Tissue injury and inflammation are tightly linked and can cause each other.1 Infectious, toxic, or traumatic injuries usually result in tissue inflammation, and in turn, autoimmune inflammation causes tissue injury. However, what are the molecular mechanisms underlying these bidirectional causalities? Immunity-related cell death involves the complement system or cytotoxic T cells as effector mechanisms of infection and autoimmunity.2,3Vice versa, cell death–related immunity is best explained by the recently discovered paradigm of dying cells releasing danger-associated molecular patterns (DAMPs) that trigger innate immunity via pattern recognition receptors (PRR) (Supplemental Material).4 However, somehow these two concepts coexist in parallel and still lack a unifying theory on injury and inflammation.5–9 The very recent discovery of the numerous forms of regulated necrosis gives rise to such a theory, herein referred to as necroinflammation.1,6,10 Necroinflammation integrates several concepts on the causal links between tissue injury and necrosis. In this review, we discuss the definition of necroinflammation and its functions, molecular pathways involved, contribution to kidney disease, and strategies to therapeutically disrupt necroinflammation to limit kidney injury and failure.
What Is Necroinflammation?
Necroinflammation describes an autoamplification loop driven by necrosis (defined by cell death involving rupture of the plasma membrane) and inflammation (defined by cytokine release, increased vascular permeability, and recruitment of immune effector cells).10 The autoamplification loop of necroinflammation is accelerated by immunity-related cellular necrosis and necrosis-related immune activation. Necroinflammation can be initiated by a few necrotic cells that activate the innate immune system, which subsequently leads to necrosis of more cells triggering more inflammation in a process that eventually may lead to organ failure.10 The same process can be initiated by inflammation but often the initial insult is not as obvious as in a gout attack. Monosodium urate crystals trigger IL-1β secretion and induce death in neutrophils.11,12 Neutrophil death implies the release of proteases, DNA, and histones that trigger inflammation of the joint structures, which recruits more neutrophils that die and so on.11,12 Clinically, this process presents as a sudden onset of arthritis and sometimes even as fever and acute illness, when inflammation reaches systemic dimensions.11 Similarly, in stroke, myocardial infarction, or acute tubular necrosis, the number of cells dying from the initial insult may be few, whereas the subsequent inflammatory response contributes to further cell death (i.e., unnecessary collateral tissue damage). However, why did evolution favor such a devastating mechanism?
Janeway and Medzhitov proposed the concept that pathogens activate innate immunity,13 which was subsequently confirmed on the discovery of the various types of PRRs and their pathogen-associated molecular patterns (PAMPs).14 From this example it is obvious that the danger control program of inflammation was selected during evolution to at first combat pathogens. Pathogen entry implies a disrupted barrier to the outside (e.g., wounded skin or a corneal, oral, or intestinal ulceration). In this setting, inflammation not only kills invaded pathogens but also provides a functional barrier to prevent further pathogen entry until re-epithelialization regenerates a structural barrier to the outside.15,16 Inflammation kills host cells at the site of infection to attack intracellular pathogens,7 which despite some collateral tissue injury, as a net impact, usually helps host survival.7 Matzinger insisted that also sterile dangers alert the innate immune system,14,17,18 which was confirmed by the discovery of dying cell-released DAMPs during sterile injuries (Table 1).4 PAMPs and DAMPs are integrated at the level of the same PRRs that translate danger recognition into innate immune activation.7 This explains why, for example, gouty arthritis is clinically indistinguishable from bacterial arthritis.4,19,20 Together, this suggests that necroinflammation is an autoamplification loop of necrosis and inflammation that evolved as a life-saving mechanism of host defense but causes unnecessary tissue damage in sterile diseases.
Table 1: Necrosis-related DAMPs and alarmins and their PRRs
Necroinflammation Involves Multiple Molecular Pathways
Categories of Regulated Necrosis
Both apoptosis and necrosis are executed in a regulated manner; however, necrosis especially alerts innate immunity because the ruptured plasma membrane involves massive DAMP release into the extracellular space. Many pathways of regulated necrosis have been implicated, but the following six appear as the most appropriate to be distinguished for the purpose of this review. It is of note that in failing organs, a single one of these pathways may predominate, but often they overlap and trigger each other. For more detailed information on the different categories of regulated cell death, we refer the reader to excellent recent reviews.1,21
Necroptosis is defined by the receptor-interacting protein kinase 3 (RIPK3)–mediated phosphorylation of mixed-lineage kinase domain-like (MLKL),22 which by unknown means leads to plasma membrane rupture.23 Numerous signaling events can trigger RIPK3 phosphorylation, especially a change of the RIPK1 polyubiquitination pattern that may be initiated downstream of several cell surface receptors. Necroptosis has been well described to kill parenchymal cells of all tissues, including renal tubular epithelial cells.22 TNF receptor 1 activation can trigger apoptosis and necroptosis, but the latter is favored in case of caspase deactivation.24 Meanwhile, the first negative regulator of necroptosis has been described. Protein phosphatase 1b dephosphorylates phosphorylated RIPK3, which thereby loses its ability to activate MLKL-mediated membrane rupture.25
Ferroptosis is characterized by a defined lipid peroxidation signature in oxylipidomics analysis caused by a failure of glutathione-peroxidase 4 function.26,27 Ferroptosis involves ER stress and dysfunction caused by a lack of glutathione,28,29 which may be induced by inhibition of the cellular Cys/Glu-antiporter System Xc-, which may be controlled by p53 via SLC7A11 or heat shock protein beta 1.30,31 Ferroptosis can occur in any glutathione-depleted cell and has also been described in renal tubular epithelial cells.27,32
Mitochondrial-permeability transition-mediated regulated necrosis (MPT-RN) is independent of ferroptosis or necroptosis because it depends on cyclophilin D and may partially overlap with a poly (ADP-ribose) polymerase 1–mediated pathway of regulated necrosis, previously referred to as parthanatos. Both MPT-RN32-35 and parthanatos36–39 have been particularly worked out in renal cells and have been reviewed elsewhere.40,41
Pyroptosis is a consequence of inflammasome-driven caspase-11 activation.42,43 Concomitant caspase-1 activity induces mature IL-1β and IL-18 secretion, rendering pyroptosis particularly inflammatory.42 Pyroptosis has been clearly documented in infected macrophages and dendritic cells, and if pyroptosis can occur in renal cells is under debate.44,45
NETosis is a controlled and often suicidal act of activated neutrophils, which results in the formation of neutrophil extracellular traps (NETs), consisting of expelled chromatin loaded with lysosomal and cytosolic proteases.46 The involved signaling pathways have not yet been fully understood but include NADPH-dependent ROS production and RIPK1 signaling.47
Another avenue of cell death is mitotic catastrophe. When cells are forced to overcome the G2/M arrest of the cell cycle despite significant DNA damage, aberrant division of chromosomes (i.e., aneuploidy) renders the cell to death (i.e., often necrosis).48–53 This is obvious in podocytes that impair their capacity to maintain foot processes and to adhere to the filtration barrier once forced to retract their cytoskeleton from the foot processes to form the mitotic spindle.49–52 Another example is the necessity to delete cells with significant cell damage in the early injury phase of AKI.53
How Necrosis Induces Inflammation
Necrotic cells release DAMPs and alarmins from several intracellular compartments (Figure 1, Table 1). Alarmins are a heterogeneous group of preformed proinflammatory molecules that are released by cell death from stores inside the cell.54,55 By contrast, DAMPs are molecules with other proinflammatory functions under normal conditions that turn into danger signals only once being released by cell death and by alerting the innate immune system via a group of PRRs on the surface or inside other cells.
Figure 1: Molecular pathways involved in necroinflammation. Necrotic renal cells release DAMPs and alarmins that activate DAMP or alarmin receptors on immune (and parenchymal) cells, respectively (
Table 1). Activation of immune (and parenchymal) cells induces the secretion of numerous proinflammatory cytokines that in turn can induce several forms of regulated necrosis (
e.g., necroptosis, pyroptosis). The necroptosis signaling pathway involves auto- and transphosphorylation of RIPK1 and RIPK3 and the recruitment of MLKL. Activation of caspase-1 induces release of IL-1
β and IL-18, which causes inflammation. Certain DAMPs and proinflammatory cytokines, such as TNF-
α or IL-8, directly activate neutrophils for the formation of NETs. NET formation expels large amounts of histones into the extracellular space. Extracellular histones have direct cytotoxic impact on renal cells and in addition induce TLR2-/TLR4- and NLRP3-related immune cell activation. GPX-4, glutathione peroxidase; MOMP, mitochondrial outer membrane permeabilization; p-MLKL, phospho-mixed lineage kinase domain-like; Ripk1, receptor-activating protein kinase 1; Ripk3, receptor-activating protein kinase 3.
Toll-like Receptors
Toll-like receptors (TLRs) are single transmembrane receptors containing a cytosolic Toll/IL-1 receptor interaction domain. On DAMP binding, TLR homo- or heterodimerization supports the recruitment of specific cytosolic adapter molecules, such as myeloid differentiation primary response gene 88, TRIF, TRAM, or TIRAP, to the Toll/IL-1 receptor domain, which activates a set of kinases and transcription factors, including NF-κB, activating protein-1, or IFN-related factors.56
NOD-like Receptors
NOD-like receptors (NLRs) are intracellular PRRs detecting danger signals in the cytosol. Among all NLRs, NOD-like receptor protein 3 (NLRP3) is specifically involved in DAMP recognition. For example, uric acid and ATP activate NLRP3 to bind to its adapter molecule, adapter protein apoptosis associated speck-like protein containing a CARD, which both form a caspase-1–activating complex, named the inflammasome.57 Caspase-1 activation cleaves pro–IL-1β and pro–IL-18 and induces the secretion of the mature cytokines. Both cytokines further induce innate immunity via their respective cytokine receptors (i.e., IL-1R type 1, IL-18Rs).56
Retinoic Acid Inducible Gene–like Receptors
Retinoic acid inducible gene–like receptors (RLRs) are intracellular receptors for viral nucleic acids. RLRs signal through adapter protein IFNβ promoter stimulator-1, resulting in IRF3 and NF-κB activation.56 RLRs detect mostly replicating viruses58; therefore, their role in the recognition of endogenous nucleic acids is questionable.
C-type Lectin Receptors
C-type lectin receptors (CLRs) are surface receptors that can induce two different pathways: CLEC4C, CLEC4E, CLEC5A, and CLEC6A are associated with and induce signaling pathways through ITAM-containing adaptor molecules, such as FcRγ or DAP12. Other CLRs (e.g., dectin-1, DC-SIGN, CLECL12A) induce signaling pathways through the activation of protein kinases or phosphatases. CLR signaling mainly activates or modulates NF-κB functions.59
How Inflammation Induces Necrosis
DAMPs released by dying cells activate the PRRs on infiltrating immune cells, such as dendritic cells, macrophages, neutrophils, and lymphocytes, and intrinsic renal parenchymal cells, and induce the expression and local release of numerous proinflammatory mediators (Figure 1).60 In particular, TNF-α and IFN-γ can induce necroptosis via two distinct pathways. TNFR1 activation by TNF-α or TRIF activation via TLRs affects the polyubiquitination of RIPK1, which activates the kinase domain and induces the phosphorylation of RIPK3 and subsequently of MLKL,61–63 whereas IFN-γ signals use a STAT3-protein kinase R-dependent pathway to induce necroptosis.22 In this process RIPK1 serves as a negative regulator to TNF-α–mediated cell death via caspase-8.21 TNF-α and IL-8 can also induce NETosis in neutrophils.46,64 NET formation involves the release of histones, which elicit cytotoxic impact on endothelial cells, mesangial cells, and podocytes, a process that is poorly defined at the molecular level.65 Extracellular histones do not seem to kill cells via a specific surface receptor but rather in a charge-dependent manner.65 Furthermore, PAMP- or DAMP-induced NLRP3 activation not only triggers cytokine release but also pyroptosis.42 An inactive pro–caspase-1 is synthesized on TLR activation, and activated NLR inflammasome complexes cleave pro–caspase-1 into its active form. Hence, it is considered as an infection-induced cell death.42,66 Together, several proinflammatory signals induce regulated necrosis.
Necroinflammation in Kidney Disease
Sepsis Urosepsis
Local immune responses disrupting epithelial and mesenchymal barriers can facilitate pathogen spreading into the circulation (i.e., sepsis).67,68 For example, when ascending urinary tract infection reaches the kidney, infective pyelonephritis triggers a potent inflammatory response for host defense.69,70 Pathogen entry into the renal parenchyma involves LPS-/TLR4-mediated pathogen recognition and chemokine (C-X-C motif) ligand 12–driven neutrophil recruitment, which can cause abscess formation and massive renal cell necrosis.71 Necrosis disrupts internal barriers and promotes pathogen spreading and urosepsis.71 Circulating bacterial endotoxin is a potent stimulus for cytokine release from monocytes and other immune and nonimmune cells, which has been referred to as a cytokine storm.67,72 In this process, cytokine-induced NETosis and histone release can occur everywhere in the microvasculature, which is indicated by increased levels of histones in the circulation of septic patients.73,74 Circulating histones cause cytotoxic impact to endothelial cells and activates platelets, which imply disseminated intravascular coagulation, tissue hypoperfusion, and organ dysfunction (e.g., of the lung, intestine, heart, kidneys).65,73,74 These mechanisms of remote disease exacerbation apply to any source of sepsis or trauma.75,76 Massive local DAMP or histone release can cause remote microvascular injuries and clotting (e.g., in the lung), which serves as a pathophysiologic explanation to acute respiratory distress syndrome in such settings.76
Acute Tubular Necrosis
Ex vivo microscopy (high-resolution microscopy of freshly isolated primary kidney tubules) revealed how single-cell necrosis can spread on neighboring cells so that the entire tubule segment undergoes a synchronized cell death.35,77,78 Whether synchronized necrosis occurs in vivo has yet to be demonstrated. Regulated tubule necrosis is not limited to post-ischemic AKI35,79 or delayed graft function,77,80 but cisplatin nephrotoxicity involves necroptosis of tubular cells, triggering necroinflammation35,81 (Table 2). The same process causes tubular necrosis in rhabdomyolysis.82 In addition, contrast media–related renal dysfunction is entirely reversible by necrostatin-1.83 Moderate-dose adriamycin-induced tubular necrosis and survival is attenuated in Ripk3-deficient mice,84 but whether tubular cells themselves directly die via necroptosis, or succumb to some other cell death secondary to initial RIPK3-mediated hypoperfusion, is still a matter of debate. First, tubules cannot be sensitized to necroptosis by the loss of Fas-associated protein with death domain or caspase-835; second, necrostatins do not protect isolated renal tubules from ischemic injury. In contrast, inhibition of ferroptosis is very effective in protection from ischemia-reperfusion injury in vivo and hydroxychloroquine-/iron-induced tubular damage ex vivo.32 Therefore, the primary mode of tubular cell death in these models is still uncertain (Figure 2). Even MPT-RN appears to contribute to tubular injury, either directly or indirectly. Combined deficiency of cyclophilin D and RIPK3 led to highly significant survival benefits, even in severe models of post-ischemic tubular necrosis, suggesting that MPT-RN and necroptosis additively contribute to tubular necrosis.33,34 In turn, inflammation is an important accelerator of tubular injury15,85 and involves potential triggers of necroptosis such as TNF-α.86,87 Oxalate crystal formation inside tubules induces RIPK3- and MLKL-dependent tubular cell death (S.R. Mulay and H.-J. Anders, unpublished data), and the associated ATP release from dying tubular cells triggers NLRP3 inflammasome activation, IL-1β–dependent renal inflammation, and renal failure in mice.88 Blocking either necroptosis with necrostatin-1 or IL-1 with anakinra can abrogate kidney injury and dysfunction88 (S.R. Mulay and H.-J. Anders unpublished data). ATN-associated release of proinflammatory cytokines, DAMPs, and other components may also cause remote tissue injury.80,89 These authors detected parthanatos, necroptosis, and MPT-RN in the transplanted organ after reperfusion and in the lungs of the recipients.
Table 2: Regulated necrosis pathways tested in different models of acute tubular necrosis
Figure 2: Necroinflammation in glomerular and tubulointerstitial inflammation. (A) Neutrophil extracellular trap formation in glomerular capillaries (e.g., in ANCA vasculitis) causes histone-related endothelial cell death and subsequent capillary loop necrosis. DAMP release activates other glomerular cells to produce cytokines and chemokines, which recruit more cytokine-producing leukocytes, including neutrophils undergoing NETosis. TNF-α and histones drive further necrosis of glomerular cells. The associated rupture of the glomerular basement membrane implies not only podocyte injury but also plasma leakage into Bowman’s space. Plasma components such as fibrinogen activate parietal epithelial cell hyperplasia and crescent formation. (B) In tubule injury, regulated necrosis of any kind may be the initial event. Histones released by tubular cells and netting neutrophils elicit direct cytotoxicity. The associated release of DAMPs and alarmins induces inflammation, which implies the recruitment of cytokine-producing leukocytes into the peritubular interstitium. Also, TNF-α and possibly other cytokines drive necroptosis as a secondary cell death category contributing to tubular necrosis and renal dysfunction. This sets up the auto-amplification loop of necroinflammation.
Rapidly Progressive GN
Rapidly progressive GN usually presents as necrotizing and crescentic GN, which are classic presentations of ANCA-associated vasculitis, lupus nephritis, antiglomerular basement membrane disease, or Henoch–Schonlein purpura.90 Mechanistically, necrotizing GN develops from glomerular endothelial cell necrosis (Figure 2).91 For example, in experimental antiglomerular basement membrane disease, neutrophils undergo NETosis inside the glomerular capillaries. It is the release of histones that induces glomerular endothelial cell necrosis.92 This became obvious because either the specific inhibition of NETosis or the neutralization of extracellular histones abrogated crescentic GN.92 Similarly, NETs are also found in necrotic glomerular lesions in human renal vasculitis.93 Complement-mediated cytotoxicity is another trigger for glomerular necroinflammation.94 Glomerular cell necrosis induces DAMP release that further drives cytokine and chemokine release, leukocyte recruitment, and inflammation.92 Infiltrating immune cells, in turn, further contribute to necroinflammation by NETosis (neutrophils),92 cytokine-induced cell death (all proinflammatory leukocytes),95 or direct T cell–related cytotoxicity.96 Vascular wall rupture not only facilitates hematuria but also plasma leakage as a major driver of parietal epithelial cell hyperplasia and crescent formation.97 Therefore, NETosis and histone or complement cytotoxicity triggering glomerular necroinflammation are a central pathomechanism of rapidly progressive GN related to ANCA-associated vasculitis and the other forms of necrotizing GN.98,99
Thrombotic Microangiopathy
Thrombotic microangiopathy (TMA) can be initiated by various triggers, which all share endothelial cell injury-related microvascular thrombosis and organ dysfunction.100 In some disorders, endothelial cell injury is the primary event (e.g., Shiga toxin-induced in hemolytic uremic syndrome, uncontrolled complement activation-induced atypical hemolytic uremic syndrome, preeclampsia, systemic sclerosis, malignant hypertension).100 In other forms of TMA, an inappropriate activation of clotting initiates the process (e.g., antiphospholipid syndrome, ADAMTS13 deficiency).100 Independent of the triggering mechanism, TMA involves an autoamplification loop of tissue necrosis and inflammation.100 Thrombosis involves a massive release of cytokines and chemokines from platelets that activate endothelial cells and recruit leukocytes to the site of injury, a process referred to as immunothrombosis.101 Circulating neutrophils get increasingly involved and contribute to thrombus formation via NET formation,102 which is associated with local histone release. Extracellular histones elicit three different impacts: they kill microvascular endothelial cells,74,92,103 trigger platelet activation and further thrombosis,104 and activate innate immunity via TLRs and the NLRP3 inflammasome.103,105 As a proof of principle, histone injection into the renal artery induces widespread TMA and renal cell necrosis.103 Currently, this autoamplification loop of microvascular injury, thrombosis, and inflammation can only be terminated by abrogating the initial trigger of TMA.100 Long-term outcomes of renal TMA depend on each cell type’s capacity to repair from intrinsic or extrinsic progenitor reservoirs.106
Targeting Renal Necroinflammation for Prophylaxis and Therapy
If necroinflammation is an autoamplification loop, blockade of either component should be sufficient to decelerate and potentially abrogate it. This is well documented by numerous experimental studies of inhibiting different molecular targets that still fully abrogate experimental AKI (Table 3).107 For example, prophylactic therapy with a combination of three different cell death inhibitors (ferrostatins, necrostatin-1, sanglifehrin A) prevents kidney injury, inflammation, and dysfunction in models of ultra-severe duration of ischemia before reperfusion.35 Along these lines, preventing NETosis with a PAD4 inhibitor or extracellular histone neutralization abrogates crescentic GN.92 Autoacceleration of necroinflammation implies that the time point of intervention is of critical importance. Only preemptive blockade of necroinflammation can be maximally effective, and vice versa, most experimental studies reporting prophylactic treatments hardly predict efficacy of delayed therapy. Currently, it remains unknown if delayed cell death inhibition still can prevent kidney injury. In general, few studies report the protective impact of late-onset interventions, such as in histone neutralization preventing crescent formation in overt GN.92 GSK-3 inhibition and recombinant pentraxin-3 can both limit necroinflammation in AKI.108,109 A single-dose application of the GSK-3 inhibitor lithium was reported to attenuate post-ischemic AKI up to 3 hours after renal pedicle clamping108 and recombinant pentraxin-3 injection for up to 6 hours.109 Timing of interventions is essential for the design of clinical trials, and an earlier-the-better strategy is generally recommended. However, the current definition of AKI that is on the basis of functional markers (urinary output, serum creatinine) and not on injury markers (e.g., KIM-1) remains a major conceptual drawback. Any early therapy may arrive way too late to affect necroinflammation as a mechanism of AKI. However, late onset of therapy may still affect long-term outcomes of AKI. For example, TNF-α blockade starting 5 days after AKI or endothelin blockade up to 12 days after AKI prevents nephron loss and subsequent kidney atrophy in experimental AKI.110,111
Table 3: Some molecular targets to therapeutically interrupt necroinflammation
Summary
Necroinflammation is a theory that combines previously established, coexisting concepts of kidney injury. Necroinflammation is a serial event of regulated necrosis triggering inflammation, secondary-regulated necrosis, and so forth. If not opposed at an early stage, necroinflammation can lead to organ failure or even systemic inflammation and remote organ injury. Following this concept, either anti-inflammatory agents or cell death inhibitors can abrogate this process. However, numerous questions remain, some of which are listed in Table 4. We conclude that the various aspects of necroinflammation offer great opportunities for novel discoveries and eventually also for novel treatment options for patients with kidney disease.
Table 4: Necroinflammation
Disclosures
None.
The authors are supported by the Deutsche Forschungsgemeinschaft (MU3906/1-1, AN372/21-1, AN372/14-3, EXC 306, project W TP1).
Published online ahead of print. Publication date available at www.jasn.org.
This article contains supplemental material online at http://jasn.asnjournals.org/lookup/suppl/doi:10.1681/ASN.2015040405/-/DCSupplemental.
References
1. Wallach D, Kang TB, Kovalenko A: Concepts of tissue injury and cell death in inflammation: A historical perspective. Nat Rev Immunol 14: 51–59, 2014
2. Barry M, Bleackley RC: Cytotoxic T lymphocytes: All roads lead to death. Nat Rev Immunol 2: 401–409, 2002
3. Zipfel PF, Skerka C: Complement regulators and inhibitory proteins. Nat Rev Immunol 9: 729–740, 2009
4. Rock KL, Latz E, Ontiveros F, Kono H: The sterile inflammatory response. Annu Rev Immunol 28: 321–342, 2010
5. Allam R, Anders HJ: The role of innate immunity in autoimmune tissue injury. Curr Opin Rheumatol 20: 538–544, 2008
6. Blander JM: A long-awaited merger of the pathways mediating host defence and programmed cell death. Nat Rev Immunol 14: 601–618, 2014
7. Medzhitov R: Origin and physiological roles of inflammation. Nature 454: 428–435, 2008
8. Kurts C, Panzer U, Anders HJ, Rees AJ: The immune system and kidney disease: Basic concepts and clinical implications. Nat Rev Immunol 13: 738–753, 2013
9. Anders HJ, Banas B, Schlöndorff D: Signaling danger: Toll-like receptors and their potential roles in kidney disease. J Am Soc Nephrol 15: 854–867, 2004
10. Linkermann A, Stockwell BR, Krautwald S, Anders HJ: Regulated cell death and inflammation: An auto-amplification loop causes organ failure. Nat Rev Immunol 14: 759–767, 2014
11. Neogi T: Clinical practice. Gout. N Engl J Med 364: 443–452, 2011
12. Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J: Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440: 237–241, 2006
13. Janeway CA Jr, Medzhitov R: Innate immune recognition. Annu Rev Immunol 20: 197–216, 2002
14. Lemaitre B: The road to Toll. Nat Rev Immunol 4: 521–527, 2004
15. Anders HJ: Immune system modulation of kidney regeneration--mechanisms and implications. Nat Rev Nephrol 10: 347–358, 2014
16. Gurtner GC, Werner S, Barrandon Y, Longaker MT: Wound repair and regeneration. Nature 453: 314–321, 2008
17. Matzinger P: Tolerance, danger, and the extended family. Annu Rev Immunol 12: 991–1045, 1994
18. Matzinger P: The danger model: A renewed sense of self. Science 296: 301–305, 2002
19. Anders HJ: Toll-like receptors and danger signaling in kidney injury. J Am Soc Nephrol 21: 1270–1274, 2010
20. Anders HJ, Muruve DA: The inflammasomes in kidney disease. J Am Soc Nephrol 22: 1007–1018, 2011
21. Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P: Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol 15: 135–147, 2014
22. Linkermann A, Green D: Mechanisms of disease: Necroptosis. N Engl J Med 2014, in press
23. Sun L, Wang H, Wang Z, He S, Chen S, Liao D, Wang L, Yan J, Liu W, Lei X, Wang X: Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148: 213–227, 2012
24. Brenner D, Blaser H, Mak TW: Regulation of tumour necrosis factor signalling: Live or let die. Nat Rev Immunol 15: 362–374, 2015
25. Chen W, Wu J, Li L, Zhang Z, Ren J, Liang Y, Chen F, Yang C, Zhou Z, Su SS, Zheng X, Zhang Z, Zhong CQ, Wan H, Xiao M, Lin X, Feng XH, Han J: Ppm1b negatively regulates necroptosis through dephosphorylating Rip3. Nat Cell Biol 17: 434–444, 2015
26. Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, Brown LM, Girotti AW, Cornish VW, Schreiber SL, Stockwell BR: Regulation of ferroptotic cancer cell death by GPX4. Cell 156: 317–331, 2014
27. Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, Aichler M, Walch A, Eggenhofer E, Basavarajappa D, Rådmark O, Kobayashi S, Seibt T, Beck H, Neff F, Esposito I, Wanke R, Förster H, Yefremova O, Heinrichmeyer M, Bornkamm GW, Geissler EK, Thomas SB, Stockwell BR, O’Donnell VB, Kagan VE, Schick JA, Conrad M: Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol 16: 1180–1191, 2014
28. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, Morrison B 3rd, Stockwell BR: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 149: 1060–1072, 2012
29. Dixon SJ, Patel DN, Welsch M, Skouta R, Lee ED, Hayano M, Thomas AG, Gleason CE, Tatonetti NP, Slusher BS, Stockwell BR: Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. eLife 3: e02523, 2014
30. Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R, Gu W: Ferroptosis as a p53-mediated activity during tumour suppression. Nature 520: 57–62, 2015
31. Sun X, Ou Z, Xie M, Kang R, Fan Y, Niu X, Wang H, Cao L, Tang D: HSPB1 as a novel regulator of ferroptotic cancer cell death. Oncogene 2015, in press
32. Linkermann A, Skouta R, Himmerkus N, Mulay SR, Dewitz C, De Zen F, Prokai A, Zuchtriegel G, Krombach F, Welz PS, Weinlich R, Vanden Berghe T, Vandenabeele P, Pasparakis M, Bleich M, Weinberg JM, Reichel CA, Bräsen JH, Kunzendorf U, Anders HJ, Stockwell BR, Green DR, Krautwald S: Synchronized renal tubular cell death involves ferroptosis. Proc Natl Acad Sci U S A 111: 16836–16841, 2014
33. Devalaraja-Narashimha K, Diener AM, Padanilam BJ: Cyclophilin D gene ablation protects mice from ischemic renal injury. Am J Physiol Renal Physiol 297: F749–F759, 2009
34. Park JS, Pasupulati R, Feldkamp T, Roeser NF, Weinberg JM: Cyclophilin D and the mitochondrial permeability transition in kidney proximal tubules after hypoxic and ischemic injury. Am J Physiol Renal Physiol 301: F134–F150, 2011
35. Linkermann A, Bräsen JH, Darding M, Jin MK, Sanz AB, Heller JO, De Zen F, Weinlich R, Ortiz A, Walczak H, Weinberg JM, Green DR, Kunzendorf U, Krautwald S: Two independent pathways of regulated necrosis mediate ischemia-reperfusion injury. Proc Natl Acad Sci U S A 110: 12024–12029, 2013
36. Kim J, Long KE, Tang K, Padanilam BJ: Poly(ADP-ribose) polymerase 1 activation is required for cisplatin nephrotoxicity. Kidney Int 82: 193–203, 2012
37. Kim J, Padanilam BJ: Loss of poly(ADP-ribose) polymerase 1 attenuates renal fibrosis and inflammation during unilateral ureteral obstruction. Am J Physiol Renal Physiol 301: F450–F459, 2011
38. Martin DR, Lewington AJ, Hammerman MR, Padanilam BJ: Inhibition of poly(ADP-ribose) polymerase attenuates ischemic renal injury in rats. Am J Physiol Regul Integr Comp Physiol 279: R1834–R1840, 2000
39. Zheng J, Devalaraja-Narashimha K, Singaravelu K, Padanilam BJ: Poly(ADP-ribose) polymerase-1 gene ablation protects mice from ischemic renal injury. Am J Physiol Renal Physiol 288: F387–F398, 2005
40. Devalaraja-Narashimha K, Singaravelu K, Padanilam BJ: Poly(ADP-ribose) polymerase-mediated cell injury in acute renal failure. Pharmacol Res 52: 44–59, 2005
41. Linkermann A, De Zen F, Weinberg J, Kunzendorf U, Krautwald S: Programmed necrosis in acute kidney injury. Nephrol Dial Transplant 27: 3412–3419, 2012
42. Bergsbaken T, Fink SL, Cookson BT: Pyroptosis: Host cell death and inflammation. Nat Rev Microbiol 7: 99–109, 2009
43. Case CL, Kohler LJ, Lima JB, Strowig T, de Zoete MR, Flavell RA, Zamboni DS, Roy CR: Caspase-11 stimulates rapid flagellin-independent pyroptosis in response to Legionella pneumophila. Proc Natl Acad Sci U S A 110: 1851–1856, 2013
44. Krautwald S, Linkermann A: The fire within: Pyroptosis in the kidney. Am J Physiol Renal Physiol 306: F168–F169, 2014
45. Yang JR, Yao FH, Zhang JG, Ji ZY, Li KL, Zhan J, Tong YN, Lin LR, He YN: Ischemia-reperfusion induces renal tubule pyroptosis via the CHOP-caspase-11 pathway. Am J Physiol Renal Physiol 306: F75–F84, 2014
46. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A: Neutrophil extracellular traps kill bacteria. Science 303: 1532–1535, 2004
47. Branzk N, Papayannopoulos V: Molecular mechanisms regulating NETosis in infection and disease. Semin Immunopathol 35: 513–530, 2013
48. Galluzzi L, Maiuri MC, Vitale I, Zischka H, Castedo M, Zitvogel L, Kroemer G: Cell death modalities: Classification and pathophysiological implications. Cell Death Differ 14: 1237–1243, 2007
49. Lasagni L, Lazzeri E, Shankland SJ, Anders HJ, Romagnani P: Podocyte mitosis - a catastrophe. Curr Mol Med 13: 13–23, 2013
50. Liapis H, Romagnani P, Anders HJ: New insights into the pathology of podocyte loss: mitotic catastrophe. Am J Pathol 183: 1364–1374, 2013
51. Mulay SR, Thomasova D, Ryu M, Kulkarni OP, Migliorini A, Bruns H, Gröbmayr R, Lazzeri E, Lasagni L, Liapis H, Romagnani P, Anders HJ: Podocyte loss involves MDM2-driven mitotic catastrophe. J Pathol 230: 322–335, 2013
52. Thomasova D, Anders HJ: Cell cycle control in the kidney. Nephrol Dial Transplant 2014, in press
53. DiRocco DP, Bisi J, Roberts P, Strum J, Wong KK, Sharpless N, Humphreys BD: CDK4/6 inhibition induces epithelial cell cycle arrest and ameliorates acute kidney injury. Am J Physiol Renal Physiol 306: F379–F388, 2014
54. Oppenheim JJ, Yang D: Alarmins: Chemotactic activators of immune responses. Curr Opin Immunol 17: 359–365, 2005
55. Chan JK, Roth J, Oppenheim JJ, Tracey KJ, Vogl T, Feldmann M, Horwood N, Nanchahal J: Alarmins: Awaiting a clinical response. J Clin Invest 122: 2711–2719, 2012
56. Creagh EM, O’Neill LA: TLRs, NLRs and RLRs: A trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol 27: 352–357, 2006
57. Schroder K, Tschopp J: The inflammasomes. Cell 140: 821–832, 2010
58. Kumagai Y, Kumar H, Koyama S, Kawai T, Takeuchi O, Akira S: Cutting edge: TLR-dependent viral recognition along with type I IFN positive feedback signaling masks the requirement of viral replication for IFN-alpha production in plasmacytoid dendritic cells. J Immunol 182: 3960–3964, 2009
59. Geijtenbeek TB, Gringhuis SI: Signalling through C-type lectin receptors: Shaping immune responses. Nat Rev Immunol 9: 465–479, 2009
60. Vielhauer V, Kulkarni O, Reichel CA, Anders HJ: Targeting the recruitment of monocytes and macrophages in renal disease. Semin Nephrol 30: 318–333, 2010
61. Dannappel M, Vlantis K, Kumari S, Polykratis A, Kim C, Wachsmuth L, Eftychi C, Lin J, Corona T, Hermance N, Zelic M, Kirsch P, Basic M, Bleich A, Kelliher M, Pasparakis M: RIPK1 maintains epithelial homeostasis by inhibiting apoptosis and necroptosis. Nature 513: 90–94, 2014
62. Kearney CJ, Cullen SP, Clancy D, Martin SJ: RIPK1 can function as an inhibitor rather than an initiator of RIPK3-dependent necroptosis. FEBS J 281: 4921–4934, 2014
63. Takahashi N, Vereecke L, Bertrand MJ, Duprez L, Berger SB, Divert T, Gonçalves A, Sze M, Gilbert B, Kourula S, Goossens V, Lefebvre S, Günther C, Becker C, Bertin J, Gough PJ, Declercq W, van Loo G, Vandenabeele P: RIPK1 ensures intestinal homeostasis by protecting the epithelium against apoptosis. Nature 513: 95–99, 2014
64. Lapponi MJ, Carestia A, Landoni VI, Rivadeneyra L, Etulain J, Negrotto S, Pozner RG, Schattner M: Regulation of neutrophil extracellular trap formation by anti-inflammatory drugs. J Pharmacol Exp Ther 345: 430–437, 2013
65. Allam R, Kumar SV, Darisipudi MN, Anders HJ: Extracellular histones in tissue injury and inflammation. J Mol Med (Berl) 92: 465–472, 2014
66. Bortoluci KR, Medzhitov R: Control of infection by pyroptosis and autophagy: Role of TLR and NLR. Cell Mol Life Sci 67: 1643–1651, 2010
67. Stearns-Kurosawa DJ, Osuchowski MF, Valentine C, Kurosawa S, Remick DG: The pathogenesis of sepsis. Annu Rev Pathol 6: 19–48, 2011
68. Hotchkiss RS, Nicholson DW: Apoptosis and caspases regulate death and inflammation in sepsis. Nat Rev Immunol 6: 813–822, 2006
69. Tittel AP, Heuser C, Ohliger C, Knolle PA, Engel DR, Kurts C: Kidney dendritic cells induce innate immunity against bacterial pyelonephritis. J Am Soc Nephrol 22: 1435–1441, 2011
70. Wagenlehner FM, Lichtenstern C, Rolfes C, Mayer K, Uhle F, Weidner W, Weigand MA: Diagnosis and management for urosepsis. Int J Urol 20: 963–970, 2013
71. Patole PS, Schubert S, Hildinger K, Khandoga S, Khandoga A, Segerer S, Henger A, Kretzler M, Werner M, Krombach F, Schlöndorff D, Anders HJ: Toll-like receptor-4: Renal cells and bone marrow cells signal for neutrophil recruitment during pyelonephritis. Kidney Int 68: 2582–2587, 2005
72. Hotchkiss RS, Coopersmith CM, McDunn JE, Ferguson TA: The sepsis seesaw: Tilting toward immunosuppression. Nat Med 15: 496–497, 2009
73. Chaput C, Zychlinsky A: Sepsis: The dark side of histones. Nat Med 15: 1245–1246, 2009
74. Xu J, Zhang X, Pelayo R, Monestier M, Ammollo CT, Semeraro F, Taylor FB, Esmon NL, Lupu F, Esmon CT: Extracellular histones are major mediators of death in sepsis. Nat Med 15: 1318–1321, 2009
75. Ekaney ML, Otto GP, Sossdorf M, Sponholz C, Boehringer M, Loesche W, Rittirsch D, Wilharm A, Kurzai O, Bauer M, Claus RA: Impact of plasma histones in human sepsis and their contribution to cellular injury and inflammation. Crit Care 18: 543, 2014
76. Abrams ST, Zhang N, Manson J, Liu T, Dart C, Baluwa F, Wang SS, Brohi K, Kipar A, Yu W, Wang G, Toh CH: Circulating histones are mediators of trauma-associated lung injury. Am J Respir Crit Care Med 187: 160–169, 2013
77. Lau A, Wang S, Jiang J, Haig A, Pavlosky A, Linkermann A, Zhang ZX, Jevnikar AM: RIPK3-mediated necroptosis promotes donor kidney inflammatory injury and reduces allograft survival. Am J Transplant 13: 2805–2818, 2013
78. Linkermann A, Hackl MJ, Kunzendorf U, Walczak H, Krautwald S, Jevnikar AM: Necroptosis in immunity and ischemia-reperfusion injury. Am J Transplant 13: 2797–2804, 2013
79. Linkermann A, Bräsen JH, Himmerkus N, Liu S, Huber TB, Kunzendorf U, Krautwald S: Rip1 (receptor-interacting protein kinase 1) mediates necroptosis and contributes to renal ischemia/reperfusion injury. Kidney Int 81: 751–761, 2012
80. Zhao H, Ning J, Lemaire A, Koumpa FS, Sun JJ, Fung A, Gu J, Yi B, Lu K, Ma D: Necroptosis and parthanatos are involved in remote lung injury after receiving ischemic renal allografts in rats. Kidney Int 87: 738–748, 2015
81. Xu Y, Ma H, Shao J, Wu J, Zhou L, Zhang Z, Wang Y, Huang Z, Ren J, Liu S, Chen X, Han J: A role for tubular necroptosis in cisplatin-induced AKI. J Am Soc Nephrol 2015, in press
82. Homsi E, Andreazzi DD, Faria JB, Janino P: TNF-α-mediated cardiorenal injury after rhabdomyolysis in rats. Am J Physiol Renal Physiol 308: F1259–F1267, 2014
83. Linkermann A, Heller JO, Prókai A, Weinberg JM, De Zen F, Himmerkus N, Szabó AJ, Bräsen JH, Kunzendorf U, Krautwald S: The RIP1-kinase inhibitor necrostatin-1 prevents osmotic nephrosis and contrast-induced AKI in mice. J Am Soc Nephrol 24: 1545–1557, 2013
84. Hakroush S, Cebulla A, Schaldecker T, Behr D, Mundel P, Weins A: Extensive podocyte loss triggers a rapid parietal epithelial cell response. J Am Soc Nephrol 25: 927–938, 2014
85. Jang HR, Rabb H: Immune cells in experimental acute kidney injury. Nat Rev Nephrol 11: 88–101, 2015
86. Summers SA, Chan J, Gan PY, Dewage L, Nozaki Y, Steinmetz OM, Nikolic-Paterson DJ, Kitching AR, Holdsworth SR: Mast cells mediate acute kidney injury through the production of TNF. J Am Soc Nephrol 22: 2226–2236, 2011
87. Xu C, Chang A, Hack BK, Eadon MT, Alper SL, Cunningham PN: TNF-mediated damage to glomerular endothelium is an important determinant of acute kidney injury in sepsis. Kidney Int 85: 72–81, 2014
88. Mulay SR, Kulkarni OP, Rupanagudi KV, Migliorini A, Darisipudi MN, Vilaysane A, Muruve D, Shi Y, Munro F, Liapis H, Anders HJ: Calcium oxalate crystals induce renal inflammation by NLRP3-mediated IL-1β secretion. J Clin Invest 123: 236–246, 2013
89. Vanden Berghe T, Linkermann A: Take my breath away: Necrosis in kidney transplants kills the lungs! Kidney Int 87: 680–682, 2015
90. McAdoo SP, Tanna A, Randone O, Tam FW, Tarzi RM, Levy JB, Griffith M, Lightstone L, Cook HT, Cairns T, Pusey CD: Necrotizing and crescentic glomerulonephritis presenting with preserved renal function in patients with underlying multisystem autoimmune disease: A retrospective case series. Rheumatology (Oxford) 54: 1025–1032, 2014
91. Fujita E, Nagahama K, Shimizu A, Aoki M, Higo S, Yasuda F, Mii A, Fukui M, Kaneko T, Tsuruoka S: Glomerular capillary and endothelial cell injury is associated with the formation of necrotizing and crescentic lesions in crescentic glomerulonephritis. J Nippon Med Sch 82: 27–35, 2015
92. Kumar SV, Kulkarni OP, Mulay SR, Darisipudi MN, Romoli S, Thomasova D, Scherbaum CR, Hohenstein B, Hugo C, Müller S, Liapis H, Anders HJ: Neutrophil extracellular trap-related extracellular histones cause vascular necrosis in severe GN. J Am Soc Nephrol 2015, in press
93. Kessenbrock K, Krumbholz M, Schönermarck U, Back W, Gross WL, Werb Z, Gröne HJ, Brinkmann V, Jenne DE: Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med 15: 623–625, 2009
94. Schreiber A, Xiao H, Jennette JC, Schneider W, Luft FC, Kettritz R: C5a receptor mediates neutrophil activation and ANCA-induced glomerulonephritis. J Am Soc Nephrol 20: 289–298, 2009
95. Vielhauer V, Stavrakis G, Mayadas TN: Renal cell-expressed TNF receptor 2, not receptor 1, is essential for the development of glomerulonephritis. J Clin Invest 115: 1199–1209, 2005
96. Tsumiyama K, Hashiramoto A, Takimoto M, Tsuji-Kawahara S, Miyazawa M, Shiozawa S: IFN-γ-producing effector CD8 T lymphocytes cause immune glomerular injury by recognizing antigen presented as immune complex on target tissue. J Immunol 191: 91–96, 2013
97. Ryu M, Migliorini A, Miosge N, Gross O, Shankland S, Brinkkoetter PT, Hagmann H, Romagnani P, Liapis H, Anders HJ: Plasma leakage through glomerular basement membrane ruptures triggers the proliferation of parietal epithelial cells and crescent formation in non-inflammatory glomerular injury. J Pathol 228: 482–494, 2012
98. Jennette JC, Falk RJ: Pathogenesis of antineutrophil cytoplasmic autoantibody-mediated disease. Nat Rev Rheumatol 10: 463–473, 2014
99. Charles Jennette J, Xiao H, Hu P: Complement in ANCA-associated vasculitis. Semin Nephrol 33: 557–564, 2013
100. George JN, Nester CM: Syndromes of thrombotic microangiopathy. N Engl J Med 371: 654–666, 2014
101. Engelmann B, Massberg S: Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol 13: 34–45, 2013
102. von Brühl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, Khandoga A, Tirniceriu A, Coletti R, Köllnberger M, Byrne RA, Laitinen I, Walch A, Brill A, Pfeiler S, Manukyan D, Braun S, Lange P, Riegger J, Ware J, Eckart A, Haidari S, Rudelius M, Schulz C, Echtler K, Brinkmann V, Schwaiger M, Preissner KT, Wagner DD, Mackman N, Engelmann B, Massberg S: Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 209: 819–835, 2012
103. Allam R, Scherbaum CR, Darisipudi MN, Mulay SR, Hägele H, Lichtnekert J, Hagemann JH, Rupanagudi KV, Ryu M, Schwarzenberger C, Hohenstein B, Hugo C, Uhl B, Reichel CA, Krombach F, Monestier M, Liapis H, Moreth K, Schaefer L, Anders HJ: Histones from dying renal cells aggravate kidney injury via TLR2 and TLR4. J Am Soc Nephrol 23: 1375–1388, 2012
104. Martinod K, Wagner DD: Thrombosis: Tangled up in NETs. Blood 123: 2768–2776, 2014
105. Allam R, Darisipudi MN, Tschopp J, Anders HJ: Histones trigger sterile inflammation by activating the NLRP3 inflammasome. Eur J Immunol 43: 3336–3342, 2013
106. Schirutschke H, Vogelbacher R, Stief A, Parmentier S, Daniel C, Hugo C: Injured kidney endothelium is only marginally repopulated by cells of extrarenal origin. Am J Physiol Renal Physiol 305: F1042–F1052, 2013
107. Bonventre JV, Yang L: Cellular pathophysiology of ischemic acute kidney injury. J Clin Invest 121: 4210–4221, 2011
108. Bao H, Ge Y, Wang Z, Zhuang S, Dworkin L, Peng A, Gong R: Delayed administration of a single dose of lithium promotes recovery from AKI. J Am Soc Nephrol 25: 488–500, 2014
109. Lech M, Römmele C, Gröbmayr R, Eka Susanti H, Kulkarni OP, Wang S, Gröne HJ, Uhl B, Reichel C, Krombach F, Garlanda C, Mantovani A, Anders HJ: Endogenous and exogenous pentraxin-3 limits postischemic acute and chronic kidney injury. Kidney Int 83: 647–661, 2013
110. Lech M, Gröbmayr R, Ryu M, Lorenz G, Hartter I, Mulay SR, Susanti HE, Kobayashi KS, Flavell RA, Anders HJ: Macrophage phenotype controls long-term AKI outcomes--kidney regeneration versus atrophy. J Am Soc Nephrol 25: 292–304, 2014
111. Zager RA, Johnson AC, Andress D, Becker K: Progressive endothelin-1 gene activation initiates chronic/end-stage renal disease following experimental ischemic/reperfusion injury. Kidney Int 84: 703–712, 2013
112. Eigenbrod T, Park JH, Harder J, Iwakura Y, Núñez G: Cutting edge: Critical role for mesothelial cells in necrosis-induced inflammation through the recognition of IL-1 alpha released from dying cells. J Immunol 181: 8194–8198, 2008
113. Lukens JR, Vogel P, Johnson GR, Kelliher MA, Iwakura Y, Lamkanfi M, Kanneganti TD: RIP1-driven autoinflammation targets IL-1α independently of inflammasomes and RIP3. Nature 498: 224–227, 2013
114. Vabulas RM, Ahmad-Nejad P, da Costa C, Miethke T, Kirschning CJ, Häcker H, Wagner H: Endocytosed HSP60s use toll-like receptor 2 (TLR2) and TLR4 to activate the toll/interleukin-1 receptor signaling pathway in innate immune cells. J Biol Chem 276: 31332–31339, 2001
115. Kakkar R, Lee RT: The IL-33/ST2 pathway: Therapeutic target and novel biomarker. Nat Rev Drug Discov 7: 827–840, 2008
116. Moussion C, Ortega N, Girard JP: The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: A novel ‘alarmin’? PLoS One 3: e3331, 2008
117. Wang C, Pan Y, Zhang QY, Wang FM, Kong LD: Quercetin and allopurinol ameliorate kidney injury in STZ-treated rats with regulation of renal NLRP3 inflammasome activation and lipid accumulation. PLoS One 7: e38285, 2012
118. Neumann K, Castiñeiras-Vilariño M, Höckendorf U, Hannesschläger N, Lemeer S, Kupka D, Meyermann S, Lech M, Anders HJ, Kuster B, Busch DH, Gewies A, Naumann R, Groß O, Ruland J: Clec12a is an inhibitory receptor for uric acid crystals that regulates inflammation in response to cell death. Immunity 40: 389–399, 2014
119. Biton S, Ashkenazi A: NEMO and RIP1 control cell fate in response to extensive DNA damage via TNF-α feedforward signaling. Cell 145: 92–103, 2011
120. Pobezinskaya YL, Liu Z: The role of TRADD in death receptor signaling. Cell Cycle 11: 871–876, 2012
121. Penfold SA, Coughlan MT, Patel SK, Srivastava PM, Sourris KC, Steer D, Webster DE, Thomas MC, MacIsaac RJ, Jerums G, Burrell LM, Cooper ME, Forbes JM: Circulating high-molecular-weight RAGE ligands activate pathways implicated in the development of diabetic nephropathy. Kidney Int 78: 287–295, 2010
122. Nawa Y, Kawahara K, Tancharoen S, Meng X, Sameshima H, Ito T, Masuda Y, Imaizumi H, Hashiguchi T, Maruyama I: Nucleophosmin may act as an alarmin: Implications for severe sepsis. J Leukoc Biol 86: 645–653, 2009
123. Tsai SY, Segovia JA, Chang TH, Morris IR, Berton MT, Tessier PA, Tardif MR, Cesaro A, Bose S: DAMP molecule S100A9 acts as a molecular pattern to enhance inflammation during influenza A virus infection: Role of DDX21-TRIF-TLR4-MyD88 pathway. PLoS Pathog 10: e1003848, 2014
124. Vogl T, Tenbrock K, Ludwig S, Leukert N, Ehrhardt C, van Zoelen MA, Nacken W, Foell D, van der Poll T, Sorg C, Roth J: Mrp8 and Mrp14 are endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-induced shock. Nat Med 13: 1042–1049, 2007
125. Yamasaki S, Ishikawa E, Sakuma M, Hara H, Ogata K, Saito T: Mincle is an ITAM-coupled activating receptor that senses damaged cells. Nat Immunol 9: 1179–1188, 2008
126. Ahrens S, Zelenay S, Sancho D, Hanč P, Kjær S, Feest C, Fletcher G, Durkin C, Postigo A, Skehel M, Batista F, Thompson B, Way M, Reis e Sousa C, Schulz O: F-actin is an evolutionarily conserved damage-associated molecular pattern recognized by DNGR-1, a receptor for dead cells. Immunity 36: 635–645, 2012
127. Lu CC, Wu TS, Hsu YJ, Chang CJ, Lin CS, Chia JH, Wu TL, Huang TT, Martel J, Ojcius DM, Young JD, Lai HC: NK cells kill mycobacteria directly by releasing perforin and granulysin. J Leukoc Biol 96: 1119–1129, 2014
128. Steinhoff M, Vergnolle N, Young SH, Tognetto M, Amadesi S, Ennes HS, Trevisani M, Hollenberg MD, Wallace JL, Caughey GH, Mitchell SE, Williams LM, Geppetti P, Mayer EA, Bunnett NW: Agonists of proteinase-activated receptor 2 induce inflammation by a neurogenic mechanism. Nat Med 6: 151–158, 2000
129. Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, Brohi K, Itagaki K, Hauser CJ: Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 464: 104–107, 2010
130. He S, Peng Q, Walls AF: Potent induction of a neutrophil and eosinophil-rich infiltrate in vivo by human mast cell tryptase: Selective enhancement of eosinophil recruitment by histamine. J Immunol 159: 6216–6225, 1997
131. Solini A, Menini S, Rossi C, Ricci C, Santini E, Blasetti Fantauzzi C, Iacobini C, Pugliese G: The purinergic 2X7 receptor participates in renal inflammation and injury induced by high-fat diet: Possible role of NLRP3 inflammasome activation. J Pathol 231: 342–353, 2013
132. McDonald B, Pittman K, Menezes GB, Hirota SA, Slaba I, Waterhouse CC, Beck PL, Muruve DA, Kubes P: Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science 330: 362–366, 2010
133. Pullerits R, Bokarewa M, Jonsson IM, Verdrengh M, Tarkowski A: Extracellular cytochrome c, a mitochondrial apoptosis-related protein, induces arthritis. Rheumatology (Oxford) 44: 32–39, 2005
134. Savarese E, Chae OW, Trowitzsch S, Weber G, Kastner B, Akira S, Wagner H, Schmid RM, Bauer S, Krug A: U1 small nuclear ribonucleoprotein immune complexes induce type I interferon in plasmacytoid dendritic cells through TLR7. Blood 107: 3229–3234, 2006
135. Christensen SR, Kashgarian M, Alexopoulou L, Flavell RA, Akira S, Shlomchik MJ: Toll-like receptor 9 controls anti-DNA autoantibody production in murine lupus. J Exp Med 202: 321–331, 2005
136. Lau CM, Broughton C, Tabor AS, Akira S, Flavell RA, Mamula MJ, Christensen SR, Shlomchik MJ, Viglianti GA, Rifkin IR, Marshak-Rothstein A: RNA-associated autoantigens activate B cells by combined B cell antigen receptor/Toll-like receptor 7 engagement. J Exp Med 202: 1171–1177, 2005
137. Hägele H, Allam R, Pawar RD, Reichel CA, Krombach F, Anders HJ: Double-stranded DNA activates glomerular endothelial cells and enhances albumin permeability via a toll-like receptor-independent cytosolic DNA recognition pathway. Am J Pathol 175: 1896–1904, 2009
138. Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, Latz E, Fitzgerald KA: AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 458: 514–518, 2009
139. Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J, Frazier A, Yang H, Ivanova S, Borovikova L, Manogue KR, Faist E, Abraham E, Andersson J, Andersson U, Molina PE, Abumrad NN, Sama A, Tracey KJ: HMG-1 as a late mediator of endotoxin lethality in mice. Science 285: 248–251, 1999
140. Julian MW, Shao G, Bao S, Knoell DL, Papenfuss TL, VanGundy ZC, Crouser ED: Mitochondrial transcription factor A serves as a danger signal by augmenting plasmacytoid dendritic cell responses to DNA. J Immunol 189: 433–443, 2012
141. Darisipudi MN, Thomasova D, Mulay SR, Brech D, Noessner E, Liapis H, Anders HJ: Uromodulin triggers IL-1β-dependent innate immunity via the NLRP3 inflammasome. J Am Soc Nephrol 23: 1783–1789, 2012
142. Säemann MD, Weichhart T, Zeyda M, Staffler G, Schunn M, Stuhlmeier KM, Sobanov Y, Stulnig TM, Akira S, von Gabain A, von Ahsen U, Hörl WH, Zlabinger GJ: Tamm-Horsfall glycoprotein links innate immune cell activation with adaptive immunity via a Toll-like receptor-4-dependent mechanism. J Clin Invest 115: 468–475, 2005
143. Najjar M, Suebsuwong C, Ray SS, Thapa RJ, Maki JL, Nogusa S, Shah S, Saleh D, Gough PJ, Bertin J, Yuan J, Balachandran S, Cuny GD, Degterev A: Structure guided design of potent and selective ponatinib-based hybrid inhibitors for RIPK1. Cell Rep 10: 1850–1860, 2015
144. Mandal P, Berger SB, Pillay S, Moriwaki K, Huang C, Guo H, Lich JD, Finger J, Kasparcova V, Votta B, Ouellette M, King BW, Wisnoski D, Lakdawala AS, DeMartino MP, Casillas LN, Haile PA, Sehon CA, Marquis RW, Upton J, Daley-Bauer LP, Roback L, Ramia N, Dovey CM, Carette JE, Chan FK, Bertin J, Gough PJ, Mocarski ES, Kaiser WJ: RIP3 induces apoptosis independent of pronecrotic kinase activity. Mol Cell 56: 481–495, 2014
145. Li JX, Feng JM, Wang Y, Li XH, Chen XX, Su Y, Shen YY, Chen Y, Xiong B, Yang CH, Ding J, Miao ZH: The B-Raf(V600E) inhibitor dabrafenib selectively inhibits RIP3 and alleviates acetaminophen-induced liver injury. Cell Death Dis 5: e1278, 2014
146. Su L, Quade B, Wang H, Sun L, Wang X, Rizo J: A plug release mechanism for membrane permeation by MLKL. Structure 22: 1489–1500, 2014
147. Kapoor K, Singla E, Sahu B, Naura AS: PARP inhibitor, olaparib ameliorates acute lung and kidney injury upon intratracheal administration of LPS in mice. Mol Cell Biochem 400: 153–162, 2015
148. Arumugam TV, Shiels IA, Strachan AJ, Abbenante G, Fairlie DP, Taylor SM: A small molecule C5a receptor antagonist protects kidneys from ischemia/reperfusion injury in rats. Kidney Int 63: 134–142, 2003
149. Cofiell R, Kukreja A, Bedard K, Yan Y, Mickle AP, Ogawa M, Bedrosian CL, Faas SJ: Eculizumab reduces complement activation, inflammation, endothelial damage, thrombosis, and renal injury markers in aHUS. Blood 125: 3253–3262, 2015
150. Hoehlig K, Maasch C, Shushakova N, Buchner K, Huber-Lang M, Purschke WG, Vater A, Klussmann S: A novel C5a-neutralizing mirror-image (l-)aptamer prevents organ failure and improves survival in experimental sepsis. Mol Ther 21: 2236–2246, 2013
151. Khan MA, Maasch C, Vater A, Klussmann S, Morser J, Leung LL, Atkinson C, Tomlinson S, Heeger PS, Nicolls MR: Targeting complement component 5a promotes vascular integrity and limits airway remodeling. Proc Natl Acad Sci U S A 110: 6061–6066, 2013
152. Pauly D, Nagel BM, Reinders J, Killian T, Wulf M, Ackermann S, Ehrenstein B, Zipfel PF, Skerka C, Weber BH: A novel antibody against human properdin inhibits the alternative complement system and specifically detects properdin from blood samples. PLoS One 9: e96371, 2014
153. Kulkarni O, Eulberg D, Selve N, Zöllner S, Allam R, Pawar RD, Pfeiffer S, Segerer S, Klussmann S, Anders HJ: Anti-Ccl2 Spiegelmer permits 75% dose reduction of cyclophosphamide to control diffuse proliferative lupus nephritis and pneumonitis in MRL-Fas(lpr) mice. J Pharmacol Exp Ther 328: 371–377, 2009
154. Furuichi K, Wada T, Iwata Y, Kitagawa K, Kobayashi K, Hashimoto H, Ishiwata Y, Asano M, Wang H, Matsushima K, Takeya M, Kuziel WA, Mukaida N, Yokoyama H: CCR2 signaling contributes to ischemia-reperfusion injury in kidney. J Am Soc Nephrol 14: 2503–2515, 2003
155. Nedjai B, Viney JM, Li H, Hull C, Anderson CA, Horie T, Horuk R, Vaidehi N, Pease JE: CXCR3 antagonist VUF10085 binds to an intrahelical site distinct from that of the broad spectrum antagonist TAK-779. Br J Pharmacol 172: 1822–1833, 2015
156. Naidu BV, Farivar AS, Woolley SM, Grainger D, Verrier ED, Mulligan MS: Novel broad-spectrum chemokine inhibitor protects against lung ischemia-reperfusion injury. J Heart Lung Transplant 23: 128–134, 2004
157. Ni J, Zhu YN, Zhong XG, Ding Y, Hou LF, Tong XK, Tang W, Ono S, Yang YF, Zuo JP: The chemokine receptor antagonist, TAK-779, decreased experimental autoimmune encephalomyelitis by reducing inflammatory cell migration into the central nervous system, without affecting T cell function. Br J Pharmacol 158: 2046–2056, 2009
158. Reckless J, Tatalick LM, Grainger DJ: The pan-chemokine inhibitor NR58-3.14.3 abolishes tumour necrosis factor-alpha accumulation and leucocyte recruitment induced by lipopolysaccharide in vivo. Immunology 103: 244–254, 2001
159. Aringer M, Steiner G, Graninger WB, Höfler E, Steiner CW, Smolen JS: Effects of short-term infliximab therapy on autoantibodies in systemic lupus erythematosus. Arthritis Rheum 56: 274–279, 2007
160. Scemla A, Gerber S, Duquesne A, Parize P, Martinez F, Anglicheau D, Snanoudj R, Zuber M, Bougnoux ME, Legendre C, Lortholary O: Dramatic improvement of severe cryptococcosis-induced immune reconstitution syndrome with adalimumab in a renal transplant recipient. Am J Transplant 15: 560–564, 2015
161. Shealy DJ, Cai A, Staquet K, Baker A, Lacy ER, Johns L, Vafa O, Gunn G 3rd, Tam S, Sague S, Wang D, Brigham-Burke M, Dalmonte P, Emmell E, Pikounis B, Bugelski PJ, Zhou H, Scallon BJ, Giles-Komar J: Characterization of golimumab, a human monoclonal antibody specific for human tumor necrosis factor α. MAbs 2: 428–439, 2010
162. Bihorel S, Fiedler-Kelly J, Ludwig E, Sloan-Lancaster J, Raddad E: Population pharmacokinetic modeling of LY2189102 after multiple intravenous and subcutaneous administrations. AAPS J 16: 1009–1017, 2014
163. Chakraborty A, Tannenbaum S, Rordorf C, Lowe PJ, Floch D, Gram H, Roy S: Pharmacokinetic and pharmacodynamic properties of canakinumab, a human anti-interleukin-1β monoclonal antibody. Clin Pharmacokinet 51: e1–e18, 2012
164. Geiler J, McDermott MF: Gevokizumab, an anti-IL-1β mAb for the potential treatment of type 1 and 2 diabetes, rheumatoid arthritis and cardiovascular disease. Curr Opin Mol Ther 12: 755–769, 2010
165. Goh AX, Bertin-Maghit S, Ping Yeo S, Ho AW, Derks H, Mortellaro A, Wang CI: A novel human anti-interleukin-1β neutralizing monoclonal antibody showing in vivo efficacy. MAbs 6: 765–773, 2014
166. McDermott MF: Rilonacept in the treatment of chronic inflammatory disorders. Drugs Today (Barc) 45: 423–430, 2009
167. Ando K, Takahashi F, Kato M, Kaneko N, Doi T, Ohe Y, Koizumi F, Nishio K, Takahashi K: Tocilizumab, a proposed therapy for the cachexia of Interleukin6-expressing lung cancer. PLoS One 9: e102436, 2014
168. Arima H, Hanada M, Hayasaka T, Masaki N, Omura T, Xu D, Hasegawa T, Togawa D, Yamato Y, Kobayashi S, Yasuda T, Matsuyama Y, Setou M: Blockade of IL-6 signaling by MR16-1 inhibits reduction of docosahexaenoic acid-containing phosphatidylcholine levels in a mouse model of spinal cord injury. Neuroscience 269: 1–10, 2014
169. Liang B, Gardner DB, Griswold DE, Bugelski PJ, Song XY: Anti-interleukin-6 monoclonal antibody inhibits autoimmune responses in a murine model of systemic lupus erythematosus. Immunology 119: 296–305, 2006
170. Straub RH, Härle P, Yamana S, Matsuda T, Takasugi K, Kishimoto T, Nishimoto N: Anti-interleukin-6 receptor antibody therapy favors adrenal androgen secretion in patients with rheumatoid arthritis: A randomized, double-blind, placebo-controlled study. Arthritis Rheum 54: 1778–1785, 2006
171. Langley RG, Elewski BE, Lebwohl M, Reich K, Griffiths CE, Papp K, Puig L, Nakagawa H, Spelman L, Sigurgeirsson B, Rivas E, Tsai TF, Wasel N, Tyring S, Salko T, Hampele I, Notter M, Karpov A, Helou S, Papavassilis CERASURE Study GroupFIXTURE Study Group: Secukinumab in plaque psoriasis--results of two phase 3 trials. N Engl J Med 371: 326–338, 2014
172. Leonardi C, Matheson R, Zachariae C, Cameron G, Li L, Edson-Heredia E, Braun D, Banerjee S: Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. N Engl J Med 366: 1190–1199, 2012
173. Patel DD, Lee DM, Kolbinger F, Antoni C: Effect of IL-17A blockade with secukinumab in autoimmune diseases. Ann Rheum Dis 72[Suppl 2]: ii116–ii123, 2013
174. Lassen UN, Meulendijks D, Siu LL, Karanikas V, Mau-Sorensen M, Schellens JH, Jonker DJ, Hansen AR, Simcox ME, Schostack KJ, Bottino D, Zhong H, Roessler M, Vega-Harring SM, Jarutat T, Geho D, Wang K, DeMario M, Goss GD: A phase I monotherapy study of RG7212, a first-in-class monoclonal antibody targeting TWEAK signaling in patients with advanced cancers. Clin Cancer Res 21: 258–266, 2015
175. Xia Y, Campbell SR, Broder A, Herlitz L, Abadi M, Wu P, Michaelson JS, Burkly LC, Putterman C: Inhibition of the TWEAK/Fn14 pathway attenuates renal disease in nephrotoxic serum nephritis. Clin Immunol 145: 108–121, 2012
176. Michaelson JS, Wisniacki N, Burkly LC, Putterman C: Role of TWEAK in lupus nephritis: A bench-to-bedside review. J Autoimmun 39: 130–142, 2012
177. Neria F, Castilla MA, Sanchez RF, Gonzalez Pacheco FR, Deudero JJ, Calabia O, Tejedor A, Manzarbeitia F, Ortiz A, Caramelo C: Inhibition of JAK2 protects renal endothelial and epithelial cells from oxidative stress and cyclosporin A toxicity. Kidney Int 75: 227–234, 2009
178. Pang M, Ma L, Gong R, Tolbert E, Mao H, Ponnusamy M, Chin YE, Yan H, Dworkin LD, Zhuang S: A novel STAT3 inhibitor, S3I-201, attenuates renal interstitial fibroblast activation and interstitial fibrosis in obstructive nephropathy. Kidney Int 78: 257–268, 2010
179. Mascarenhas J, Hoffman R: Ruxolitinib: The first FDA approved therapy for the treatment of myelofibrosis. Clin Cancer Res 18: 3008–3014, 2012
180. Chiricozzi A, Faleri S, Saraceno R, Bianchi L, Buonomo O, Chimenti S, Chimenti MS: Tofacitinib for the treatment of moderate-to-severe psoriasis. Expert Rev Clin Immunol 11: 443–455, 2015
181. Arslan F, Houtgraaf JH, Keogh B, Kazemi K, de Jong R, McCormack WJ, O’Neill LA, McGuirk P, Timmers L, Smeets MB, Akeroyd L, Reilly M, Pasterkamp G, de Kleijn DP: Treatment with OPN-305, a humanized anti-Toll-Like receptor-2 antibody, reduces myocardial ischemia/reperfusion injury in pigs. Circ Cardiovasc Interv 5: 279–287, 2012
182. Reilly M, Miller RM, Thomson MH, Patris V, Ryle P, McLoughlin L, Mutch P, Gilboy P, Miller C, Broekema M, Keogh B, McCormack W, van de Wetering de Rooij J: Randomized, double-blind, placebo-controlled, dose-escalating phase I, healthy subjects study of intravenous OPN-305, a humanized anti-TLR2 antibody. Clin Pharmacol Ther 94: 593–600, 2013
183. Suárez-Fariñas M, Arbeit R, Jiang W, Ortenzio FS, Sullivan T, Krueger JG: Suppression of molecular inflammatory pathways by Toll-like receptor 7, 8, and 9 antagonists in a model of IL-23-induced skin inflammation. PLoS One 8: e84634, 2013
184. Lamkanfi M, Mueller JL, Vitari AC, Misaghi S, Fedorova A, Deshayes K, Lee WP, Hoffman HM, Dixit VM: Glyburide inhibits the Cryopyrin/Nalp3 inflammasome. J Cell Biol 187: 61–70, 2009
185. Stack JH, Beaumont K, Larsen PD, Straley KS, Henkel GW, Randle JC, Hoffman HM: IL-converting enzyme/caspase-1 inhibitor VX-765 blocks the hypersensitive response to an inflammatory stimulus in monocytes from familial cold autoinflammatory syndrome patients. J Immunol 175: 2630–2634, 2005
186. Coll RC, Robertson AA, Chae JJ, Higgins SC, Muñoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, Croker DE, Butler MS, Haneklaus M, Sutton CE, Núñez G, Latz E, Kastner DL, Mills KH, Masters SL, Schroder K, Cooper MA, O’Neill LA: A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med 21: 248–255, 2015
187. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D’Agostino D, Planavsky N, Lupfer C, Kanneganti TD, Kang S, Horvath TL, Fahmy TM, Crawford PA, Biragyn A, Alnemri E, Dixit VD: The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med 21: 263–269, 2015