You are on page 1of 13

CHAPTER 32

Ubiquitin-Proteasome Pathway 32
in the Pathogenesis of Liver Disease
Samuel W. French, Fawzia Bardag-Gorce

the chamber of the proteasome to undergo digestion.


32.1 Ubiquitin (shown as Ub) exists as a pool of free mol-
Introduction ecules (upper left of Fig. 32.1), which can be attached
either singly or in polyubiquitin chains to the sub-
The discovery of the ubiquitin-proteasome pathway strate protein (bottom center). The polyubiquitinat-
as a regulated system of protein digestion within all ed protein substrate is then degraded by the 26S pro-
cells [32] has led to an appreciation of the impor- teasome to smaller peptides, which can then either
tance of protein turnover control. This control is a be degraded to amino acids by other proteinases or
mechanism for regulation of cellular processes and be presented on the surface of the cell membrane in
quality control of intracellular proteins. Many liver the immune cells. The beta subunits in the middle
cell functions are regulated by this mechanism of of the 20S proteasome include chymotrypsin-like
protein degradation. These include cell cycle check and trypsin-like catalytic enzymes, which digest
points and activation of transcription factors such the proteins that enter the chamber. The entrance of
as nuclear factor-κB (NF-κB) (Chapter 29), and hy- the proteins into the digestion chamber is gated by
poxia inducible factor-1α (HIF-1α) (Chapter 26) alpha subunits. In this way the digestion of proteins
[28, 67, 71]. The loss of proteasomes or the inhibi- is selectively regulated. Proteins that are digested by
tion of the ubiquitin-proteasome pathway can lead the 26S proteasomes must first be targeted to pro-
to hepatocellular injury including proliferation and teasomes by at least a 4 polyubiquitin chain where
apoptosis [74, 107], and hepatic inclusions of ag- the C-terminal glycine is covalently bound to lysine
gregated cytokeratins [24]. Liver cell gene expres- residues in the protein destined to be digested by the
sions, dependent on transcription factor activation proteasome.
by the proteasome, could impede the inflammatory
response of the liver and the response to hypoxic in-
jury. The importance of the ubiquitin-proteasome
pathway to the homeostatic mechanisms involved 32.3
in liver injury is the focus of this review. Ubiquitination of Proteins Destined
for Digestion by the Proteasome

Initially, in the conjugation phase of ubiquitin to the


32.2 protein destined for proteolysis by the proteasome,
Ubiquitin-Proteasome Pathway ubiquitin is first activated through the ATP-depend-
ent formation of thiol ester with a cysteine residue of
The enzymes that catalyze ubiquitin activation, the ubiquitination-activating enzyme (E1) (Fig. 32.1).
conjugation and ligation are depicted as E1, E2, and The carboxyl group of the last amino acid of ubiqui-
E3, respectively in Fig. 32.1. Ubiquitin is indicated tin, a glycine-glycine dipeptide, is first activated by
as a shaded circle. Note that the polyubiquitinated adenylation. A thiol group in the activating enzyme
protein indicated by Cn+1 docks at the 19S protea- (E1), which initiates the ubiquitination enzyme cas-
some. Alpha (α) units of the 20S proteasome par- cade, attaches the ubiquitin-carboxyl-adenosine 5’-
ticle are shown in gray, while the beta (β) subunits monophosphate to form a E1-ubiquitin thiol ester.
are shown in white. Both types of subunits make up Ubiquitin is then transferred to a cysteine residue
the 20S catalytic core of the proteasome. The arrows on a ubiquitin-conjugating enzyme (E2) (Fig. 32.1).
shown at the 19S proteasome indicate that the ubiq- Ubiquitin is then transferred to a lysine residue of
uitinated protein is deubiquitinated by proteasomal the protein substrate in a reaction catalyzed by a
deubiquitinase before the protein unfolds and enters ubiquitin-protein ligase (E3) (Fig. 32.1) [38, 40]. All
378 PART II: Specific Signaling Pathways

ATP Fig. 32.1. Scheme


of the ubiquitin-proteasome
pathway

Deubiquitination

known E3s utilize one of two catalytic domains, only tenfold when eight more ubiquitin molecules
HECT domains or a RING finger [81]. are added [100]. Therefore, the length of the polyu-
The ubiquitin-proteasome system of regulated biquitin chain determines the rate of substrate deg-
protein digestion is highly selective where signal- radation by the proteasome, after the chain binds to
dependent proteolysis directs the time and location the 19S proteasome, possibly because of the nature
of protein turnover in the cell [38, 40]. The selection of the deubiquitinating enzyme chain reaction. If
of proteins to be degraded by the proteasome oc- the chain is too short before degradation is initiat-
curs at the point of conjugation, though the E2s and ed, the substrate will be deubiquitinated, but not de-
E3s have different substrate specificities. Each E3, graded. A longer ubiquitin chain would allow more
in conjunction with its cognate E2, binds substrates time for unfolding of the substrate and opening the
that share a particular ubiquitination signal, usual- gate to allow entrance of the substrate into the cata-
ly a small primary sequence motif [53, 62, 80]. Rec- lytic chamber of the 20S proteasome for digestion.
ognition of the primary signal targets the substrate The ubiquitin-proteasome system is regulated
to be labeled with a second signal, i.e. the polyu- at the level of ubiquitination or at the level of the
biquitin chain formation. Only the polyubiquitin proteasome activity. The targeting motif can be the
chain signal is recognized by the proteasome. The N-terminal residue, a sequence, a domain, or a post-
phenomenon of polyubiquitinated secondary signal translational change such as phosphorylation that
formation provides a large number of substrates for results from externally generated signal transduc-
the proteasome, which are predestined for selective tion. Phosphorylation of either the substrate or the
digestion. ligase can activate binding of the ligase. Binding can
The recognition of the secondary signal by the be enhanced by ubiquitin-like proteins (UBL) by in-
proteasome is true only for the proteasome and creasing the affinity of the ligases to the E2 compo-
differs in this way from other ATP-dependent pro- nent of the conjugating apparatus, especially in the
teases in the cell [100]. The ubiquitin molecules are case of phosphorylated substrates. UBL proteins in-
linked covalently to one another through K48-G76 teract with the polyubiquitinin domain (UBA). The
isopeptide bonds. The first ubiquitin binds a lysine UBL domain probably acts to facilitate the interac-
in the substrate. This supports residues in the sub- tion with UB-containing proteins, targeting them to
strate to generate an isopeptide bond [16]. The af- the proteasome [17]. Linking elements on proteins
finity of binding of the polyubiquitin chain to the such as UBA and UBL may increase the efficiency of
proteasome increases 100-fold as the chain length the proteasome process.
increases from two to four ubiquitins, but increases
CHAPTER 32: Ubiquitin-Proteasome Pathway in the Pathogenesis of Liver Disease 379

[69]. The concept of liver injury through this mech-


32.4 anism applies to both alcoholic and non-alcoholic
Ubiquitin-Proteasome Mechanics liver disease in man [98, 99], as well as experimental
ethanol-induced liver disease in rats and mice [4, 6,
The processing of short-lived regulatory and struc- 21, 23, 28].
tural proteins within the cell for degradation by
the ubiquitin-proteasome system is extraordinarily
complex and has recently been reviewed elsewhere
in detail [32]. The process involves the folding of 32.6
intracellular proteins by heat shock proteins and Functional Consequences
chaperones [35, 37]; polyubiquitination by three of Ethanol Inhibition
enzymes in sequence (ubiquitin-activating enzyme, of the Proteasome in the Liver
E1; ubiquitin-conjugating enzyme, E2s; and ubiqui-
tin-protein ligase, E3s); assembly of the 20S protea- Donohue et al. [21] first showed that the activities
some containing the catalytic subunits; assembly of three proteolytic enzymes located in the 20S pro-
of the 19S proteasome including the base and lid; teasome, i.e., chymotrypsin-like, trypsin-like, and
assembly of the 26S subunit; ATP, and deubiquiti- peptidyl-glutamyl-peptide hydrolase, were inhibited
nating enzyme. There are thousands of proteasomes in rats fed ethanol intragastrically for 2 months but
in hepatocytes that are mobile in the cytosol and not in rats fed ethanol ad lib. The inhibition of these
localized to the nuclear pore, endoplasmic reticu- proteasome enzyme activities correlated with an in-
lum, and the nucleus [33]. In pathologic conditions, crease in liver weight, compared to pair-fed controls.
where proteins accumulate to form an aggresome, The liver total proteins also increased significantly
the proteasomes localize in the aggresomes [42]. compared to pair-fed controls, suggesting that the
When isolated from the liver they are found as 20S, increase in liver weight and protein content was a
20S with one 19S, and 20S with two 19S attachments result of the inhibition of protein proteolysis asso-
(26S) [28]. The 20S proteasome does not require ciated with ethanol intake. The relative amounts of
ubiquitination of the proteins or ATP to digest pro- proteasome subunits did not change with ethanol,
teins. This includes oxidized proteins [91, 97]. Thus, suggesting that the inhibition of catalytic activity
the digestion of proteins by the 26S proteasome is was not due to reduced enzyme protein levels. The
regulated by the polyubiquitinated protein and proteasome inhibition was shown in the standard
ATP-dependent mechanisms. activities, i.e. in the SDS-stimulated 20S proteasome
Other proteins involved in delivering the ubiq- activities, and in the ATP-stimulated 26S protea-
uitinated proteins to the proteasome for digestion some activities, indicating that the defect involved
include CHIP, Rad 23, AG-1, and P62 (ZIP). These the 26S proteasome as well as the 20S proteasome.
proteins dimerize and bind to polyubiquitin chains The inhibition of proteasome activity corre-
and the proteasome to facilitate proteolysis through lated with an increase in malondialdehyde in liver
a hypothetical UBA/UBL [18, 30, 35–37, 83, 102]. The homogenates, suggesting that malondialdehyde ad-
protein substrates for proteasome digestion are nu- ducts resulting from increased lipid peroxidation
merous and the list is still growing [24, 101]. These caused by alcohol could be involved in proteasome
include p53, Cdc 25, Cdc 18, IKBα, β-catenin, CD4, inhibition. None of these changes was noted in the
HIF-α, EGF-R, PDGF-R, Mdm2, and MATα2, which rats fed ethanol ad lib, suggesting that a dose-re-
are involved in liver diseases. Each has a different sponse relationship is responsible for proteasome
ubiquitin ligase. inhibition [21]. The studies of Fataccioli et al. [23]
corroborated those of Donohue et al. [21]. In addi-
tion, when CYP2E1 inhibitors were fed with ethanol,
inhibition of the proteasome chymotrypsin-like ac-
32.5 tivity was significantly reduced, suggesting that free
Experimental Alcoholic Liver Disease radical damage may be the mechanism of inhibition
and Proteasomal Catalytic Activity of the proteasome caused by ethanol feeding. Li-
pid peroxidation correlated inversely with chymot-
Donohue [22] has recently reviewed the literature rypsin-like activity. Again, ad lib feeding of ethanol
summarizing the evidence implicating the ubiq- did not cause these associations with proteasome
uitin-proteasome system in ethanol-induced liver inhibition, nor was there an increase in formation of
disease. He first introduced the concept [21] but, to malondialdehyde, protein thiols, or protein carbon-
date, it has largely been ignored in recent reviews yls. Also, ad lib feeding of ethanol did not increase
of the mechanisms of alcohol-induced liver injury the total liver protein levels.
380 PART II: Specific Signaling Pathways

Rouach et al. [88] found that rats fed ethanol would be a likely mechanism of inhibition. Indeed,
intragastrically had increased levels of malondial- transgenic mice, in which the gene for CYP2E1 had
dehyde, protein thiols, and protein carbonyls in the been knocked out, failed to develop proteasome
liver. The increase in oxidized proteins correlated activity inhibition even after 1 month of ethanol
positively with the severity of the liver pathology. feeding [4]. One candidate product is 4-hydrox-
Thus, the inhibition of proteasome activity and the ynonenal (4-HNE), an end product of lipid peroxi-
increase in protein oxidation may be related because dation, which results from the hydroxyethyl radical
the proteasome eliminates oxidized proteins [91]. generated by CYP2E1 during ethanol oxidation [2].
Indeed, 4-HNE is increased in the liver of rats fed
ethanol intragastrically [26]. 4-HNE progressively
increased in the liver during 1 month of ethanol
32.7 feeding, peaking when the proteasome catalytic ac-
Other Functional Consequences tivity had become inhibited [10]. 4-HNE adducts are
of Proteasome Inhibition formed in the liver in alcoholic liver disease. Their
presence correlated with the morphologic severity
Ethanol stabilizes proteins normally degraded by of liver disease [89].
the proteasome. Hypothetically, this would include The possibility of adduct forming between a pro-
CYP2E1, NF-κB, and HIF-1α. In the case of CYP2E1, teasome subunit and 4-HNE accounting for the loss
it has been shown that ethanol withdrawal leads to an of proteolytic activity was investigated. When the
overnight reduction in the ethanol-induced CYP2E1 20S proteasome was isolated from the livers of rats
and that this rapid loss in CYP2E1 in the liver can be fed ethanol and controls, no loss of catalytic activity
prevented by injecting a potent proteasome inhibi- was found. Also, no 4-HNE adduct was found when
tor, PS-341 [7]. This change in CYP2E1 levels cor- the 20S proteasome was studied by two-dimension-
related with a return of the proteasome, inhibited al (2D)-electrophoresis and Western blot. However,
by ethanol, to normal levels of activity 24 h after when the 26S proteasome was isolated, and studied
ethanol withdrawal. This is compelling evidence by 2D-electrophoresis and Western blot, there was
that CYP2E1 was stabilized by ethanol inhibition of loss of proteasome activity, and a 4-HNE adduct to
the proteasome. Proteasome inhibitor treatment of a 44-kDa subunit was found in the alcohol-fed rats.
HepG2 cells in vitro increases the level of CYP2E1 The 2D membrane, stripped of the 4-HNE antibody
threefold by preventing degradation by the protea- and stained with an antibody to Rpt4, an ATPase
some (stabilizes CYP2E1) [79]. In the case of NF-κB subunit of the regulatory complex 19S part of the
stabilization, it was shown that ethanol fed intragas- 26S proteasome, showed that 4-HNE was adducted
trically for 2 months was associated with the lack of to this subunit in the ethanol-fed rat liver. Thus,
NF-κB activation [34], which could be the result of proteasomal catalytic activity could be lost because
inhibition of proteasome digestion of IκBα that pre- the subunit that controls the opening of the catalytic
vents NF-κB activation. NF-κB would be expected to channel was altered by 4-HNE adduction [10].
be activated by oxidative stress caused by ethanol in Another possible mechanism that could explain
this model. In the case of HIF-1α, the nuclear activa- the loss of proteasome catalytic activity after ethanol
tion of this transcription factor is increased at high feeding is phosphorylation of a regulatory subunit of
blood alcohol levels and expression of genes regu- the proteasome. Previous in vitro studies had shown
lated by HIF-1α is also increased, including eryth- that ethanol stimulated phosphorylation of proteins
ropoietin, iNOS, and VEGF gene expression [7, 70], in liver cells through a protein kinase C (PKC) route
suggesting that the normal elimination of HIF-1α by [49]. Okadaic acid, which inhibits phosphatase ac-
the proteasome is inhibited by ethanol feeding. tivity and increases phosphorylated proteins in the
liver, caused profound inhibition of proteasome
activity in vivo [8]. Proteasomes isolated from the
32.8 liver of rats fed ethanol and controls were studied
The Mechanism of Ethanol-Induced Inhibition for subunit phosphorylation using an antibody to
of Proteasome Activity in the Liver phosphothreonine. Using 2D-electrophoresis and
Western blotting, a hyperphosphorylated subunit,
Ethanol feeding does not inhibit proteasome cata- alpha 7, was found in the proteasomes isolated from
lytic activity directly [10]. The inhibition does not the liver. Inhibition of proteolytic activity of the
occur after 2 weeks of ethanol feeding. Inhibition of 20S proteasome could result from phosphorylation
the proteasome was not found until 1 month of feed- of this subunit because the α subunits are also in-
ing rats ethanol. This suggested that an oxidized volved in opening the mouth of the catalytic cham-
product of CYP2E1 generated by ethanol CYP2E1 ber of the 26S proteasome.
CHAPTER 32: Ubiquitin-Proteasome Pathway in the Pathogenesis of Liver Disease 381

Fig. 32.2a–f. Comparison of ethanol feeding


and proteasome inhibition effects on liver cells.
a–d Hematoxylin and eosin-stained sections
from the liver of rats fed ethanol for 1 month
(a), or fed ethanol for 1 month and given PS-
341 (b) or rats given PS-341 only (c, d). Note
that both ethanol and PS-341 caused apoptosis
(a, c). e, f Hepatocytes stained by TUNEL from
the liver of a rat fed ethanol for 1 month (e) or
a rat fed ethanol for 1 month and given PS-
341 (f). PS-341 treatment caused apoptosis (f)
mainly when combined with ethanol (e)

Bd-2, and NF-κB. Thus, the ubiquitin-proteasome


32.9 system is an essential player in the regulation of
Some Consequences of Inhibition proteins involved in programmed cell death in the
of the Proteasome: Apoptosis liver and other organs.

Apoptosis in the liver is increased in alcoholic liver


disease [27]. Apoptosis is also increased in the liver
from viral hepatitis and drug hepatitis. Inhibition 32.10
of the proteasome is one mechanism involved in Role of the Ubiquitin-Proteasome Pathway
apoptosis. When rats were injected with the potent in Liver Cell Aggresome (Mallory Body)
proteasome inhibitor PS-341 (IP 0.5 mg/kg), apop- Formation
tosis increased compared to controls [28] (Fig. 32.2).
The livers were examined for apoptosis 4 h post in- Mallory bodies are formed by damaged liver cells
jection using TUNEL staining. Positive-stained in a great variety of different chronic liver diseases
nuclei were increased, as were cells that stained [41]. Thus, they are a sign of more severe liver injury
positive for activated caspase-3 (hepatocytes and [3, 90]. In alcoholic liver disease, ubiquitin conju-
sinusoidal cells). Alanine aminotransferase (ALT) gates (polyubiquitinated proteins) are increased in
was increased. Proteasome chymotrypsin-like ac- the blood [99] and ubiquitin in the liver correlates
tivity was decreased in liver homogenates (~ 85% with the presence of Mallory bodies in non-alco-
decrease). The way in which proteasome inhibition holic steatohepatitis [3]. Thus, there is a general in-
leads to apoptosis is complex. Many pro- and anti- crease in polyubiquitinated proteins in an environ-
apoptosis proteins are regulated by the proteasome ment in which Mallory bodies form. The association
[28]. These include Bax, Bcl-2, p53, mdm 2, IKBα, of Mallory bodies with ubiquitinated cytokeratins
caspase 3, caspase 8 c-1AP1, XiAP, NF-κB precursor, in liver cells has been known since 1988 [64, 75].
and Bid [74]. Pro-apoptosis proteins are p53, Bax, Only recently has the role of the ubiquitin-protea-
and Bid. Anti-apoptosis proteins are c-IAPI, XIAP, some pathway in the pathogenesis of Mallory body
382 PART II: Specific Signaling Pathways

Fig. 32.3a, b. Theoretical model of the role of heat shock pro- ubiquitin, hsp 70 heat shock protein 70, hsp 90 heat shock protein
teins (hsp) in normal protein degradation and MB formation. (a) 90, MB Mallory body, UBB+1 mutant ubiquitin, cytokeratin. (Re-
Normal cytokeratin degradation. (b) Cytokeratin aggregation printed from [86], with permission from Elsevier)
and formation of MBs in the presence of stress and/or UBB+1. Ub

formation been appreciated. The new concept is by ubiquitination. Proteolysis of the cytokeratins by
based on the aggresome phenomenon, which biolo- the proteasome then occurs [56].
gists have described in a variety of tissues where To test the hypothesis that hyperphosphorylation
protein aggregates accumulate around the centro- induces Mallory body formation, drug-primed mice
some [42, 104, 106]. The idea was first formulated as were given an acute dose of okadaic acid and the liv-
a consequence of events in the degradation pathway ers were monitored for the appearance of Mallory
of liver cell cytokeratins. It started with a conforma- bodies. Fifteen minutes after okadaic acid injection,
tional change in the cytokeratin proteins caused by phosphorylated protein aggregates formed in liver
phosphorylation, where α helices decreased and β cells. The aggregates stained positive with cytokera-
sheets increased, causing the cytokeratins to stick tin antibody [109]. True Mallory bodies formed by
to each other [109]. According to the concept of ag- day 2 and progressed with daily injections of oka-
gresome formation, β-sheet conformational change daic acid. The Mallory bodies stained positive for
is an important mechanism for protein aggregation phosphothreonine in tissue slices and by Western
[54]. Conformational changes in Mallory bodies blot in the Mallory body band. Human liver cell
have been documented by infrared spectroscopy Mallory bodies also stained positive with the phos-
[44]. Hyperphosphorylation of cytokeratin 8 by phothreonine antibody. Similar cytokeratin aggre-
hepatocytes from mouse livers forming Mallory gates formed in hepatocytes in tissue culture when
bodies was documented by 32P incorporation into exposed to okadaic acid [13, 92].
cytokeratins of hepatocytes in tissue culture using
2D-electrophoresis [15]. Mallory bodies containing
hyperphosphorylated proteins have been found in
a model of Mallory body formation in hepatocytes. 32.11
Phosphothreonine antibody was used in Western Role of Heat Shock Proteins
blots of Mallory body protein [72]. Hyperphosphor- in Mallory Body Formation
ylation of cytokeratins in Mallory-body-forming liv-
ers has been characterized further by finding phos- Heat shock proteins act as chaperones for altered
phorylation of multiple sites on amino acid residues cytoplasmic proteins that have been targeted for
in the cytokeratin proteins [94]. The turnover of cy- digestion by the proteasome after polyubiquitina-
tokeratins is signaled by phosphorylation, followed tion (Fig. 32.3) [105]. Heat shock treatment of drug-
CHAPTER 32: Ubiquitin-Proteasome Pathway in the Pathogenesis of Liver Disease 383

Fig. 32.4a–c. Human liver cells with numerous Mallory bodies a confocal microscope. a, and b images were merged in c. Note
due to alcohol abuse. The liver section was double stained with the merged image shows the Mallory bodies, indicating colo-
antibodies to cytokeratin 18 (a) and to p62 (b) and viewed with calization of p62 and cytokeratin 18 (×2,500)

primed mice induces Mallory bodies after upregula- zole prevent aggresome formation [42, 104, 106].
tion of heat shock protein expression [108]. When Miniaggregates are transported on microtubules
human or mice livers containing Mallory bodies towards the microtubule organizing center at the
were stained with antibodies to heat shock proteins centrosome, driven by the minus-end motor protein
70 and 90, they stained positive, indicating that both dynein [29, 43]. Using a new tissue culture model
heat shock proteins were pathogenetically linked to of Mallory body formation, microtubule inhibitors
Mallory body formation [86, 95]. The expression of totally blocked Mallory body formation [87]. Only
heat shock protein 70 and 90 as determined by West- small cytokeratin-ubiquitin positive aggregates
ern blot correlated with the number of Mallory bod- formed at the cell periphery, whereas the control liv-
ies expressed [9]. er cultures formed numerous Mallory bodies in the
absence of the microtubular inhibitors (Fig. 32.5).
Tubulin and Tau isoforms located in microtubules
localized in the Mallory body [51, 84].
32.12
Role of p62
in Mallory Body Formation
32.14
P62 (Z1P) links polyubiquitinated proteins to the Role of Transglutaminase
proteasome. P62 is a major component of hyaline in Mallory Body Formation
bodies in hepatocellular carcinomas [93], Mallory
bodies [96], as well as Alzheimer’s disease neurofi- Transglutaminase was found in the Mallory bodies
brillary tangles and synucleipathies [59, 60] are in- and in Western blots and was induced when Mallory
volved in p62-ubiquitin binding in aggresome for- bodies were induced [9]. Northern blots showed that
mation. ZIP/p62 is a scaffolding protein that binds transglutaminase gene expression was upregulated
to polyubiquitin at its UBA domain [30, 82]. Human when Mallory bodies were formed at the same time
Mallory bodies stain strongly with p62 antibody that cytokeratin 8 was overexpressed. Increased
(Fig. 32.4). transglutaminase activity and cross-linking of cy-
tokeratins were reported earlier [19, 110, 111]. Cross-
linking of cytokeratin by transglutaminase in hepa-
tocytes may account for the insolubility of Mallory
32.13 bodies in the SDS cocktail used in Western blots
Role of Microtubules [78].
in Mallory Body Formation

Aggresomes require microtubules to be present,


in order for them to form. Colchicine and nocoda-
384 PART II: Specific Signaling Pathways

Fig. 32.5a–c. Primary culture of mouse liver cells forming Mal- image shows the Mallory bodies in yellow, indicating colocaliza-
lory bodies. The liver cells were double stained for cytokeratin tion of ubiquitin and cytokeratin 18. Confocal fluorescent micro-
18 (green, a) and for ubiquitin (red, b) and viewed with a confocal scopy (×2,500)
microscope. c a merged picture from a and b. Note the merged

man and mouse livers [24, 68]. The UBB+1 mutant


32.15 was first reported to be present in neuronal cell ag-
Role of Proteasome Inhibition gresomes in Alzheimer’s disease [103]. This mutant
in the Formation of Mallory Bodies is a result of molecular misreading of the ubiquitin
B gene during transcription (a dinucleotide dele-
Proteasome inhibitors induced aggresome forma- tion), which results in a mutant ubiquitin that has
tion in vitro, presumably because the inhibition of lost the binding site in the C prime end of ubiquitin.
proteolysis causes an accumulation of proteins in It therefore cannot function to bind misfolded pro-
the cell [42, 106]. This means that inhibition of the teins destined for proteolysis by the ubiquitin-pro-
proteasome is central to Mallory body formation teasome pathway. This is not a functional ubiquitin.
[109]. In fact, injection of a potent proteasome in- Worse, UBB+1 is polyubiquitinated by wild-type
hibitor in drug-primed mice induces Mallory body ubiquitin, but the resultant polyubiquitin chains
formation after 2 days [24]. The proteasome inhibi- are refractory to disassembly by deubiquitinating
tor PS-341 is selective in its action on the protea- enzymes (Fig. 32.6). This potentially inhibits the
some and is an effective anticancer drug because degradation of a polyubiquitinated substrate by pu-
proteasome inhibition causes apoptosis of cancer rified 26S proteasomes [61]. In vitro studies using
cells [1]. PS-341 also induces aggresome formation primary liver cultures showed that UBB+1 transfec-
in hepatocytes in vitro after going through a stage of tion induced cytokeratin-ubiquitin aggresomes in
collapse of cytokeratin from the plasma membrane hepatocytes within 4 h of incubation [11]. In vitro
followed by retraction of the cytokeratin around studies used a ubiquitin-proteasome pathway mix-
the nucleus [85] at 5 h. Later (24 h), aggresomes are ture to form a reconstituted ubiquitin-proteasome,
formed in place of the contracted cytokeratin. When which includes a cytokeratin immunoprecipitate.
Mallory bodies form in mouse liver, the cytokeratin This mixture contained Mallory bodies, added
disappears in the cytoplasm (ghost cell formation). ubiquitin, a ubiquitination system with the enzymes
Proteasomes are localized at the edge of the Mallory E1, E2, and E3, purified proteasomes, and an enzyme
body and the proteasomes in the cytoplasm are di- mixture for generating ATP. The mixture was in-
minished [5]. cubated for 4 and 24 h [9]. The Mallory body band
(insoluble protein at the top of the gel located in the
loading gel) increased when the mixture was added.
Likewise, the ubiquitinated protein fraction was in-
32.16 creased. Adding UBB+1 to the mixture increased the
Role of the Frameshift Mutant Mallory body band further [11]. Adding PS-341 also
of Ubiquitin in Mallory Body Formation increased the Mallory body band. The combination
of UBB+1 and PS-341 did not increase the Mallory
Recently, a frameshift mutant of ubiquitin, UBB+1, body band further, suggesting that the two treat-
was shown to be present in Mallory bodies in hu- ments were acting through the same mechanism.
CHAPTER 32: Ubiquitin-Proteasome Pathway in the Pathogenesis of Liver Disease 385

Fig. 32.6. Molecular misreading and Mallory


body formation. Representation of how target
proteins can be tagged efficiently for protea-
somal destruction (right side) and how this oc-
curs less efficiently (left side) when the mutant
ubiquitin UBB+1 is involved in the ubiquitina-
tion. UBB+1 has lost its ability to ubiquitinate
because it does not have a terminal Gly resi-
due. It is refractory to deubiquitination by the
deubiquitinase (isopeptidase) enzyme. In fact,
the mutant ubiquitin becomes ubiquitinated
itself, and inhibits the proteasome in a domi-
nant-negative way. (Reprinted from [68], with
permission from Elsevier)

Ubiquitination of UBB+1 occurred. The proteasomes tin 18 to cause overexpression, led to the formation
localized within the Mallory body, indicating that, of cytokeratin aggresomes. But these aggresomes
as occurs in aggresomes in vivo, the proteasomes failed to form the characteristic filaments seen in
were bound to the aggresome. This might explain Mallory bodies [14, 39, 73].
how UBB+1 interferes with the ubiquitin-proteasome Mallory bodies incorporate newly synthesized
pathway of proteolysis. cytokeratin directly in tissue culture as shown by
The question arises: does UBB+1 expression lead [35S]methionine pulse labeling using Western blots
to cell death? In the case of neurons, UBB+1 expres- and electron microscopy autoradiography of deter-
sion does lead to cell death [20]. Aggresome forma- gent-extracted hepatocytes in culture where only the
tion under many differing circumstances may cause cytoskeleton remained [45]. In Western blots, the
death of the cell [12, 63]. radiolabel was found in the Mallory band as well as
in normal cytokeratin 8 and cytokeratin 18 bands.
Similarly, the radiolabel was incorporated into the
Mallory filaments as well as the cytokeratin fila-
32.17 ments. Cytokeratin 18 cDNA was used to transfect
Role of Cytokeratin 8 and 18 Overexpression Mallory-body-forming cells in mouse liver tissue
in Mallory Body Formation culture using a green fluorescent protein tag. The
cytokeratin localized in both the normally distrib-
During primary tissue culture of liver cells from uted cytokeratin but was also mainly concentrated
Mallory-body-forming livers, there was an increase in the Mallory body. This suggested that the nascent
in synthesis of cytokeratin 8 and cytokeratin 18 as human cytokeratin went both to the normal fila-
measured by Western blot [15]. Cytokeratin 8 gene ments as well as directly to the Mallory body after
expression was also markedly upregulated in Mal- synthesis [84, 85].
lory-body-forming livers [72]. The increase of cy-
tokeratins in hepatocytes may overload the ubiqui-
tin-proteasome pathway of cytokeratin proteolysis,
which then may cause cytokeratin accumulation to
form the Mallory body. Several studies, where tissue
culture cells were transfected with plasmids con-
taining cDNA coded for cytokeratin 8 or cytokera-
386 PART II: Specific Signaling Pathways

to FAS-induced apoptosis is increased. Cytokeratin


32.18 aggresome formation (Mallory bodies) may further
Functional Consequence reduce proteolysis by the proteasome.
of the Loss of Cytokeratin Organization
Due to Aggresome (Mallory Body) Formation

The function of liver cell cytokeratins and the vul- References


nerability of liver cells exposed to toxins and viral
damage have been reviewed elsewhere [52, 57, 76, 1. Adams J. Development of the proteasome inhibitor PS-341.
77]. Mutations in cytokeratin 8 and cytokeratin 18 Oncologist 2002;7:9–16.
were found in patients with cryptogenic cirrhosis, 2. Albano E, Clot P, Morimoto M et al. Role of cytochrome
hepatitis C, and other liver diseases. The question P4502E1-dependent formation of hydroxyethyl free radical
is: do changes in the cytokeratin organization due in the development of liver damage in rats intragastrically
to mutations or other etiologies interfere with liver fed with ethanol. Hepatology 1996;23:155–163.
function and increase vulnerability of the liver to in- 3. Banner BF, Savas L, Zivny J et al. Ubiquitin as a marker of
jury? For instance, hyperphosphorylation of the liv- cell injury in non alcoholic steatohepatitis. Am J Clin Pathol
er cell cytokeratins changes their organization, but 2000;114:860–866.
does this lead to loss of function [55]? Cytokeratin 4. Bardag-Gorce F, Yuan QX, Li J et al. The effect of ethanol-
reorganizes extensively during mitoses, apoptosis induced cytochrome P450 2E1 on the inhibition of protea-
and polarization of the cell [66]. In in vitro studies some activity by alcohol. Biochem Biophys Res Commun
using transfection of cells with mutant cytokeratins, 2000;279:23–29.
the cells were more vulnerable to injury. Cytokerat- 5. Bardag-Gorce F, French BA, Lue YH et al. Mallory bodies
ins have viscoelastic properties, which make cells formed in proteasome-depleted hepatocytes: an immuno-
resistant to physical stress. Lack of cytokeratins in histochemical study. Exp Molec Pathol 2001;70:7–18.
K18/8-deficient mice hepatocytes disturbs the pro- 6. Bardag-Gorce F, French BA, Li J et al. The importance of cy-
gression of the cell cycle. These cells are more likely cling of blood alcohol levels in the pathogenesis of experi-
to develop apoptosis in response to FAS [31, 58, 65]. mental alcoholic liver disease in rats fed ethanol intragastri-
The cytokeratins of the liver anchor and support the cally. Gastroenterology 2002;123:325–335.
bile canaliculus, forming the pericanalicular sheath 7. Bardag-Gorce F, Li J, French BA, French SW. Ethanol with-
supporting bile flow [47, 48]. Likewise the cytokera- drawal induced CYP2E1 degradation in vivo, blocked by pro-
tin filaments of the liver cell are involved in transhe- teasomal inhibitor PS-341. Free Radical Biol Med 2002;32:17–
patic transport of vesicles [50]. 21.
The cytokeratin cytoskeleton of liver cells that 8. Bardag-Gorce F, Li J, French BA et al. Chronic alcohol-in-
form Mallory bodies is profoundly altered so that duced phosphorylation of proteasome subunits proteolysis
only the Mallory body stains for keratin, leaving the of the proteasome. FASEB J 2002;16:A245.
rest of the cell unstained, the so-called “empty cell” 9. Bardag-Gorce F, van Leeuwen FW, Nguyen V et al. The role of
or “ghost cell” [46]. Mallory body cells lose their bile the ubiquitin-proteasome pathway in the formation of Mal-
canaliculi and cytokeratin in the cytoplasm. This is lory bodies. Exp Molec Pathol 2002;73:75–83.
associated with cell enlargement and rounding. The 10. Bardag-Gorce F, Li J, French BA, French SW. The effect of
cytokeratin network supports the nucleus, the nu- ethanol-induced CYP2E1 lipid peroxidation by product 4-
clear pores and the centrioles. The Mallory-body- hydroxynonenal on the 26S proteasome. FASEB J 2003;17:
containing cells in primary tissue culture were un- A325.
able to form bile canaliculi or secrete fluorescent 11. Bardag-Gorce F, Riley N, Nguyen V et al. The mechanism of
diacetate, compared with controls [25]. cytokeratin aggresome formation: the role of mutant ubiq-
uitin (UBB+1). Exp Molec Pathol 2003;74:160–167.
12. Bence NF, Sampat RM, Kopito RR. Impairment of the ubiq-
uitin-proteasome system by protein aggregation. Science
32.19 2001;292:1552–1555.
Concluding Remarks 13. Blankson H, Holen I, Seglen PO. Disruption of the cytokera-
tin cytoskeleton and inhibition of hepatocyte autophage by
It can be concluded that the inhibition of the ubiq- okadaic acid. Exp Cell Res 1995;218:522–530.
uitin-proteasome pathway of protein degradation 14. Blouin R, Kawahara H, French SW, Marceau N. Selective ac-
has profound effects on the structure and function cumulation of IF proteins at a focal juxtanuclear site in COS-1
of liver cells, including loss of cell shape and inter- cells transfected with mouse keratin 18 cDNA. Exp Cell Res
nal organization of organelles, polarity, and the bile 1990;187:234–242.
secretory apparatus. Vulnerability of the liver cell
CHAPTER 32: Ubiquitin-Proteasome Pathway in the Pathogenesis of Liver Disease 387

15. Cadrin M, Anderson M, Aasheim LH et al. Modifications in 32. Glickman MH, Ciechanover A. The ubiquitin-proteasome
cytokeratin and actin in cultured liver cells derived from gri- proteolytic pathway: destruction for the sake of construc-
seofulvin-fed mice. Lab Invest 1995;72:453–460. tion. Physiol Rev 2002;82:373–428.
16. Chan V, Tobias JW, Bachmair A et al. A multiubiquitin chain is 33. Gordon C. The intracellular localization of the proteasome.
confined to specific lysone in a targeted short-lived protein. Curr Topics Microbiol Immunol 2002;268:175–184.
Science 1989;243:1576–1584. 34. Gouillon ZQ, Miyamoto K, Donohue TM et al. Role of CYP2E1
17. Ciechanover A, Brundin P. The ubiquitin proteasome sys- in the pathogenesis of alcoholic liver disease: modifications
tem in neurodegenerative diseases: sometimes the chicken, by cAMP and ubiquitin-proteasome pathway. Frontiers Bio-
sometimes the egg. Neuron 2003;40:427–446. sci 1999;4:16–25.
18. Connell P, Ballinger CA, Jiang J et al. The co-chaperone CHIP 35. Hartl FU, Hayer-Hartl M. Molecular chaperones in the
regulates protein triage decisions mediated by heat-shock cytosol: from nascent chain to folded protein. Science
proteins. Nature Cell Biol 2001;3:93–96. 2002;295:1852–1858.
19. Denk H, Bernklau G, Krepler R. Effect of griseofulvin treat- 36. Hartmann-Petersen R, Seeger M, Gordon C. Transferring
ment and neoplastic transformation on transglutaminase substrates to the 26S proteasome. Trends Biochem Sci
activity in mouse liver. Liver 1984;4:208–213. 2003;28:26–31.
20. De Vrij FM, Sluijs JA, Gregori L et al. Mutant ubiquitin ex- 37. Hartmann-Petersen R, Semple CA, Ponting CP et al. UBA do-
pressed in Alzheimer’s disease causes neuronal death. main containing proteins in fission yeast. Intern J Biochem
FASEB J 2001;15:2680—2688. Cell Biol 2003;35:629–636.
21. Donohue Jr TM, Zetterman RK, Zhang-Gouillon Z-Q, French 38. Hershko A, Ciechanover A. The ubiquitin system. Annu Rev
SW. Peptidase activities of the multicatalytic protease in rat Biochem 1998;67s:425–480.
liver after voluntary and intragastric ethanol administration. 39. Hirano K, Roth J, Zuber C, Ziak M. Expression of a mutant ER-
Hepatology 1998;28:486–491. retained polytope membrane protein in cultured rat hepa-
22. Donohue TM. The ubiquitin-proteasome system and its role tocytes results in Mallory body formation. Histochem Cell
in ethanol-induced disorders. Addict Biol 2002;7(1):15–28 Biol 2002;117:41–53.
23. Fataccioli V, Andraud E, Gentil M et al. Effects of chronic 40. Hochstrasser M. Ubiquitin-dependent protein degradation.
ethanol administration on rat liver proteasome activities: re- Annu Rev Genet 1996;30:405–439.
lationship with oxidative stress. Hepatology 1999;29:14–20. 41. Jensen K, Gluud C. The Mallory body: morphological, clini-
24. French, BA, van Leeuwen F, Riley NE et al. Aggresome for- cal and experimental studies (Part 1 of a literature survey).
mation in liver cells in response to different toxic mecha- Hepatology 1994;20:1061–1077.
nisms. Role of the ubiquitin-proteasome pathway and 42. Johnston JA, Ward CL, Kopito RR. Aggresomes: a cellular re-
the frameshift mutant of ubiquitin. Exp Molec Pathol sponse to misfolded protein. J Cell Biol 1998;143:1883–1889.
2001;71:241–246. 43. Johnston JA, Illing ME, Kopito RR. Cytoplasmic dynein/dyn-
25. French SW, Kawahara H, Katsuma Y et al. Interaction of in- actin mediates the assembly of aggresomes. Cell Motil Cy-
termediate filaments with nuclear lamina and cell periphery. toskeleton 2002;53:26–38.
Electron Microsc Rev 1989;2:17–51. 44. Kachi K, Wong TT, French SW. Molecular structural changes
26. French SW, Wong KW, Jui L et al. Effect of ethanol on cyto- in Mallory body proteins in human and mouse livers: an in-
chrome P450 2E1 (CYP2E1) lipid peroxidation and serum frared spectroscopy study. Exp Molec Pathol 1993;59:197–
protein adduct formation in relation to liver pathology 210.
pathogenesis. Exp Molec Pathol 1993;58:61–75. 45. Kachi K, Cadrin M, French SW. Synthesis of Mallory body, in-
27. French SW, Nash J, Shitabata P et al. and the VA Coop- termediate filament, and microfilament proteins in liver cell
erative Study Group. Pathology of Alcoholic Liver Disease primary cultures. Lab Invest 1993;68:71–81.
1993;13:154–169. 46. Katsuma Y, Swierenga SH, Khettry U et al. Changes in the
28. French SW, Bardag-Gorce F. The ubiquitin-proteasome 26S cytokeratin intermediate filament cytoskeleton associated
pathway in liver cell protein turnover: effect of ethanol and with Mallory body formation in mouse and human liver.
drugs. Alcoholism: Clin Exp Res 2001;25:225s–229s. Hepatology 1987;7:1215–1223.
29. Garcia-Mata R, Gao Y-S, Sztul E. Hassles with taking out the 47. Katsuma Y, Marceau N, Ohta M, French SW. Cytokeratin inter-
garbage: aggravating aggresomes. Traffic 2002;3:388–396. mediated filaments of rat hepatocytes: different cytoskele-
30. Geetha T, Wooten MW. Structure and functional properties tal domains and their three dimensional structure. Hepatol-
of the ubiquitin binding protein p62. FEBS Lett 2002;512:19– ogy 1988;8:559–568.
24. 48. Kawahara H, Marceau N, French SW. Effect of agents which
31. Gilbert S, Loranger A, Daigle N, Marceau N. Simple epithe- rearrange the cytoskeleton in vitro on the structure and
lium keratins 8 and 18 provide resistance to FAS-mediated function of hepatocyte canaliculi. Lab Invest 1989;60:692–
apoptosis. The protection occurs through a receptor-target- 704.
ing modulation. J Cell Biol 2001;154:763–773. 49. Kawahara H, Cadrin M, French SW. Ethanol-induced phos-
phorylation of cytokeratin in cultured hepatocytes. Life Sci
1990;47:859–863.
388 PART II: Specific Signaling Pathways

50. Kawahara H, Cadrin M, Perry L et al. Role of the cytokeratin 68. McPhaul LW, Wang J, Hol EM et al. Molecular misreading of
intermediate filaments in transhepatic transport and canal- the ubiquitin B gene and hepatic Mallory body formation.
icular secretion. Hepatology 1990;11:435–448. Gastroenterology 2002;122:1878–1885.
51. Kenner L, el Shabrawi Y, Hutter H et al. Expression of three- 69. Molina PE, Hoek JB, Nelson S et al. Mechanisms of alcohol-in-
and four-repeat tau isoforms in mouse liver. Hepatology duced tissue injury. Alcohol Clin Exp Res 2003;27:563–575.
1994;20:1086–1089. 70. Montgomery R, French SW. Alcohol-induced liver hypoxia in
52. Kirfel J, Magin TM, Reichelt J. Keratins: a structural scaffold rats: role of HIF-1 induction. FASEB J 2003;17:A1283.
with emerging functions. Cell Mol Life Sci 2003;60:56–71. 71. Muratani M, Tansey WP. How the ubiquitin-proteasome sys-
53. Koepp DM, Harper JW, Elledge SJ. How the cyclin be- tem controls transcription. Nat Rev Mol Cell Biol 2003;4:192–
came a cyclin: regulated proteolysis in the cell cycle. Cell 201.
1999;97:431–434. 72. Nagao Y, Yuan Q-X, Wan Y-JY et al. Pathogenesis of Mal-
54. Kopito RR, Ron D. Conformational disease. Nat Cell Biol lory body formation. Studies using the drug-primed mouse
2000;2:E207–E209. model. Hepatol Res 1998;13:15–24.
55. Ku NO, Michie SA, Soetikno RM et al. Susceptibility to hepato- 73. Nakamichi I, Hatakeyama S, Nakayama KI. Formation of Mal-
toxicity in transgenic mice that express dominant-negative lory body-like inclusions and cell death induced by deregu-
human keratin 18 mutant. J Clin Invest 1996;98:1034–1046. lated expression of keratin 18. Mol Biol Cell 2002;13:3441–
56. Ku NO, Omary MB. Keratins turn over by ubiquitina- 3451.
tion in a phosphorylation-modulated fashion. J Cell Biol 74. Naujokat C, Hoffmann S. Role and function of the 26S
2000;149:547–552. proteasome in proliferation and apoptosis. Lab Invest
57. Ku NO, Gish R, Wright TL, Omary MB. Keratin 8 mutations 2002;82:965–980.
in patients with cryptogenic liver disease. N Engl J Med 75. Ohta M, Marceau N, Perry G et al. Ubiquitin is present on the
2001;344:1580–1587. cytokeratin intermediate filaments and Mallory bodies of
58. Ku NO, Soetikno RM, Omary ME. Keratin mutation in trans- hepatocytes. Lab Invest 1988;58:848–856.
genic mice predisposes Fas but not TNF-induced apoptosis 76. Omary MB, Ku NO. Intermediate filament proteins of the liv-
and massive liver injury. Hepatology 2003;37:1006–1014. er: emerging disease association and functions. Hepatology
59. Kuusisto E, Salminen A, Alafuzoff I. Ubiquitin-binding pro- 1997;25:1043–1048.
tein p62 is present in neuronal and glial inclusions in hu- 77. Omary MB, Ku NO, Toivola DM. Keratins: guardians of the
man tauopathies and synucleinopathies. Neuroreport liver. Hepatology 2002;35:251–257.
2001;12:2085–2090. 78. Osborn M, Weber K. Keratins, transglutaminase, and Mallory
60. Kuusisto E, Salminen A, Alafuzoff I. Early accumulation of bodies. The really insoluble stuff. Lab Invest 1989;61:585–
p62 in neurofibrillary tangles in Alzheimer’s disease: pos- 587.
sible role in tangle formation. Neuropathol Appl Neurobiol 79. Perez MJ, Cederbaum AI. Proteasome inhibition potenti-
2002;28:228–237. ates CYP2E1-mediated toxicity in HepG2 cells. Hepatology
61. Lam YA, Pickart CM, Alban A et al. Inhibition of the ubiquitin- 2003;37(6):1395–1404.
proteasome system in Alzheimer’s disease. Proc Natl Acad 80. Pickart CM. Ubiquitin in chains. Trends Biochem Sci 2000;25:
Sci USA 2000;97:9902–9906. 544–548.
62. Laney JD, Hochstrasser M. Substrate targeting in the ubiqui- 81. Pickart CM. Mechanisms underlying ubiquitination. Annu
tin system. Cell 1999;97:427–430. Rev Biochem 2001;70:503–533.
63. Lang-Rollin I, Rideout H, Stefanis L. Ubiquitinated inclusions 82. Puls A, Schmidt S, Grawe F, Stabel S. Interaction of protein
and neuronal cell death. Histol Histopathol 2003;18:509– kinase C with ZIP, a novel protein kinase C-binding protein.
517. Proc Natl Acad Sci USA 1997;94:6191–6196.
64. Lowe J, Blanchard A, Morrell K et al. Ubiquitin is a common 83. Raasi S, Pickart CM. Rad23 ubiquitin-associated domains
factor in intermediate filament inclusion bodies of diverse (UBA) inhibit 26S proteasome-catalyzed proteolysis by
type in man, including those of Parkinson’s disease, Pick’s sequestering lysine 48-linked polyubiquitin chains. J Biol
disease, and Alzheimer’s disease as well as Rosenthal fib- Chem 2003;278:8951–8959.
ers in cerebellar astrocytomas, cytoplasmic bodies in mus- 84. Riley NE, Li J, Worrall S et al. Mallory body as an aggresome:
cle, and Mallory bodies in alcoholic liver disease. J Pathol in vitro studies. Exp Molec Pathol 2002;72:17–23.
1988;155:9–15. 85. Riley NE, Bardag-Gorce F, French SW. Proteasome inhibi-
65. Marceau N, Loranger A, Gilbert S et al. Keratin-medi- tor PS-341 causes aggresome formation, and loss of cell
ated resistance to stress and apoptosis in simple epithe- cohesion in a mouse hepatoma cell line. Molec Biol Cell
lial cells in relation to health and disease. Biochem Cell Biol 2002;13s:265A.
2001;79:543–555. 86. Riley NE, Li J, McPhaul LW et al. Heat shock proteins are
66. Marceau N. Keratin 8 mutations in patients with cryptogenic present in Mallory bodies (cytokeratin aggresomes) in hu-
liver disease. J Hepatol 2002;36:710–711. man liver biopsy specimens. Exp Molec Pathol 2003;74:168–
67. Mayer JR. The meteoric rise of regulated intracellular prote- 172.
olysis. Nature Rev: Molec Cell Biol 2000;1:145–148.
CHAPTER 32: Ubiquitin-Proteasome Pathway in the Pathogenesis of Liver Disease 389

87. Riley NE, Bardag-Gorce F, Montgomery RO et al. Microtu- alcoholic liver diseases: immunoaffinity isolation and elec-
bules are required for cytokeratin aggresome formation in trophoretic mobility. Alcohol Clin Exp Res 2002;26:1692–
hepatocytes (Mallory bodies): an in vitro study. Exp Molec 1696.
Pathol 2003;74:173–179. 100. Thrower JS, Hoffman L, Rechsteiner M, Pickart CM. Rec-
88. Rouach H, Fataccioli V, Gentil M et al. Effect of chronic etha- ognition of the polyubiquitin proteolytic signal. EMBO J
nol feeding on lipid peroxidation and protein oxidation in 2000;19:94–104.
relation to liver pathology. Hepatology 1997;25(2):351–355. 101. Ulrich HD. Natural substrates of the proteasome and their
89. Seki S, Kitada T, Sakaguchi H et al. Pathological significance recognition by the ubiquitin system. Curr Topics Microbiol
of oxidative cellular damage in human alcoholic liver dis- Immunol 2002;268:137–174.
ease. Histopathology 2003;42:365–371. 102. Vadlamudi RK, Joung I, Strominger JL, Shin J. P62, a phos-
90. Shimada M, Hashimoto E, Kaneda H et al. Nonalcoholic photyrosine-independent ligand of the SH2 domain of
steatohepatitis: risk factors for liver fibrosis. Hepatol Res 56lck , belongs to a new class of ubiquitin-binding proteins.
2002;24(4):429–438. J Biol Chem 1996;271:20235–20237.
91. Shringarpure R, Grune T, Mehlhase J, Davies KJ. Ubiquitin 103. van Leeuwen FW, de Kleijn DP, van den Hurk HH et al.
conjugation is not required for the degradation of oxidized Frameshift mutants of β-amyloid precursor protein
proteins by proteasome. J Biol Chem 2003;278:311–318. and ubiquitin-B in Alzheimer's down patients. Science
92. Strnad P, Windoffer R, Leube RE. In vivo detection of cytok- 1998;279:242–247.
eratin filament network breakdown in cells treated with 104. Wigley WC, Fabunmi RP, Lee MG et al. Dynamic association
the phosphatase inhibitor okadaic acid. Cell Tissue Res of proteasomal machinery with the centrosome. J Cell Biol
2001;306:277–293. 1999;145:481–490.
93. Stumptner C, Heid H, Fuchsbichler A et al. Analysis of intra- 105. Wilkinson KD. Ubiquitination and deubiquitination: target-
cytoplasmic hyaline bodies in a hepatocellular carcinoma. ing of proteins for degradation by the proteasome. Semin
Am J Pathol 1999;154:1701–1710. Cell Develop Biol 2000;11:141–148.
94. Stumptner C, Omary MB, Fickert P et al. Hepatocyte cytok- 106. Wojcik C, Shroeter D, Wilk S et al. Ubiquitin-mediated prote-
eratins are hyperphosphorylated at multiple sites in human olysis centers in HeLa cells: indication from studies of an in-
alcoholic hepatitis and in a Mallory body mouse model. Am hibitor of the chymotrypsin-like activity of the proteasome.
J Pathol 2001;156:77–90. Eur J Cell Biol 1996;7:311–318.
95. Stumptner C, Fuchsbichler M, Lehner M et al. Sequence 107. Wojcik C. Regulation of apoptosis by the ubiquitin and pro-
of events in the assembly of Mallory body components in teasome pathway. J Cell Mol Med 2002;6:25–48.
mouse liver: clues to the pathogenesis and significance of 108. Yuan QX, Marceau N, French BA, French SW. Heat shock in
Mallory body formation. J Hepatol 2001;34:665–675. vivo induces Mallory body formation in drug primed mouse
96. Stumptner C, Fuchsbichler A, Heid H et al. Mallory body: liver. Exp Molec Pathol 1995;63:63–76.
a disease-associated type of sequestosome. Hepatology 109. Yuan QX, Nagao Y, Gaal K et al. Mechanisms of Mallory body
2002;35:1035–1062. formation induced by Okadaic acid in drug-primed mice.
97. Szweda PA, Friguet B, Szweda LI. Proteolysis, free radicals, Exp Molec Pathol 1998;65:87–103.
and aging. Free Radical Biol Med 2002;33:29–36. 110. Zatloukal K, Denk H, Lackinger E, Rainer I. Hepatocellular
98. Takagi M, Yamauchi M, Toda G et al. Serum ubiquitin levels cytokeratins as substrates of transglutaminases. Lab Invest
in patients with alcoholic liver disease. Alcohol Clin Exp Res 1989;61:603–608.
1999;23:76s–80s. 111. Zatloukal K, Fesus L, Denk H et al. High amount of Σ-(γ-
99. Takagi M, Yamauchi M, Takada K, Ohkawa K. Serum ubiqui- glutamyl)lysine cross-links in Mallory bodies. Lab Invest
tin-protein conjugates in normal subjects and patients with 1992;66:774–777.

You might also like