You are on page 1of 10

View Article Online / Journal Homepage / Table of Contents for this issue

Toxicology Research Dynamic Article Links

Cite this: Toxicol. Res., 2012, 1, 161


www.rsc.org/tx REVIEW
Role of innate and adaptive immunity during drug-induced liver injury
C. David Williams and Hartmut Jaeschke*
Received 23rd May 2012, Accepted 24th August 2012
DOI: 10.1039/c2tx20032e

Drug-induced liver injury (DILI) is a major human health concern and is the most frequent cause of FDA
boxed warnings and the removal of drugs from the market. Idiosyncratic DILI (IDILI) is highly variable
Published on 28 August 2012. Downloaded on 27/10/2014 12:55:05.

in its time to onset and no one clear hypothesis exists to explain the mechanism. The general belief is
most cases of IDILI involve some immune mediated component; however no animal models can
recapitulate human IDILI. Despite numerous drugs that have IDILI potential, the most common cause of
DILI is acetaminophen (APAP) overdose. APAP in animal models and in patients is a dose-dependent
hepatotoxicant resulting in severe centrilobular necrosis and a robust inflammatory response. In this
review we will compare the existing hypotheses for potential causes of IDILI and discuss the potential
roles of immune involvement in DILI. Additionally we will focus on what we have learned from the
mechanisms of APAP toxicity ( protein adduction, mitochondrial dysfunction, oxidant stress, DNA
damage, release of damage associated molecular patterns (DAMPs)) and useful interventions to alleviate
APAP-toxicity (reduced protein binding, scavenging of reactive oxygen, induction of autophagy).
Mechanistically APAP-induced liver injury appears to be fundamentally different from IDILI, however,
there are potential critical events shared between APAP-induced liver injury and IDILI. The strategies and
methods currently being used to study APAP-induced liver injury are described in this review. This
improved insight into mechanisms of APAP-induced injury with initiation, propagation and inflammation
may also help to better understand IDILI.

Introduction
The liver is generally considered a target of drug toxicity
Department of Pharmacology, Toxicology and Therapeutics, University because of its first-pass exposure to orally administered drugs
of Kansas Medical Center, Kansas City, KS, USA.
E-mail: hjaeschke@kumc.edu; Fax: +1 913 588 7501; and its high capacity for xenobiotic metabolism. Phase I (oxi-
Tel: +1 913 588 7969 dation-reduction) and phase II (conjugation) reactions occur

Dr Clarence David Williams Dr Hartmut Jaeschke is cur-


received his doctorate degree in rently Professor and Chairman
toxicology from the University of the Department of Pharma-
of Kansas Medical Center and cology, Toxicology and Thera-
currently works in the labora- peutics at the University of
tory of Dr Jaeschke. Dave has Kansas Medical Center in
previously studied, in an indus- Kansas City. He received a
try setting, the mechanisms of MSc and PhD degree in bio-
immunogenicity to biologic chemistry and toxicology from
therapeutics. Currently his the University of Tübingen,
work focuses on liver injury Germany. Since 1988 he held
and in particular the role of faculty positions at Baylor
inflammation in the progression College of Medicine in Hoston,
Clarence David Williams and resolution of injury. The Hartmut Jaeschke TX, The University of Arkansas
primary model that Dave focuses on is acetaminophen-induced for Medical Sciences in Little Rock and The University of
hepatotoxicity in both rodents and humans. Arizona in Tucson and was a scientist at the Upjohn Company.
Dr Jaeschke has published more than 270 original manuscripts
and invited reviews and book chapters on mechanisms of liver
pathophysiology and drug hepatotoxicity.

This journal is © The Royal Society of Chemistry 2012 Toxicol. Res., 2012, 1, 161–170 | 161
View Article Online

within hepatocytes to detoxify and facilitate the removal of xeno- of IDILI which would indicate an adaptive immune response.
biotics, however in this process toxic metabolites can be gener- Additionally elevated transaminase activities can be observed
ated and accumulate within hepatocytes. Additionally, many with some drugs up to one month following discontinuation of
phase III (transporter) reactions lead to the accumulation of drug.7 Some IDILI also involves the generation of antidrug anti-
xenobiotics and their metabolites within the liver. These meta- bodies or autoantibodies.8,9
bolic capacities, in combination with the liver’s portal blood In most cases IDILI is regarded as dose-independent, however
supply, and unique immune system make it vulnerable to drug this is most likely an incorrect statement. IDILI is very rarely
toxicity. seen at doses of <10 mg per day and more than three fourth of
Despite the relative infrequency of idiosyncratic drug-induced IDILI cases occur when the drug is given at >50 mg per day.10
liver injury (IDILI) it poses a very serious health concern with The notion that IDILI is dose independent probably arises from
potentially fatal consequences. In a retrospective study from the fact that the overwhelming majority of patients taking the
1998 to 2007 it was reported that of all causes of acute liver drug are non-responders in regard to toxicity.8 In this review we
failure, APAP was the predominant cause (46%) and DILI from will compare and contrast what is known about mechanisms of
other drugs accounted for 11%.1 Additionally, the spontaneous APAP-induced liver injury and IDILI.
patient survival is quite different between etiologies; APAP
Published on 28 August 2012. Downloaded on 27/10/2014 12:55:05.

showed a 65% spontaneous survival rate while the spontaneous


survival rate from other drugs was only 29%.1 In many cases, Why is the liver a target of drug toxicity?
IDILI cannot be detected in preclinical testing or clinical trials
because the occurrence is so infrequent that the study number is Drug disposition alone cannot explain IDILI because there is no
insufficient to detect it (Table 1), however guidelines have been association with the accumulation of excess drug or metabolite
implemented to minimize these risks. If a drug is suspected of that would result in concentrations that are toxic to the liver.11
IDILI the decision making process if the drug should be discon- This is in contrast to what is seen in APAP overdose. It is inter-
tinued is generally determined by the frequency of patients that esting however that deletion of certain drug detoxification
fall into “Hy’s Law” (Table 2). If a patient meets the three cri- enzymes cause an increased risk for IDILI. Patient with gluta-
teria there is a 10–50% chance of patient mortality.2 thione S-transferase M1 and T1 null mutations were 2.7-fold
Drugs with the highest IDILI concern are antineoplastic more likely to develop IDILI; these odds ratios increased to
agents, NSAIDs, antivirals, antidepressants, and antimicrobials,3 3.5-fold for antibacterial and 5.6-fold for NSAIDs.12 Also of
and in the US, antimicrobials account for 46% of IDILI.4 In a interest in this study was the predominance of women with this
majority of cases (73%) a single prescription was implicated as double null genotype as there is generally a female predomi-
the cause of IDILI.5 Clinically, the only effective treatment for nance of IDILI cases.12 IDILI is classified by its bizarre and, as
DILI is to stop the administration of the drug and supportive care to date, unexplainable toxicity unlike APAP which is explainable
(with the exception of N-acetyl-cysteine for APAP overdose and based on its chemical structure and generation of excess reactive
potentially carnitine for valproic acid).6 metabolite.
There are several reasons to assume IDILI is immune Liver toxicity is the leading cause of removing drugs from the
mediated. The most striking is the time to onset; depending on market and issuance of boxed warnings.11 There is no one single
the drug it could take months to over one year prior to initiation genetic, environmental or other factor that can be the sole cause
of IDILI. This is illustrated in the case of flucloxacillin. A
genome wide association study showed patients with the
Table 1 Frequencies of drug-induced altered liver functiona HLA-B*5701 genotype were at a much higher risk of developing
IDILI to flucloxacillin with an incredibly high odds ratio (OR) of
Severity Clinical manifestation Frequency rate 80.6.13 This genotype was in no means the sole determinant of
DILI, however. Despite this very strong association, only 1 out
Mild <3-fold ULN ALT 1/10 to 1/1000
Moderate Impaired hepatic function 1/100 to 1/10 000 of every 500 to 1000 patients treated with flucloxacillin who
(>3-fold ULN ALT) have the HLA-B*5701 genotype develop IDILI. The
Severe Acute liver failure or death 1/10 000 to 1/1 000 000 HLA-B*5701 genotype is also associated with abacavir hyper-
a
The frequency of IDILI is variable due to failures in reporting and sensitivity.10 This is a clear illustration of the multifactorial
potential conflicting causes of liver injury (i.e. autoimmune hepatitis, nature of IDILI and certain genotypes and environmental queues
viral hepatitis and others), however the individual susceptibility for are risk factors rather than causative. This study also demon-
altered liver function due to a drug is shown. Content adapted with
permission from Robert J. Fontana, MD, University of Michigan.
strates a direct link to risk of IDILI with the adaptive immune
system as HLA-B is an MHC class I (MHC-I) molecule that
presents self-antigen to CD8+ T cells. For additional detail of
MHC-related risk factors for IDILI we recommend a current
Table 2 “Hy’s Law”: Determining the potential for serious drug- critical review by Daly and Day.14
induced liver injury The liver is considered a site of immune tolerance, and
Hepatocellular Injury 3-fold ULN for ALT or AST perhaps in IDILI this tolerance is lost. Tolerance in the liver has
Hepatic Impairment 2-fold ULN for total bilirubin been demonstrated in several ways.15 Systemic donor-specific T
Exclusion of other No evidence of viral hepatitis, confounding cell tolerance can be achieved after orthotopic liver transplan-
causes drugs, acute liver disease or cholestasis (should tation. Introduction of antigen via portal vein can promote toler-
not present elevated serum ALP)
ance. Liver transplantations can be performed between patients

162 | Toxicol. Res., 2012, 1, 161–170 This journal is © The Royal Society of Chemistry 2012
View Article Online

with MHC mismatches. The liver can function as a site for enzymes. The reactive metabolite then adducts cellular proteins;
clonal deletion of T cells and has been referred to as a “sink” for of particular interest is the adduction of mitochondrial proteins.28
activated T cells.16,17 This mitochondrial binding propagates injury by initiating an
Perhaps this tolerance is due in part to altered leukocyte distri- oxidant stress that leads to mitochondrial failure, the formation
bution relative to other organs, blood and secondary lymphoid of ROS and the mitochondrial release of endonucleases.29 These
tissue. In the human liver the ratio of CD4+/CD8+ T cells is endonucleases translocate to the nucleus and damage DNA.30
1 : 3.5 but 2 : 1 in peripheral blood.18 Additionally the liver has a All of these events ultimately lead to cellular oncotic necrosis.31
much higher number of cells that blur the line between innate
and adaptive immune response. This includes the ‘unconven-
tional’ γδ T cells with frequency in the liver five-times that NK and NK T cells
found in peripheral blood,18 natural killer (NK) T cells
Despite the liver’s role in immune tolerance, it is also critical
(CD3+CD56+) which account from 5% to 25% of liver lympho-
during inflammation and capable of very quick and robust
cytes but are rarely seen in blood19 and NK cells which account
inflammatory response. Of particular interest in this regard are
for approximately one third of human liver lymphocytes.20
the liver NK and NKT cells that have the capacity to become
Additionally, between species the frequencies of these cell types
Published on 28 August 2012. Downloaded on 27/10/2014 12:55:05.

activated very quickly and produce large amounts of cytokines,


can differ dramatically with invariant NK T cells being relatively
in particular IFN-γ. Examples of this massive inflammatory
rare in the human liver.21
response can be seen in hepatitis A and E infections where these
In the liver the accumulation of leukocytes occurs in a manner
viral vectors are delivered to the liver via the portal vein.32 Inter-
substantially different from other organs mediated at least in part
estingly, depletion of Kupffer cells (KCs) results in decreased
by the blood supply and architecture of the liver. The sinusoidal
NK cell numbers in the liver.21
vasculature is small in diameter, low pressure and fenestrated,
NK and NK T cells were suspected to cause increased injury
and leukocyte accumulation is not dependent on selectins or
during APAP overdose via IFN-γ production and enhanced neu-
integrins.22,23 It was reported that leukocyte accumulation into
trophil recruitment.33 It was later demonstrated that the DMSO
the liver in the endotoxin model was dependent on CD44-hyal-
vehicle in these studies using C57BL/6 mice artificially recruited
uronan interaction,24 however this does not appear to be the
and activated these cells types.34 It is known that even between
mechanism of neutrophil accumulation during APAP overdose
different strains of mice the surface marker expression of NK
(Williams and Jaeschke, unpublished data). Most leukocytes that
cells is variable.21 It is therefore reasonable to conclude that
leave the vasculature and enter into the parenchyma do so via the
function or activation could be different between strains as well,
sinusoids rather than postcapillary venules.25,26
and it was shown that elimination of NK and NKT cells reduced
APAP-induced injury in IL-13 deficient mice.35 While these cell
types in C57BL/6 mice do not cause additional injury in APAP
Liver immunity and its role during APAP overdose overdose they are still suspected as potential participants in other
forms of DILI primarily because these cells function as innate
The most common model to study drug-induced hepatotoxicity
immune cells but perform these functions in ways similar to the
is APAP. APAP overdose results in centrilobular necrosis that is
adaptive immune system. It was shown in a case report of two
not exacerbated by the recruitment of inflammatory cells.27
patients with IDILI-induced hepatic failure that NK and NKT
Despite extensive evidence of the benign role of inflammation in
cell quantity and expression of the costimulatory factor CD28
regard to injury several groups still report that inflammatory cells
was different than control patients.36 Additionally, of potential
can increase injury, and this will be discussed in more detail in
importance is the Fas/Fas ligand interaction that can be mediated
later sections and has previously been reviewed in detail.27 If
by these cells on hepatocytes.27 Generally, hepatocytes are resist-
biopsy is performed in IDILI patients the mode of cell death
ant to perforin/granzyme-induced cell death which means T cell
appears to necrosis with inflammatory infiltrates,11 however the
mediated killing could involve Fas/FasL interaction or TNF-α.37
mechanisms of T cell mediated death is generally apoptosis.
From these data it is very difficult to determine the mode of cell
death in IDILI because it is possible that at the time of biopsy Neutrophils
the injury has already degraded into secondary necrosis. Clearly
there are striking differences between IDILI and APAP toxicity. Neutrophils can occasionally be seen in normal, healthy liver,
APAP toxicity is dose dependent with a very rapid onset of however upon inflammatory insult these cells are rapidly
injury (within hours). Therefore the mechanisms of toxicity and recruited into the liver and the number of total hepatic neutro-
cell death are most likely different between idiosyncratic hepato- phils can be increased several orders of magnitude.38 If proper
toxic drugs and APAP, but understanding of intracellular events stimuli are present, these cells will extravasate from the vascula-
of APAP toxicity (i.e. formation of reactive metabolite, covalent ture into the hepatic parenchyma and cause injury.39 Without
binding of cellular proteins, mitochondrial dysfunction, release these signals the neutrophils will remain in the sinusoids,
of mitochondrial components, nuclear DNA damage) could lead undergo apoptosis and be cleared by KCs. Normally the half-life
to more clues regarding susceptibility or outcome of IDILI. of a neutrophil is 6–12 h but during active inflammation this can
The initiation of APAP toxicity is dependent on the metabolic be increased to 48 h.40
conversion of APAP to N-acetyl-p-benzoquinone imine It has been extensively demonstrated that neutrophils do not
(NAPQI) in hepatocytes predominantly by CYP2E1 and to a participate in APAP induced injury,27 however, some controversy
lesser extent by CYP1A2, CYP2D6 and potentially other P450 still exists. A neutropenia-inducing antibody results in protection

This journal is © The Royal Society of Chemistry 2012 Toxicol. Res., 2012, 1, 161–170 | 163
View Article Online

against APAP toxicity but only if given 24 h prior to APAP,41–43 via liposome/clodronate. Additionally this tolerance could be
but not if given after APAP overdose despite functional inacti- induced upon adoptive transfer of KCs from tolerized mice.55
vation of neutrophils.44–46 Also, neutrophils show no enhanced KCs are also important in attenuating this inflammatory response
activation status during the APAP-induced injury phase,47 by producing high levels of IL-10.37
priming and activating neutrophils during APAP overdose by During APAP overdose distinct macrophage populations can
IL-1β or endotoxin does not increase injury,47,48 CD18-47 and be observed during the injury phase and injury resolution. These
ICAM-1-deficient mice44 are not protected, an anti-CD 18 anti- different populations have unique phenotypes which can be
body did not affect injury,49 and mice that have inhibited determined by surface markers. Kupffer cells are often referred
NADPH oxidase or lack NADPH oxidase ( phagocytic respirat- to as M1 (classically activated) macrophages are typically
ory burst) have no difference in oxidant stress or APAP-induced thought of as pro-inflammatory and drive cells toward a Th1
injury.44,50 Often times neutrophils are thought to be the cause of phenotype through high IL-12 production; traditionally these
enhanced injury because in genetic or pharmacologic interven- cells are activated by IFN-γ or LPS. M2 (alternatively activated)
tions mice with reduced injury will have lower hepatic neutrophil are typically anti-inflammatory, activated by IL-4 or IL-13,
counts or reduced myeloperoxidase staining,43,51 however, this is highly phagocytic and promote tissue repair.56 During injury
most like due to the fact that mice with less injury, release less resolution monocytes are recruited into the liver and become
Published on 28 August 2012. Downloaded on 27/10/2014 12:55:05.

DAMPs, and therefore recruit fewer neutrophils. In simple M2-biased. Depletion of KCs prior to APAP overdose actually
terms, blocking upstream events (injury) will reduce downstream results in increased injury which is mediated by the loss of IL-10
events (inflammation). A caveat of most immunological inter- production.57,58 Also of importance during APAP overdose is
ventions is these studies often times do not assess drug meta- IL-6 which is also hepatoprotective and generally considered
bolism, protein binding or other intracellular events. Without pro-regenerative.59 IDILI most likely involves the loss of
these control experiments results are difficult if not impossible to immune tolerance. Alteration of the cytokine production profile
interpret and a variety of off-target effects could occur. This could cause of a shift away from immune tolerance which would
should be a particular concern when the data contradict many most likely begin with the resident tissue macrophages of the
published findings. liver.
Most theories and models of IDILI do not have neutrophils
being a major contributor to injury with a few exceptions. If neu-
trophils are participating in IDILI then it would most likely Dendritic cells
occur as a secondary effect to damage caused by an adaptive
As stated previously, KCs produce fairly high levels of IL-10
immune response. One exception is the case of halothane.
which is capable of altering the function of the liver’s other
Halothane was used clinically as an inhaled anesthetic, but
antigen presenting cell type, dendritic cells (DCs). The liver con-
patients upon reexposure had an increased risk of potentially
tains both plasmacytoid and myeloid DCs which are most com-
fatal hepatotoxicity.9 Halothane is converted to the reactive
monly found within portal tracts.32 High levels of IL-10 tend to
metabolite trifluoroacetic acid which adducts hepatic protein and
drive DCs away from effector pathways, and DCs of the liver are
initiates liver injury.9 It was shown that depletion of neutrophils
generally immature and express low levels of co-stimulatory
in this model could attenuate injury.52 It was later demonstrated
molecules, which tend to produce regulatory T cells.37
that NK T cells were critical for the recruitment of neutrophils in
It was reported that dendritic cell depletion results in enhanced
this model.53
APAP-induced injury.60 DC depletion was induced in CD11c.
DTR mice via diphtheria toxin prior to APAP; this resulted in
enhanced inflammatory cytokine and chemokine profiles and
Kupffer cells (KCs) increased liver injury. Conversely, expansion of DC populations
via Fms-related tyrosine kinase 3 ligand (Flt3L) decreased
Administration of LPS or other inflammatory insults results in
APAP-induced injury. Additionally, neutrophil or NK cell
massive cytokine formation and accumulation of leukocytes in
depletion in addition to DC depletion did not modulate the
the liver. KCs are instrumental in the removal of gut-derived
injury further confirming these innate immune cells do not con-
endotoxin and the phagocytosis and killing of bacteria. Within
tribute to APAP-induced injury.60 The immature phenotype of
minutes of the inflammatory stimuli KCs produce numerous
DCs in the liver almost certainly participates in the maintenance
cytokines, chemokines and subsequently stimulate hepatocytes
of tolerance and loss of tolerance could induce IDILI. It is not
( predominantly through IL-6) to produce acute phase proteins
clear if loss of tolerance would be mediated by DCs directly or
which include pentraxins, protease inhibitors, coagulation
modulation of the microenvironment (i.e. exposure to cytokines
factors, complement components and others.37 Interestingly,
and stimulatory factors).
inactivation of KCs using gadolinium chloride (GdCl3) prevents
portal vein tolerance.54 This was determined by the adminis-
tration of alloantigen (spleen cells from different mouse strains) Nalp3 inflammasome
with or without GdCl3 treatment and measurement of delayed
hypersensitivity reaction by foot swelling.54 In another model of The NACHT, LRR and PYD domains containing protein 3
T cell mediated delayed hypersensitivity it was shown that KCs (Nalp3) inflammasome is assembled after the initiation of sterile
were involved in the induction of tolerance for protein adducts inflammation and is responsible for the maturation of IL-1β and
or haptens.55 This was determined by sensitization of mice to IL-18 through activation of caspase-1.61 It was reported that
2,4-dinitrochlorobenzene (DCNB) with or without KC depletion mice deficient for each component of the Nalp3 are protected

164 | Toxicol. Res., 2012, 1, 161–170 This journal is © The Royal Society of Chemistry 2012
View Article Online

from APAP-induced injury62 and ATP released during early factors described previously. Additionally, this theory proposes
injury signals through the P2X7 purinergic receptor to activate that only professional APCs (i.e. DCs) can prime naïve T cells
the Nalp3 inflammasome.63 These studies were repeated and no where subsequent activation can be performed by other APCs
protection was observed in mice deficient for each component of (macrophages and B cells). Effector T cells that are presented
the Nalp3 inflammasome.64 Additionally, inhibiting caspase-1 antigen without the required co-stimulatory factors will become
prevented IL-1β maturation without altering liver injury, and senescent. Initially, it is thought that cells dying by apoptosis are
pharmacologic addition of recombinant IL-1β during APAP non-inflammatory and therefore tolerogenic and necrotic cells
overdose did not modulate liver injury.48 In addition, the puriner- are pro-inflammatory and therefore immunogenic, however it has
gic receptor antagonist can directly act on hepatocytes and been shown that this is not always the case. Antigens from apop-
inhibit protein adduct formation, oxidant stress and c-Jun N- totic cells can be cross-presented to CD8+ T cells and prime an
terminal kinase (JNK) activation (Williams and Jaeschke, unpub- immune response, and cells that die at the peak of an immune
lished data). These data suggest that this compound alters the response can be immunogenic but cells dying as inflammation
mechanisms of intracellular cell death, which occur upstream wanes can induce tolerance.67 It was also shown that DCs that
and independently of the Nalp3 inflammasome. From these data phagocytosed necrotic cells debris presented antigen to both
it is highly unlikely that the Nalp3 inflammasome contributes to CD4+ and CD8+ T cells, but DCs that phagocytosed apoptotic
Published on 28 August 2012. Downloaded on 27/10/2014 12:55:05.

APAP-induced injury but its role in IDILI is yet to be deter- cells only presented antigen to CD8+ T cells.68
mined. This is just one of many examples where data from The first step of the danger hypothesis involves some initial
immunological or pharmacological interventions are misinter- cell injury or at the very least some cell stress to cause the initial
preted because effects of these agents on intracellular signaling danger signal. Potentially these danger signals could be cyto-
events are not considered. kines released by innate immune cells or DAMPs released by
the injured cells. High mobility group box-1 (HMGB-1) protein
currently is one of the most commonly studied and measured
Major theories of IDILI DAMPs. APAP causes the release of HMGB-1.64,69 In agree-
ment with one of Matzinger’s original hypotheses, oxidation of
There are a number of hypotheses regarding the cause of IDILI.
HMGB-1 which occurs in apoptotic but not necrotic cells was
Most likely no one hypothesis alone can explain the mechanism
critical to neutralizing its stimulatory activity and blocking oxi-
of all cases of IDILI and several theories might occur sequen-
dation sites prevented tolerance.70 Potentially the oxidation state
tially or concurrently to explain the mechanism. As described
of HMGB-1 could be of prognostic value during drug-induced
previously IDILI most likely involves an adaptive immune
liver injury.71
response which is fundamentally different from what is observed
in APAP-induced injury. The accepted dogma of adaptive
immune activation requires presentation of antigen loaded on an
MHC molecule to the T cell receptor (TCR) of a T cell. “Hapten hypothesis”
Additionally, a second costimulatory signal is required which
A hapten is a small molecule (typically incapable of initiating an
could be a B7 molecule (CD80 or CD86) on antigen presenting
immune response) that covalently binds to a larger molecule
cell (APC) interacting with CD28 on the T cell. When both of
(cellular protein) and becomes immunogenic. This theory of
these signals occur an immune response is mounted.65 Without
IDILI involves the concept of a drug or more-likely the drug’s
this second costimulatory signal the T cell becomes anergic for
reactive metabolite binding to a cellular protein and triggering
the maintenance of immune tolerance. This central dogma of
the initiation of an immune response. This is a rational theory
immunology gives rise to the “danger hypothesis”.65 APCs
because of the high expression of CYP450’s (drug metabolizing
present antigen to CD4+ T cells via MHC class II (MHC-II); all
enzymes) and drug transporters found in the liver, therefore most
other cells present antigen to CD8+ T cells via MHC class I
of these reactive metabolites are present within the liver as is the
(MHC-I). Classically this means exogenous antigens are pres-
case with the conversion of APAP to the reactive metabolite
ented via the MHC-II and endogenous antigens are presented via
NAPQI. Adduction of cellular proteins by the reactive meta-
MHC-I. There are exceptions to this rule however with the most
bolite then can initiate an adaptive immune response following
predominant being that of “cross-presentation”.66 In this process
antigen processing and MHC presentation; this new immuno-
antigens from the extracellular environment can be processed,
logical entity is referred to as a hapten. The haptenized protein
loaded into MHC-I and presented to CD8+ T cells.
can elicit a T cell response to the drug, the protein itself or a
mixed response can occur depending on the antigen presen-
“Danger hypothesis in IDILI” tation.9,72 If the drug metabolite adducts a cellular protein and
results in cell death then these aberrant antigens during an
Due to the environment and diet we are constantly exposed to inflammatory response (increase costimulatory factors) could be
countless exogenous substances through multiple routes of deli- presented by APCs and trigger a CD4+ response, so this theory
very yet mount immune response to very few of these things. generally involves the danger hypothesis to propagate the injury.
For this reason the “danger hypothesis” was proposed.8,9 It is Evidence for the pathogenic role of haptens can be seen with
believed that the immune system responds to “danger” (i.e. cell drugs like tienilic acid, dihydralazine, halothane, phenytoin, car-
injury) rather than specifically to “non-self”.64 In this theory bamazepine and others.9 In these cases, common targets of
necrotic cell death is referred to as “bad death” and only bad haptenation are the enzymes responsible for metabolism of the
death triggers immune response mediated by the co-stimulatory drug as well as other liver-specific proteins.73 It has been

This journal is © The Royal Society of Chemistry 2012 Toxicol. Res., 2012, 1, 161–170 | 165
View Article Online

demonstrated that these antibodies or autoantibodies can react because T cell activation in this hypothesis does not require co-
with P450 enzymes (CYP2D6, CYP2C9, CYP1A2, and others) stimulatory factors. This theory was first used to describe the
and UDP glucuronyltransferases.74,75 It is unclear if these anti- IDILI potential of sulfamethoxazole. In this study T cell clones
bodies exert a direct toxicity. However, it does clearly demon- could be activated by drug even after the fixation of APCs indi-
strate that immune cell activation has occurred, and in the case cating antigen processing was not required.81 While this theory
of autoantibodies immune tolerance has been compromised.8,9 appears to be plausible for some drugs it does not apply for all
An argument against the hapten hypothesis is some IDILI IDILI drugs. It has been shown that the P-I hypothesis can be
drugs do not form reactive metabolites to covalently bind pro- applied to other drugs like carbamazepine, lamotrigine, lidocaine
teins.8,76 Additionally, it should be noted, that covalent binding and others;80 however, these drugs also can produce reactive
should be regarded as a bioactivation event and not one of overt metabolites, so perhaps P-I hypothesis occurs in concert with
toxicity, and conversely not all drugs must be metabolized to another model of IDILI.
trigger an adverse drug reaction.77 Additionally, the efficacy of
some drugs is dependent on covalent binding like aspirin and “Mitochondria hypothesis” (SOD2+/− model)
proton-pump inhibitors.78
Not all drugs have to form a reactive metabolite to cause This hypothesis involves a subclinical mitochondrial dysfunction
Published on 28 August 2012. Downloaded on 27/10/2014 12:55:05.

IDILI and generation of reactive metabolite does not mean an that gradually accumulates until a critical threshold is met at
immune response will be generated even in the presence of sub- which time liver injury rapidly occurs.82 Mitochondria lack
stantial injury and inflammation which is the case with APAP. It DNA repair mechanisms, and cumulative damage over time
was hypothesized that APAP toxicity creates a tolerance to might induce toxicity. Typically speaking DNA damage is
APAP protein adducts.79 In a mouse model of APAP overdose associated with hard electrophiles which have a high positive
lymphocyte apoptosis could be seen in the spleen, thymus and charge density; these electrophiles are also more likely to react
liver-draining lymph nodes, and APAP overdose also suppressed with amino acid residues like lysine. Soft electrophiles (i.e.
the hypersensitivity response of DNCB.79 This theory might be NAPQI) generally do not attack nucleic acids and interact with
a mechanism of how progression of IDILI is avoided in most amino acids like cysteine and are predominantly detoxified by
people with minor or subclinical drug toxicity. GSH.77,83 However during mitochondrial dysfunction ROS gen-
erated within the mitochondria is the most likely cause of mito-
“Failure to adapt hypothesis” chondrial failure. Mitochondrial DNA damage is a late event
(after drug metabolism) which can be prevented by scavenging
Another concept with IDILI is the notion of a lack of adaptation ROS in the mitochondria.84
to a toxic insult in which injury caused by the drug or metabolite The inner membrane of mitochondria contains high levels of
triggers a response or repair mechanism. This injury could be cardiolipin which has many unsaturated bonds potentially
initiated in the mitochondria (as discussed later), in the endoplas- making in vulnerable to lipid peroxidation.82 This would be an
mic reticulum as unfolded protein response or other cellular organelle-specific toxicity potentially leading to impaired mito-
stressors. chondrial respiration and damage as previously described. It is
Occasionally, patients on a drug with known IDILI potential unknown if lipid peroxidation (on a cellular level) is a relevant
will present with serum ALT >3 times upper limit of normal mechanism of cell death in IDILI, but it is not a relevant general
(ULN) but return to baseline levels even when continued on the mechanism during APAP overdose.85,86
drug.9 This concept is known as adaption. Why will some Sod2-heterozyous mice are more susceptible to mitochondrial
patients return to normal and other progress to severe liver injury, and SOD is critical for the detoxification of superoxide to
injury? The response mechanisms could involve phase I, II or III prevent peroxynitrite formation.87–89 It was shown that in
metabolic processes; the induction of anti-oxidant responses, Sod2+/− mice troglitazone caused delayed hepatic injury.90 This
enhanced autophagy, enhanced cell proliferation or other protec- injury is consistent with a delay in toxicity seen in patients,
tive measures. On the other hand, this could be indicative of the however these findings could not be repeated in a subsequent
development of immune tolerance. It is possible the loss of toler- study using a very similar model.87 Additionally, long-term
ance could be mediated by KCs. This could occur by generation administration of flutamide resulted in oxidant stress, mitochon-
of reactive metabolite within the KCs9 or cross-presentation of drial dysfunction and hepatic injury in Sod2+/− mice.91 Clini-
antigen. The mode of this initial cell death could also be impor- cally it has been shown that patients with SOD2 Val16Ala
tant. It has been shown that caspases or other cellular proteases mutation are prone to mixed cholestatic injury (OR = 2.3) and
upon activation may create new antigenic epitopes within the patients homozygous for this mutation with this type of injury
cell.67 are more susceptible to drugs with known mitochondrial hazards
(OR = 3.6).92
“Pharmacologic interaction (P-I) hypothesis” If a mitochondrial threshold effect does occur and causes toxi-
city this will not explain why patients with idiosyncratic toxici-
This theory involves the direct binding of drug to TCR and T ties are susceptible to toxicity upon re-challenge. Most likely the
cell activation upon MHC interaction with an APC, or the drug mitochondria would be an initiating event in IDILI that would
non-covalently interacts with MHC and then presents it to a then proceed to an immune response to propagate the injury.
responsive T cell.80 This concept can explain why drugs that do Thus, the Sod2+/− model could be useful to evaluate the poten-
not form reactive metabolites can initiate IDILI, however this tial of drugs to cause a mitochondrial stress but does not mimic
hypothesis is fundamentally different from the danger hypothesis the entire pathophysiology of IDILI.

166 | Toxicol. Res., 2012, 1, 161–170 This journal is © The Royal Society of Chemistry 2012
View Article Online

“Inflammagen model” immune functions.96,97 These functions include digestion and


removal of intracellular pathogens, promotion of Th1-Th2 polar-
This model uses drugs with IDILI potential and co-treats ization during intracellular infection, modulation of TLR
animals with LPS. The assumption behind this approach is that response and inflammation via pathogen associated molecular
the hepatotoxicity of idiosyncratic drugs is not detectable pattern (PAMP) delivery, presentation of endogenous peptides to
because of interfering events (death of animals). In addition an MHC-II molecules, regulation of T cell homeostasis, and
inflammagen like LPS causes a leftward shift of the dose- immune tolerance. Autophagy can result in the “non-classical”
response curve and thus makes the toxicity detectable. The goal MHC class II presentation of autophagosomal peptides98 and
of this model is to induce hepatotoxicity in a large percentage of self-antigen presentation on MHC-II by DCs might be respon-
treated animals which will allow for better statistical analysis and sible for CD4+ mediated peripheral tolerance and immature den-
provide an economical way to test for IDILI.93 The inflammagen dritic cells (like those observed in the liver) have a high rate of
model has been used for drugs like amiodarone, chlorpromazine, autophagy and are considered pro-tolerogenic.96 Following
diclofenac, trovafloxacin, sulindac and others.93 In these models pattern recognition receptor signaling, autophagy is enhanced
the dosing of drug and LPS was highly variable; occasionally thereby promoting non-classical MHC-II presentation.97 Interest-
LPS was given prior to drug, sometimes post-drug, sometimes ingly autophagy has been shown to be critical for type I inter-
Published on 28 August 2012. Downloaded on 27/10/2014 12:55:05.

drug was given multiple times.93 In these studies the liver injury feron production, however, loss of autophagy can also up-
ranged from moderate to severe liver necrosis with a relatively regulate several pro-inflammatory cytokines and generation of
rapid onset. ROS in macrophages.99 Autophagy is also critical in T cell
However, there are concerns regarding the relevance of this homeostasis and activation. It has been shown that autophagy is
model. First, the generally used dose of endotoxin (44 × 106 EU induced during T-cell activation and it has been reported that it
kg−1 or ∼4 mg kg−1) results in plasma levels approximately five is required for clonal expansion.100,101 In this regard, autophagy
orders of magnitude higher than the maximum serum concen- promotes T cell expansion. Conversely, if autophagy is blocked
tration measured in septic patients with confirmed Gram-negative the immune synapse (MHC-TCR interaction) can become hyper-
infection.94 And, sepsis has not been identified to precede or stablized. Ultimately this leads to enhanced T cell activation.102
accompany the onset of IDILI. Second, the mechanism of liver In addition to the potential implications of autophagy within
injury in the inflammagen model appears to always involve the immune system autophagy within the liver could be, equally,
TNF-α, neutrophils and the coagulation cascade93 independent if not more important. Autophagy of damaged mitochondria or
of the drug. Thus it is likely that the model mimics LPS-induced other cellular organelles could be critical to limit oxidant stress
liver injury where a subtoxic dose of LPS causes neutrophil- or ER stress as was previously described in the “failure to adapt”
mediated liver injury when additional stress (exposure to drug) is section. The role of autophagy and DILI has only very recently
applied. been evaluated.
Support of these concerns comes from the application of the Just this year several papers have shown the role of autophagy
inflammagen model to APAP toxicity. Pretreatment with a high during APAP overdose however caution must be used when
dose of LPS before a subtoxic dose of APAP caused severe but using these models as we will discuss in more detail. The
delayed liver injury.95 The interpretation was that LPS caused a primary hypothesis regarding the role of autophagy after APAP
leftward shift of toxicity implying the same mechanism of is that of hepatoprotection which is not surprising since auto-
toxicity just with a lower dose of APAP in the presence of LPS phagy is normally considered a cell survival pathway. Following
pretreatment.95 However the time course of toxicity with LPS overdose, APAP adducts cellular proteins potentially making
suggests a different mechanism. Pretreatment with LPS will some of them non-functional; of particular interest is the adduc-
cause the recruitment of primed neutrophils into the liver vascu- tion of mitochondrial proteins ultimately resulting in mitochon-
lature.25 The subtoxic dose of APAP still causes GSH depletion drial dysfunction. As some mitochondria begin to fail an oxidant
and protein adduct formation (McGill and Jaeschke, unpublished stress is generated, mitochondrial membrane potential is lost,
data), which in the centrilobular area is likely a sufficient stress and mitochondria swell then rupture releasing endonucleases;
signal for the neutrophils to extravaste and attack39 resulting in a the combination of these events ultimately lead to cell death by
neutrophil-mediated liver injury. Thus, a high dose of APAP and oncotic necrosis.29,86,89 During this process there must be some
low dose of APAP with LPS pretreatment have fundamentally critical threshold for mitochondrial loss that determines cell fate;
different mechanisms of liver injury. the most centrilobular hepatocytes which have the highest P450
enzyme activities and lowest GSH levels presumably have the
Role of autophagy in DILI highest reactive metabolite burden and therefore are the most
susceptible. Depending on the dose, the centrilobular injury can
As mentioned previously autophagy could play a major role in expand and potentially invade midzonal or even periportal areas,
the adaptation process to prevent the progression of IDILI. It is however the injury always begins with the most centrilobular
becoming more apparent that autophagy is critical for various hepatocytes. The hepatocytes at the threshold between necrotic
functions in both the innate and adaptive immune response. and healthy tissue are the cells where autophagy plays a role.
Current investigations involve the modulation of autophagy in These cells remove damaged mitochondria and adducted pro-
vaccine efficacy and chronic inflammatory disease. Dysfunction teins thereby preventing lethal oxidant stress and the release of
or altered regulation of autophagy can lead to loss of immune endonucleases allowing cell survival.
tolerance, which is most likely a critical component to the induc- The first paper investigating the role of autophagy during
tion and progression of IDILI. Autophagy has a multitude of APAP overdose demonstrated that APAP does in fact activate

This journal is © The Royal Society of Chemistry 2012 Toxicol. Res., 2012, 1, 161–170 | 167
View Article Online

autophagy and this activation is protective.103 This was deter-


mined by using both inhibitors (3-methyladenine or chloroquine)
and activators (rapamycin) of autophagy in vivo and in primary
cultured mouse hepatocytes. The predominant mechanism of
protection appears to be mitophagy (autophagy of mitochondria)
which is then capable of reducing APAP-induced injury.
Most recently it was shown that mice deficient for Atg5 in the
liver were actually resistant to APAP toxicity.104 This finding is
contradictory to the dogma that autophagy is protective, but it
was determined that the protection from APAP toxicity was due
to the aberrant phenotype seen in these mice. Atg5 was deleted
from hepatocytes under the albumin-Cre promoter. The Atg5
liver knockout mice develop mild liver injury resulting in
increased apoptosis and compensatory proliferation. Additionally
the liver becomes preconditioned and Nrf2 activation increases
Published on 28 August 2012. Downloaded on 27/10/2014 12:55:05.

the expression of target genes which increases basal GSH levels,


and therefore these mice are protected by compensatory effects Fig. 1 There are multiple hypotheses regarding the initiation and
rather than by loss of Atg5. Clinically the induction of Nrf-2 propagation of IDILI. This illustration depicts how several of these
could be indicative of a “danger signal” because it is activated events could occur and potential key events that could involve auto-
by oxidant stress or the induction of autophagy. phagy. Autophagy could clear adducted or nonfunctional proteins or
Recently, another study has demonstrated that loss of auto- could remove damaged organelles. If these processes are insufficient
phagy can promote APAP toxicity. In this study Atg7 conditional drug or metabolite could cause cell stress or death within the hepatocyte.
knockout animals were generated under Mx1-Cre by treating This could then trigger an immune response and activation. This acti-
mice with poly I:C.105 The poly I:C treatment is capable of mo- vation could then trigger various downstream effector functions in
dulating the metabolic activation of APAP, and this study shows additional immune cells. Propagation of injury can then occur through
these adaptive or innate immune responses. Autophagy could potentially
a mechanism of cell death in the Atg7-deficient mice quite
attenuate these responses through the modulation of TLR/ligand inter-
different from control mice, however. The Atg7-deficient mice action or destabilization of the immune synapse. These roles of auto-
have caspase processing and apoptosis. It has previously been phagy during IDILI are speculative however convincing evidence of the
reported that the poly I:C treatment in Atg7-Mx1-Cre mice participation of autophagy during APAP overdose has been shown. The
results in hepatomegaly and liver injury.106 The abnormal pheno- figure is a modification of the concepts previously described.72
type of these mice is a major concern, therefore, care must be
used when interpreting data from these types of models.107
The role of autophagy and DILI is now beginning to be esta- CD8+ T cell recognition; however in absence of co-stimulatory
blished. Potentially, autophagy might be critical in the clearance factors this should result in immune tolerance. If the reactive
of adducted proteins, removal of damaged organelles or modu- metabolite is released and then phagocytosed by an APC, these
lation of immune tolerance (Fig. 1). antigens will be presented to CD4+ T cells via MHC-II. These
helper T cells can then lead to B cell or CTL activation.72,109
Clearly, there will be no easy answer to explain the cause of
IDILI. As evidenced by genetic polymorphisms there are clearly
Conclusions risk factors involved with the development of IDILI but genetics
During APAP overdose there are several critical stages which alone do not explain the cause. No better example illustrates this
can be defined as initiation and propagation. The initiating event point than the HLA-B*5701 genotype with flucloxacillin, which
is reactive metabolite formation resulting in protein adduction. greatly increases the risk but is by no means the determinant of
The injury is then propagated by mitochondrial dysfunction and DILI.14 There are multiple factors involved like dose of drug,
ROS production. In APAP overdose, both of these events occur gender, age, alcohol or concomitant drug use, underlying disease
independently of immune cell infiltration27 and depletion of resi- state, environmental exposures, and potentially other as yet un-
dent immune cells (KCs and DCs) actually causes increased identified risk factors.110,111 Additional risk could be attributed
injury.57,60,108 A different situation most likely occurs in IDILI, to enhanced production or impaired detoxification of oxidant
but this toxicity probably involves initiation and propagation stress, or altered mitochondrial respiration due to inherited or
stages as well (Fig. 1). The initiation stage likely involves the acquired mitochondrial dysfunction.112 Because of these com-
generation of reactive metabolites or the parent drug impairs plexities most likely not all IDILI will share the same mechan-
some cell function (mitochondrial respiration or autophagy); this ism of toxicity. Therefore, uncovering new risk factors for
could lead to formation of a hapten or trigger mild liver injury. patients and trying to develop animal models of IDILI is critical
The propagation stage most likely involves an immune com- to reduce this human health concern.
ponent which then greatly enhances the liver injury. In theory the
generation of reactive metabolites or location of parent drug will
determine the type of immune response by the antigenic presen- References
tation via MHC-I or MHC-II. If the reactive metabolite is gener- 1 W. M. Lee, R. H. Squires Jr., S. L. Nyberg, E. Doo and J. H. Hoofnagle,
ated within the cell and then presented via MHC-I this will lead to Hepatology, 2008, 47, 1401–1415.

168 | Toxicol. Res., 2012, 1, 161–170 This journal is © The Royal Society of Chemistry 2012
View Article Online

2 H. J. Zimmerman, Hepatotoxicity: The Adverse Effects of Drugs and 40 S. Yamashiro, H. Kamohara, J. M. Wang, D. Yang, W. H. Gong and
Other Chemicals on the Liver, Lippincott Williams & Wilkins, Phila- T. Yoshimura, J. Leukocyte Biol., 2001, 69, 698–704.
delphia, 2nd edn, 1999. 41 Y. Ishida, T. Kondo, A. Kimura, K. Tsuneyama, T. Takayasu and
3 M. Chen, V. Vijay, Q. Shi, Z. Liu, H. Fang and W. Tong, Drug Discov- N. Mukaida, Eur. J. Immunol., 2006, 36, 1028–1038.
ery Today, 2011, 16, 697–703. 42 Z. X. Liu, D. Han, B. Gunawan and N. Kaplowitz, Hepatology, 2006,
4 R. J. Fontana, L. B. Seeff, R. J. Andrade, E. Bjornsson, C. P. Day, 43, 1220–1230.
J. Serrano and J. H. Hoofnagle, Hepatology, 2010, 52, 730–742. 43 P. E. Marques, S. S. Amaral, D. A. Pires, L. L. Nogueira, F. M. Soriani,
5 N. Chalasani, R. J. Fontana, H. L. Bonkovsky, P. B. Watkins, T. Davern, B. H. Freire Lima, G. A. Oliveira Lopes, R. C. Russo, T. V. Avila,
J. Serrano, H. Yang and J. Rochon, Gastroenterology, 2008, 135, 1924– J. G. Melgaco, A. G. Oliveira, M. A. Pinto, C. X. Lima, A. M. de Paula,
1934, 1934 e1921–1924. D. C. Cara, M. F. Leite, M. M. Teixeira and G. B. Menezes, Hepatology,
6 CDER, Drug-induced Liver Injury: Premarketing Clinical Evaluation, 2012, DOI: 10.1002/hep.25801.
2009. http://www.fda.gov/downloads/Drugs?GuidanceComplianceRegu- 44 C. Cover, J. Liu, A. Farhood, E. Malle, M. P. Waalkes, M. L. Bajt and
latoryInformation/Guidances/UCM174090.pdf H. Jaeschke, Toxicol. Appl. Pharmacol., 2006, 216, 98–107.
7 S. Verma and N. Kaplowitz, Gut, 2009, 58, 1555–1564. 45 H. Jaeschke, Hepatology, 2008, 48, 699–701.
8 J. Uetrecht, Annu. Rev. Pharmacol. Toxicol., 2007, 47, 513–539. 46 H. Jaeschke, M. R. McGill and C. D. Williams, Hepatology, 2012, DOI:
9 J. Uetrecht, Semin. Liver Dis., 2009, 29, 383–392. 10.1002/hep.25877.
10 X. Zhang, F. Liu, X. Chen, X. Zhu and J. Uetrecht, Drug Metab. Phar- 47 C. D. Williams, M. L. Bajt, A. Farhood and H. Jaeschke, Liver Int.,
macokinet., 2011, 26, 47–59. 2010, 30, 1280–1292.
11 N. Kaplowitz, Nat. Rev. Drug Discovery, 2005, 4, 489–499. 48 C. D. Williams, A. Farhood and H. Jaeschke, Toxicol. Appl. Pharmacol.,
Published on 28 August 2012. Downloaded on 27/10/2014 12:55:05.

12 M. I. Lucena, R. J. Andrade, C. Martinez, E. Ulzurrun, E. Garcia- 2010, 247, 169–178.


Martin, Y. Borraz, M. C. Fernandez, M. Romero-Gomez, A. Castiella, 49 J. A. Lawson, A. Farhood, R. D. Hopper, M. L. Bajt and H. Jaeschke,
R. Planas, J. Costa, S. Anzola and J. A. Agundez, Hepatology, 2008, Toxicol. Sci., 2000, 54, 509–516.
48, 588–596. 50 L. P. James, S. S. McCullough, T. R. Knight, H. Jaeschke and
13 A. K. Daly, P. T. Donaldson, P. Bhatnagar, Y. Shen, I. Pe’er, A. Floratos, J. A. Hinson, Free Radical Res., 2003, 37, 1289–1297.
M. J. Daly, D. B. Goldstein, S. John, M. R. Nelson, J. Graham, 51 X. Wang, R. Sun, H. Wei and Z. Tian, Hepatology, 2012, DOI: 10.1002/
B. K. Park, J. F. Dillon, W. Bernal, H. J. Cordell, M. Pirmohamed, hep.25982.
G. P. Aithal and C. P. Day, Nat. Genet., 2009, 41, 816–819. 52 Q. You, L. Cheng, T. P. Reilly, D. Wegmann and C. Ju, Hepatology,
14 A. K. Daly and C. P. Day, Drug Metab. Rev., 2012, 44, 116–126. 2006, 44, 1421–1431.
15 I. N. Crispe, M. Giannandrea, I. Klein, B. John, B. Sampson and 53 L. Cheng, Q. You, H. Yin, M. P. Holt and C. Ju, Biochem. Pharmacol.,
S. Wuensch, Immunol. Rev., 2006, 213, 101–118. 2010, 80, 255–261.
16 B. John and I. N. Crispe, J. Immunol, 2004, 172, 5222–5229. 54 M. P. Callery, T. Kamei and M. W. Flye, Transplantation, 1989, 47,
17 W. Z. Mehal, A. E. Juedes and I. N. Crispe, J. Immunol., 1999, 163, 1092–1094.
3202–3210. 55 C. Ju and L. R. Pohl, Toxicology, 2005, 209, 109–112.
18 S. Norris, C. Collins, D. G. Doherty, F. Smith, G. McEntee, O. Traynor, 56 M. P. Holt, L. Cheng and C. Ju, J. Leukocyte Biol., 2008, 84, 1410–
N. Nolan, J. Hegarty and C. O’Farrelly, J. Hepatol., 1998, 28, 84–90. 1421.
19 S. Norris, D. G. Doherty, C. Collins, G. McEntee, O. Traynor, 57 C. Ju, T. P. Reilly, M. Bourdi, M. F. Radonovich, J. N. Brady,
J. E. Hegarty and C. O’Farrelly, Hum. Immunol., 1999, 60, 20–31. J. W. George and L. R. Pohl, Chem. Res. Toxicol., 2002, 15, 1504–
20 D. G. Doherty and C. O’Farrelly, Immunol. Rev., 2000, 174, 5–20. 1513.
21 B. Gao, S. Radaeva and O. Park, J. Leukocyte. Biol., 2009, 86, 513– 58 M. Bourdi, Y. Masubuchi, T. P. Reilly, H. R. Amouzadeh, J. L. Martin,
528. J. W. George, A. G. Shah and L. R. Pohl, Hepatology, 2002, 35,
22 H. Jaeschke, Am. J. Physiol., 1997, 273, G602–G611. 289–298.
23 W. Y. Lee and P. Kubes, J. Hepatol., 2008, 48, 504–512. 59 Y. Masubuchi, M. Bourdi, T. P. Reilly, M. L. Graf, J. W. George and
24 B. McDonald, E. F. McAvoy, F. Lam, V. Gill, C. de la Motte, L. R. Pohl, Biochem. Biophys. Res. Commun., 2003, 304, 207–212.
R. C. Savani and P. Kubes, J. Exp. Med., 2008, 205, 915–927. 60 M. K. Connolly, D. Ayo, A. Malhotra, M. Hackman, A. S. Bedrosian,
25 J. G. Chosay, N. A. Essani, C. J. Dunn and H. Jaeschke, J. Ibrahim, N. E. Cieza-Rubio, A. H. Nguyen, J. R. Henning, M. Dorvil-
Am. J. Physiol., 1997, 272, G1195–G1200. Castro, H. L. Pachter and G. Miller, Hepatology, 2011, 54, 959–968.
26 D. H. Adams, C. Ju, S. K. Ramaiah, J. Uetrecht and H. Jaeschke, 61 J. E. Sims and D. E. Smith, Nat. Rev. Immunol., 2010, 10, 89–102.
Toxicol. Sci., 2010, 115, 307–321. 62 A. B. Imaeda, A. Watanabe, M. A. Sohail, S. Mahmood,
27 H. Jaeschke, C. D. Williams, A. Ramachandran and M. L. Bajt, Liver M. Mohamadnejad, F. S. Sutterwala, R. A. Flavell and W. Z. Mehal,
Int., 2012, 32, 8–20. J. Clin. Invest., 2009, 119, 305–314.
28 M. A. Tirmenstein and S. D. Nelson, J. Biol. Chem., 1989, 264, 9814– 63 R. Hoque, M. A. Sohail, S. Salhanick, A. F. Malik, A. Ghani,
9819. S. C. Robson and W. Z. Mehal, Am. J. Physiol.: Gastrointest. Liver
29 H. Jaeschke and M. L. Bajt, Toxicol. Sci., 2006, 89, 31–41. Physiol., 2012, 302, G1171–G1179.
30 M. L. Bajt, C. Cover, J. J. Lemasters and H. Jaeschke, Toxicol. Sci., 64 C. D. Williams, D. J. Antoine, P. J. Shaw, C. Benson, A. Farhood,
2006, 94, 217–225. D. P. Williams, T. D. Kanneganti, B. K. Park and H. Jaeschke, Toxicol.
31 J. S. Gujral, T. R. Knight, A. Farhood, M. L. Bajt and H. Jaeschke, Appl. Pharmacol., 2011, 252, 289–297.
Toxicol. Sci., 2002, 67, 322–328. 65 P. Matzinger, Annu. Rev. Immunol., 1994, 12, 991–1045.
32 D. H. Adams, B. Eksteen and S. M. Curbishley, Gut, 2008, 57, 838– 66 K. L. Rock and L. Shen, Immunol. Rev., 2005, 207, 166–183.
848. 67 D. R. Green, T. Ferguson, L. Zitvogel and G. Kroemer, Nat. Rev.
33 Z. X. Liu, S. Govindarajan and N. Kaplowitz, Gastroenterology, 2004, Immunol., 2009, 9, 353–363.
127, 1760–1774. 68 T. S. Griffith, H. Kazama, R. L. VanOosten, J. K. Earle, Jr.,
34 M. J. Masson, L. D. Carpenter, M. L. Graf and L. R. Pohl, Hepatology, J. M. Herndon, D. R. Green and T. A. Ferguson, J. Immunol., 2007,
2008, 48, 889–897. 178, 2679–2687.
35 S. B. Yee, M. Bourdi, M. J. Masson and L. R. Pohl, Chem. Res. 69 D. J. Antoine, D. P. Williams, A. Kipar, R. E. Jenkins, S. L. Regan,
Toxicol., 2007, 20, 734–744. J. G. Sathish, N. R. Kitteringham and B. K. Park, Toxicol. Sci., 2009,
36 R. Miyakawa, T. Ichida, S. Yamagiwa, C. Miyaji, H. Watanabe, Y. Sato, 112, 521–531.
H. Yokoyama, C. Tsukada, Y. Ishimoto, S. Sugahara, X. H. Yang, 70 H. Kazama, J. E. Ricci, J. M. Herndon, G. Hoppe, D. R. Green and
T. Abo and H. Asakura, J. Gastroenterol. Hepatol., 2005, 20, 1126– T. A. Ferguson, Immunity, 2008, 29, 21–32.
1130. 71 D. J. Antoine, R. E. Jenkins, J. W. Dear, D. P. Williams, M. R. McGill,
37 B. Eksteen, S. C. Afford, S. J. Wigmore, A. P. Holt and D. H. Adams, M. R. Sharpe, D. G. Craig, K. J. Simpson, H. Jaeschke and B. K. Park,
Semin. Liver Dis., 2007, 27, 351–366. J. Hepatol., 2012, 56, 1070–1079.
38 M. L. Bajt, A. Farhood and H. Jaeschke, Am. J. Physiol. Gastrointest. 72 J. P. Uetrecht, Chem. Res. Toxicol., 1999, 12, 387–395.
Liver Physiol., 2001, 281, G1188–G1195. 73 Z. X. Liu and N. Kaplowitz, Clin. Liver Dis., 2002, 6, 755–774.
39 H. Jaeschke, Am. J. Physiol.: Gastrointest. Liver Physiol., 2006, 290, 74 M. P. Manns and P. Obermayer-Straub, Hepatology, 1997, 26, 1054–
G1083–G1088. 1066.

This journal is © The Royal Society of Chemistry 2012 Toxicol. Res., 2012, 1, 161–170 | 169
View Article Online

75 T. Mizutani, M. Shinoda, Y. Tanaka, T. Kuno, A. Hattori, T. Usui, C. Guarner, G. Soriano, Y. Borraz and J. A. Agundez, Hepatology,
N. Kuno and T. Osaka, Drug Metab. Rev., 2005, 37, 235–252. 2010, 52, 303–312.
76 J. Li and J. P. Uetrecht, Handb. Exp. Pharmacol., 2010, 493– 93 R. A. Roth and P. E. Ganey, Crit. Rev. Toxicol., 2011, 41, 723–739.
509. 94 J. C. Marshall, P. M. Walker, D. M. Foster, D. Harris, M. Ribeiro,
77 B. K. Park, A. Boobis, S. Clarke, C. E. Goldring, D. Jones, J. G. Kenna, J. Paice, A. D. Romaschin and A. N. Derzko, Crit. Care, 2002, 6,
C. Lambert, H. G. Laverty, D. J. Naisbitt, S. Nelson, D. A. Nicoll- 342–348.
Griffith, R. S. Obach, P. Routledge, D. A. Smith, D. J. Tweedie, 95 J. F. Maddox, C. J. Amuzie, M. Li, S. W. Newport, E. Sparkenbaugh,
N. Vermeulen, D. P. Williams, I. D. Wilson and T. A. Baillie, Nat. Rev. C. F. Cuff, J. J. Pestka, G. H. Cantor, R. A. Roth and P. E. Ganey,
Drug Discovery, 2011, 10, 292–306. J. Toxicol. Environ. Health, Part A, 2010, 73, 58–73.
78 D. P. Williams, Toxicology, 2006, 226, 1–11. 96 D. Schmid and C. Munz, Immunity, 2007, 27, 11–21.
79 M. J. Masson, R. A. Peterson, C. J. Chung, M. L. Graf, L. D. Carpenter, 97 V. Deretic, Curr. Opin. Immunol., 2009, 21, 53–62.
J. L. Ambroso, D. L. Krull, J. Sciarrotta and L. R. Pohl, Chem. Res. 98 C. Paludan, D. Schmid, M. Landthaler, M. Vockerodt, D. Kube,
Toxicol., 2007, 20, 20–26. T. Tuschl and C. Munz, Science, 2005, 307, 593–596.
80 J. Adam, W. J. Pichler and D. Yerly, Br. J. Clin. Pharmacol., 2011, 71, 99 H. W. Virgin and B. Levine, Nat. Immunol., 2009, 10, 461–470.
701–707. 100 H. H. Pua, I. Dzhagalov, M. Chuck, N. Mizushima and Y. W. He,
81 B. Schnyder, D. Mauri-Hellweg, M. Zanni, F. Bettens and W. J. Pichler, J. Exp. Med., 2007, 204, 25–31.
J. Clin. Invest., 1997, 100, 136–141. 101 C. M. Walsh and A. L. Edinger, Immunol. Rev., 2010, 236, 95–109.
82 U. A. Boelsterli and P. L. Lim, Toxicol. Appl. Pharmacol., 2007, 220, 102 M. E. Wildenberg, A. C. Vos, S. C. Wolfkamp, M. Duijvestein,
92–107. A. P. Verhaar, A. A. Te Velde, G. R. van den Brink and D. W. Hommes,
Published on 28 August 2012. Downloaded on 27/10/2014 12:55:05.

83 A. Srivastava, J. L. Maggs, D. J. Antoine, D. P. Williams, D. A. Smith Gastroenterology, 2012, 142, 1493–1503.


and B. K. Park, Handb. Exp. Pharmacol., 2010, 165–194. 103 H. M. Ni, A. Bockus, N. Boggess, H. Jaeschke and W. X. Ding, Hepa-
84 C. Cover, A. Mansouri, T. R. Knight, M. L. Bajt, J. J. Lemasters, tology, 2012, 55, 222–232.
D. Pessayre and H. Jaeschke, J. Pharmacol. Exp. Ther., 2005, 315, 104 H. M. Ni, N. Boggess, M. R. McGill, M. Lebofsky, P. Borude, U. Apte,
879–887. H. Jaeschke and W. X. Ding, Toxicol. Sci., 2012, 127, 438–450.
85 T. R. Knight, M. W. Fariss, A. Farhood and H. Jaeschke, Toxicol. Sci., 105 Y. Igusa, S. Yamashina, K. Izumi, Y. Inami, H. Fukada, M. Komatsu,
2003, 76, 229–236. K. Tanaka, K. Ikejima and S. Watanabe, J. Gastroenterol., 2012, 47,
86 H. Jaeschke, M. R. McGill, C. D. Williams and A. Ramachandran, Life 433–443.
Sci., 2011, 88, 737–745. 106 M. Komatsu, H. Kurokawa, S. Waguri, K. Taguchi, A. Kobayashi,
87 K. Fujimoto, K. Kumagai, K. Ito, S. Arakawa, Y. Ando, S. Oda, Y. Ichimura, Y. S. Sou, I. Ueno, A. Sakamoto, K. I. Tong, M. Kim,
T. Yamoto and S. Manabe, Toxicol. Pathol., 2009, 37, 193–200. Y. Nishito, S. Iemura, T. Natsume, T. Ueno, E. Kominami,
88 A. Ramachandran, M. Lebofsky, S. A. Weinman and H. Jaeschke, H. Motohashi, K. Tanaka and M. Yamamoto, Nat. Cell. Biol., 2010, 12,
Toxicol. Appl. Pharmacol., 2011, 251, 226–233. 213–223.
89 H. Jaeschke, M. R. McGill and A. Ramachandran, Drug Metab. Rev., 107 H. Jaeschke and W. X. Ding, J. Gastroenterol., 2012, 47, 845–846.
2012, 44, 88–106. 108 C. Ju and T. Reilly, Drug Metab. Rev., 2012, 44, 107–115.
90 M. M. Ong, C. Latchoumycandane and U. A. Boelsterli, Toxicol. Sci., 109 B. K. Park, M. Pirmohamed and N. R. Kitteringham, Chem. Res.
2007, 97, 205–213. Toxicol., 1998, 11, 969–988.
91 R. Kashimshetty, V. G. Desai, V. M. Kale, T. Lee, C. L. Moland, 110 R. J. Andrade, J. A. Agundez, M. I. Lucena, C. Martinez, R. Cueto and
W. S. Branham, L. S. New, E. C. Chan, H. Younis and U. A. Boelsterli, E. Garcia-Martin, Curr. Drug Metab., 2009, 10, 956–970.
Toxicol. Appl. Pharmacol., 2009, 238, 150–159. 111 N. Chalasani and E. Bjornsson, Gastroenterology, 2010, 138, 2246–
92 M. I. Lucena, E. Garcia-Martin, R. J. Andrade, C. Martinez, 2259.
C. Stephens, J. D. Ruiz, E. Ulzurrun, M. C. Fernandez, M. Romero- 112 S. Russmann, A. Jetter and G. A. Kullak-Ublick, Hepatology, 2010, 52,
Gomez, A. Castiella, R. Planas, J. A. Duran, A. M. De Dios, 748–761.

170 | Toxicol. Res., 2012, 1, 161–170 This journal is © The Royal Society of Chemistry 2012

You might also like