanisms leading to cell death by apoptosis have been extensively reviewed recently [25, 26]. Apoptosis can be divided into intrinsic or extrinsic apoptosis, depending on the main origin of the first signal inducing the cell death.
Extrinsic apoptosis
Typically, extrinsic apoptosis is initiated when ligands such as FAS/CD95 ligand (FASL/CD95L), tumour necrosis factor α (TNFα) or TNF-related apoptosis-inducing ligand (TRAIL) bind to various transmembrane death receptors, namely FAS/CD95, TNFα receptor 1 (TNFR1) and TRAIL receptor (TRAILR)1–2, respectively [27]. When death receptors are activated, apoptosis is induced by a complex cascade of signalling pathways (recently reviewed in [25]) leading to the activation of initiator caspases (mainly caspases 8 and 10), and subsequently effector or executioner caspases (caspase 3,6,7) [28]. Extrinsic apoptosis can also be initiated in the absence of ligands, by oligomerization of death receptors.
Intrinsic apoptosis
Many intracellular stress circumstances, including DNA damage, oxidative stress, cytosolic Ca2+ overload, accumulation of unfolded proteins in the endoplasmic reticulum and many others may also activate apoptosis. Different signalling cascades distinctly initiated at specific cell locations, converge on mitochondria to activate a common mechanism of intrinsic apoptosis [29]. When lethal signals prevail, mitochondrial outer membrane permeabilization (MOMP) occurs which leads to mitochondrial transmembrane potential (ΔΨm) dissipation and inhibition of mitochondrial ATP synthesis and Dcm-dependent transport activities. The respiratory chain becomes uncoupled, leading to toxic overproduction of reactive oxygen species (ROS). Also, proteins that are normally confined within the mitochondrial intermembrane space (IMS) are released into the cytosol [25]. Most significantly, cytochrome c, once in the cytosol, binds to apoptosis protease-activating factor-1 (apaf-1) to recruit and activate initiator caspase 9. Once activated, caspase 9 activates executioner caspases and unleashes apoptosis.
Anoikis
Anoikis describes the apoptotic cell death of adherent cells in response to loss of cell-to-matrix interactions [30]. In most cases, the cell death programme triggered by anoikis is the same as described for intrinsic apoptosis [31].
Pyroptosis
Pyroptosis is a form of programmed cell death associated with antimicrobial responses during inflammation. In this process, immune cells that recognize several intracellular danger signals produce cytokines, particularly interleukin-1β (IL-1β) and IL-18, swell, burst and finally die. Pyroptotic cells can exhibit apoptotic and/or necrotic morphological features [32]. The most distinctive biochemical feature of pyroptosis is the early activation of caspase-1, which mediates the proteolytic activation of caspase-7 (rather than that of caspase-3) [33]. It is not clear whether pyroptosis is a specific form of cell death or whether it represents a particular case of caspase-dependent intrinsic apoptosis.
Regulated necrosis
Necrotic cell death is characterized by cytoplasmic and organelle swelling, followed by the loss of cell membrane integrity and release of the cellular contents into the surrounding extracellular space, that produces an inflammatory response within the tissue. Necrosis has been considered for a long time as an accidental, uncontrolled form of cell death lacking underlying signalling events. However, there is now a general agreement that necrosis can occur in a regulated manner, and that necrotic cell death has a prominent role in multiple physiological situations [34]. Several triggers can induce regulated necrosis, including alkylating DNA damage, excitotoxins and the activation of death receptors, at least in some circumstances [35]. Two special forms of regulated necrosis are necroptosis and parthanatos.
Necroptosis
Necroptosis is sometimes used as a synonym of regulated necrosis, but it was originally introduced to indicate a specific case or regulated necrosis, which is started by TNFR1 ligation and can be inhibited by the RIP1-targeting chemical necrostatin-1. Since then, an assortment of necroptosis triggers have been identified, such as FAS/CD95, TRAILR (TNF-related apoptosis-inducing ligand receptor), TLR3/4 (Toll-like receptor), etoposide and IRI (ischaemia-reperfusion injury) [102]. Necroptosis occurs as a consequence of death receptor signalling upon formation of the RIPK1/RIPK3/mixed lineage kinase domain-like protein (MLKL) containing necroptosome [34, 103]. RIPK3 is activated by phosphorylation and in turn, phosphorylates the pseudokinase MLKL, which has been suggested to be involved in the plasma-membrane rupture [102, 104]. A second necroptotic pathway involves the opening of the mitochondria permeability transition (MPT) pore. Upon MPT pore opening, along with apoptosome-forming proteins, other proteins are released from mitochondria that exert specific events contributing to the apoptotic phenotype in a caspase-independent manner. They include endonucleases such as apoptosis-inducing factor (AIF) and endonuclease G, which degrade nuclear DNA [12]. Under apoptosis inhibition conditions, the MPT pore pathway leads to a necroptotic cell death mode.
Parthanatos
Parthanatos is a caspase-independent cell death mode involving the DNA damage-responsive enzymes poly(ADP-ribose) polymerases (PARPs), and in particular PARP1 [105]. In physiological conditions, PARP1 cooperates with the DNA repair machinery to ensure genomic homeostasis upon mild DNA damage but PARP1 overactivation has toxic consequences, including NAD+ and ATP depletion, as well as the accumulation of mitochondrial-toxic PAR, which favours ΔΨm dissipation and AIF release [106, 107].
Ferroptosis
Ferroptosis is a type of cell death characterized by iron dependence, as this type of cell death is also inhibited by the iron chelator, deferoxamine [108], and iron-dependent accumulation of lipid peroxides [104, 109]. Although the aetiology of the iron-dependency of ferroptosis is not yet known, cellular iron may be the most important factor in lipid peroxide generation during ferroptosis. Lipid peroxidation and ferroptosis are inhibited physiologically by antioxidant mechanisms including glutathione peroxidase 4 (GPX4), an enzyme, whose function depends on the glu/cys antiporter in the plasma membrane known as system Xc- [108, 110]. Ferroptosis can be inhibited ‘in vitro’ by ferrostatin-1 (Fer-1), a synthetic, potent antioxidant molecule [108, 109, 111]. Morphologically, ferroptosis is characterized by the presence of small mitochondria with condensed membrane densities, and is not associated with chromatin condensation, plasma membrane rupture, swelling of cytoplasmic organelles, or the formation of cytoplasmic vesicles/vacuoles [108].
Autophagic cell death
Autophagic cell death is characterized by massive cytoplasmic vacuolization suggesting that autophagy would actually execute cell death [112]. However, in most cases autophagy is a cytoprotective response activated by dying cells in the attempt to manage cell stress, and its inhibition accelerates, rather than prevents, cell death [113]. Thus, the term autophagic cell death should be used when death can be suppressed by the inhibition of autophagia [114, 115].
Mitotic catastrophe
Mitotic catastrophe is a mechanism of cell death initiated by perturbations of the mitotic apparatus during the M phase of the cell cycle and that is paralleled by some degree of mitotic arrest and ultimately causes cell death or senescence [116].
Entosis
Entosis is a cell death mode that occurs in epithelial cells linked to the invasion of one living cell into another homotypic or heterotypic cell [117], a phenomenon also called cell cannibalism [118]. In most cases, internalized cells appear virtually normal and later disappear, as they are degraded by lysosomal hydrolases. Entosis would be provoked by the loss of ECM interaction, but, in contrast to anoikis, it does not involve the activation of executioner caspases.
NETosis
NETosis is a regulated form of necrosis that is restricted to immune cells like neutrophils (NETosis) and other granulocytes or macrophages (then called ETosis). Pro-inflammatory cytokines such as IL-8 or TNF-α activate neutrophils to undergo a regulated cell death that spreads all chromatin outside the cells in a net-like structure (NETs = neutrophil extracellular traps). TLR2, TLR4, complement, and platelet activation all trigger NETosis whereas increased reactive oxygen species production facilitates NETosis [119]. NETosis spreads histones out of the cell at sites of infection as well as in sterile inflammation. In a similar process, dying renal tubular epithelial cells release histones locally, which promote microvascular and parenchymal injury [119].
Evidence of different cell death modes in animal models and human types of acute kidney injury
Table 3 shows selected references reporting the best evidence found in the literature on the occurrence of distinct modes of cell death in different types of AKI types. Many other studies report evidence for different modes of cell death in many models of AKI. In general, in vivo evidence for specific death phenotypes in animal models and human biopsy material is scarce, weak and superficial. Very few studies convincingly determine the mode or modes of cell death occurring in the kidneys. The two most reported and best documented cell death modes in vivo are necrosis and apoptosis followed distantly by autophagic death. A handicap for in vivo determination of the cell mode is that it is difficult to obtain a body of manifold evidence provided by morphological, biochemical and molecular details. This manifold body of evidence is necessary to unambiguously determine the mode of cell death, because single pieces of evidence are not exclusive to one mode of cell death. Studies are very heterogeneous in their way of addressing the study of cell death mode. They rarely combine morphological, with biochemical and signalling information, but mostly concentrate on one or two of these aspects, typically with superficial probatory depth.
Table 3:
Types of cell death involved in the most frequently used experimental models of Acute Kidney Injury
![]() |
When epithelial cell necrosis is reported, no proof of true ‘cell necrosis’ is given in most studies, but deduced from simple histological observations. The situation with necrosis is complicated for several reasons. First, because of the initial association between cell death and necrosis, when (before the 1970s) only one form of cell death was known, and termed necrosis. Second, because at that time the pathological pattern characterized by tubule epithelial cell death in acute cases of renal damage, was logically termed ‘acute tubular necrosis’ in the 1940–50s [120]. The first new mode of cell death identified different from necrosis was apoptosis [13]. Many articles produced in that intermediate period consolidated the association between ATN and cell necrosis, and both were mistakenly used somewhat indistinctly in many publications, even to the present. Third, the term necrosis has been widened in the last decade, when it was progressively realized that specific forms of necrotic-seeming phenotypes were in fact the result of specific cellular death programmes [12]. The first consequence was the distinction between passive and programmed necrosis (or necroptosis). In vivo, this is complicated, as necroptosis shares signalling pathways with other forms of death [121]. In vivo, phenotypical evaluation of cell morphology within tissue architecture is less explicit than that in cell cultures, with isolated cells.
Apoptosis is also poorly documented in numerous in vivo studies. In many papers, there is a mixture of a few in vivo data and many in vitro measurements, and conclusions of what occurs in vivo are derived from this mixture of results. However, this probatory argument is weak. Cellular models, both primary cultures of renal cells and renal cell lines, are to an undetermined extent phenotypically adrift from their physiological and pathophysiological condition. In addition, they are devoid of undetermined conditionings and determinants only found in vivo. Both differences make in vitro findings ambiguously predictive of in vivo events. A rich body of experimental tools exists to study and manipulate cell models in order to evidence apoptosis. However, in vivo, most of these techniques are limited by the need to study whole kidneys for specific molecular events (as from tissue extracts), or to study events on histological samples that provide localization evidence, but more ambiguous molecular specificity. Detection of DNA nicks by terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) is one of the most used tools to study apoptosis in vivo, as DNA fragmentation is considered a hallmark of apoptosis. However, even this signature mark of apoptosis has proved relatively unspecific, since TUNEL-positive cells can also be observed in cells undergoing necroptosis [122]. This exemplifies the need for multifactorial evidence and stresses the difficulty of assessing cell death mode in vivo.
Autophagic cell death has even been put into question [123]. A debate exists over whether it constitutes a distinct cell death programme, or merely a defence mechanism of protection from stress that not only does not contribute to cell demise, but rather its inhibition accelerates cell death [124]. Similarly to necrosis and apoptosis, its demonstration in vivo is a difficult task. To this difficulty, the uncertainty on its pathophysiological role must be added, when marks of autophagia (i.e. cell vacuolation, activation of specific signalling pathways, etc.) are detected in vivo. Other forms of cell death are even less well described or more doubts are cast on their existence as independent forms of cell death or as epiphenomenons of necrosis or apoptosis.
As a conclusion, sound evidence of most if not all cell death modes is available in renal cellular models, but very weak and low level evidence is available from in vivo studies and human tissue. Clearly, more focused investigation is necessary to prove the occurrence of specific modes of cell death in different AKI scenarios, their extent in each pathological circumstance and, thus, their repercussion in the overall pathological process. Finally, beyond understanding the pathophysiological role of cell death modes in AKI, markers of these processes in easy-to-obtain biological samples will contribute to perform a progressively more accurate etiopathogenic diagnosis of individual AKI episodes.
Pathophysiological role, significance and repercussion of different cell death modes in acute kidney injury
Many forms of AKI are characterized by tubular epithelial cell death [125], which is a cause of tubular dysfunction and tubular activation. Tubular dysfunction activates the tubuloglomerular feedback mechanism, which reduces glomerular filtration to prevent water and electrolyte loss. Tubular cell death also activates extant cells to proliferate and replace dead cells and to attract immune system cells to aid in repair. Tubule cell activation involves the production of pro-inflammatory and vasoactive mediators (cytokines, reactive oxygen species, etc.) which act in a paracrine and autocrine manner to contract mesangial cells and arterioles, to maintain filtration low. In addition, cell debris also produces tubular obstruction in more distal segments of the nephron, thus reducing glomerular filtration rate and the excretory capacity of the kidney [9].
In addition, tubule cell death is also a cause of further damage, which closes a vicious degenerative circle of injury amplification [9, 126]. Various forms of cell death have been reported to occur simultaneously to different relative and absolute extent depending on AKI type (ischaemic, toxic, septic, etc.), the stage of injury and the level of repair. Each form of cell death produces specific functional and histological consequences. An important reason for these differences is that various cell death modes distinctly stimulate the innate immune and inflammatory responses [127]. Different cell death modes result in a different degree of release of the so-called damage-associated molecular patterns (DAMPs), which stimulate and amplify inflammation and tissue damage [128].
The most studied and best described forms of cell death in AKI are apoptosis and necrosis, but several forms of regulated necrosis and autophagic death have been also described recently. As a general concept, apoptosis results in less aggravation of renal function than necrosis, because apoptosis is far less immunogenic than necrosis [127]. This is because, during apoptosis, release of the cellular content to the medium is limited. In vivo, apoptotic cells and their membrane-bound remains (apoptotic bodies) are rapidly removed by phagocytic and neighbouring cells [129]. In contrast, in necrosis and necroptosis, the cell membrane is broken and the cell content is released to the medium [130]. Necrotic debris and poured intracellular content attract immune system cells with the result of increased release of inflammatory cytokines and increased production of reactive oxygen species, all in turn resulting in subsequent or further renal damage [9, 126, 131] (Figure 1). Thus, any manoeuvre that prevents cell death by necrosis, even by transforming it into apoptosis, should have beneficial results for the severity of AKI. However, it should also be noted that the apoptotic signalling pathway, when intercepted or in absence of enough energy supply, may be diverted to necrosis [132].
Pyroptosis is a necrotic-like cell death mode that was thought to occur exclusively in macrophages and leucocytes, but that it has been also described in tubular epithelial cell [133–135]. The major characteristic of pyroptosis compared with other pathways of regulated necrosis is the maturation of pro-inflammatory cytokines such as IL-1 β and IL-18 during the cell death process. Cytokine maturation is cleavage-dependent, and it is mediated by non-apoptotic caspases such as caspase-1 [136]. Membrane rupture leads to the release of these cytokines to the interstitium, inflammation and worsened AKI. Then, pyroptosis shows maximal immunogenic effect among the different types of necrosis. Its role in AKI and its underlying signalling pathways should be further investigated with therapeutic aims.
Therapeutic perspectives and conclusions
There is reasonably solid evidence that apoptosis and necrosis (i.e. necrotic-looking) is involved in the pathophysiology of AKI. Although some evidence that other modes of cell death may take place in the diseased kidneys of AKI, more focused research is necessary to both clearly define distinct cell death modes in vivo (including necrosis and apoptosis), and their implication in AKI pathophysiology. Unravelling the occurrence of different cell death modes present in each AKI case, their pathological consequences and the underlying mechanisms and signalling pathways is crucial for a specific and personalized therapy and handling, and a research endeavour for the immediate future [137]. Furthermore, new diagnostic tools in the form of more sensitive imaging techniques and cell death mode-specific biomarkers are needed for an effective theranostic approach to AKI.
Despite the participation of apoptosis in AKI, apoptosis inhibition as a therapy for AKI has been questioned because caspase inhibitors are not successful in improving AKI development and outcome [103]. Despite being the core enzymes responsible for the apoptotic phenotype, caspase activation is a downstream level of apoptosis (in the execution phase). More upstream, initiating apoptotic events, such as mitochondria targeting, result in inevitable cell death [138].
After the discovery of necroptosis and necrostatin-1 (Nec-1) as an inhibitor of necroptosis [139], new strategies were designed for AKI treatment. It was therefore disappointing to realize that Nec-1 could only partially protect from ischaemic AKI [140]. Furthermore, in necroptosis, plasma membrane rupture occurs as early as 20 min after RIPK3 dimerization [141], and administration of Nec-1, 30 min after the beginning of reperfusion, has no detectable protective effect [140]. Therefore, targeting regulated necrosis may be limited to such disorders in which AKI may be anticipated, like heart surgery-associated AKI, contrast-induced AKI, or kidney transplantation.
Regarding other cell death modes, pyroptosis may be targeted by caspase inhibitors or cytokine response modifier A [142]. In ferroptosis, ferrostatin-1 has been demonstrated to be useful in reducing AKI [111]. The utility of inhibiting these events is yet uncertain. In addition, targeting autophagy results in impairment of renal function in several models of AKI [100].
Thus, therapy that combines apoptotic, necroptotic and ferroptotic inhibitors might be potentially useful, but more research is necessary. Finally, translation of such results into clinical trials is highly problematic. Control groups are required for any single- and double-therapeutic strategy, and support of such studies might become long-winded in the absence of strong, convincing preclinical evidence.
Conflict of interest statement
None declared.
References
- 1.Thomas ME, Blaine C, Dawnay A, et al. The definition of acute kidney injury and its use in practice. Kidney Int 2015; 87: 62–73 [DOI] [PubMed] [Google Scholar]
- 2.Waikar SS, Liu KD, Chertow GM. Diagnosis, epidemiology and outcomes of acute kidney injury. Clin J Am Soc Nephrol 2008; 3: 844–861 [DOI] [PubMed] [Google Scholar]
- 3.Kerr M, Bedford M, Matthews B, et al. The economic impact of acute kidney injury in England. Nephrol Dial Transplant 2014; 29: 1362–1368 [DOI] [PubMed] [Google Scholar]
- 4.Vandijck DM, Oeyen S, Decruyenaere JM, et al. Acute kidney injury, length of stay, and costs in patients hospitalized in the intensive care unit. Acta Clin Belg Suppl 2007: 341–345 [DOI] [PubMed] [Google Scholar]
- 5.Kaufman J, Dhakal M, Patel B, et al. Community-acquired acute renal failure. Am J Kidney Dis 1991; 17: 191–198 [DOI] [PubMed] [Google Scholar]
- 6.Koyner JL, Garg AX, Thiessen-Philbrook H, et al. TRIBE-AKI Consortium. Adjudication of etiology of acute kidney injury: experience from the TRIBE-AKI multi-center study. BMC Nephrol 2014; 15: 105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Uchino S. The meaning of transient azotemia. Contrib Nephrol 2010; 165: 337–344 [DOI] [PubMed] [Google Scholar]
- 8.Endre ZH, Kellum JA, Di Somma S, et al. Differential diagnosis of AKI in clinical practice by functional and damage biomarkers: workgroup statements from the tenth Acute Dialysis Quality Initiative Consensus Conference. Contrib Nephrol 2013; 182: 30–44 [DOI] [PubMed] [Google Scholar]
- 9.Lopez-Novoa JM, Quiros Y, Vicente L, et al. New insights into the mechanism of aminoglycoside nephrotoxicity: an integrative point of view. Kidney Int 2011; 79: 33–45 [DOI] [PubMed] [Google Scholar]
- 10.Ucero AC, Benito-Martin A, Izquierdo MC, et al. Unilateral ureteral obstruction: beyond obstruction. Int Urol Nephrol 2014; 46: 765–776 [DOI] [PubMed] [Google Scholar]
- 11.Heyman SN, Rosenberger C, Rosen S. Acute kidney injury: lessons from experimental models. Contrib Nephrol 2011; 169: 286–296 [DOI] [PubMed] [Google Scholar]
- 12.Galluzzi L, Bravo-San Pedro JM, Vitale I, et al. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015. Cell Death Differ 2015; 22: 58–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 1972; 26: 239–257 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Efrati S, Berman S, Ben Aharon G, et al. Application of normobaric hyperoxia therapy for amelioration of haemorrhagic shock-induced acute renal failure. Nephrol Dial Transplant 2008; 23: 2213–2222 [DOI] [PubMed] [Google Scholar]
- 15.Jackson EK, Kost CK, Jr, Herzer WA, et al. A(1) receptor blockade induces natriuresis with a favorable renal hemodynamic profile in SHHF/Mcc-fa(cp) rats chronically treated with salt and furosemide. J Pharmacol Exp Ther 2001; 299: 978–987 [PubMed] [Google Scholar]
- 16.Ferreira L, Quiros Y, Sancho-Martínez SM, et al. Urinary levels of regenerating islet-derived protein III β and gelsolin differentiate gentamicin from cisplatin-induced acute kidney injury in rats. Kidney Int 2011; 79: 518–528 [DOI] [PubMed] [Google Scholar]
- 17.Ohmachi Y, Imamura T, Ikeda M, et al. Sodium bicarbonate protects uranium-induced acute nephrotoxicity through uranium-decorporation by urinary alkalinization in rats. J Toxicol Pathol 2015; 28: 65–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Quiros Y, Ferreira L, Sancho-Martínez SM, et al. Sub-nephrotoxic doses of gentamicin predispose animals to developing acute kidney injury and to excrete ganglioside M2 activator protein. Kidney Int 2010; 78: 1006–1015 [DOI] [PubMed] [Google Scholar]
- 19.Speir RW, Stallings JD, Andrews JM, et al. Effects of valproic acid and dexamethasone administration on early bio-markers and gene expression profile in acute kidney ischemia-reperfusion injury in the rat. PLoS One 2015; 10: e0126622 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Seely KA, Holthoff JH, Burns ST, et al. Hemodynamic changes in the kidney in a pediatric rat model of sepsis-induced acute kidney injury. Am J Physiol Renal Physiol 2011; 301: F209–F217 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Nakamura A, Niimi R, Yanagawa Y. Protection from sepsis-induced acute renal failure by adenoviral-mediated gene transfer of beta2-adrenoceptor. Nephrol Dial Transplant 2010; 25: 730–737 [DOI] [PubMed] [Google Scholar]
- 22.de Jesus Soares T, Volpini RA, Francescato HD, et al. Effects of resveratrol on glycerol-induced renal injury. Life Sci 2007; 81: 647–656 [DOI] [PubMed] [Google Scholar]
- 23.Wan B, Hao L, Qiu Y, et al. Blocking tumor necrosis factor-alpha inhibits folic acid-induced acute renal failure. Exp Mol Pathol 2006; 81: 211–216 [DOI] [PubMed] [Google Scholar]
- 24.Sperandio S, de Belle I, Bredesen DE. An alternative, nonapoptotic form of programmed cell death. Proc Natl Acad Sci USA 2000; 97: 14376–14381 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kroemer G, Galluzzi L, Brenner C. Mitochondrial membrane permeabilization in cell death. Physiol Rev 2007; 87: 99–163 [DOI] [PubMed] [Google Scholar]
- 26.Galluzzi L, Joza N, Tasdemir E, et al. No death without life: vital functions of apoptotic effectors. Cell Death Differ 2008; 15: 1113–1123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Schutze S, Tchikov V, Schneider-Brachert W. Regulation of TNFR1 and CD95 signalling by receptor compartmentalization. Nat Rev Mol Cell Biol 2008; 9: 655–662 [DOI] [PubMed] [Google Scholar]
- 28.Riedl SJ, Shi Y. Molecular mechanisms of caspase regulation during apoptosis. Nat Rev Mol Cell Biol 2004; 5: 897–907 [DOI] [PubMed] [Google Scholar]
- 29.Kroemer G, Galluzzi L, Vandenabeele P, et al. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ 2009; 16: 3–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Frisch SM, Francis H. Disruption of epithelial cell-matrix interactions induces apoptosis. J Cell Biol 1994; 124: 619–626 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Reginato MJ, Mills KR, Paulus JK, et al. Integrins and EGFR coordinately regulate the pro-apoptotic protein Bim to prevent anoikis. Nat Cell Biol 2003; 5: 733–740 [DOI] [PubMed] [Google Scholar]
- 32.Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol 2009; 7: 99–109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Rintahaka J, Lietzen N, Ohman T, et al. Recognition of cytoplasmic RNA results in cathepsin-dependent inflammasome activation and apoptosis in human macrophages. J Immunol 2011; 186: 3085–3092 [DOI] [PubMed] [Google Scholar]
- 34.Vandenabeele P, Galluzzi L, Vanden Berghe T, et al. Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol 2010; 11: 700–714 [DOI] [PubMed] [Google Scholar]
- 35.He S, Wang L, Miao L, et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell 2009; 137: 1100–1111 [DOI] [PubMed] [Google Scholar]
- 36.Lee IC, Kim SH, Lee SM, et al. Melatonin attenuates gentamicin-induced nephrotoxicity and oxidative stress in rats. Arch Toxicol 2012; 86: 1527–1536 [DOI] [PubMed] [Google Scholar]
- 37.El Mouedden M, Laurent G, Mingeot-Leclercq MP, et al. Apoptosis in renal proximal tubules of rats treated with low doses of aminoglycosides. Antimicrob Agents Chemother 2000; 44: 665–675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Martínez-Salgado C, Eleno N, Morales AI, et al. Gentamicin treatment induces simultaneous mesangial proliferation and apoptosis in rats. Kidney Int 2004; 65: 2161–2171 [DOI] [PubMed] [Google Scholar]
- 39.Sahu BD, Tatireddy S, Koneru M, et al. Naringin ameliorates gentamicin-induced nephrotoxicity and associated mitochondrial dysfunction, apoptosis and inflammation in rats: possible mechanism of nephroprotection. Toxicol Appl Pharmacol 2014; 277: 8–20 [DOI] [PubMed] [Google Scholar]
- 40.Megyesi J, Safirstein RL, Price PM. Induction of p21WAF1/CIP1/SDI1 in kidney tubule cells affects the course of cisplatin-induced acute renal failure. J Clin Invest 1998; 101: 777–782 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wei Q, Dong G, Franklin J, et al. The pathological role of Bax in cisplatin nephrotoxicity. Kidney Int 2007; 72: 53–62 [DOI] [PubMed] [Google Scholar]
- 42.Sheikh-Hamad D, Cacini W, Buckley AR, et al. Cellular and molecular studies on cisplatin-induced apoptotic cell death in rat kidney. Arch Toxicol 2004; 78: 147–155 [DOI] [PubMed] [Google Scholar]
- 43.Li S, Basnakian A, Bhatt R, et al. PPAR-alpha ligand ameliorates acute renal failure by reducing cisplatin-induced increased expression of renal endonuclease G. Am J Physiol Renal Physiol 2004; 287: F990–F998 [DOI] [PubMed] [Google Scholar]
- 44.Wei Q, Dong G, Yang T, et al. Activation and involvement of p53 in cisplatin-induced nephrotoxicity. Am J Physiol Renal Physiol 2007; 293: F1282–F1291 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Tsuruya K, Ninomiya T, Tokumoto M, et al. Direct involvement of the receptor-mediated apoptotic pathways in cisplatin-induced renal tubular cell death. Kidney Int 2003; 63: 72–82 [DOI] [PubMed] [Google Scholar]
- 46.Zhou Y, Xu H, Xu W, et al. Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res Ther 2013; 4: 34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wu CH, Chen AT, Yen GY. Protective effects of glycyrrhizic acid and 18β-glycyrrhetinic acid against cisplatin-induced nephrotoxicity in BALB/c mice. J Agric Food Chem 2015; 63: 1200–1209 [DOI] [PubMed] [Google Scholar]
- 48.Lee HC, Chang JG, Yen HW, et al. Ionic contrast media induced more apoptosis in diabetic kidney than nonionic contrast media. J Nephrol 2011; 24: 376–380 [DOI] [PubMed] [Google Scholar]
- 49.Liu TQ, Luo WL, Tan X, et al. A novel contrast-induced acute kidney injury model based on the 5/6-nephrectomy rat and nephrotoxicological evaluation of iohexol and iodixanol in vivo. Oxid Med Cell Longev 2014; 2014: 427560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hsu SP, Tsai TJ, Chien CT. Ioxitalamate induces renal tubular apoptosis via activation of renal efferent nerve-mediated adrenergic signaling, renin activity, and reactive oxygen species production in rats. Toxicol Sci 2010; 114: 149–158 [DOI] [PubMed] [Google Scholar]
- 51.Buyuklu M, Kandemir FM, Ozkaraca M, et al. Benefical effects of lycopene against contrast medium-induced oxidative stress, inflammation, autophagy, and apoptosis in rat kidney. Hum Exp Toxicol 2015; 34: 487–496 [DOI] [PubMed] [Google Scholar]
- 52.Zhao Y, Tao Z, Xu Z, et al. Toxic effects of a high dose of non-ionic iodinated contrast media on renal glomerular and aortic endothelial cells in aged rats in vivo. Toxicol Lett 2011; 202: 253–260 [DOI] [PubMed] [Google Scholar]
- 53.Jiang G, Wang M, Wang L, et al. The protective effect of nesfatin-1 against renal ischemia-reperfusion injury in rats. Ren Fail 2015; 1–8 [DOI] [PubMed] [Google Scholar]
- 54.Zang XJ, An SX, Feng Z, et al. In vivo mechanism study of NGAL in rat renal ischemia-reperfusion injury. Genet Mol Res 2014; 13: 8740–8748 [DOI] [PubMed] [Google Scholar]
- 55.An S, Zang X, Yuan W, et al. Neutrophil gelatinase-associated lipocalin (NGAL) may play a protective role against rats ischemia/reperfusion renal injury via inhibiting tubular epithelial cell apoptosis. Ren Fail 2013; 35: 143–149 [DOI] [PubMed] [Google Scholar]
- 56.Yoshida T, Shimizu A, Masuda Y, et al. Caspase-3-independent internucleosomal DNA fragmentation in ischemic acute kidney injury. Nephron Exp Nephrol 2012; 120: e103–e113 [DOI] [PubMed] [Google Scholar]
- 57.Messaris E, Memos N, Chatzigianni E, et al. Apoptotic death of renal tubular cells in experimental sepsis. Surg Infect (Larchmt) 2008; 9: 377–388 [DOI] [PubMed] [Google Scholar]
- 58.Stoyanoff TR, Todaro JS, Aguirre MV, et al. Amelioration of lipopolysaccharide-induced acute kidney injury by erythropoietin: involvement of mitochondria-regulated apoptosis. Toxicology 2014; 318: 13–21 [DOI] [PubMed] [Google Scholar]
- 59.Nakajima Y, Mikami O, Yoshioka M, et al. Involvement of apoptosis in the endotoxemic lesions of the liver and kidneys of piglets. J Vet Med Sci 2000; 62: 621–626 [DOI] [PubMed] [Google Scholar]
- 60.Lerolle N, Nochy D, Guérot E, et al. Histopathology of septic shock induced acute kidney injury: apoptosis and leukocytic infiltration. Intensive Care Med 2010; 36: 471–478 [DOI] [PubMed] [Google Scholar]
- 61.Taulan M, Paquet F, Argiles A, et al. Comprehensive analysis of the renal transcriptional response to acute uranyl nitrate exposure. BMC Genomics 2006; 7: 2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Taulan M, Paquet F, Maubert C, et al. Renal toxicogenomic response to chronic uranyl nitrate insult in mice. Environ Health Perspect 2004; 112: 1628–1635 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Sánchez DJ, Bellés M, Albina ML, et al. Nephrotoxicity of simultaneous exposure to mercury and uranium in comparison to individual effects of these metals in rats. Biol Trace Elem Res 2001; 84: 139–154 [DOI] [PubMed] [Google Scholar]
- 64.Haley DP. Morphologic changes in uranyl nitrate-induced acute renal failure in saline- and water-drinking rats. Lab Invest 1982; 46: 196–208 [PubMed] [Google Scholar]
- 65.Kanda H, Kikushima M, Homma-Takeda S, et al. Downregulation of arginase II and renal apoptosis by inorganic mercury: overexpression of arginase II reduces its apoptosis. Arch Toxicol 2008; 82: 67–73 [DOI] [PubMed] [Google Scholar]
- 66.Edwards JR, Diamantakos EA, Peuler JD, et al. A novel method for the evaluation of proximal tubule epithelial cellular necrosis in the intact rat kidney using ethidium homodimer. BMC Physiol 2007; 7: 1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Goering PL, Fisher BR, Noren BT, et al. Mercury induces regional and cell-specific stress protein expression in rat kidney. Toxicol Sci 2000; 53: 447–457 [DOI] [PubMed] [Google Scholar]
- 68.Yuan G, Dai S, Yin Z, et al. Sub-chronic lead and cadmium co-induce apoptosis protein expression in liver and kidney of rats. Int J Clin Exp Pathol 2014; 7: 2905–2914 [PMC free article] [PubMed] [Google Scholar]
- 69.Fujiwara Y, Lee JY, Tokumoto M, et al. Cadmium renal toxicity via apoptotic pathways. Biol Pharm Bull 2012; 35: 1892–1897 [DOI] [PubMed] [Google Scholar]
- 70.Prozialeck WC, Edwards JR, Lamar PC, et al. Expression of kidney injury molecule-1 (Kim-1) in relation to necrosis and apoptosis during the early stages of Cd-induced proximal tubule injury. Toxicol Appl Pharmacol 2009; 238: 306–314 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Wang X, Zhao H, Shao Y, et al. Nephroprotective effect of astaxanthin against trivalent inorganic arsenic-induced renal injury in wistar rats. Nutr Res Pract 2014; 8: 46–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wei T, Tian W, Liu F, et al. Protective effects of exogenous β-hydroxybutyrate on paraquat toxicity in rat kidney. Biochem Biophys Res Commun 2014; 447: 666–671 [DOI] [PubMed] [Google Scholar]
- 73.Ben Rejeb A, Maillet M, Bescol-Liversaac J, et al. Ultrastructure of the kidney in paraquat-poisoned rats. Comparative study with literature data on man and animal. Arch Anat Cytol Pathol 1997; 45: 199–207 [PubMed] [Google Scholar]
- 74.Wei Q, Hill WD, Su Y, et al. Heme oxygenase-1 induction contributes to renoprotection by G-CSF during rhabdomyolysis-associated acute kidney injury. Am J Physiol Renal Physiol 2011; 301: F162–F170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kim YS, Jung MH, Choi MY, et al. Glutamine attenuates tubular cell apoptosis in acute kidney injury via inhibition of the c-Jun N-terminal kinase phosphorylation of 14-3-3. Crit Care Med 2009; 37: 2033–2044 [DOI] [PubMed] [Google Scholar]
- 76.Kim JH, Lee SS, Jung MH, et al. N-acetylcysteine attenuates glycerol-induced acute kidney injury by regulating MAPKs and Bcl-2 family proteins. Nephrol Dial Transplant 2010; 25: 1435–1443 [DOI] [PubMed] [Google Scholar]
- 77.Bengatta S, Arnould C, Letavernier E, et al. MMP9 and SCF protect from apoptosis in acute kidney injury. J Am Soc Nephrol 2009; 20: 787–797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kindt N, Menzebach A, Van de Wouwer M, et al. Protective role of the inhibitor of apoptosis protein, survivin, in toxin-induced acute renal failure. FASEB J 2008; 22: 510–521 [DOI] [PubMed] [Google Scholar]
- 79.Kumar D, Singla SK, Puri V, et al. The restrained expression of NF-kB in renal tissue ameliorates folic acid induced acute kidney injury in mice. PLoS One 2015; 10: e115947 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Whiting PH, Brown PA. The relationship between enzymuria and kidney enzyme activities in experimental gentamicin nephrotoxicity. Ren Fail 1996; 18: 899–909 [DOI] [PubMed] [Google Scholar]
- 81.Kandemir FM, Ozkaraca M, Yildirim BA, et al. Rutin attenuates gentamicin-induced renal damage by reducing oxidative stress, inflammation, apoptosis, and autophagy in rats. Ren Fail 2015; 37: 518–525 [DOI] [PubMed] [Google Scholar]
- 82.Linkermann A, Bräsen JH, Darding M, et al. Two independent pathways of regulated necrosis mediate ischemia-reperfusion injury. Proc Natl Acad Sci U S A 2013; 110: 12024–12029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Bolisetty S, Traylor AM, Kim J, et al. Heme oxygenase-1 inhibits renal tubular macroautophagy in acute kidney injury. J Am Soc Nephrol 2010; 21: 1702–1712 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Periyasamy-Thandavan S, Jiang M, Wei Q, et al. Autophagy is cytoprotective during cisplatin injury of renal proximal tubular cells. Kidney Int 2008; 74: 631–640 [DOI] [PubMed] [Google Scholar]
- 85.Jiang M, Wei Q, Dong G, et al. Autophagy in proximal tubules protects against acute kidney injury. Kidney Int 2012; 82: 1271–1283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Takahashi A, Kimura T, Takabatake Y, et al. Autophagy guards against cisplatin-induced acute kidney injury. Am J Pathol 2012; 180: 517–525 [DOI] [PubMed] [Google Scholar]
- 87.Inoue K, Kuwana H, Shimamura Y, et al. Cisplatin-induced macroautophagy occurs prior to apoptosis in proximal tubules in vivo. Clin Exp Nephrol 2010; 14: 112–122 [DOI] [PubMed] [Google Scholar]
- 88.Linkermann A, Bräsen JH, Himmerkus N, et al. Rip1 (receptor-interacting protein kinase 1) mediates necroptosis and contributes to renal ischemia/reperfusion injury. Kidney Int 2012; 81: 751–761 [DOI] [PubMed] [Google Scholar]
- 89.Chien CT, Shyue SK, Lai MK. Bcl-xL augmentation potentially reduces ischemia/reperfusion induced proximal and distal tubular apoptosis and autophagy. Transplantation 2007; 84: 1183–1190 [DOI] [PubMed] [Google Scholar]
- 90.Suzuki C, Isaka Y, Takabatake Y, et al. Participation of autophagy in renal ischemia/reperfusion injury. Biochem Biophys Res Commun 2008; 368: 100–106 [DOI] [PubMed] [Google Scholar]
- 91.Lempiäinen J, Finckenberg P, Mervaala EE, et al. Caloric restriction ameliorates kidney ischaemia/reperfusion injury through PGC-1α-eNOS pathway and enhanced autophagy. Acta Physiol (Oxf) 2013; 208: 410–421 [DOI] [PubMed] [Google Scholar]
- 92.Liu S, Hartleben B, Kretz O, et al. Autophagy plays a critical role in kidney tubule maintenance, aging and ischemia-reperfusion injury. Autophagy 2012; 8: 826–837 [DOI] [PubMed] [Google Scholar]
- 93.Kimura T, Takabatake Y, Takahashi A, et al. Autophagy protects the proximal tubule from degeneration and acute ischemic injury. J Am Soc Nephrol 2011; 22: 902–913 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lee S, Lee SJ, Coronata AA, et al. Carbon monoxide confers protection in sepsis by enhancing beclin 1-dependent autophagy and phagocytosis. Antioxid Redox Signal 2014; 20: 432–442 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Hsiao HW, Tsai KL, Wang LF, et al. The decline of autophagy contributes to proximal tubular dysfunction during sepsis. Shock 2012; 37: 289–296 [DOI] [PubMed] [Google Scholar]
- 96.Howell GM, Gomez H, Collage RD, et al. Augmenting autophagy to treat acute kidney injury during endotoxemia in mice. PLoS One 2013; 8: e69520 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Chargui A, Zekri S, Jacquillet G, et al. Cadmium-induced autophagy in rat kidney: an early biomarker of subtoxic exposure. Toxicol Sci 2011; 121: 31–42 [DOI] [PubMed] [Google Scholar]
- 98.Brown MM, Rhyne BC, Goyer RA. Intracellular effects of chronic arsenic administration on renal proximal tubule cells. J Toxicol Environ Health 1976; 1: 505–514 [DOI] [PubMed] [Google Scholar]
- 99.Homsi E, Andreazzi DD, Faria JB, et al. TNF-α-mediated cardiorenal injury after rhabdomyolysis in rats. Am J Physiol Renal Physiol 2015; 308: F1259–F1267 [DOI] [PubMed] [Google Scholar]
- 100.Fähling M, Mathia S, Paliege A, et al. Tubular von Hippel-Lindau knockout protects against rhabdomyolysis-induced AKI. J Am Soc Nephrol 2013; 24: 1806–1819 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Allam R, Scherbaum CR, Darisipudi MN, et al. Histones from dying renal cells aggravate kidney injury via TLR2 and TLR4. J Am Soc Nephrol 2012; 23: 1375–1388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, et al. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol 2014; 15: 135–147 [DOI] [PubMed] [Google Scholar]
- 103.Linkermann A, Bräsen JH, Darding M, et al. Two independent pathways of regulated necrosis mediate ischemia-reperfusion injury. Proc Natl Acad Sci USA 2013; 110: 12024–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Linkermann A, Chen G, Dong G, et al. Regulated cell death in AKI. J Am Soc Nephrol 2014; 25: 2689–2701 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Ame JC, Spenlehauer C, de Murcia G. The PARP superfamily. Bioessays 2004; 26: 882–893 [DOI] [PubMed] [Google Scholar]
- 106.Bürkle A, Virág L. Poly (ADP-ribose): PARadigms and PARadoxes. Mol Aspects Med 2013; 34: 1046–1065 [DOI] [PubMed] [Google Scholar]
- 107.Virág L, Robaszkiewicz A, Rodriguez-Vargas JM, et al. Poly (ADP-ribose) signaling in cell death. Mol Aspects Med 2013; 34: 1153–1167 [DOI] [PubMed] [Google Scholar]
- 108.Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012; 149: 1060–1072 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Linkermann A, Skouta R, Himmerkus N, et al. Synchronized renal tubular cell death involves ferroptosis. Proc Natl Acad Sci USA 2014; 111: 16836–16841 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Galluzzi L, Kepp O, Krautwald S, et al. Molecular mechanisms of regulated necrosis. Semin Cell Dev Biol 2014; 35: 24–32 [DOI] [PubMed] [Google Scholar]
- 111.Skouta R, Dixon SJ, Wang J, et al. Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J Am Chem Soc 2014; 136: 4551–4556 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kroemer G, Levine B. Autophagic cell death: the story of a misnomer. Nat Rev Mol Cell Biol 2008; 9: 1004–1010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Boya P, Gonzalez-Polo RA, Casares N, et al. Inhibition of macroautophagy triggers apoptosis. Mol Cell Biol 2005; 25: 1025–1040 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Liang XH, Jackson S, Seaman M, et al. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 1999; 402: 672–676 [DOI] [PubMed] [Google Scholar]
- 115.Fimia GM, Stoykova A, Romagnoli A, et al. Ambra1 regulates autophagy and development of the nervous system. Nature 2007; 447: 1121–1125 [DOI] [PubMed] [Google Scholar]
- 116.Vitale I, Galluzzi L, Castedo M, et al. Mitotic catastrophe: a mechanism for avoiding of genomic instability. Nat Rev Mol Cell Biol 2011; 12: 385–392 [DOI] [PubMed] [Google Scholar]
- 117.Overholtzer M, Mailleux AA, Mouneimne G, et al. A nonapoptotic cell death process, entosis, that occurs by cell-in-cell invasion. Cell 2007; 131: 966–979 [DOI] [PubMed] [Google Scholar]
- 118.Matarrese P, Ciarlo L, Tinari A, et al. Xeno-cannibalism as an exacerbation of self-cannibalism: a possible fruitful survival strategy for cancer cells. Curr Pharm Des 2008; 14: 245–252 [DOI] [PubMed] [Google Scholar]
- 119.Allam R, Kumar SV, Darisipudi MN, et al. Extracellular histones in tissue injury and inflammation. J Mol Med (Berl) 2014; 92: 465–472 [DOI] [PubMed] [Google Scholar]
- 120.Dible JH, Bull GM, Darmady EM. Acute tubular necrosis. Br Med J 1950; 1: 1262–1264 [PMC free article] [PubMed] [Google Scholar]
- 121.Jouan-Lanhouet S, Riquet F, Duprez L, et al. Necroptosis, in vivo detection in experimental disease models. Semin Cell Dev Biol 2014; 35: 2–13 [DOI] [PubMed] [Google Scholar]
- 122.Yoshida T, Shimizu A, Masuda Y, et al. Caspase-3-independent internucleosomal DNA fragmentation in ischemic acute kidney injury. Nephron Exp Nephrol 2012; 120: e103–e113 [DOI] [PubMed] [Google Scholar]
- 123.Shen S, Kepp O, Kroemer G. The end of autophagic cell death? Autophagy 2012; 8: 1–3 [DOI] [PubMed] [Google Scholar]
- 124.Clarke PG, Puyal J. Autophagic cell death exists. Autophagy 2012; 8: 867–869 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Bonventre JV, Yang L. Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest 2011; 121: 4210–4221 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Quiros Y, Vicente-Vicente L, Morales AI, et al. An integrative overview on the mechanisms underlying the renal tubular cytotoxicity of gentamicin. Toxicol Sci 2011; 119: 245–256 [DOI] [PubMed] [Google Scholar]
- 127.Tait SW, Ichim G, Green DR. Die another way—non-apoptotic mechanisms of cell death. J Cell Sci 2014; 127: 2135–2144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ratliff BB, Rabadi MM, Vasko R, et al. Messengers without borders: mediators of systemic inflammatory response in AKI. J Am Soc Nephrol 2013; 24: 529–536 [DOI] [PubMed] [Google Scholar]
- 129.Núñez R, Sancho-Martínez SM, Novoa JM, et al. Apoptotic volume decrease as a geometric determinant for cell dismantling into apoptotic bodies. Cell Death Differ 2010; 17: 1665–1671 [DOI] [PubMed] [Google Scholar]
- 130.Vanden Berghe T, Vanlangenakker N, Parthoens E, et al. Necroptosis, necrosis and secondary necrosis converge on similar cellular disintegration features. Cell Death Differ 2010; 17: 922–930 [DOI] [PubMed] [Google Scholar]
- 131.Sanchez-González PD, López-Hernández FJ, López-Novoa JM, et al. An integrative view of the pathophysiological events leading to cisplatin nephrotoxicity. Crit Rev Toxicol 2011; 41: 803–821 [DOI] [PubMed] [Google Scholar]
- 132.Sancho-Martínez SM, Piedrafita FJ, Cannata JB, et al. Necrotic concentrations of cisplatin activate the apoptotic machinery but inhibit effector caspases and interfere with the execution of apoptosis. Toxicol Sci 2011; 122: 73–85 [DOI] [PubMed] [Google Scholar]
- 133.Yang JR, Yao FH, Zhang JG, et al. Ischemia-reperfusion induces renal tubule pyroptosis via the CHOP-caspase-11 pathway. Am J Physiol Renal Physiol 2014; 306: F75–F84 [DOI] [PubMed] [Google Scholar]
- 134.Lorenz G, Darisipudi MN, Anders HJ. Canonical and non-canonical effects of the NLRP3 inflammasome in kidney inflammation and fibrosis. Nephrol Dial Transplant 2014; 29: 41–48 [DOI] [PubMed] [Google Scholar]
- 135.Krautwald S, Linkermann A. The fire within: pyroptosis in the kidney. Am J Physiol Renal Physiol 2014; 306: F168–F169 [DOI] [PubMed] [Google Scholar]
- 136.Miao EA, Leaf IA, Treuting PM, et al. Caspase-1-induced pyroptosis is an innate immune effector mechanism against intracellular bacteria. Nat Immunol 2010; 11: 1136–1142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Anders HJ, Schaefer L. Beyond tissue injury-damage-associated molecular patterns, toll-like receptors, and inflammasomes also drive regeneration and fibrosis. J Am Soc Nephrol 2014; 25: 1387–1400 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Keeble JA, Gilmore AP. Apoptosis commitment—translating survival signals into decisions on mitochondria. Cell Res 2007; 17: 976–984 [DOI] [PubMed] [Google Scholar]
- 139.Degterev A, Huang Z, Boyce M, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol 2005; 1: 112–119 [DOI] [PubMed] [Google Scholar]
- 140.Linkermann A, Bräsen JH, Himmerkus N, et al. Rip1 (receptor-interacting protein kinase 1) mediates necroptosis and contributes to renal ischemia/reperfusion injury. Kidney Int 2012; 81: 751–761 [DOI] [PubMed] [Google Scholar]
- 141.Tait SW, Oberst A, Quarato G, et al. Widespread mitochondrial depletion via mitophagy does not compromise necroptosis. Cell Rep 2013; 5: 878–885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Krautwald S, Ziegler E, Rölver L, et al. Effective blockage of both the extrinsic and intrinsic pathways of apoptosis in mice by TAT-crmA. J Biol Chem 2010; 285: 19997–20005 [DOI] [PMC free article] [PubMed] [Google Scholar]


