Abstract
Background: The liver has a number of functions in innate immunity. These functions predispose the liver to innate immune-mediated liver injury when inflammation goes unchecked. Significant progress has been made in the last 25 years on sterile inflammatory liver injury in a number of models; however, a great deal of controversy and many questions about the nature of sterile inflammation still exist.
Aim: The goal of this article is to review sterile inflammatory liver injury using both a basic approach to what constitutes the inflammatory injury, and through examination of current models of liver injury and inflammation. This information will be tied to human patient conditions when appropriate.
Relevance for patients: Inflammation is one of the most critical factors for managing in-patient liver disease in a number of scenarios. More information is needed for both scientists and clinicians to develop rational treatments.
Keywords: sterile inflammation, liver, neutrophil, monocyte, mechanisms, cholestasis, ischemia, reperfusion, acetaminophen
1. Introduction
Liver associated morbidity and mortality is a major concern globally. The liver has a number of important functions in the body. One of the major functions of the liver is related to innate immunity, as the liver holds the largest macrophage population in the body, termed Kupffer cells (KCs). Diseases that result in acute liver injury, activate the liver’s innate immune functions, sometimes pathologically, even in the absence of an infection. Activation of KCs in the liver results in release of reactive oxygen species (ROS), and secretion of an array of cytokines that recruit other potentially cytotoxic inflammatory cells, including neutrophils, monocytes, T-cells, and NK/NKT cells [1]. The idea of innate immune-mediated liver injury in the absence of an infection has become a highly studied topic in liver biology over the last 25 years. While in some models there is nearly universal agreement that inflammation is a direct, pathological component of the injury process, there are other models wherein there is considerable debate over the role of sterile inflammation [2-5]. In a number of diseases, it is becoming increasingly understood that inflammation may actually play a beneficial, or at least a necessary role, as both acute and chronic liver failure can dramatically increase susceptibility to infection. The purpose of this review is to discuss mechanisms of sterile inflammatory liver injury and further discuss the role of sterile inflammation in clinically relevant models of acute liver injury.
2. Sterile inflammation – mechanisms of inflammation and injury
2.1. Release of damage associated molecular patterns
Sterile inflammation is a common outcome of a number of different clinical liver disorders. Sterile inflammation occurs in solid organs, such as the liver, when an organized inflammatory response occurs in the absence of any infection. While infectious inflammatory responses are driven by the detection and engulfment of infectious species, or pathogen associated molecular patterns (PAMPs), such as lipopolysaccharide and flagellin, sterile inflammatory responses are driven by local release of damage associated molecular patterns (DAMPs) from dying, necrotic cells [1, 6]. As cells die, their plasma membrane becomes permeable to intracellular contents [7]. In the case of outright necrosis, rupture of the cell occurs, and intracellular contents, or DAMPs, spill into the surrounding area. In the classic model of sterile inflammation these molecules then bind to pattern recognition receptors such as Toll-like receptors (TLRs), which induce pro-inflammatory cytokine formation [3,6,8]. Other DAMPs can either prime, or activate the multimeric protein complex called the inflammasome [9,10]. A number of damage associated molecular patterns have been identified including: ATP [11], high mobility group box-1 (HMGB1) [12], mitochondrial DNA and nuclear DNA [13,14], uric acid [15], hyaluronan [16], histones [17, 18] and more. Additionally, some molecules, such as bile acids (BA), have a number of similar qualities to known DAMPs, and are released during cell death [19]. Moreover, as necrosis results in total cellular breakdown, any number of cellular components could also be functioning as DAMPs. Thus, the sterile inflammatory response during liver injury is initiated by breakdown of the hepatocyte membrane and release of these constituents. The presence and the detection of these molecules drives the inflammatory response.
2.2. Activation of Kupffer cells
KCs are an endogenous macrophage population present in the sinusoidal space of the liver vasculature. Release of DAMPs activates a number of potential receptors on KCs. KCs express TLRs including TLR3, TLR4, TLR9 and more [20]. Additionally, KCs express purinergic receptors [21], the hyaluronan receptor CD44 [22], the receptor for advanced glycation end products (RAGE), which acts as a receptor for HMGB1 [23], and potentially other unknown receptors that also mediate these effects. Binding of these receptors serves to activate KCs resulting in local production of a number of cytokines and chemokines, in addition to the NF-κB pathway which causes upregulation of a multiple pro-inflammatory proteins [20]. Release of TNF-α from KCs can also activate NF-κB in hepatocytes, which upregulates adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) in models of liver injury [24]. In addition, in mice, many of these cytokines and chemokines such as macrophage inflammatory protein-2 (MIP-2), mouse keratinocyte factor (mKC), and macrophage chemoattractant protein-1 (MCP-1) have their own receptors i.e. CXC chemokine receptor 1 (CXCR1), CXC chemokine receptor 2 (CXCR2), and C-C chemokine receptor 2 (CCR2), respectively, all of which have been implicated in specific liver diseases [25-30]. Parallel activation of a number of these pathways likely occurs and amplifies the signals. All of these agents serve to increase recruitment of other inflammatory cells such as neutrophils and monocytes in addition to stimulating other resident macrophages.
Activation of TLR signaling on KCs also initiates the two-step process responsible for activating the inflammasome [31]. The inflammasome is a multimeric complex of proteins that initiates the activation of caspase-1 from pro-caspase 1, and subsequent cleavage of pro-interleukin-1ß (pro-IL-1ß) to IL-1ß, which serves as a potent pro-inflammatory cytokine [9]. While a number of different proteins can initiate formation of an inflammasome-like complex, the classical and most studied example of the inflammasome in the liver is the Nalp3 inflammasome [3]. Activation of TLR4, as an example, increases expression of pro-IL-1ß through the NF-κB pathway [8]. When KCs encounter DAMP signals, such as ATP, which activates the purinergic receptor 2X7 (P2X7), a ligand-gated ion channel, it causes release of intracellular potassium and activation of caspase-1 by the NLRP3 inflammasome. The activated caspase-1 cleaves pro-IL-1ß to form IL-1ß and then IL-1ß is released into the blood [9]. The NLRP3 inflammasome has been implicated in a number of models of liver injury, and will be discussed further in the context of specific diseases.
2.3. Initiation of inflammatory cell death – cytotoxic mediators of inflammation
Increased blood levels of cytokines and chemokines released from KCs result in rapid recruitment of multiple innate immune cells to the liver. The discussion will be mainly focused on the two cell populations that accumulate most commonly and in the highest numbers: neutrophils and monocytes/monocyte-derived macrophages. A brief section will highlight potential roles of other inflammatory cells as well. The liver recruits high numbers of both cell populations rapidly in response to injury. In models such as cholestatic liver injury [32,33], acetaminophen- (APAP) induced liver injury [34], hepatic ischemia-reperfusion liver injury [35], binge alcohol administration to chronically alcohol fed mice [36], and endotoxemia [37] there is rapid recruitment of neutrophils to the liver within 3-9 hours post initiation of toxicity. Monocytes typically follow shortly afterwards in some of these models [28]; however, it should be noted that KCs can also initiate toxicity and are endogenously present. Specific toxicity mechanisms of these two populations will be discussed in detail.
2.3.1. Kupffer cells and monocyte-derived macrophages
KCs are endogenously present in the liver and are a primary innate immune cell type responsible for considerable bacterial, fungal, and viral clearance [38,39]. However, over-activation of KCs can be detrimental to hepatocyte function. KCs produce ROS through the enzyme NADPH oxidase (NOX2). Upon stimulation, KCs induce a vascular oxidant stress as evidenced by major increases in extracellular glutathione disulfide [40]. This is directly linked to hepatocyte cell death through activation of complement [41]. In support of these data, inactivation or removal of KCs with a number of differrent methods including administration of methyl palmitate, gadolinium chloride, or clodronate liposomes are protective against injury [40,42-46]. Given its established role in numerous diseases, release of ROS remains the most likely mechanism of toxicity from KCs.
In addition to production of ROS, KCs are frequently noted as potent producer of tumor necrosis factor-α (TNF-α), despite the fact that in a majority of liver diseases the actual increase in serum TNF-α levels is very limited. While there are models that are unequivocally mediated by release of TNF-α from KCs [47,48], in a majority of models, the release of TNF-α alone is insufficient to induce hepatocyte apoptosis. This is due to the fact that hepatocytes are highly resistant to TNF-α, and only undergo TNF-α mediated cell death in the presence of inhibition of nuclear factor kappa light chain enhancer of B cells (NF-κB) [49]. Human hepatocytes and human hepatocyte-like cell lines may be even more resistant to TNF-α, as they require even higher concentrations of the cytokine to induce apoptosis [50,51]. As such, TNF-α is likely less than oxidative stress to be relevant as a cytotoxic mediator of KC.
Monocyte-derived macrophages (MoMF) are a well-studied cellular population in the liver. During liver injury, outside monocytes get recruited to the liver, wherein they take on a macrophage-like phenotype with similar functions. Increasingly, it is being recognized that these monocytes fit one of two very different roles. Classically activated M1 or proinflammatory macrophages generally secrete pro-inflammatory cytokines, are Ly6Chi, exacerbate inflammation, and in some cases contribute to cell death [52]. Alternately activated M2 or anti-inflammatory type macrophages secrete large amounts of anti-inflammatory and pro-resolving cytokines like interleukin-10 (IL-10), are stimulated by IL-4, and serve to remove necrotic debris and resolve inflammation in preparation for regeneration [52,53]. Accumulation of M1 type macrophages in the liver would indicate monocytes could potentially cause inflammatory liver injury; whereas accumulation of M2 macrophages would suggest wound healing is beginning to occur and the injury phase is declining [54]. MoMF express the same subset of enzymes necessary for production of oxidative stress as KCs, and are hypothesized to produce toxicity in the same fashion.
Finally, it should be noted that despite its acknowledged roles in multiple disease states, modulation of the innate immune system, especially endogenous KCs, is risky as a therapeutic target. The primary purpose for KCs and other innate immune cells remains the clearance of bacteria. In patients with alcoholic hepatitis, blockade of TNF-α signaling resulted in increased risk of sepsis, and caused excess mortality [55,56]. Similarly, despite the fact that acute liver failure patients suffer from dramatically reduced liver function, a large percentage of patients do not die of hepatic insufficiency, but rather from sepsis associated with liver failure. Given this information, blockade of inflammation remains a controversial target at best in liver disease, and should only be pursued in patients without an active infection, or evidence of systemic inflammatory response syndrome (SIRS). Moreover, more specific therapeutic targets are needed that could potentially reduce the adverse effects of major immune activation, without inhibiting host defense. In spite of this, understanding sterile inflammation in different models is imperative to understanding disease progression and mortality in patients.
2.3.2. Neutrophils
Neutrophil-mediated solid organ injury has been studied extensively in liver and other organs, and requires multiple steps [2,57]. Neutrophil recruitment is necessary, and in high numbers neutrophils can generate sufficient oxidative stress to kill hepatocytes [58]. This is carried out by production of cytokines and chemokines in both mice and humans. Next, neutrophils have to adhere to the vasculature and extravasate into the parenchyma [59,60]. In most organs, neutrophils roll along post-capillary venules through interactions between selectins on endothelial cells and their ligands expressed on neutrophils [61]. Expression of these molecules is regulated by cytokines, and thus cytokines serve not only to recruit neutrophils, but to promote their attachment to the endothelium for transendothelial migration. However, in the liver, only limited neutrophil rolling is present in post-sinusoidal venules [62,63]. In contrast, most neutrophil extravasation occurs from the sinusoids [64]. Both E-selectin [36,65], and their ligands [66], have been suggested to be involved in neutrophil mediated liver injury in different models, indicating E-selectin may still be involved in neutrophil transendothelial migration in sinusoids. Alternatively, the hepatoprotection by blocking selectins may be a secondary effect caused by reduced injury in the intestine in certain models such as ischemia-reperfusion injury [67]. Furthermore, it was hypothesized that P-selectin-dependent platelet accumulation during hepatic ischemia-reperfusion may cause microvascular perfusion failure and facilitate neutrophil accumulation in sinusoids [68,69]. However, the pathophysio-logical role of platelet-neutrophil interactions has recently been questioned [70].
Independent of the controversial role of selectins in the liver, neutrophil transmigration involves mainly interactions of LFA-1 (CD11a/CD18) or Mac-1 (CD11b/CD18) on neutrophils and ICAM-1 on sinusoidal endothelial cells [71]. Once neutrophils transmigrate past the endothelium, interactions between β2 integrins (LFA-1/Mac-1) and ICAM-1 on the target cells mediate neutrophil adhesion to target cells [72,73] including hepatocytes [74]. Firm adhesion to hepatocytes is required for toxicity as this triggers the long-lasting oxidant stress and the diffusion of ROS into hepatocytes [2,57]. Furthermore, local anti-proteases nullify the toxic potential of neutrophil-derived proteases without very close physical proximity [75]. Neutrophil cytotoxicity requires upregulation of CD11b on neutrophils, and expression of the Mac-1 complex on the cellular surface of neutrophils [72], in addition to ICAM-1 upregulation on hepatocytes [76,77]. Blockade or deletion of either adhesion component results in a significant decrease in neutrophil toxicity in the liver [32,35,41] as does deletion of ICAM-1 [76-78]. As such, CD11b expression at the cellular surface is a common indicator of neutrophil activation [79]. Finally, when firmly adhered, neutrophils are capable of killing hepatocytes through degranulation and release of a number of cytotoxic species [80,81]. Neutrophils produce ROS through the NADPH oxidase system, which catalyzes the formation of superoxide by transferring electrons from NADPH to molecular oxygen and initiates toxicity [81]. Superoxide dismutates spontaneously or catalyzed by superoxide dismutase (SOD) into molecular oxygen and hydrogen peroxide, which can be toxic to hepatocytes [58]. In addition, neutrophils are the sole producer in mammalian biology of the enzyme myeloperoxidase.
Myeloperoxidase is present in neutrophil granules in the interior of the cell. Upon neutrophil activation, this enzyme is released into the local space where it reacts with chloride anion and hydrogen peroxide produced from NADPH oxidase to generate hypochlorous acid, a potent oxidant [82]. Hypochlorous acid is highly toxic and rapidly adducts tyrosine, which causes significant oxidative stress and protein dysfunction [83]. These adducts are readily identifiable with antibodies against chlorotyrosine, and are present in a number of models of neutrophil mediated liver injury [32,78,81,84]. Additionally, neutrophils express elastase in their azuophilic granules, which can result in cellular dysfunction in some disease, such as emphysema of the lungs [85]. Elastase is a serine-protease thought to be involved in breakdown of extracellular matrix for neutrophil movement. Neutrophil elastase has been suggested to potentially be involved in liver injury [86]; although, mechanisms of how elastase could damage hepatocytes have not been elucidated, nor has there been any update on how elastase escapes the effect of local anti-proteases in the liver [75].
Recently it was identified that neutrophils are capable of secreting a mixture of granule proteins and neutrophil DNA, that form a mesh-like fibrous network known as a neutrophil extracellular trap (NET) [87]. These traps have a clear role in pathogen removal, wherein pathogens are trapped within the fibrous network and are subsequently cleared [87,88]. While this process is unequivocally important in immune function, whether or not NETs can serve as a cause of tissue injury or tissue pathology is poorly characterized. NET formation during sterile inflammation has been noted in multiple models [89,90]. NET formation may have a role in ischemic liver injury, as NETs are formed in ischemic liver lobes and treatment with DNase I both reduced liver injury, and reduced NET formation [91]. This likely occurs through a HMGB1- TLR4 mediated pathway, indicating sterile inflammation can serve as an activator of NET formation independent of bacteria [91]. More research is required in this area, along with more specific ways of targeting NET formation, to fully understand how NETs themselves might drive pathogenesis and damage hepatocytes.
Given these well-established mechanisms of neutrophil cytotoxicity in the liver [57,92], whether or not neutrophils directly cause injury in a model can be assessed by interventions that target these mechanisms. If neutrophils were causing injury, the above steps would happen prior to the major development of injury, biomarkers of neutrophil-mediated liver injury such as chlorotyrosine staining, CD11b upregulation, and localized neutrophil recruitment would be prevalent, and knockout of the critical components such as CD11a, CD11b, CD18, ICAM-1, LFA, or other adhesion molecules would provide protection in relevant models.
2.3.3. Other inflammatory liver cells
In addition to the populations of macrophages and recruited neutrophils, the liver also contains a large population of natural killer (NK) cells of various types including: NK cells, NKT cells, and γδT cells [93]. Natural killer cells produce a number of cytokines that can exacerbate inflammation, but primarily are known for secreting interferon-gamma (IFN-γ) and TNF-α [93]. NK cells have cytotoxic activity as their granules contain a number of cytotoxic proteases that can directly perforate cells causing cell death, and they can also present pro-apoptotic proteins such as Fas ligand and CD40L [94]. NK cells have been implicated as potential players in the inflammatory environment of multiple types of acute liver injury including cholestatic liver diseases [95], APAP-induced liver injury [96], and hepatic ischemia-reperfusion injury [97], although the most common explanation is not as an effector cell, but rather as a stimulant for increased pro-inflammatory cytokine production. The role of NK cells in APAP induced liver injury has been particularly controversial with groups attaining opposite results. Jα18 mice that are deficient in Vα14iNKT cells are also resistant to APAP induced liver injury but this was attributed to changes in metabolism of APAP by the Jα18 mice that preferentially detoxified APAP [98]. These results were also challenged though as studies with CD1d knockout and Jα18 mice later indicated these mice underwent more severe injury due to stabilization and subsequent increases in CYP2E1 levels and thus greater biotransformation of APAP to its reactive metabolite [99]. As such, there is considerable debate in the field as to what the role of NK/NKT/iNKT cells truly is. Additional research is ongoing in this area as to whether or not subpopulations of these cells can potentially modulate the inflammatory environment during acute liver injury.
Dendritic cells have also been implicated in sterile inflammation in the liver. Dendritic cells are antigen presenting cells associated with the adaptive immune system that bridge the gap between innate immunity and adaptive immunity by presenting antigen material to local T cells [100]. The role of dendritic cells in acute sterile inflammation has not been well explored. There is some evidence that depletion of dendritic cells exacerbates APAP-induced liver injury although the mechanism through which this occurs is not confirmed [101].
3. Sterile inflammation – models
It is difficult to exactly duplicate a clinical liver disease in mice. Despite this, a number of models have been developed that approximate the clinical liver disease present in patients. Many of these, such as APAP-induced liver injury and hepatic ischemia-reperfusion injury are high fidelity models that have informed clinical practice and dramatically sped up the rate of identifying potential therapeutic targets. These models remain imperative to our understanding of the associated human conditions and, as such, defining mechanisms of injury and inflammation in these models accurately is necessary for clinical advancement. Herein, we will discuss sterile inflammation in the context of multiple diseases in an attempt to understand how inflammation affects liver injury.
4. Hepatic ischemia-reperfusion injury
Liver transplantation and liver resection are becoming increasingly common interventions for patients with advanced liver disease or liver tumors, respectively. Both transplantation and resection require clamping the hepatic portal triad, and thus induce ischemia, followed by unclamping and reperfusion of the liver. This procedure can cause cell injury during ischemia (due to lack of oxygen and absence of blood flow) and during reperfusion (due to the reintroduction of oxygen and blood flow). The overall pathophysiology of hepatic ischemia-reperfusion injury is complex [39,102-104]. Hepatic ischemia-reperfusion injury is a classical and widely accepted example of sterile inflammation and inflammatory liver injury [57,102,103] (Figure 1). The basic mechanisms that control liver injury in this model serve as a guide.
Figure 1. A simple model of ischemia-reperfusion injury. In the ischemia phase (top), occlusion of the blood flow causes hepatocyte swelling and release of intracellular contents that activate local Kupffer cells that express TLR4.
This causes release of ROS from KCs. In the reperfusion phase, localized production of chemokines and cytokines by KCs recruit and activate neutrophils. These neutrophils exacerbate the ROS produced by KCs. KC – Kupffer cell, PMN – neutrophil, HMGB1 – high mobility group box-1, ROS – reactive oxygen species, DAMPs – damage associated molecular patterns, ATP – adenosine triphosphate,
4.1. Ischemic liver injury
Clamping of the vasculature leads to immediate changes in hepatocytes. Primarily, the lack of oxygen shifts metabolism away from oxidative phosphorylation and towards glycolysis. This results in depletion of ATP, and a dramatic increase in intracellular constituents such as lactate, increased intracellular osmolarity, and reduced pH due to accumulation of H+ cations [100,105]. Eventually, these cells will undergo necrosis due to accumulation of calcium, and breakdown of membrane potential, however, there is no formation of reactive oxygen species [107,108]. Under realistic clinical conditions during elective surgery, the ischemic time will be limited in order to avoid severe ischemic injury. Nevertheless, hepatic ischemia together with early events during reperfusion, which include cell swelling caused by hyperosmolarity in the cytosol due to the accumulated metabolites, will lead to cellular stress and release of intracellular content [109]. Once the accumulated metabolites are either metabolized or flushed out, the swelling will subside. If there was no mitochondrial damage during ischemia, there will be limited intracellular oxidant stress during reperfusion [108]. On the other hand, if there was ischemic damage, the intracellular post-ischemic oxidant stress will be derived from xanthine oxidase, and more importantly, from damaged mitochondria [110]. In patients, prolonged hepatic ischemia can trigger ischemic hepatitis, which has a very high mortality rate [111]. Biomarkers indicate that ischemic hepatitis involves severe necrosis with extensive mitochondrial damage [112].
4.2. Reperfusion injury – initiation
A majority of cell death occurs after reperfusion [113]. Hepatocytes are generally resistant to ischemia for long periods, in part due to the fact that the mitochondrial membrane permeability transition pore (MPTP) opening can be reversible [114]. However, once re-oxygenation begins, hepatocyte leakage and mild necrosis releases a number of DAMPs such as HMGB1 [115], and mitochondrial DNA [14]. HMGB1 release occurs as early as one hour post-reperfusion, and continues to increase for up to twelve hours later [116]. HMGB1 activates TLR4 on KCs [115] and hepatocytes [117]. In addition, complement is activated during this period which directly activates KCs to generate reactive oxygen and complement components are potent neutrophils activators [41]. Thus, inhibition of complement activation or depletion of complement factors is highly protective against the injury [41]. KCs then produce ROS that can damage hepatocytes [41,118]. In addition, cytokines such as TNF-α and chemokines promote neutrophil recruitment [119,120]. In murine models, this also includes activation of the Nalp3 inflammasome through an apoptosis-associated speck-like protein (ASC), which exacerbates further inflammation [121]. Inflammasome activation may occur through TLR9 in this model, although a better characterization of inflammasome activating factors such as histones are needed [122]. A number of experimental interventions support the idea that KCs are critical for the initial injury and the progression of the inflammatory response [102]. The predominant oxidant stress occurs largely in the vasculature and not in parenchymal cells during reperfusion, indicating that hepatocytes do not contribute directly to the oxidative stress [40]. Inactivation of KCs dramatically reduces injury and vascular oxidant stress [40]. Preconditioning of the liver by pulsing short bursts of ischemia and reperfusion induces a number of anti-oxidant genes that are protective [123]. Administration of glutathione is also protective through the scavenging of the vascular oxidant stress [124-126]. A number of different pharmacological agents have been shown to act through either reduction of KC- mediated oxidative stress or through a direct action on KC activation [39,127]. After the initial oxidative stress from KCs, secreted cytokines, chemokines and activated complement factors further recruit neutrophils which produce more cytokines formation and more oxidant stress, leading to a vicious cycle of continued inflammation and injury [41,118-120,128,129].
4.3. Reperfusion injury – a second stage
Neutrophils exacerbate ischemia-reperfusion injury in the liver [35]. Neutrophil accumulation in the post-ischemic liver lobes is obvious and dramatic as early as three hours post-reperfusion [41]. Of note, neutrophils begin to upregulate CD11b as early as one hour post-reperfusion, consistent with a neutrophil mediated liver injury [130]. Inactivation of neutrophils with a Mac-1 monoclonal antibody is protective against the injury, even when administered only 30 minutes before the onset of ischemia [130]. Treatment of rats with an antibody against ICAM-1 is also protective [77], even in steatotic livers that are more prone to cell death after transplantation [126]. Protection against hepatic ischemia reperfusion with a number of agents is correlated strongly with a reduction in neutrophil recruitment or function [39]. Generally, inhibition of upstream mediators of inflammation such as HGMB1 [115], or mitochondrial DAMPs [14, 131], prevents downstream neutrophil accumulation and is highly protective against injury. As such, neutrophils are the likely critical mediator of the second phase of injury. In addition, it has been noted that other inflammatory cells might be involved in the second phase. CD4+ T cells also accumulate rapidly in the liver [132]. The primary mechanism through which these cells injure hepatocytes and exacerbate injury is likely through enhanced recruitment of neutrophils in an IL-17 dependent fashion [133]. Interferon regulatory factor 3 (IRF3) might be the critical link between recruitment of T-cells and the initial oxidant stress, as IRF3-/- mice are strongly protected against hepatic ischemia reperfusion injury in an IL-17 dependent mechanism [134].
Cytokine expression profile during this period is integral to the injury as activation of CXCR4 [135], and CXCR2 [29] are both detrimental to liver health; whereas, a number of other interleukins and cytokines such as IL-33 [136], and IL-37 [137], as well as other KC-derived mediators such as secreted leukocyte protease inhibitor (SLPI) reduce and control excessive inflammation [138]. The inflammation is eventually resolved through these anti-inflammatory processes, largely via production of IL-10 and activation of NF-κB [139] as well as production of IL-4 and activation of signal activator and transducer 6 (STAT6) [140] and production of IL-6 [141]. Resolution of hepatic ischemia-reperfusion injury is critical to recovery from both resection and liver transplantation, which is apparent in clinical practice by the substantial number of anti-inflammatory and immuno-modulatory drugs given in both settings [142].
5. Acetaminophen-induced liver injury
By far the most controversial topic in the field of sterile inflammation is the role of inflammation in APAP-induced liver injury. Despite the fact that there has been an effective antidote for over 30 years to prevent APAP-induced liver injury in early presenting patients, there still is no antidote for preventing late-stage APAP hepatotoxicity, or APAP-induced acute liver failure (ALF). Administration of N-acetylcysteine (NAC) replenishes glutathione (GSH) levels, which can first scavenge the reactive metabolite N-acetyl p-benzoquinone imine (NAPQI) and later support ROS detoxification [143]. NAC is most effective at early time points. After APAP metabolism is complete, the efficacy of NAC is diminished [144]. To address this concern, many have proposed the idea that there is a second phase of injury associated with APAP overdose that is dependent on inflammatory cells, predominantly focused on KCs, neutrophils and monocytes [30,86,96,145-147]. Despite the obvious appeal and some experimental support, there is a considerable amount of evidence that also argues for the contrary [5,148].
5.1. Acetaminophen induced liver injury – intracellular oxidative stress and the initial phase of toxicity
The initial phase of toxicity has been extensively studied over the last 40+ years. Foundational work was performed by Mitchell and colleagues when they demonstrated that APAP overdose rapidly depletes GSH, causes APAP protein adducts and leads to hepatocellular necrosis [149]. Subsequent work has demonstrated a number of the intracellular signaling mechanisms responsible for both metabolism and the toxicity.
APAP is rapidly glucuronidated by UDP-glucuronosyltransferase 1A1 and sulfated through sulfotransferase 2A1 [150]. During an overdose, only the sulfation but not glucuronidation pathway is saturated [151]. While neither of these processes produces a toxic metabolite, oxidation by cytochrome P4502E1 results in the formation of NAPQI [152], which can adduct proteins and trigger a profound mitochondrial oxidative stress [153]. The exact nature of which proteins become adducted remains a question in the field, but what is understood is that this intracellular oxidative stress mediates a substantial portion of the subsequent injury through a number of mechanisms. Foremost, the oxidative stress triggers activation of the c-Jun N-terminal kinase (JNK) pathway, and results in phosphorylation of JNK, and translocation of JNK to the mitochondria [154]. A number of other pathway members are also activated including Sab, MLK3, and Ask1, and also contribute to the injury process [155-157]. Knockout or pharmacological inhibition of these proteins is protective, as is knockout or pharmacological inhibition of JNK itself [158]. In addition to JNK activation, the oxidative stress activates the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signaling pathway [159]. This results in upregulation of a number of cytoprotective molecules that mediate autoprotection upon further APAP dosing [159]. Knockout of Nrf2 dramatically exacerbates injury [159], and knockout of the Nrf2 binding factor Kelch like-ECH-associated protein 1 (Keap1) is highly protective against APAP-induced liver injury [160]. This same oxidative stress also activates a number of different B-cell lymphoma-2 (Bcl-2) family proteins including Bcl-2 associated X-protein (bax) and BH3 interacting death domain agonist (bid) [161,162]. Both of these factors translocate to the mitochondria where they form pores in the outer mitochondrial membrane and help to initiate the mitochondrial permeability transition (MPT). Simultaneous to these events, is activation of the receptor interacting kinase 1/receptor interacting kinase 3 (RIP1/RIP3) pathway [163-165]. In the classical necroptotic pathway, these proteins form a complex called the ripoptosome that can activate mixed lineage kinase domain-like (MLKL), which then translocates to the cell membrane and forms membrane-disrupting pores leading to necrotic cell death [166]. However, recent studies showed that MLKL-/- mice are not protected against APAP hepatotoxicity suggesting that APAP-induced cell death is not a necroptotic mechanism [165,167]. Although there seems to be no role for MLKL and thus direct necroptosis in APAP-induced liver injury, yet knockout of either RIP1 or RIP3 seems to be protective [163,165]. While the current role of these proteins is not well understood in APAP toxicity, it is believed that this complex might have other functions that promote cytotoxicity. Regard-less, the primary event that occurs subsequent to all these events is MPT formation in the mitochondria [168]. JNK translocation to the mitochondria dramatically enhances the mitochondrial oxidative stress and stimulates mitochondrial dysfunction and pore opening [169,170]. This causes complex cellular dysfunction and reduces ATP stores for the cell [171]. Moreover, the mitochondrial oxidative stress results in leakage of mitochondrial membrane proteins such as apoptosis-inducing factor (AIF), and endonuclease G [172,173]. Both proteins are nucleases and translocate the nucleus where they directly attack DNA and eventually cause nuclear DNA lysis and cell death [173]. The primary oxidative stress and GSH depletion occurs within the first two hours, and the secondary oxidative stress occurs for the following 4-12 hours. During this secondary period, stimulation of autophagy also occurs, and autophagy can protect against APAP induced liver injury by removing damaged mitochondria that leak cytotoxic proteins [174,175]. Autophagy also removes protein adducts which reduces the initial harmful stimuli and further protects against future cell death [176]. This is especially apparent in cells distal from the central vein area where the primary site of toxicity is, as these cells express lower quantities of CYP2E1, generate fewer adducts, and undergo less oxidative cell death [177]. This initial phase of toxicity is supported by dozens of papers, interventions, and mouse models that all point towards the idea that protein adducts serve as a central source of oxidative stress that targets the mitochondria and eventually causes cellular necrosis due to DNA lysis and failed mitochondria.
5.2. Sterile inflammation – a second phase of injury after APAP overdose?
5.2.1. Neutrophils
The idea of a second phase of injury has been controversial in the literature. The primary hypothesis that has been put forth many times is that recruited inflammatory cells, predominantly monocytes and neutrophils, exacerbate the initial injury through the release of reactive oxygen species and other cytotoxic compounds consistent with sterile inflammatory liver injury. This occurs after the initial oxidative stress, typically reported between 6 and 12 h after the initial overdose of APAP. Studies in favor of this hypothesis have presented a number of points that indicate depletion of inflammatory cells, depletion of specific inflammatory mediators and chemotactic factors, or pharmacological inhibition of specific mediators of the injury is protective against acetaminophen-induced liver injury [30,86,96,142,178-182]. Moreover, during the inflammatory injury phase there is accumulation of a number of these factors including the release of DAMPs, chemokine and cytokine formation, substantial neutrophil and monocyte recruitment, and co-localization of these factors with areas of necrosis [30,86,96,142,178-182]. As such, at first glance, the combination of these studies strongly supports this idea and argues in favor of a sequential sterile inflammatory response that exacerbates APAP-induced liver injury.
Despite the considerable evidence presented in these studies, there are a number of unresolved issues with this hypothesis. Foremost, there is still a substantial controversy over the obtained results in this field, with many studies not being repeatable by others laboratories [5]. The first report of a potential neutrophil-mediated cytotoxicity after APAP overdose roundly rejected the hypothesis as there was no evidence in favor of upregulation of CD11b, shedding of L-selectin, or other evidence of neutrophil activation during the toxicity phase, nor was there protection when a CD18 antibody was administered to reduce recruitment, transmigration and cytotoxicity of neutrophils [34]. This was challenged by a subsequent study that found that depletion of NK/NKT cells reduced liver injury [96], with the same authors later proposing that the neutrophil was the main mediator of the second phase of APAP-induced liver injury [96]. Both of these studies have contrary reports indicating that the results depend on a questionable experimental design. The protective effect of NK/NKT cell depletion appeared to depend on the presence of dimethyl sulfoxide (DMSO), i.e. in the absence of DMSO as solvent for APAP no effect of NK/NKT cells was detectable [183]. In addition, neutrophil depletion is only effective against APAP toxicity when the neutropenia-inducing antibody is given as a 24 hour pre-treatment [142,145,184], but not when the antibody is administered shortly after APAP [184]. Importantly, it takes only 30 minutes to an hour to completely deplete neutrophils using the Gr-1 antibody that was used in these studies [185]. Interestingly, pre-treatment with the neutrophil-depleting antibody causes a pre-conditioning effect which activates a number of cyto-protective genes [186]. Use of these neutrophil-depleting antibodies has been repeated in subsequent studies, with the authors coming to the same conclusions despite the obvious problems present in this system [182]. If the Gr-1 antibody was truly protective due to its neutropenia effect, then administration just before the initial toxicity would also be effective; however, the Gr-1 antibody is clearly not protective when administered shortly after APAP administration but before any evidence of liver injury [184]. It has been noted though, that neutrophil activation as measured by CD11b expression and priming for ROS occurs largely after the development of liver injury, both in mice and more importantly, in man [184,187,188]. Moreover, inhibition of NADPH oxidase activity by either chemical inhibitors or deficiency of the gp91phox subunit, which completely inhibits hypochlorous acid formation as it stops the production of superoxide, is not protective [184,188]. In addition to anti-CD18 antibody administration not being effective [34], gene deficiency of CD18 is also ineffective, once again showing that there is no neutrophil-mediated cytotoxicity [187]. Similarly, it was proposed that the deletion of the CXC receptor 2, which binds neutrophil chemoattractant chemokines, was protected [189], although this was supposedly not through inhibition of neutrophils but expression of anti-apoptotic genes. However, apoptosis as mode of cell death during APAP hepatotoxicity has been roundly rejected on multiple occasions [190,191]. It has also been proposed that neutrophils mediate toxicity after APAP overdose through release of elastase [86], a serine protease whose primary purpose is to enhance transendothelial migration through the breakdown of extracellular matrix components such as elastin. Elastase is largely present in azurophilic granules present in neutrophils. The authors of this manuscript failed to assess neutrophil activation, a required component of degranulation, and only reported that mice with elastase-deficient bone marrow cells were protected with no explanation as to how the actual cytotoxicity might occur [86]. Furthermore, in our hands, elastase-deficient mice were not protected against APAP toxicity, nor was a pharmacological inhibitor of elastase protective (Jaeschke and Woolbright, unpublished). In addition to these direct interventions against neutrophils, other studies that affected the availability of various DAMPs such as uric acid, or complement, had a substantial effect on hepatic neutrophil accumulation but did not attenuate liver injury [192,193]. Given the totality of this data, it is clear that there has yet to be a study to conclusively demonstrate the role of neutrophils as a feed-forward amplification loop of APAP- induced liver injury. The current data surrounding the role of neutrophils is largely based on experimental approaches that have been questioned. At the present time, there is no conclusive evidence for a relevant role of neutrophil cytotoxicity in this model and most importantly in patients [188].
5.2.2. Macrophages and monocytes
Similarly, there are multiple reports on the efficacy of reducing either endogenous macrophage function or recruited monocyte function; however, these reports came to drastically different conclusions despite the fact the same strains of mice and same chemicals were used to derive these results. It was reported that pre-treatment with gadolinium chloride, a compound that inactivates KCs and attenuates ROS formation [96], prevented almost 100% of APAP toxicity [178]. However, these findings have never been confirmed by others [194-196]. Gadolinium chloride treatment may have additional off-target effects and its use has caused controversy in other models [43,197]. Moreover, using a separate approach to eliminate KC activity, clodronate liposomes, caused the opposite result, i.e., aggravated APAP hepatotoxicity [195]. In addition, mice deficient in the gp91phox gene, a component of the NADPH oxidase, showed the same oxidant stress and liver injury as wild type animals [188,198]. Taken together, these findings suggest that KCs are not directly involved in the injury.
Of note, monocytes and MoMF have also been investigated repeatedly with highly variable results [27,28,145,199-201]. Despite using the same mice, these groups obtained opposite results in regard to the role of the macrophage recruitment receptor CCR2 in APAP-induced liver injury with groups finding no differences in injury between CCR2-/- and wild type mice [27,28], groups finding exacerbation of injury in CCR2-/- mice compared to wild type animals [145,199] and one group finding protection against the injury in CCR2-/- mice [30]. Variable results such as this remain unexplained in the literature. Importantly, results obtained with MCP-1-/- mice matched the findings in the CCR2-/- mice in one of the studies, indicating that CCR2/MCP-1 signaling is likely not involved in the injury [27]. In support of the animal studies, the role of macrophages has been investigated in patients [202-204]. Macrophages display a distinct M2 phenotype in APAP overdose patients [204]. This applies to both endogenous macrophages, which are proliferative after injury, and bone marrow-derived monocytes that are recruited via CCL2/MCP-1 [204]. These data correlate with data from the mouse, wherein it was shown that recruited macrophages are also of the M2 phenotype and more importantly, tissue regeneration and repair after APAP overdose was delayed in CCR2-/- mice [27,28,200]. Moreover, high levels of secretory leukocyte protease inhibitor released from hepatocytes and macrophages inhibit immune function and increase susceptibility to sepsis and infection in APAP overdose patients [204]. Given this anti-inflammatory, pro-regenerative phenotype, macrophages are likely a key mediator of recovery, and preventing their recruitment and inactivation would be ill-advised. Even still, more reproducible effects in preclinical models are needed before any reliable conclusions can be drawn, and pitfalls in the experimental systems need to be identified and avoided.
5.2.3. The inflammasome
Although the evidence in favor of any specific inflammatory cell being the mediator of APAP-induced liver injury is limited in comparison to the evidence against it, there is also considerable evidence both for and against the role of specific mediators of inflammation, including a number of chemokines and cytokines. One of the foremost ideas in the field is that the inflammasome is primed through DAMPs binding to toll like receptors, e.g. TLR4 and TLR9, and actually activated by other DAMPs such as ATP, and that this activation mediates the injury through production of IL-1ß [3,28,29,181,182]. In support of this, there are obvious increases in serum mitochondrial DNA [13], and serum ATP [205], after APAP overdose, and there is a modest increase in pro-IL-1ß expression and serum IL-1ß levels after APAP overdose -179, 180, 206]. In addition, the formation of IL1β but not pro-IL-1β (IL-1β mRNA) is inhibited by a caspase inhibitor [206]. Furthermore, the supposed inhibition of TLR4 by benzyl alcohol can suppress IL-1ß signaling and protect against the injury [180]. Finally, there is reported protection in a number of models including NLRP3-/-, ASC-/-, P2XR7-/-, and TLR9-/- mice in addition to protection by pharmacological inhibition of TLR9, or treatment with apyrase, or treatment with DNase I, to breakdown extracellular DNA and prevent TLR9 activation [21,179-182,205]. However, some of these reports also are controversial as attempts to repeat much of these data have failed. Foremost, protection against injury with NLRP3-/-, ASC-/-, caspase-1-/-, IL-1R-/- mice and treatment with caspase inhibitors failed to provide protection in subsequent studies [206,207]. Administration of IL-1ß directly in high doses did not yield any increase in toxicity, despite an increase in neutrophil recruitment [206,207]. Treatment with P2XR7 pharmacological antagonist yielded more protection than the corresponding gene knockout of P2XR7 [21], and the inhibitor was subsequently shown to inhibit APAP metabolism, and thus its protective effects were independent of its alleged pharmacological effects on inflammasome activation [208]. Furthermore, evidence that extracellular ATP directly exacerbates APAP- induced cell death [205] has also been un-repeatable in both rodent and human hepatocytes [209]. Most importantly, human plasma samples from APAP overdose patients did not have any evidence of enhanced IL-1ß formation at any point over the first seven days of injury [5]. Thus, the inflammasome is not a likely a therapeutic target for preventing APAP toxicity, and its role in human patients is limited at best.
5.2.4. Interleukins
In addition to IL-1β, APAP exposure causes upregulation of several additional pro- and anti-inflammatory members of the interleukin family. Knockout of the IL-10 gene exacerbated APAP-induced liver injury [210]; however, this increased injury was linked to induction of inducible nitric oxide synthase in the absence of IL-10 and not an immune cell-mediated injury, indicating the increased injury was due to an effect on intracellular signaling mechanisms as APAP overdose causes extensive peroxynitrite formation in the mitochondria [211,212]. Knockout of IL-6 did not affect the injury, but did increase the rate of hepatocyte regeneration [213]. This may be due to signaling interactions with macrophages as inhibition of monocyte recruitment slows the rate of regeneration [28]. The role of IL-4 has become controversial as groups have attained opposite results in different strains of mice [214,215]. IL-4 is protective in C57BL/6J mice as it sustains increases in glutathione synthesis and recovery that are responsible for detoxification of oxidant stress in APAP toxicity [214], and thus IL-4-/- mice develop more severe injury. IL-4 administration in this model reversed the deleterious effects [214]. Others have recently reported that in the Balb/C mouse IL-4 exacerbates injury by enhancing inflammation and glutathione depletion [215]. This was in direct contrast to the previous results, although no attempts were made to explain these opposite findings. Since the article by Ryan and coworkers used multiple approaches to assess the role of IL-4, it is likely that this cytokines is beneficial by promoting hepatic GSH recovery [214]. Nevertheless, it needs to be kept in mind that the effect of IL-4 may be strain-dependent.
6. Liver injury during obstructive cholestasis
Cholestasis is caused by a blockage in bile flow from the liver to the intestine. It occurs either within the internal bile ducts of the liver, or in the external bile duct that connects the liver to the intestine. Regardless of the location, substantial liver injury occurs when animals or human patients undergo severe obstructive cholestasis. Inflammation is a noted aspect of murine models such as bile duct ligation (BDL) in rats [216], or mice [32], the multi-drug resistance protein 2 (MDR2) knockout mouse [217], and in human cholestasis patients of various etiologies [51]. Previous studies have indicated that cholestatic liver injury is mediated by both bile acid accumulation [218], and through inflammation, with both KCs [43], and neutrophils [32] being critically involved. One of the remaining questions in the field is which of these processes stimulates the initial cell death, and which mechanisms and cell types are then responsible for cell death. Herein, we will discuss these mechanisms with a focus on the BDL model, due to its prevalence in the literature, while also focusing on human patients.
6.1. Cholestatic liver injury – bile acid induced toxicity
When hepatocytes are exposed to sufficient concentrations of specific bile acids, they can undergo a variety of changes including apoptosis [218], necrosis [51,219], dramatic upregulation of cytokines such as MIP-2 and mKC [25], and activation of signaling pathways through G-protein coupled bile acid receptor (TGR) and farnesoid X receptor (FXR) [220,221]. With the exception of the activation of the above signaling pathways, which seem to occur in both human and rodent cells, many of these other changes are largely dependent on the origin of hepatocytes and the associated, individual bile acid species. Hepatocytes derived from the rat rapidly undergo apoptosis upon exposure to high concentrations of glycochenodeoxycholic acid (GCDC), a bile acid that is predominantly found in humans [51,222]. While apoptosis is not considered as pro-inflammatory as necrotic cell death, due to the lack of release of DAMPs, it has been noted that apoptosis can cause inflammation through serving as a danger signal itself, and through activation of local phagocytic macrophages [43,52]. In contrast, a number of other mammalian cells, including humans, are highly resistant to GCDC-induced apoptosis and require high concentrations of bile acids to initiate cell death [51,219,223]. Instead, hepatocytes from both humans and mice undergo frank necrosis after exposure to high concentrations of bile acids and release DAMPs such as HMGB1 [51]. Moreover, the relative toxicity of individual bile acids is highly variable, and dependent on their side chains, and to which amino acid they are conjugated [224]. The overwhelming majority of bile acids in cholestatic mammals are conjugated to either taurine or glycine [225]; however, the degree to which mammals use each amino acid varies greatly [51,226]. This has a number of biological effects, as conjugation to taurine reduces toxicity, but seems to promote inflammation, at least in the mouse [25]. Many of the less toxic taurine-conjugated bile acids, such as taurocholic acid, accumulate significantly in both mice and humans [226], and strongly upregulate and promote a number of pro-inflammatory cytokines in the mouse [25]. Of note though, this interaction is less prominent in either primary human hepatocytes [51], or human HepaRG cells [227]. As such, accumulation of bile acids can affect sterile inflammation in a number of ways: 1) bile acidinduced apoptosis can stimulate inflammation by activating KCs through phagocytosis of apoptotic bodies [43]; 2) bile acid-induced necrosis can stimulate inflammation through DAMP release [51]; 3) bile acids can directly stimulate inflammation without causing cell death through an early growth response factor-1 (Egr-1)-dependent signaling pathway [25]; 4) accumulation of bile acids and biliary pressure can activate signaling molecules such as osteopontin that result in neutrophil recruitment [228]. All of these pathways and mechanisms are contextual, based both on which bile acids are administered or which bile acids accumulate in the model or human patient, and also what sort of rodent model or human patient is being examined.
6.2. Bile duct ligation – a consensus model of inflammatory liver injury
BDL results in an immediate increase in biliary pressure that initiates overload of the biliary tracts and causes rupture of the bile ducts and release of bile onto the hepatic parenchyma within 6 hours [229,230]. Neutrophil counts in the liver rise about this same time, and are largely localized to the area immediately surrounding the area of biliary infarction [33]. Concomitantly, neutrophil activity rises acutely after BDL, and neutrophils from BDL animals have elevated NADPH oxidase activity [216,231]. Previous work from our laboratory has demonstrated that deficiency of the adhesion molecules CD18 or ICAM-1 is 60-80% protective against BDL injury [32,78]. Presumably, this is due to the lack of observable neutrophil extravasation present in the knockout animals [32,78]. Mice deficient in the Fas receptor are also protected against BDL via a substantial reduction in immune activity [232]. Supporting these data, P-selectin glycoprotein-1-/- mice with deficient neutrophil attachment are also protected against BDL [66]. Knockout of the pro-inflammatory factor osteopontin is highly protective at early time points suggesting that cleaved osteopontin may be the initial critical chemotactic factor for neutrophil accumulation after BDL [228]. Osteopontin is cleaved by matrix metalloproteinases, and inhibition of these enzymes is also protective [228,233,234]. Expansion of this inflammatory response over time appears to involve Th17 cells as administration of IL-17 worsens inflammation and injury [235], and knockout of cluster of differentiation 279 results in a dramatic decrease in IL-17 producing cells which correlated with protection from inflammation and injury [236]. Multiple models of mice with an attenuated inflammatory response are protected including the lpr mouse [232,237], and the Na+/H+ exchanger regulatory factor-/- mouse [238]. All of these studies point towards a single hypothesis – the inflammation associated with bile duct ligation mediates a significant portion of the injury [19,239].
Despite this, the driving agent for inflammation in the model is still poorly characterized [239] Inflammation after BDL largely occurs at, and around, the point of biliary infarction, that is, the point of rupture of the biliary ducts due to the increased mechanical pressure [33,240]. Given that these points of rupture are also the site of the overwhelming degree of the associated necrosis, it has been hypothesized that bile acids released from the bile ducts directly damage hepatocytes, resulting in DAMP release and inflammation [237]. Initially, the hypothesis was that bile acid accumulation in the BDL model resulted in apoptosis, which drove the inflammatory response and subsequent neutrophil infiltration which exacerbated the injury and the fibrosis [237]. This was subsequently questioned as there is a lack of apoptosis in the BDL model, and the injury proceeds predominantly through necrosis [33,240]. Apoptosis of normal hepatocytes has only been convincingly shown in rat hepatocytes exposed to high concentrations of GCDC [51,222], however, GCDC is not typically found in rodent models [226]. Attempts at defining a bile acid-dependent cell death in the mouse showed conclusively that concentrations of bile acids that did accumulate in the mouse were largely non-toxic, as taurine conjugated bile acids did not elicit toxicity at concentrations of up to 10 mM, far higher than what was present in serum or bile of cholestatic mice [25,226]. Instead, exposure of primary murine hepatocytes to taurine-conjugated bile acids such as taurocholic acid (TCA) elicited a dramatic increase in CXC chemokine expression and upregulation of ICAM-1 [25], similar to what was observed in vivo after BDL [232,241]. In addition, deficiency of osteopontin resulted in nearly complete protection against necrosis at early time points during BDL, but the protection was not sustained at distal time points [226]. This suggests that cleaved osteopontin is an early chemotactic factor for neutrophils that is released into the parenchyma after BDL. Subsequently, the chemotaxis may be maintained by CXC chemokines generated by bile acid-exposed hepatocytes [239]. Exposure to hydrophobic bile acid does cause significant cellular stress as indicated by mitochondrial dysfunction [242, 243]. This may result in the release of additional local factors responsible for pro-inflammatory mediator generation, as there is substantial HMGB1 release as early as 6 hours post BDL [33]. More studies are needed to better define the initiating factors why neutrophils target damaged or dying hepatocytes during cholestasis, especially in humans.
6.3. Obstructive cholestasis in human patients
Cholestasis in humans occurs in various pathophysiologies. Acute obstructive cholestasis induced by gallstones is rarely a chronic concern as patients will commonly present to the hospital rapidly with abdominal pain. The obstruction can be removed by endoscopic retrograde cholangiopancreatography. However, patient populations exist that cannot safely undergo endoscopy, and a greater understanding of cholestasis in patients is required to generate therapeutic options. Acute obstructive cholestasis presents with dramatic increases in alkaline phosphatase, and bilirubin, in addition to increases in serum transaminases. Furthermore, very early studies understood that serum bile acid levels were also dramatically elevated in cholestatic patients [225]. The idea that elevated serum bile acids might induce toxicity was proposed and tested, whereupon it was noted that bile acids induce toxicity in human hepatocytes at concentrations between 500μM and 1mM GCDC, far higher than what is necessary in rats or mice [51]. Human hepatocytes are also highly resistant to taurine-conjugated bile acids, and more importantly, they do not upregulate ICAM-1 or chemokines upon exposure to TCA [51]. A number of subsequent studies have conclusively shown the concentration necessary for toxicity is multiple orders of magnitude above human serum values achieved during cholestasis [51,244,245]; however, biliary concentrations for GCDC and other glycine-conjugated bile acids are commonly in the mM range [51, 246]. As such, it is possible that biliary concentrations of bile acids can directly elicit toxicity. As the characteristic “foamy necrosis” and biliary infarcts present in BDL are also present in human patients, biliary bile acid concentrations may be responsible for the initial toxicity in human patients and subsequent release of pro-inflammatory factors such as HMGB1 [51]. While a sterile inflammatory response during obstructive cholestasis is clearly present in laboratory animal and in human patients, there are obvious differences between the current rodent models and human patients. Further studies are also needed in human patients to more fully define if neutrophils, or other inflammatory mediators, exacerbate the disease.
7. Conclusions
A sterile inflammatory response is an important aspect of acute liver injury. As the liver is an innate immune organ by nature, there is a delicate balance between activation, over-activation and hyper-stimulation that is constantly ongoing. Future studies are needed to better define the mechanisms that drive a sterile inflammatory response in various models and the pathophysiological relevance for the progression of the injury and the critical involvement in the process of regeneration and recovery. In addition, the vital importance of inflammation as defense against infection needs to be taken into account. Only a more detailed understanding of these mechanisms in relevant animal models and patients will allow the identification of potential therapeutic targets that can be used to translate these findings in animals to patients without impacting normal host defense functions.
Acknowledgements
Work in our laboratory was supported by a CTSA grant from NCATS awarded to the University of Kansas Medical Center for Frontiers: The Heartland Institute for Clinical and Translational Research # UL1TR000001 (formerly #UL1RR033179). This work was also supported in part by the National Institutes of Health grants R01 DK102142, DK070195, AA12916, and by grants from the National Institute of General Medical Sciences (P20 GM103549 and P30 GM118247) of the National Institutes of Health. Additional support came from an award (to BLW) from the ‘Training Program in Environmental Toxicology’ T32 ES007079-26A2 from the National Institute of Environmental Health Sciences.
Disclosure
The authors declare no conflict of interest.
References
- [1].Krenkel O, Mossanen JC, Tacke F. Immune mechanisms in acetaminophen-induced acute liver failure. Hepatobiliary Surg Nutr. 2014;3:331–343. doi: 10.3978/j.issn.2304-3881.2014.11.01. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Jaeschke H. Reactive oxygen and mechanisms of inflammatory liver injury: Present concepts. J Gastroenterol Hepatol. 2011;26(Suppl 1):173–179. doi: 10.1111/j.1440-1746.2010.06592.x. [DOI] [PubMed] [Google Scholar]
- [3].Kubes P, Mehal WZ. Sterile inflammation in the liver. Gastroenterology. 2012;143:1158–1172. doi: 10.1053/j.gastro.2012.09.008. [DOI] [PubMed] [Google Scholar]
- [4].Woolbright BL, Jaeschke H. Sterile inflammation in acute liver injury: myth or mystery? Expert Rev Gastroenterol Hepatol. 2015;9:1027–1029. doi: 10.1586/17474124.2015.1060855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Woolbright BL, Jaeschke H. Role of the Inflammasome in Acetaminophen-induced Liver Injury and Acute Liver Failure. J Hepatol. 2017 doi: 10.1016/j.jhep.2016.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–195. doi: 10.1038/nature00858. [DOI] [PubMed] [Google Scholar]
- [7].Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am J Pathol. 1995;146:3–15. [PMC free article] [PubMed] [Google Scholar]
- [8].Chow JC, Young DW, Golenbock DT, Christ WJ, Gusovsky F. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. J Biol Chem. 1999;274:10689–10692. doi: 10.1074/jbc.274.16.10689. [DOI] [PubMed] [Google Scholar]
- [9].Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10:4171–426. doi: 10.1016/s1097-2765(02)00599-3. [DOI] [PubMed] [Google Scholar]
- [10].Kayagaki N, Warming S, Lamkanfi M, Vande Walle L, Louie S, Dong J, Newton K, Qu Y, Liu J, Heldens S, Zhang J, Lee WP, Roose-Girma M, Dixit VM. Non-canonical inflammasome activation targets caspase-11. Nature. 2011;479:117–121. doi: 10.1038/nature10558. [DOI] [PubMed] [Google Scholar]
- [11].Andrei C, Dazzi C, Lotti L, Torrisi MR, Chimini G, Rubartelli A. The secretory route of the leaderless protein interleukin 1beta involves exocytosis of endolysosome-related vesicles. Mol Biol Cell. 1999;10:1463–1475. doi: 10.1091/mbc.10.5.1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Gardella S, Andrei C, Ferrera D, Lotti LV, Torrisi MR, Bianchi ME, Rubartelli A. The nuclear protein HMGB1 is secreted by monocytes via a non-classical, vesicle-mediated secretory pathway. EMBO Rep. 2002;3:995–1001. doi: 10.1093/embo-reports/kvf198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].McGill MR, Sharpe MR, Williams CD, Taha M, Curry SC, Jaeschke H. The mechanism underlying acetaminophen-induced hepatotoxicity in humans and mice involves mitochondrial damage and nuclear DNA fragmentation. J Clin Invest. 2012;122:1574–1583. doi: 10.1172/JCI59755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].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. 2010;464:104–107. doi: 10.1038/nature08780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–241. doi: 10.1038/nature04516. [DOI] [PubMed] [Google Scholar]
- [16].Yamasaki K, Muto J, Taylor KR, Cogen AL, Audish D, Bertin J, Grant EP, Coyle AJ, Misaghi A, Hoffman HM, Gallo RL. NLRP3/cryopyrin is necessary for interleukin-1ß (IL-1ß) release in response to hyaluronan, an endogenous trigger of inflammation in response to injury. J Biol Chem. 2009;284:12762–12771. doi: 10.1074/jbc.M806084200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Huang H, Evankovich J, Yan W, Nace G, Zhang L, Ross M, Liao X, Billiar T, Xu J, Esmon CT, Tsung A. Endogenous histones function as alarmins in sterile inflammatory liver injury through Toll-like receptor 9 in mice. Hepatology. 2011;54:999–1008. doi: 10.1002/hep.24501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Wen Z, Lei Z, Yao L, Jiang P, Gu T, Ren F, Liu Y, Gou C, Li X, Wen T. Circulating histones are major mediators of systemic inflammation and cellular injury in patients with acute liver failure. Cell Death Dis. 2016;7:e2391. doi: 10.1038/cddis.2016.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Woolbright BL, Jaeschke H. Therapeutic targets for cholestatic liver injury. Expert Opin Ther Targets. 2016;20:463–475. doi: 10.1517/14728222.2016.1103735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Seki E, Brenner DA. Toll-like receptors and adaptor molecules in liver disease: update. Hepatology. 2008;48:322–335. doi: 10.1002/hep.22306. [DOI] [PubMed] [Google Scholar]
- [21].Hoque R, Sohail MA, Salhanick S, Malik AF, Ghani A, Robson SC, Mehal WZ. P2X7 receptor-mediated purinergic signaling promotes liver injury in acetaminophen hepatotoxicity in mice. Am J Physiol Gastrointest Liver Physiol. 2012;302:G1171–G1179. doi: 10.1152/ajpgi.00352.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Kennel SJ, Lankford TK, Foote LJ, Shinpock SG, Stringer C. CD44 expression on murine tissues. J Cell Sci. 1993;104:373–382. doi: 10.1242/jcs.104.2.373. [DOI] [PubMed] [Google Scholar]
- [23].Smedsrad B, Melkko J, Araki N, Sano H, Horiuchi S. Advanced glycation end products are eliminated by scavenger-receptor-mediated endocytosis in hepatic sinusoidal Kupffer and endothelial cells. Biochem J. 1997;322:567–573. doi: 10.1042/bj3220567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Jaeschke H. Cellular adhesion molecules: regulation and functional significance in the pathogenesis of liver diseases. Am J Physiol. 1997;273:G602–11. doi: 10.1152/ajpgi.1997.273.3.G602. [DOI] [PubMed] [Google Scholar]
- [25].Allen K, Jaeschke H, Copple BL. Bile acids induce inflammatory genes in hepatocytes: a novel mechanism of inflammation during obstructive cholestasis. Am J Pathol. 2011;178:175–186. doi: 10.1016/j.ajpath.2010.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Clarke C, Kuboki S, Sakai N, Kasten KR, Tevar AD, Schuster R, Blanchard J, Caldwell CC, Edwards MJ, Lentsch AB. CXC chemokine receptor-1 is expressed by hepatocytes and regulates liver recovery after hepatic ischemia/reperfusion injury. Hepatology. 2011;53:261–271. doi: 10.1002/hep.24028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Dambach DM, Watson LM, Gray KR, Durham SK, Laskin DL. Role of CCR2 in macrophage migration into the liver during acetaminophen-induced hepatotoxicity in the mouse. Hepatology. 2002;35:1093–1103. doi: 10.1053/jhep.2002.33162. [DOI] [PubMed] [Google Scholar]
- [28].Holt MP, Cheng L, Ju C. Identification and characterization of infiltrating macrophages in acetaminophen-induced liver injury. J Leukoc Biol. 2008;84:1410–1421. doi: 10.1189/jlb.0308173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Kuboki S, Shin T, Huber N, Eismann T, Galloway E, Schuster R, Blanchard J, Edwards MJ, Lentsch AB. Hepatocyte signaling through CXC chemokine receptor-2 is detrimental to liver recovery after ischemia/reperfusion in mice. Hepatology. 2008;48:1213–1223. doi: 10.1002/hep.22471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Mossanen JC, Krenkel O, Ergen C, Govaere O, Liepelt A, Puengel T, Heymann F, Kalthoff S, Lefebvre E, Eulberg D, Luedde T, Marx G, Strassburg CP, Roskams T, Trautwein C, Tacke F. Chemokine (C-C motif) receptor 2-positive monocytes aggravate the early phase of acetaminophen-induced acute liver injury. Hepatology. 2016;64:1667–1682. doi: 10.1002/hep.28682. [DOI] [PubMed] [Google Scholar]
- [31].Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling. Nat Rev Immunol. 2016;16:407–420. doi: 10.1038/nri.2016.58. [DOI] [PubMed] [Google Scholar]
- [32].Gujral JS, Farhood A, Bajt ML, Jaeschke H. Neutrophils aggravate acute liver injury during obstructive cholestasis in bile duct-ligated mice. Hepatology. 2003;38:355–363. doi: 10.1053/jhep.2003.50341. [DOI] [PubMed] [Google Scholar]
- [33].Woolbright BL, Antoine DJ, Jenkins RE, Bajt ML, Park BK, Jaeschke H. Plasma biomarkers of liver injury and inflammation demonstrate a lack of apoptosis during obstructive cholestasis in mice. Toxicol Appl Pharmacol. 2013;273:524–531. doi: 10.1016/j.taap.2013.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Lawson JA, Farhood A, Hopper RD, Bajt ML, Jaeschke H. The hepatic inflammatory response after acetaminophen overdose: role of neutrophils. Toxicol Sci. 2000;54:509–516. doi: 10.1093/toxsci/54.2.509. [DOI] [PubMed] [Google Scholar]
- [35].Jaeschke H, Farhood A, Smith CW. Neutrophils contribute to ischemia/reperfusion injury in rat liver in vivo. FASEB J. 1990;4:3355–3359. [PubMed] [Google Scholar]
- [36].Bertola A, Park O, Gao B. Chronic plus binge ethanol feeding synergistically induces neutrophil infiltration and liver injury in mice: a critical role for E-selectin. Hepatology. 2013;58:1814–23. doi: 10.1002/hep.26419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Jaeschke H, Farhood A, Smith CW. Neutrophil-induced liver cell injury in endotoxin shock is a CD11b/CD18-dependent mechanism. Am J Physiol. 1991;261:G1051–1056. doi: 10.1152/ajpgi.1991.261.6.G1051. [DOI] [PubMed] [Google Scholar]
- [38].Bilzer M, Roggel F, Gerbes AL. Role of Kupffer cells in host defense and liver disease. Liver Int. 2006;26:1175–1186. doi: 10.1111/j.1478-3231.2006.01342.x. [DOI] [PubMed] [Google Scholar]
- [39].Jaeschke H, Woolbright BL. Current strategies to minimize hepatic ischemia-reperfusion injury by targeting reactive oxygen species. Transplant Rev (Orlando) 2012;26:103–114. doi: 10.1016/j.trre.2011.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Jaeschke H, Farhood A. Neutrophil and Kupffer cell-induced oxidant stress and ischemia-reperfusion injury in rat liver. Am J Physiol. 1991;260:G355–362. doi: 10.1152/ajpgi.1991.260.3.G355. [DOI] [PubMed] [Google Scholar]
- [41].Jaeschke H, Farhood A, Bautista AP, Spolarics Z, Spitzer JJ. Complement activates Kupffer cells and neutrophils during reperfusion after hepatic ischemia. Am J Physiol. 1993;264:G801–G809. doi: 10.1152/ajpgi.1993.264.4.G801. [DOI] [PubMed] [Google Scholar]
- [42].Adachi Y, Bradford BU, Gao W, Bojes HK, Thurman RG. Inactivation of Kupffer cells prevents early alcohol-induced liver injury. Hepatology. 1994;20:453–460. [PubMed] [Google Scholar]
- [43].Canbay A, Feldstein AE, Higuchi H, Werneburg N, Grambihler A, Bronk SF, Gores GJ. Kupffer cell engulfment of apoptotic bodies stimulates death ligand and cytokine expression. Hepatology. 2003;38:1188–1198. doi: 10.1053/jhep.2003.50472. [DOI] [PubMed] [Google Scholar]
- [44].Liu P, McGuire GM, Fisher MA, Farhood A, Smith CW, Jaeschke H. Activation of Kupffer cells and neutrophils for reactive oxygen formation is responsible for endotoxin-enhanced liver injury after hepatic ischemia. Shock. 1995;3:56–62. [PubMed] [Google Scholar]
- [45].Marzi I, Cowper K, Takei Y, Lindert K, Lemasters JJ, Thurman RG. Methyl palmitate prevents Kupffer cell activation and improves survival after orthotopic liver transplantation in the rat. Transpl Int. 1991;4:215–220. doi: 10.1007/BF00649106. [DOI] [PubMed] [Google Scholar]
- [46].Zhong Z, Connor H, Mason RP, Qu W, Stachlewitz RF, Gao W, Lemasters JJ, Thurman RG. Destruction of Kupffer cells increases survival and reduces graft injury after transplantation of fatty livers from ethanol-treated rats. Liver Transpl Surg. 1996;2:383–387. doi: 10.1002/lt.500020509. [DOI] [PubMed] [Google Scholar]
- [47].Tiegs G, Wolter M, Wendel A. Tumor necrosis factor is a terminal mediator in galactosamine/endotoxin-induced hepatitis in mice. Biochem Pharmacol. 1989;38:627–631. doi: 10.1016/0006-2952(89)90208-6. [DOI] [PubMed] [Google Scholar]
- [48].Bautista AP, Skrepnik N, Niesman MR, Bagby GJ. Elimination of macrophages by liposome-encapsulated dichloromethylene diphosphonate suppresses the endotoxin-induced priming of Kupffer cells. J Leukoc Biol. 1994a;55:321–7. doi: 10.1002/jlb.55.3.321. [DOI] [PubMed] [Google Scholar]
- [49].Barkett M, Gilmore TD. Control of apoptosis by Rel/NF-kappaB transcription factors. Oncogene. 1999;18:6910–6924. doi: 10.1038/sj.onc.1203238. [DOI] [PubMed] [Google Scholar]
- [50].McGill MR, Yan HM, Ramachandran A, Murray GJ, Rollins DE, Jaeschke H. HepaRG cells: a human model to study mechanisms of acetaminophen hepatotoxicity. Hepatology. 2011;53:974–982. doi: 10.1002/hep.24132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Woolbright BL, Dorko K, Antoine DJ, Clarke JI, Gholami P, Li F, Kumer SC, Schmitt TM, Forster J, Fan F, Jenkins RE, Park BK, Hagenbuch B, Olyaee M, Jaeschke H. Bile acid-induced necrosis in primary human hepatocytes and in patients with obstructive cholestasis. Toxicol Appl Pharmacol. 2015;283:168–177. doi: 10.1016/j.taap.2015.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Wang M, You Q, Lor K, Chen F, Gao B, Ju C. Chronic alcohol ingestion modulates hepatic macrophage populations and functions in mice. J Leukoc Biol. 2014;96:657–665. doi: 10.1189/jlb.6A0114-004RR. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Heymann F, Trautwein C, Tacke F. Monocytes and macrophages as cellular targets in liver fibrosis. Inflamm Allergy Drug Targets. 2009;8:307–318. doi: 10.2174/187152809789352230. [DOI] [PubMed] [Google Scholar]
- [54].Ju C, Tacke F. Hepatic macrophages in homeostasis and liver diseases: from pathogenesis to novel therapeutic strategies. Cell Mol Immunol. 2016;13:316–327. doi: 10.1038/cmi.2015.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Naveau S, Chollet-Martin S, Dharancy S, Mathurin P, Jouet P, Piquet MA, Davion T, Oberti F, Broët P, Emilie D. Foie-Alcool group of the Association Française pour lEtude du Foie. A double-blind randomized controlled trial of infliximab associated with prednisolone in acute alcoholic hepatitis. Hepatology. 2004;39:1390–1397. doi: 10.1002/hep.20206. [DOI] [PubMed] [Google Scholar]
- [56].Boetticher NC, Peine CJ, Kwo P, Abrams GA, Patel T, Aqel B, Boardman L, Gores GJ, Harmsen WS, McClain CJ, Kamath PS, Shah VH. A randomized, double-blinded, placebo-controlled multicenter trial of etanercept in the treatment of alcoholic hepatitis. Gastroenterology. 2008;135:1953–1960. doi: 10.1053/j.gastro.2008.08.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Jaeschke H. Mechanisms of Liver Injury. II. Mechanisms of neutrophil-induced liver cell injury during hepatic ischemia-reperfusion and other acute inflammatory conditions. Am J Physiol Gastrointest Liver Physiol. 2006;290:G1083–G1088. doi: 10.1152/ajpgi.00568.2005. [DOI] [PubMed] [Google Scholar]
- [58].Jaeschke H, Ho YS, Fisher MA, Lawson JA, Farhood A. Glutathione peroxidase-deficient mice are more susceptible to neutrophil-mediated hepatic parenchymal cell injury during endotoxmia: importance of an intracellular oxidant stress. Hepatology. 1999;29:443–450. doi: 10.1002/hep.510290222. [DOI] [PubMed] [Google Scholar]
- [59].Jaeschke H, Farhood A, Fisher MA, Smith CW. Sequestration of neutrophils in the hepatic vasculature during endotoxemia is independent of beta 2 integrins and intercellular adhesion molecule-1. Shock. 1996;6:351–356. doi: 10.1097/00024382-199611000-00009. [DOI] [PubMed] [Google Scholar]
- [60].Jaeschke H, Fisher MA, Lawson JA, Simmons CA, Farhood A, Jones DA. Activation of caspase 3 (CPP32)-like proteases is essential for TNF-alpha-induced hepatic parenchymal cell apoptosis and neutrophil-mediated necrosis in a murine endo-toxin shock model. J Immunol. 1998;160:3480–3486. [PubMed] [Google Scholar]
- [61].McEver RP. Selectins: lectins that initiate cell adhesion under flow. Curr Opin Cell Biol. 2002;14:581–586. doi: 10.1016/s0955-0674(02)00367-8. [DOI] [PubMed] [Google Scholar]
- [62].Wong J, Johnston B, Lee SS, Bullard DC, Smith CW, Beaudet AL, Kubes P. A minimal role for selectins in the recruitment of leukocytes into the inflamed liver microvasculature. J Clin Invest. 1997;99:2782–2790. doi: 10.1172/JCI119468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Essani NA, Fisher MA, Simmons CA, Hoover JL, Farhood A, Jaeschke H. Increased P-selectin gene expression in the liver vasculature and its role in the pathophysiology of neutrophil-induced liver injury in murine endotoxin shock. Leukoc Biol. 1998;63:288–296. doi: 10.1002/jlb.63.3.288. [DOI] [PubMed] [Google Scholar]
- [64].Chosay JG, Essani NA, Dunn CJ, Jaeschke H. Neutrophil margination and extravasation in sinusoids and venules of liver during endotoxin-induced injury. Am J Physiol. 1997;272:G1195–G1200. doi: 10.1152/ajpgi.1997.272.5.G1195. [DOI] [PubMed] [Google Scholar]
- [65].Lawson JA, Burns AR, Farhood A, Lynn Bajt M, Collins RG, Smith CW, Jaeschke H. Pathophysiologic importance of E-and L-selectin for neutrophil-induced liver injury during en-dotoxemia in mice. Hepatology. 2000;32:990–998. doi: 10.1053/jhep.2000.19068. [DOI] [PubMed] [Google Scholar]
- [66].Dold S, Laschke MW, Zhau Y, Schilling M, Menger MD, Jeppsson B, Thorlacius H. P-selectin glycoprotein ligand-1-mediated leukocyte recruitment regulates hepatocellular damage in acute obstructive cholestasis in mice. Inflamm Res. 2010;59:291–298. doi: 10.1007/s00011-009-0099-2. [DOI] [PubMed] [Google Scholar]
- [67].Kubes P, Payne D, Woodman RC. Molecular mechanisms of leukocyte recruitment in postischemic liver microcirculation. Am J Physiol Gastrointest Liver Physiol. 2002;283:G139–G147. doi: 10.1152/ajpgi.00058.2002. [DOI] [PubMed] [Google Scholar]
- [68].Yadav SS, Howell DN, Steeber DA, Harland RC, Tedder TF, Clavien PA. P-Selectin mediates reperfusion injury through neutrophil and platelet sequestration in the warm ischemic mouse liver. Hepatology. 1999;29:1494–1502. doi: 10.1002/hep.510290505. [DOI] [PubMed] [Google Scholar]
- [69].Khandoga A, Biberthaler P, Messmer K, Krombach F. Plate-let-endothelial cell interactions during hepatic ischemia-reperfusion in vivo: a systematic analysis. Microvasc Res. 2003;65:71–77. doi: 10.1016/s0026-2862(02)00018-3. [DOI] [PubMed] [Google Scholar]
- [70].van Golen RF, Stevens KM, Colarusso P, Jaeschke H, Heger M. Platelet aggregation but not activation and degranulation during the acute post-ischemic reperfusion phase in livers with no underlying disease. J Clin Transl Res. 2015;1:107–115. doi: 10.18053/jctres.201502.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Jaeschke H, Smith CW. Mechanisms of neutrophil-induced parenchymal cell injury. J Leukoc Biol. 1997;61:647–653. doi: 10.1002/jlb.61.6.647. [DOI] [PubMed] [Google Scholar]
- [72].Entman ML, Youker K, Shoji T, Kukielka G, Shappell SB, Taylor AA, Smith CW. Neutrophil induced oxidative injury of cardiac myocytes. A compartmented system requiring CD11b/CD18-ICAM-1 adherence. J Clin Invest. 1992;90:1335–1345. doi: 10.1172/JCI115999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Diamond MS, Staunton DE, de Fougerolles AR, Stacker SA, Garcia-Aguilar J, Hibbs ML, Springer TA. ICAM-1 (CD54): a counter-receptor for Mac-1 (CD11b/CD18) J Cell Biol. 1990;111:3129–3139. doi: 10.1083/jcb.111.6.3129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Nagendra AR, Mickelson JK, Smith CW. CD18 integrin and CD54-dependent neutrophil adhesion to cytokine-stimulated human hepatocytes. Am J Physiol. 1997;272:G408–G416. doi: 10.1152/ajpgi.1997.272.3.G408. [DOI] [PubMed] [Google Scholar]
- [75].Weiss SJ. Tissue destruction by neutrophils. N Engl J Med. 1989;320:365–376. doi: 10.1056/NEJM198902093200606. [DOI] [PubMed] [Google Scholar]
- [76].Xu H, Gonzalo JA, St Pierre Y, Williams IR, Kupper TS, Co-tran RS, Springer TA, Gutierrez-Ramos JC. Leukocytosis and resistance to septic shock in intercellular adhesion molecule 1-deficient mice. J Exp Med. 1994;180:95–109. doi: 10.1084/jem.180.1.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Farhood A, McGuire GM, Manning AM, Miyasaka M, Smith CW, Jaeschke H. Intercellular adhesion molecule 1 (ICAM-1) expression and its role in neutrophil-induced ischemia-reperfusion injury in rat liver. J Leukoc Biol. 1995;57:368–374. [PubMed] [Google Scholar]
- [78].Gujral JS, Liu J, Farhood A, Hinson JA, Jaeschke H. Functional importance of ICAM-1 in the mechanism of neutrophil-induced liver injury in bile duct-ligated mice. Am J Physiol Gastrointest Liver Physiol. 2004;286:G499–G507. doi: 10.1152/ajpgi.00318.2003. [DOI] [PubMed] [Google Scholar]
- [79].Zimmerman GA1. McIntyre TM. Neutrophil adherence to human endothelium in vitro occurs by CDw18 (Mo1, MAC-1/LFA-1/GP 150, 95) glycoprotein-dependent and-indepen-dent mechanisms. J Clin Invest. 1988;81:531–537. doi: 10.1172/JCI113351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Lehnert M, Arteel GE, Smutney OM, Conzelmann LO, Zhong Z, Thurman RG, Lemasters JJ. Dependence of liver injury after hemorrhage/resuscitation in mice on NADPH oxidase-derived superoxide. Shock. 2003;19:345–351. doi: 10.1097/00024382-200304000-00009. [DOI] [PubMed] [Google Scholar]
- [81].Gujral JS, Hinson JA, Jaeschke H. Chlorotyrosine protein adducts are reliable biomarkers of neutrophil-induced cyto-toxicity in vivo. Comp Hepatol. 2004;3(Suppl 1):S48. doi: 10.1186/1476-5926-2-S1-S48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Kato Y. Neutrophil myeloperoxidase and its substrates: formation of specific markers and reactive compounds during inflammation. J Clin Biochem Nutr. 2016;58:99–104. doi: 10.3164/jcbn.15-104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Hazell LJ, Arnold L, Flowers D, Waeg G, Malle E, Stocker R. Presence of hypochlorite-modified proteins in human atherosclerotic lesions. J Clin Invest. 1996;97:1535–44. doi: 10.1172/JCI118576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Gujral JS, Hinson JA, Farhood A, Jaeschke H. NADPH oxidase-derived oxidant stress is critical for neutrophil cytotoxicity during endotoxemia. Am J Physiol Gastrointest Liver Physiol. 2004;287:G243–G252. doi: 10.1152/ajpgi.00287.2003. [DOI] [PubMed] [Google Scholar]
- [85].Sandhaus RA, Turino G. Neutrophil elastase-mediated lung disease. COPD. 2013;10(Suppl 1):60–3. doi: 10.3109/15412555.2013.764403. [DOI] [PubMed] [Google Scholar]
- [86].Huebener P, Pradere JP, Hernandez C, Gwak GY, Caviglia JM, Mu X, Loike JD, Jenkins RE, Antoine DJ, Schwabe RF. The HMGB1/RAGE axis triggers neutrophil-mediated injury amplification following necrosis. J Clin Invest. 2015;125:539–550. doi: 10.1172/JCI76887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–1535. doi: 10.1126/science.1092385. [DOI] [PubMed] [Google Scholar]
- [88].Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, Keys EM, Allen-Vercoe E, Devinney R, Doig CJ, Green FH, Kubes P. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 2007;13:463–469. doi: 10.1038/nm1565. [DOI] [PubMed] [Google Scholar]
- [89].Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V. Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science. 2015;349:316–320. doi: 10.1126/science.aaa8064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Ge L, Zhou X, Ji WJ, Lu RY, Zhang Y, Zhang YD, Ma YQ, Zhao JH, Li YM. Neutrophil extracellular traps in ischemia-reperfusion injury-induced myocardial no-reflow: therapeutic potential of DNase-based reperfusion strategy. Am J Physiol Heart Circ Physiol. 2015;308:H500–H509. doi: 10.1152/ajpheart.00381.2014. [DOI] [PubMed] [Google Scholar]
- [91].Huang H, Tohme S, Al-Khafaji AB, Tai S, Loughran P, Chen L, Wang S, Kim J, Billiar T, Wang Y, Tsung A. Damage-associated molecular pattern-activated neutrophil extracellular trap exacerbates sterile inflammatory liver injury. Hepatology. 2015;62:600–614. doi: 10.1002/hep.27841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Jaeschke H, Hasegawa T. Role of neutrophils in acute inflammatory liver injury. Liver Int. 2006;26:912–9. doi: 10.1111/j.1478-3231.2006.01327.x. [DOI] [PubMed] [Google Scholar]
- [93].Godfrey DI, Hammond KJ, Poulton LD, Smyth MJ, Baxter AG. NKT cells: facts, functions and fallacies. Immunol Today. 2000;21:573–583. doi: 10.1016/s0167-5699(00)01735-7. [DOI] [PubMed] [Google Scholar]
- [94].Swain MG. Hepatic NKT cells: friend or foe? Clin Sci (Lond) 2008;114:457–466. doi: 10.1042/CS20070328. [DOI] [PubMed] [Google Scholar]
- [95].Shimoda S, Harada K, Niiro H, Shirabe K, Taketomi A, Maehara Y, Tsuneyama K, Nakanuma Y, Leung P, Ansari AA, Gershwin ME, Akashi K. Interaction between Toll-like receptors and natural killer cells in the destruction of bile ducts in primary biliary cirrhosis. Hepatology. 2011;53:1270–1281. doi: 10.1002/hep.24194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Liu ZX, Govindarajan S, Kaplowitz N. Innate immune system plays a critical role in determining the progression and severity of acetaminophen hepatotoxicity. Gastroenterology. 2004;127:1760–1774. doi: 10.1053/j.gastro.2004.08.053. [DOI] [PubMed] [Google Scholar]
- [97].Feng M, Li G, Qian X, Fan Y, Huang X, Zhang F, Lu L. IL-17A-producing NK cells were implicated in liver injury induced by ischemia and reperfusion. Int Immunopharmacol. 2012;13:135–140. doi: 10.1016/j.intimp.2012.03.007. [DOI] [PubMed] [Google Scholar]
- [98].Downs I, Aw TY, Liu J, Adegboyega P, Ajuebor MN. Va14iNKT cell deficiency prevents acetaminophen-induced acute liver failure by enhancing hepatic glutathione and altering APAP metabolism. Biochem Biophys Res Commun. 2012;428:245–251. doi: 10.1016/j.bbrc.2012.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Martin-Murphy BV, Kominsky DJ, Orlicky DJ, Donohue TM, Jr, Ju C. Increased susceptibility of natural killer T-cell-defic-ient mice to acetaminophen-induced liver injury. Hepatology. 2013;57:1575–1584. doi: 10.1002/hep.26134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Plitas G, Burt BM, Stableford JA, Nguyen HM, Welles AP, DeMatteo RP. Dendritic cells are required for effective cross-presentation in the murine liver. Hepatology. 2008;47:1343–1351. doi: 10.1002/hep.22167. [DOI] [PubMed] [Google Scholar]
- [101].Connolly MK, Ayo D, Malhotra A, Hackman M, Bedrosian AS, Ibrahim J, Cieza-Rubio NE, Nguyen AH, Henning JR, Dorvil-Castro M, Pachter HL, Miller G. Dendritic cell depletion exacerbates acetaminophen hepatotoxicity. Hepatology. 2011;54:959–968. doi: 10.1002/hep.24429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Jaeschke H. Molecular mechanisms of hepatic ischemia-reperfusion injury and preconditioning. Am J Physiol Gastrointest Liver Physiol. 2003;284:G15–26. doi: 10.1152/ajpgi.00342.2002. [DOI] [PubMed] [Google Scholar]
- [103].van Golen RF, Reiniers MJ, Olthof PB, van Gulik TM, Heger M. Sterile inflammation in hepatic ischemia/reperfusion injury: present concepts and potential therapeutics. J Gastroenterol Hepatol. 2013;28:394–400. doi: 10.1111/jgh.12072. [DOI] [PubMed] [Google Scholar]
- [104].Zhai Y, Petrowsky H, Hong JC, Busuttil RW, Kupiec-Weglinski JW. Ischaemia-reperfusion injury in liver transplantation--from bench to bedside. Nat Rev Gastroenterol Hepatol. 2013;10:79–89. doi: 10.1038/nrgastro.2012.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Herman B, Gores GJ, Nieminen AL, Kawanishi T, Harman A, Lemasters JJ. Calcium and pH in anoxic and toxic injury. Crit Rev Toxicol. 1990;21:127–48. doi: 10.3109/10408449009089876. [DOI] [PubMed] [Google Scholar]
- [106].Kehrer JP, Jones DP, Lemasters JJ, Farber JL, Jaeschke H. Mechanisms of hypoxic cell injury. Toxicol Appl Pharmacol; Summary of the symposium presented at the 1990 annual meeting of the Society of Toxicology; 1990. pp. 165–78. [DOI] [PubMed] [Google Scholar]
- [107].Anundi I, King J, Owen DA, Schneider H, Lemasters JJ, Thurman RG. Fructose prevents hypoxic cell death in liver. Am J Physiol. 1987;253:G390–G396. doi: 10.1152/ajpgi.1987.253.3.G390. [DOI] [PubMed] [Google Scholar]
- [108].Jaeschke H, Smith CV, Mitchell JR. Hypoxic damage generates reactive oxygen species in isolated perfused rat liver. Biochem Biophys Res Commun. 1988;150:568–574. doi: 10.1016/0006-291x(88)90431-7. [DOI] [PubMed] [Google Scholar]
- [109].Jaeschke H, Smith CV, Mitchell JR. Reactive oxygen species during ischemia-reflow injury in isolated perfused rat liver. J Clin Invest. 1988;81:1240–1246. doi: 10.1172/JCI113441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Jaeschke H, Mitchell JR. Mitochondria and xanthine oxidase both generate reactive oxygen species in isolated perfused rat liver after hypoxic injury. Biochem Biophys Res Commun. 1989;160:140–147. doi: 10.1016/0006-291x(89)91632-x. [DOI] [PubMed] [Google Scholar]
- [111].Horvatits T, Trauner M, Fuhrmann V. Hypoxic liver injury and cholestasis in critically ill patients. Curr Opin Crit Care. 2013;19:128–132. doi: 10.1097/MCC.0b013e32835ec9e6. [DOI] [PubMed] [Google Scholar]
- [112].Weemhoff JL, Woolbright BL, Jenkins RE, McGill MR, Sharpe MR, Olson JC, Antoine DJ, Curry SC, Jaeschke H. Plasma biomarkers to study mechanisms ozf liver injury in patients with hypoxic hepatitis. Liver Int. 2017;37:377–384. doi: 10.1111/liv.13202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Jaeschke H, Lemasters JJ. Apoptosis versus oncotic necrosis in hepatic ischemia/reperfusion injury. Gastroenterology. 2003;125:1246–1257. doi: 10.1016/s0016-5085(03)01209-5. [DOI] [PubMed] [Google Scholar]
- [114].Hu J, Ramshesh VK, McGill MR, Jaeschke H, Lemasters JJ. Low Dose Acetaminophen Induces Reversible Mitochondrial Dysfunction Associated with Transient c-Jun N-Terminal Kinase Activation in Mouse Liver. Toxicol Sci. 2016;150:204–215. doi: 10.1093/toxsci/kfv319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Tsung A, Sahai R, Tanaka H, Nakao A, Fink MP, Lotze MT, Yang H, Li J, Tracey KJ, Geller DA, Billiar TR. The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J Exp Med. 2005;201:1135–1143. doi: 10.1084/jem.20042614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Yang M, Antoine DJ, Weemhoff JL, Jenkins RE, Farhood A, Park BK, Jaeschke H. Biomarkers distinguish apoptotic and necrotic cell death during hepatic ischemia/reperfusion injury in mice. Liver Transpl. 2014a;20:1372–1382. doi: 10.1002/lt.23958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Nace GW, Huang H, Klune JR, Eid RE, Rosborough BR, Korff S, Li S, Shapiro RA, Stolz DB, Sodhi CP, Hackam DJ, Geller DA, Billiar TR, Tsung A. Cellular-specific role of toll-like receptor 4 in hepatic ischemia-reperfusion injury in mice. Hepatology. 2013;58:374–387. doi: 10.1002/hep.26346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Jaeschke H, Bautista AP, Spolarics Z, Spitzer JJ. Superoxide generation by Kupffer cells and priming of neutrophils during reperfusion after hepatic ischemia. Free Radic Res Commun. 1991;15:277–284. doi: 10.3109/10715769109105223. [DOI] [PubMed] [Google Scholar]
- [119].Colletti LM, Remick DG, Burtch GD, Kunkel SL, Strieter RM, Campbell DA., Jr Role of tumor necrosis factor-alpha in the pathophysiologic alterations after hepatic ischemia/reperfusion injury in the rat. J Clin Invest. 1990;85:1936–1943. doi: 10.1172/JCI114656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Lentsch AB, Yoshidome H, Cheadle WG, Miller FN, Edwards MJ. Chemokine involvement in hepatic ischemia/reperfusion injury in mice: roles for macrophage inflammatory protein-2 and KC. Hepatology. 1998;27:1172–1177. doi: 10.1002/hep.510270440. [DOI] [PubMed] [Google Scholar]
- [121].Kamo N, Ke B, Ghaffari AA, Shen XD, Busuttil RW, Cheng G, Kupiec-Weglinski JW. ASC/caspase-1/IL-1ß signaling triggers inflammatory responses by promoting HMGB1 induction in liver ischemia/reperfusion injury. Hepatology. 2013;58:351–362. doi: 10.1002/hep.26320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Huang H, Chen HW, Evankovich J, Yan W, Rosborough BR, Nace GW, Ding Q, Loughran P, Beer-Stolz D, Billiar TR, Esmon CT, Tsung A. Histones activate the NLRP3 inflammasome in Kupffer cells during sterile inflammatory liver injury. J Immunol. 2013;191:2665–2679. doi: 10.4049/jimmunol.1202733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Kukan M, Vajdovà K, Horecky J, Nagyovà A, Mehendale HM, Trnovec T. Effects of blockade of Kupffer cells by gadolinium chloride on hepatobiliary function in cold ischemia-reperfusion injury of rat liver. Hepatology. 1997;26:1250–1257. doi: 10.1002/hep.510260524. [DOI] [PubMed] [Google Scholar]
- [124].Bilzer M, Paumgartner G, Gerbes AL. Glutathione protects the rat liver against reperfusion injury after hypothermic preservation. Gastroenterology. 1999;117:200–210. doi: 10.1016/s0016-5085(99)70568-8. [DOI] [PubMed] [Google Scholar]
- [125].Liu P, Fisher MA, Farhood A, Smith CW, Jaeschke H. Beneficial effects of extracellular glutathione against endotox-in-induced liver injury during ischemia and reperfusion. Circ Shock. 1994;43:64–70. [PubMed] [Google Scholar]
- [126].Nakano H, Nagasaki H, Barama A, Boudjema K, Jaeck D, Kumada K, Tatsuno M, Baek Y, Kitamura N, Suzuki T, Yamaguchi M. The effects of N-acetylcysteine and anti-intercellular adhesion molecule-1 monoclonal antibody against ischemia-reperfusion injury of the rat steatotic liver produced by a choline-methionine-deficient diet. Hepatology. 1997;26:670–678. doi: 10.1053/jhep.1997.v26.pm0009303498. [DOI] [PubMed] [Google Scholar]
- [127].Chu MJ, Vather R, Hickey AJ, Phillips AR, Bartlett AS. Impact of ischaemic preconditioning on experimental steatotic livers following hepatic ischaemia-reperfusion injury: a systematic review. HPB (Oxford) 2015;17:1–10. doi: 10.1111/hpb.12258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Jaeschke H, Bautista AP, Spolarics Z, Spitzer JJ. Superoxide generation by neutrophils and Kupffer cells during in vivo reperfusion after hepatic ischemia in rats. J Leukoc Biol. 1992;52:377–382. doi: 10.1002/jlb.52.4.377. [DOI] [PubMed] [Google Scholar]
- [129].Okaya T, Lentsch AB. Cytokine cascades and the hepatic inflammatory response to ischemia and reperfusion. J Invest Surg. 2003;16:141–147. [PubMed] [Google Scholar]
- [130].Jaeschke H, Farhood A, Bautista AP, Spolarics Z, Spitzer JJ, Smith CW. Functional inactivation of neutrophils with a Mac-1 (CD11b/CD18) monoclonal antibody protects against ischemia-reperfusion injury in rat liver. Hepatology. 1993;17:915–923. [PubMed] [Google Scholar]
- [131].Bamboat ZM, Balachandran VP, Ocuin LM, Obaid H, Plitas G, DeMatteo RP. Toll-like receptor 9 inhibition confers protection from liver ischemia-reperfusion injury. Hepatology. 2010;51:621–632. doi: 10.1002/hep.23365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Zwacka RM, Zhang Y, Halldorson J, Schlossberg H, Dudus L, Engelhardt JF. CD4(+) T-lymphocytes mediate ischemia/reperfusion-induced inflammatory responses in mouse liver. J Clin Invest. 1997;100:279–289. doi: 10.1172/JCI119533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Caldwell CC, Okaya T, Martignoni A, Husted T, Schuster R, Lentsch AB. Divergent functions of CD4+ T lymphocytes in acute liver inflammation and injury after ischemia-reperfusion. Am J Physiol Gastrointest Liver Physiol. 2005;289:G969–G976. doi: 10.1152/ajpgi.00223.2005. [DOI] [PubMed] [Google Scholar]
- [134].Loi P, Yuan Q, Torres D, Delbauve S, Laute MA, Lalmand MC, Pétein M, Goriely S, Goldman M, Flamand V. Interferon regulatory factor 3 deficiency leads to interleukin-17-mediated liver ischemia-reperfusion injury. Hepatology. 2013;57:351–361. doi: 10.1002/hep.26022. [DOI] [PubMed] [Google Scholar]
- [135].Wilson GC, Freeman CM, Kuethe JW, Quillin RC, 3rd, Nojima H, Schuster R, Blanchard J, Edwards MJ, Caldwell CC, Lentsch AB. CXC chemokine receptor-4 signaling limits hepatocyte proliferation after hepatic ischemia-reperfusion in mice. Am J Physiol Gastrointest Liver Physiol. 2015;308:G702–G709. doi: 10.1152/ajpgi.00257.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Sakai N, Van Sweringen HL, Belizaire RM, Quillin RC, Schuster R, Blanchard J, Burns JM, Tevar AD, Edwards MJ, Lentsch AB. Interleukin-37 reduces liver inflammatory injury via effects on hepatocytes and non-parenchymal cells. J Gastroenterol Hepatol. 2012;27:1609–16. doi: 10.1111/j.1440-1746.2012.07187.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Sakai N, Van Sweringen HL, Quillin RC, Schuster R, Blanchard J, Burns JM, Tevar AD, Edwards MJ, Lentsch AB. Interleukin-33 is hepatoprotective during liver ischemia/reperfusion in mice. Hepatology. 2012;56:1468–1478. doi: 10.1002/hep.25768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Lentsch AB, Yoshidome H, Warner RL, Ward PA, Edwards MJ. Secretory leukocyte protease inhibitor in mice regulates local and remote organ inflammatory injury induced by hepatic ischemia/reperfusion. Gastroenterology. 1999;117:953–961. doi: 10.1016/s0016-5085(99)70355-0. [DOI] [PubMed] [Google Scholar]
- [139].Yoshidome H, Kato A, Edwards MJ, Lentsch AB. Interleukin-10 suppresses hepatic ischemia/reperfusion injury in mice: implications of a central role for nuclear factor kappaB. Hepatology. 1999;30:203–208. doi: 10.1002/hep.510300120. [DOI] [PubMed] [Google Scholar]
- [140].Kato A, Yoshidome H, Edwards MJ, Lentsch AB. Regulation of liver inflammatory injury by signal transducer and activator of transcription-6. Am J Pathol. 2000;157:297–302. doi: 10.1016/S0002-9440(10)64540-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Camargo CA, Jr, Madden JF, Gao W, Selvan RS, Clavien PA. Interleukin-6 protects liver against warm ischemia/reperfusion injury and promotes hepatocyte proliferation in the rodent. Hepatology. 1997;26:1513–1520. doi: 10.1002/hep.510260619. [DOI] [PubMed] [Google Scholar]
- [142].Schmidt SC, Hamann S, Langrehr JM, Höflich C, Mittler J, Jacob D, Neuhaus P. Preoperative high-dose steroid administration attenuates the surgical stress response following liver resection: results of a prospective randomized study. J Hepatobiliary Pancreat Surg. 2007;14:484–492. doi: 10.1007/s00534-006-1200-7. [DOI] [PubMed] [Google Scholar]
- [143].Saito C, Zwingmann C, Jaeschke H. Novel mechanisms of protection against acetaminophen hepatotoxicity in mice by glutathione and N-acetylcysteine. Hepatology. 2010b;51:246–254. doi: 10.1002/hep.23267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Smilkstein MJ, Knapp GL, Kulig KW, Rumack BH. Efficacy of oral N-acetylcysteine in the treatment of acetaminophen overdose. Analysis of the national multicenter study. N Engl J Med. 1976 to 1985;1988;319:1557–1562. doi: 10.1056/NEJM198812153192401. [DOI] [PubMed] [Google Scholar]
- [145].Ishida Y, Kondo T, Kimura A, Tsuneyama K, Takayasu T, Mukaida N. Opposite roles of neutrophils and macrophages in the pathogenesis of acetaminophen-induced acute liver injury. Eur J Immunol. 2006;36:1028–1038. doi: 10.1002/eji.200535261. [DOI] [PubMed] [Google Scholar]
- [146].Laskin DL, Gardner CR, Price VF, Jollow DJ. Modulation of macrophage functioning abrogates the acute hepatotoxicity of acetaminophen. Hepatology. 1995;21:1045–1050. [PubMed] [Google Scholar]
- [147].Liu ZX, Han D, Gunawan B, Kaplowitz N. Neutrophil depletion protects against murine acetaminophen hepatotoxicity. Hepatology. 2006;43:1220–1230. doi: 10.1002/hep.21175. [DOI] [PubMed] [Google Scholar]
- [148].Jaeschke H, Williams CD, Ramachandran A, Bajt ML. Acetaminophen hepatotoxicity and repair: the role of sterile inflammation and innate immunity. Liver Int. 2012;32:8–20. doi: 10.1111/j.1478-3231.2011.02501.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Mitchell JR, Jollow DJ, Potter WZ, Gillette JR, Brodie BB. Acetaminophen-induced hepatic necrosis. IV. Protective role of glutathione. J Pharmacol Exp Ther. 1973 Oct;187:211–217. [PubMed] [Google Scholar]
- [150].Hjelle JJ, Klaassen CD. Glucuronidation and biliary excretion of acetaminophen in rats. J Pharmacol Exp Ther. 1984;228:407–413. [PubMed] [Google Scholar]
- [151].Xie Y, McGill MR, Cook SF, Sharpe MR, Winefield RD, Wilkins DG, Rollins DE, Jaeschke H. Time course of acetamino-phen-protein adducts and acetaminophen metabolites in circulation of overdose patients and in HepaRG cells. Xenobiotica. 2015;45:921–929. doi: 10.3109/00498254.2015.1026426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Dahlin DC, Miwa GT, Lu AY, Nelson SD. N-acetyl-p-benzoquinone imine: a cytochrome P-450-mediated oxidation product of acetaminophen. Proc Natl Acad Sci USA. 1984;81:1327–1331. doi: 10.1073/pnas.81.5.1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Jaeschke H, McGill MR, Ramachandran A. Oxidant stress, mitochondria, and cell death mechanisms in drug-induced liver injury: lessons learned from acetaminophen hepatotoxicity. Drug Metab Rev. 2012;44:88–106. doi: 10.3109/03602532.2011.602688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Hanawa N, Shinohara M, Saberi B, Gaarde WA, Han D, Kaplowitz N. Role of JNK translocation to mitochondria leading to inhibition of mitochondria bioenergetics in acetaminophen-induced liver injury. J Biol Chem. 2008;283:13565–13577. doi: 10.1074/jbc.M708916200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Sharma M, Gadang V, Jaeschke A. Critical role for mixed-lineage kinase 3 in acetaminophen-induced hepatotoxicity. Mol Pharmacol. 2012;82:1001–1007. doi: 10.1124/mol.112.079863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Xie Y, Ramachandran A, Breckenridge DG, Liles JT, Lebofsky M, Farhood A, Jaeschke H. Inhibitor of apoptosis signal-regulating kinase 1 protects against acetaminophen-induced liver injury. Toxicol Appl Pharmacol. 2015;286:1–9. doi: 10.1016/j.taap.2015.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Win S, Than TA, Min RW, Aghajan M, Kaplowitz N. c-Jun N-terminal kinase mediates mouse liver injury through a novel Sab (SH3BP5)-dependent pathway leading to inactivation of intramitochondrial Src. Hepatology. 2016;63:1987–2003. doi: 10.1002/hep.28486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Du K, Xie Y, McGill MR, Jaeschke H. Pathophysiological significance of c-jun N-terminal kinase in acetaminophen hepatotoxicity. Expert Opin Drug Metab Toxicol. 2015;11:1769–1779. doi: 10.1517/17425255.2015.1071353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Enomoto A, Itoh K, Nagayoshi E, Haruta J, Kimura T, O'Connor T, Harada T, Yamamoto M. High sensitivity of Nrf2 knockout mice to acetaminophen hepatotoxicity associated with decreased expression of ARE-regulated drug metabolizing enzymes and antioxidant genes. Toxicol Sci. 2001;59:169–177. doi: 10.1093/toxsci/59.1.169. [DOI] [PubMed] [Google Scholar]
- [160].Okawa H, Motohashi H, Kobayashi A, Aburatani H, Kensler TW, Yamamoto M. Hepatocyte-specific deletion of the keap1 gene activates Nrf2 and confers potent resistance against acute drug toxicity. Biochem Biophys Res Commun. 2006;339:79–88. doi: 10.1016/j.bbrc.2005.10.185. [DOI] [PubMed] [Google Scholar]
- [161].El-Hassan H, Anwar K, Macanas-Pirard P, Crabtree M, Chow SC, Johnson VL, Lee PC, Hinton RH, Price SC, Kass GE. Involvement of mitochondria in acetaminophen-induced apoptosis and hepatic injury: roles of cytochrome c, Bax, Bid, and caspases. Toxicol Appl Pharmacol. 2003;191:118–129. doi: 10.1016/s0041-008x(03)00240-0. [DOI] [PubMed] [Google Scholar]
- [162].Bajt ML, Farhood A, Lemasters JJ, Jaeschke H. Mitochondrial bax translocation accelerates DNA fragmentation and cell necrosis in a murine model of acetaminophen hepatotoxicity. J Pharmacol Exp Ther. 2008;324:8–14. doi: 10.1124/jpet.107.129445. [DOI] [PubMed] [Google Scholar]
- [163].Ramachandran A, McGill MR, Xie Y, Ni HM, Ding WX, Jaeschke H. Receptor interacting protein kinase 3 is a critical early mediator of acetaminophen-induced hepatocyte necrosis in mice. Hepatology. 2013;58:2099–2108. doi: 10.1002/hep.26547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Zhang YF, He W, Zhang C, Liu XJ, Lu Y, Wang H, Zhang ZH, Chen X, Xu DX. Role of receptor interacting protein (RIP)1 on apoptosis-inducing factor-mediated necroptosis during ac-etaminophen-evoked acute liver failure in mice. Toxicol Lett. 2014;225:445–453. doi: 10.1016/j.toxlet.2014.01.005. [DOI] [PubMed] [Google Scholar]
- [165].Dara L, Johnson H, Suda J, Win S, Gaarde W, Han D, Kaplowitz N. Receptor interacting protein kinase 1 mediates murine acetaminophen toxicity independent of the necrosome and not through necroptosis. Hepatology. 2015;62:1847–1857. doi: 10.1002/hep.27939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [166].Zhang J, Yang Y, He W, Sun L. Necrosome core machinery: MLKL. Cell Mol Life Sci. 2016;73:2153–2163. doi: 10.1007/s00018-016-2190-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [167].Günther C, He GW, Kremer AE, Murphy JM, Petrie EJ, Amann K, Vandenabeele P, Linkermann A, Poremba C, Schleicher U, Dewitz C, Krautwald S, Neurath MF, Becker C, Wirtz S. The pseudokinase MLKL mediates programmed hepatocellular necrosis independently of RIPK3 during hepatitis. J Clin Invest. 2016;126:4346–4360. doi: 10.1172/JCI87545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Kon K, Kim JS, Jaeschke H, Lemasters JJ. Mitochondrial permeability transition in acetaminophen-induced necrosis and apoptosis of cultured mouse hepatocytes. Hepatology. 2004;40:1170–1179. doi: 10.1002/hep.20437. [DOI] [PubMed] [Google Scholar]
- [169].Gunawan BK, Liu ZX, Han D, Hanawa N, Gaarde WA, Kaplowitz N. c-Jun N-terminal kinase plays a major role in murineacetaminophen hepatotoxicity. Gastroenterology. 2006;131:165–178. doi: 10.1053/j.gastro.2006.03.045. [DOI] [PubMed] [Google Scholar]
- [170].Saito C, Lemasters JJ, Jaeschke H. c-Jun N-terminal kinase modulates oxidant stress and peroxynitrite formation independent of inducible nitric oxide synthase in acetaminophen hepatotoxicity. Toxicol Appl Pharmacol. 2010;246:8–17. doi: 10.1016/j.taap.2010.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [171].Williams CD, Koerner MR, Lampe JN, Farhood A, Jaeschke H. Mouse strain-dependent caspase activation during acetaminophen hepatotoxicity does not result in apoptosis or modulation of inflammation. Toxicol Appl Pharmacol. 2011;257:449–458. doi: 10.1016/j.taap.2011.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [172].Bajt ML, Cover C, Lemasters JJ, Jaeschke H. Nuclear translocation of endonuclease G and apoptosis-inducing factor during acetaminophen-induced liver cell injury. Toxicol Sci. 2006;94:217–225. doi: 10.1093/toxsci/kfl077. [DOI] [PubMed] [Google Scholar]
- [173].Bajt ML, Ramachandran A, Yan HM, Lebofsky M, Farhood A, Lemasters JJ, Jaeschke H. Apoptosis-inducing factor modulates mitochondrial oxidant stress in acetaminophen hepatotoxicity. Toxicol Sci. 2011;122:598–605. doi: 10.1093/toxsci/kfr116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Ni HM, Bockus A, Boggess N, Jaeschke H, Ding WX. Activation of autophagy protects against acetaminophen-induced hepatotoxicity. Hepatology. 2012;55:222–232. doi: 10.1002/hep.24690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Williams JA, Ni HM, Haynes A, Manley S, Li Y, Jaeschke H, Ding WX. Chronic deletion and acute knockdown of parkin have differential responses to acetaminophen-induced mitophagy and liver injury in mice. J Biol Chem. 2015;290:10934–10946. doi: 10.1074/jbc.M114.602284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Ni HM, McGill MR, Chao X, Du K, Williams JA, Xie Y, Jaeschke H, Ding WX. Removal of acetaminophen protein adducts by autophagy protects against acetaminophen-induced liver injury in mice. J Hepatol. 2016;65:354–362. doi: 10.1016/j.jhep.2016.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Ni HM, Williams JA, Jaeschke H, Ding WX. Zonated induction of autophagy and mitochondrial spheroids limits acetaminophen-induced necrosis in the liver. Redox Biol. 2013;1:427–432. doi: 10.1016/j.redox.2013.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Michael SL, Pumford NR, Mayeux PR, Niesman MR, Hinson JA. Pretreatment of mice with macrophage inactivators decreases acetaminophen hepatotoxicity and the formation of reactive oxygen and nitrogen species. Hepatology. 1999;30:186–195. doi: 10.1002/hep.510300104. [DOI] [PubMed] [Google Scholar]
- [179].Imaeda AB, Watanabe A, Sohail MA, Mahmood S, Mohamadnejad M, Sutterwala FS, Flavell RA, Mehal WZ. Acetaminophen-induced hepatotoxicity in mice is dependent on Tlr9 and the Nalp3 inflammasome. J Clin Invest. 2009;119:305–314. doi: 10.1172/JCI35958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Cai C, Huang H, Whelan S, Liu L, Kautza B, Luciano J, Wang G, Chen G, Stratimirovic S, Tsung A, Billiar TR, Zuckerbraun BS. Benzyl alcohol attenuates acetaminophen-induced acute liver injury in a Toll-like receptor-4-dependent pattern in mice. Hepatology. 2014;60:990–1002. doi: 10.1002/hep.27201. [DOI] [PubMed] [Google Scholar]
- [181].Marques PE, Amaral SS, Pires DA, Nogueira LL, Soriani FM, Lima BH, Lopes GA, Russo RC, Avila TV, Melgaco JG, Oliveira AG, Pinto MA, Lima CX, De Paula AM, Cara DC, Leite MF, Teixeira MM, Menezes GB. Chemokines and mitochondrial products activate neutrophils to amplify organ injury during mouse acute liver failure. Hepatology. 2012;56:1971–1982. doi: 10.1002/hep.25801. [DOI] [PubMed] [Google Scholar]
- [182].Marques PE, Oliveira AG, Pereira RV, David BA, Gomides LF, Saraiva AM, Pires DA, Novaes JT, Patricio DO, Cisalpino D, Menezes-Garcia Z, Leevy WM, Chapman SE, Mahecha G, Marques RE, Guabiraba R, Martins VP, Souza DG, Mansur DS, Teixeira MM, Leite MF, Menezes GB. Hepatic DNA deposition drives drug-induced liver injury and inflammation in mice. Hepatology. 2015;61:348–360. doi: 10.1002/hep.27216. [DOI] [PubMed] [Google Scholar]
- [183].Masson MJ, Carpenter LD, Graf ML, Pohl LR. Pathogenic role of natural killer T and natural killer cells inacetaminophen-induced liver injury in mice is dependent on the presence of dimethyl sulfoxide. Hepatology. 2008;48:889–897. doi: 10.1002/hep.22400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Cover C, Liu J, Farhood A, Malle E, Waalkes MP, Bajt ML, Jaeschke H. Pathophysiological role of the acute inflammatory response during acetaminophen hepatotoxicity. Toxicol Appl Pharmacol. 2006;216:98–107. doi: 10.1016/j.taap.2006.04.010. [DOI] [PubMed] [Google Scholar]
- [185].Kodali P, Wu P, Lahiji PA, Brown EJ, Maher JJ. ANIT toxicity toward mouse hepatocytes in vivo is mediated primarily by neutrophils via CD18. Am J Physiol Gastrointest Liver Physiol. 2006;291:G355–63. doi: 10.1152/ajpgi.00458.2005. [DOI] [PubMed] [Google Scholar]
- [186].Jaeschke H, Liu J. Neutrophil depletion protects against murine acetaminophen hepatotoxicity: another perspective. Hepatology. 2007;45:1588–1589. doi: 10.1002/hep.21549. [DOI] [PubMed] [Google Scholar]
- [187].Williams CD, Bajt ML, Farhood A, Jaeschke H. Acetaminophen-induced hepatic neutrophil accumulation and inflammatory liver injury in CD18-deficient mice. Liver Int. 2010;30:1280–1292. doi: 10.1111/j.1478-3231.2010.02284.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Williams CD, Bajt ML, Sharpe MR, McGill MR, Farhood A, Jaeschke H. Neutrophil activation during acetaminophen hepatotoxicity and repair in mice and humans. Toxicol Appl Pharmacol. 2014;275:122–133. doi: 10.1016/j.taap.2014.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Hu B, Colletti LM. CXC receptor-2 knockout genotype increases X-linked inhibitor of apoptosis protein and protects mice from acetaminophen hepatotoxicity. Hepatology. 2010;52:691–702. doi: 10.1002/hep.23715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Gujral JS, Knight TR, Farhood A, Bajt ML, Jaeschke H. Mode of cell death after acetaminophen overdose in mice: apoptosis or oncotic necrosis? Toxicol Sci. 2002;67:322–328. doi: 10.1093/toxsci/67.2.322. [DOI] [PubMed] [Google Scholar]
- [191].Schulze-Osthoff K, Bantel H. Necrosis versus apoptosis in acetaminophen-induced hepatotoxicity. Hepatology. 2011;53:1070. doi: 10.1002/hep.24027. [DOI] [PubMed] [Google Scholar]
- [192].Kono H, Chen CJ, Ontiveros F, Rock KL. Uric acid promotes an acute inflammatory response to sterile cell death in mice. J Clin Invest. 2010;120:1939–1949. doi: 10.1172/JCI40124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Kataoka H, Kono H, Patel Z, Kimura Y, Rock KL. Evaluation of the contribution of multiple DAMPs and DAMP receptors in cell death-induced sterile inflammatory responses. PLoS One. 2014;9:e104741. doi: 10.1371/journal.pone.0104741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [194].Ito Y, Bethea NW, Abril ER, McCuskey RS. Early hepatic microvascular injury in response to acetaminophen toxicity. Microcirculation. 2003;10:391–400. doi: 10.1038/sj.mn.7800204. [DOI] [PubMed] [Google Scholar]
- [195].Ju C, Reilly TP, Bourdi M, Radonovich MF, Brady JN, George JW, Pohl LR. Protective role of Kupffer cells in acetaminophen-induced hepatic injury in mice. Chem Res Toxicol. 2002;15:1504–1513. doi: 10.1021/tx0255976. [DOI] [PubMed] [Google Scholar]
- [196].Knight TR, Jaeschke H. Peroxynitrite formation and sinusoidal endothelial cell injury during acetaminophen-induced hepatotoxicity in mice. Comp Hepatol. 2004 Jan 14;3(Suppl 1):S46. doi: 10.1186/1476-5926-2-S1-S46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [197].Gehring S, Dickson EM, San Martin ME, van Rooijen N, Papa EF, Harty MW, Tracy TF, Jr, Gregory SH. Kupffer cells abrogate cholestatic liver injury in mice. Gastroenterology. 2006;130:810–822. doi: 10.1053/j.gastro.2005.11.015. [DOI] [PubMed] [Google Scholar]
- [198].James LP, McCullough SS, Knight TR, Jaeschke H, Hinson JA. Acetaminophen toxicity in mice lacking NADPH oxidase activity: role of peroxynitrite formation and mitochondrial oxidant stress. Free Radic Res. 2003;37:1289–1297. doi: 10.1080/10715760310001617776. [DOI] [PubMed] [Google Scholar]
- [199].Hogaboam CM, Bone-Larson CL, Steinhauser ML, Matsukawa A, Gosling J, Boring L, Charo IF, Simpson KJ, Lukacs NW, Kunkel SL. Exaggerated hepatic injury due to acetaminophen challenge in mice lacking C-C chemokine receptor 2. Am J Pathol. 2000;156:1245–1252. doi: 10.1016/S0002-9440(10)64995-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [200].Holt MP, Yin H, Ju C. Exacerbation of acetaminophen-induced disturbances of liver sinusoidal endothelial cells in the absence of Kupffer cells in mice. Toxicol Lett. 2010;194:34–41. doi: 10.1016/j.toxlet.2010.01.020. [DOI] [PubMed] [Google Scholar]
- [201].You Q, Holt M, Yin H, Li G, Hu CJ, Ju C. Role of hepatic resident and infiltrating macrophages in liver repair after acute injury. Biochem Pharmacol. 2013;86:836–843. doi: 10.1016/j.bcp.2013.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [202].Antoniades CG, Berry PA, Davies ET, Hussain M, Bernal W, Vergani D, Wendon J. Reduced monocyte HLA-DR expression: a novel biomarker of disease severity and outcome in acetaminophen-induced acute liver failure. Hepatology. 2006;44:34–43. doi: 10.1002/hep.21240. [DOI] [PubMed] [Google Scholar]
- [203].Antoniades CG, Khamri W, Abeles RD, Taams LS, Triantafyllou E, Possamai LA, Bernsmeier C, Mitry RR, O'Brien A, Gilroy D, Goldin R, Heneghan M, Heaton N, Jassem W, Bernal W, Vergani D, Ma Y, Quaglia A, Wendon J, Thursz M. Secretory leukocyte protease inhibitor: a pivotal mediator of anti-inflammatory responses in acetaminophen-induced acute liver failure. Hepatology. 2014;59:1564–1576. doi: 10.1002/hep.26933. [DOI] [PubMed] [Google Scholar]
- [204].Antoniades CG, Quaglia A, Taams LS, Mitry RR, Hussain M, Abeles R, Possamai LA, Bruce M, McPhail M, Starling C, Wagner B, Barnardo A, Pomplun S, Auzinger G, Bernal W, Heaton N, Vergani D, Thursz MR, Wendon J. Source and characterization of hepatic macrophages in acetaminophen-induced acute liver failure in humans. Hepatology. 2012;56:735–746. doi: 10.1002/hep.25657. [DOI] [PubMed] [Google Scholar]
- [205].Amaral SS, Oliveira AG, Marques PE, Quintào JL, Pires DA, Resende RR, Sousa BR, Melgaco JG, Pinto MA, Russo RC, Gomes AK, Andrade LM, Zanin RF, Pereira RV, Bonorino C, Soriani FM, Lima CX, Cara DC, Teixeira MM, Leite MF, Menezes GB. Altered responsiveness to extracellular ATP enhances acetaminophen hepatotoxicity. Cell Commun Signal. 2013;11:10. doi: 10.1186/1478-811X-11-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [206].Williams CD, Farhood A, Jaeschke H. Role of caspase-1 and interleukin-1beta in acetaminophen-induced hepatic inflammation and liver injury. Toxicol Appl Pharmacol. 2010;247:169–178. doi: 10.1016/j.taap.2010.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [207].Williams CD, Antoine DJ, Shaw PJ, Benson C, Farhood A, Williams DP, Kanneganti TD, Park BK, Jaeschke H. Role of the Nalp3 inflammasome in acetaminophen-induced sterile inflammation and liver injury. Toxicol Appl Pharmacol. 2011;252:289–297. doi: 10.1016/j.taap.2011.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [208].Xie Y, Williams CD, McGill MR, Lebofsky M, Ramachandran A, Jaeschke H. Purinergic receptor antagonist A438079 protects against acetaminophen-induced liver injury by inhibiting p450 isoenzymes, not by inflammasome activation. Toxicol Sci. 2013;131:325–335. doi: 10.1093/toxsci/kfs283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [209].Xie Y, Woolbright BL, Kos M, McGill MR, Dorko K, Kumer SC, Schmitt TM, Jaeschke H. Lack of Direct Cytotoxicity of Extracellular ATP against Hepatocytes: Role in the Mechanism of Acetaminophen Hepatotoxicity. J Clin Transl Res. 2015;1:100–106. doi: 10.18053/jctres.201502.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [210].Bourdi M, Masubuchi Y, Reilly TP, Amouzadeh HR, Martin JL, George JW, Shah AG, Pohl LR. Protection against acetaminophen-induced liver injury and lethality by interleukin 10: role of inducible nitric oxide synthase. Hepatology. 2002;35:289–298. doi: 10.1053/jhep.2002.30956. [DOI] [PubMed] [Google Scholar]
- [211].Knight TR, Ho YS, Farhood A, Jaeschke H. Peroxynitrite is a critical mediator of acetaminophen hepatotoxicity in murine livers: protection by glutathione. J Pharmacol Exp Ther. 2002;303:468–475. doi: 10.1124/jpet.102.038968. [DOI] [PubMed] [Google Scholar]
- [212].Cover C, Mansouri A, Knight TR, Bajt ML, Lemasters JJ, Pessayre D, Jaeschke H. Peroxynitrite-induced mitochondrial and endonuclease-mediated nuclear DNA damage in acetaminophen hepatotoxicity. J Pharmacol Exp Ther. 2005;315:879–887. doi: 10.1124/jpet.105.088898. [DOI] [PubMed] [Google Scholar]
- [213].James LP, Lamps LW, McCullough S, Hinson JA. Interleukin 6 and hepatocyte regeneration in acetaminophen toxicity in the mouse. Biochem Biophys Res Commun. 2003a;309:857–863. doi: 10.1016/j.bbrc.2003.08.085. [DOI] [PubMed] [Google Scholar]
- [214].Ryan PM, Bourdi M, Korrapati MC, Proctor WR, Vasquez RA, Yee SB, Quinn TD, Chakraborty M, Pohl LR. Endogenous interleukin-4 regulates glutathione synthesis following acetaminophen-induced liver injury in mice. Chem Res Toxicol. 2012;25:83–93. doi: 10.1021/tx2003992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [215].Pires DA, Marques PE, Pereira RV, David BA, Gomides LF, Dias AC, Nunes-Silva A, Pinho V, Cara DC, Vieira LQ, Teixeira MM, Menezes GB. Interleukin-4 deficiency protects mice from acetaminophen-induced liver injury and inflammation by prevention of glutathione depletion. Inflamm Res. 2014;63:61–69. doi: 10.1007/s00011-013-0671-7. [DOI] [PubMed] [Google Scholar]
- [216].Levy R, Schlaeffer F, Keynan A, Nagauker O, Yaari A, Sikuler E. Increased neutrophil function induced by bile duct ligation in a rat model. Hepatology. 1993;17:908–914. [PubMed] [Google Scholar]
- [217].Van Nieuwkerk CM, Elferink RP, Groen AK, Ottenhoff R, Tytgat GN, Dingemans KP, Van Den Bergh Weerman MA, Offerhaus GJ. Effects of Ursodeoxycholate and cholate feeding on liver disease in FVB mice with a disrupted mdr2 P-glycoprotein gene. Gastroenterology. 1996;111:165–171. doi: 10.1053/gast.1996.v111.pm8698195. [DOI] [PubMed] [Google Scholar]
- [218].Spivey JR, Bronk SF, Gores GJ. Glycochenodeoxycho-late-induced lethal hepatocellular injury in rat hepatocytes. Role of ATP depletion and cytosolic free calcium. J Clin Invest. 1993;92:17–24. doi: 10.1172/JCI116546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [219].Galle PR, Theilmann L, Raedsch R, Otto G, Stiehl A. Ur-sodeoxycholate reduces hepatotoxicity of bile salts in primary human hepatocytes. Hepatology. 1990;12:486–491. doi: 10.1002/hep.1840120307. [DOI] [PubMed] [Google Scholar]
- [220].Kawamata Y, Fujii R, Hosoya M, Harada M, Yoshida H, Miwa M, Fukusumi S, Habata Y, Itoh T, Shintani Y, Hinuma S, Fujisawa Y, Fujino M. A G protein-coupled receptor responsive to bile acids. J Biol Chem. 2003;278:9435–9440. doi: 10.1074/jbc.M209706200. [DOI] [PubMed] [Google Scholar]
- [221].Wang H, Chen J, Hollister K, Sowers LC, Forman BM. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol Cell. 1999;3:543–553. doi: 10.1016/s1097-2765(00)80348-2. [DOI] [PubMed] [Google Scholar]
- [222].Patel T, Bronk SF, Gores GJ. Increases of intracellular magnesium promote glycodeoxycholate-induced apoptosis in rat hepatocytes. J Clin Invest. 1994;94:2183–92. doi: 10.1172/JCI117579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [223].Tamaki N, Hatano E, Taura K, Tada M, Kodama Y, Nitta T, Iwaisako K, Seo S, Nakajima A, Ikai I, Uemoto S. CHOP deficiency attenuates cholestasis-induced liver fibrosis by reduction of hepatocyte injury. Am J Physiol Gastrointest Liver Physiol. 2008;294:G498–G505. doi: 10.1152/ajpgi.00482.2007. [DOI] [PubMed] [Google Scholar]
- [224].Woolbright BL, Li F, Xie Y, Farhood A, Fickert P, Trauner M, Jaeschke H. Lithocholic acid feeding results in direct hepa-to-toxicity independent of neutrophil function in mice. Toxicol Lett. 2014;228:56–66. doi: 10.1016/j.toxlet.2014.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [225].Neale G, Lewis B, Weaver V, Panveliwalla D. Serum bile acids in liver disease. Gut. 1971;12:145–152. doi: 10.1136/gut.12.2.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [226].Zhang Y, Hong JY, Rockwell CE, Copple BL, Jaeschke H, Klaassen CD. Effect of bile duct ligation on bile acid composition in mouse serum and liver. Liver Int. 2012;32:58–69. doi: 10.1111/j.1478-3231.2011.02662.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [227].Woolbright BL, McGill MR, Yan H, Jaeschke H. Bile Acid-Induced Toxicity in HepaRG Cells Recapitulates the Response in Primary Human Hepatocytes. Basic Clin Pharmacol Toxicol. 2016;118:160–167. doi: 10.1111/bcpt.12449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [228].Yang M, Ramachandran A, Yan HM, Woolbright BL, Copple BL, Fickert P, Trauner M, Jaeschke H. Osteopontin is an initial mediator of inflammation and liver injury during obstructive cholestasis after bile duct ligation in mice. Toxicol Lett. 2014;224:186–195. doi: 10.1016/j.toxlet.2013.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [229].Fickert P, Zollner G, Fuchsbichler A, Stumptner C, Weiglein AH, Lammert F, Marschall HU, Tsybrovskyy O, Zatloukal K, Denk H, Trauner M. Ursodeoxycholic acid aggravates bile infarcts in bile duct-ligated and Mdr2 knockout mice via disruption of cholangioles. Gastroenterology. 2002;123:1238–1251. doi: 10.1053/gast.2002.35948. [DOI] [PubMed] [Google Scholar]
- [230].Georgiev P, Jochum W, Heinrich S, Jang JH, Nocito A, Dahm F, Clavien PA. Characterization of time-related changes after experimental bile duct ligation. Br J Surg. 2008;95:646–656. doi: 10.1002/bjs.6050. [DOI] [PubMed] [Google Scholar]
- [231].Levy R, Nagauker O, Sikuler E, Leto TL, Schlaeffer F. Elevated NADPH-oxidase activity in neutrophils from bile-duct-ligated rats: changes in the kinetic parameters and in the oxidase cytosolic factor p47. Biochim Biophys Acta. 1994;1220:261–265. doi: 10.1016/0167-4889(94)90147-3. [DOI] [PubMed] [Google Scholar]
- [232].Gujral JS, Liu J, Farhood A, Jaeschke H. Reduced oncotic necrosis in Fas receptor-deficient C57BL/6J-lpr mice after bile duct ligation. Hepatology. 2004;40:998–1007. doi: 10.1002/hep.20380. [DOI] [PubMed] [Google Scholar]
- [233].Uchinami H, Seki E, Brenner DA, D'Armiento J. Loss of MMP 13 attenuates murine hepatic injury and fibrosis during cholestasis. Hepatology. 2006;44:420–429. doi: 10.1002/hep.21268. [DOI] [PubMed] [Google Scholar]
- [234].Kahraman A, Bronk SF, Cazanave S, Werneburg NW, Mott JL, Contreras PC, Gores GJ. Matrix metalloproteinase inhibitor, CTS-1027, attenuates liver injury and fibrosis in the bile duct-ligated mouse. Hepatol Res. 2009;39:805–813. doi: 10.1111/j.1872-034X.2009.00541.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [235].O'Brien KM, Allen KM, Rockwell CE, Towery K, Luyendyk JP, Copple BL. IL-17A synergistically enhances bile acid-induced inflammation during obstructive cholestasis. Am J Pathol. 2013;183:1498–1507. doi: 10.1016/j.ajpath.2013.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [236].Licata LA, Nguyen CT, Burga RA, Falanga V, Espat NJ, Ayala A, Thorn M, Junghans RP, Katz SC. Biliary obstruction results in PD-1-dependent liver T cell dysfunction and acute inflammation mediated by Th17 cells and neutrophils. J Leukoc Biol. 2013;94:813–823. doi: 10.1189/jlb.0313137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [237].Canbay A, Higuchi H, Bronk SF, Taniai M, Sebo TJ, Gores GJ. Fas enhances fibrogenesis in the bile duct ligated mouse: a link between apoptosis and fibrosis. Gastroenterology. 2002;123:1323–1330. doi: 10.1053/gast.2002.35953. [DOI] [PubMed] [Google Scholar]
- [238].Li M, Mennone A, Soroka CJ, Hagey LR, Ouyang X, Weinman EJ, Boyer JL. Na(+)/H(+) exchanger regulatory factor 1 knockout mice have an attenuated hepatic inflammatory response and are protected from cholestatic liver injury. Hepatology. 2015;62:1227–1236. doi: 10.1002/hep.27956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [239].Woolbright BL, Jaeschke H. Novel insight into mechanisms of cholestatic liver injury. World J Gastroenterol. 2012;18:4985–4993. doi: 10.3748/wjg.v18.i36.4985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [240].Fickert P, Trauner M, Fuchsbichler A, Zollner G, Wagner M, Marschall HU, Zatloukal K, Denk H. Oncosis represents the main type of cell death in mouse models of cholestasis. J Hepatol. 2005;42:378–385. doi: 10.1016/j.jhep.2004.10.016. [DOI] [PubMed] [Google Scholar]
- [241].Kim ND, Moon JO, Slitt AL, Copple BL. Early growth response factor-1 is critical for cholestatic liver injury. Toxicol Sci. 2006;90:586–595. doi: 10.1093/toxsci/kfj111. [DOI] [PubMed] [Google Scholar]
- [242].Rolo AP, Palmeira CM, Holy JM, Wallace KB. Role of mitochondrial dysfunction in combined bile acid-induced cytotoxicity: the switch between apoptosis and necrosis. Toxicol Sci. 2004;79:196–204. doi: 10.1093/toxsci/kfh078. [DOI] [PubMed] [Google Scholar]
- [243].Sokol RJ, Dahl R, Devereaux MW, Yerushalmi B, Kobak GE, Gumpricht E. Human hepatic mitochondria generate reactive oxygen species and undergo the permeability transition in response to hydrophobic bile acids. J Pediatr Gastroenterol Nutr. 2005;41:235–243. doi: 10.1097/01.mpg.0000170600.80640.88. [DOI] [PubMed] [Google Scholar]
- [244].Trottier J, Bialek A, Caron P, Straka RJ, Heathcote J, Milkiewicz P, Barbier O. Metabolomic profiling of 17 bile acids in serum from patients with primary biliary cirrhosis and primary sclerosing cholangitis: a pilot study. Dig Liver Dis. 2012;44:303–310. doi: 10.1016/j.dld.2011.10.025. [DOI] [PubMed] [Google Scholar]
- [245].Trottier J, Bialek A, Caron P, Straka RJ, Milkiewicz P, Barbier O. Profiling circulating and urinary bile acids in patients with biliary obstruction before and after biliary stenting. PLoS One. 2011;6:e22094. doi: 10.1371/journal.pone.0022094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [246].Dilger K, Hohenester S, Winkler-Budenhofer U, Bastiaansen BA, Schaap FG, Rust C, Beuers U. Effect of ursodeoxycholic acid on bile acid profiles and intestinal detoxification machinery in primary biliary cirrhosis and health. J Hepatol. 2012;57:133–140. doi: 10.1016/j.jhep.2012.02.014. [DOI] [PubMed] [Google Scholar]