• Non ci sono risultati.

Ubiquitin-Proteasome Pathway in the Pathogenesis of Liver Disease

N/A
N/A
Protected

Academic year: 2022

Condividi "Ubiquitin-Proteasome Pathway in the Pathogenesis of Liver Disease"

Copied!
13
0
0

Testo completo

(1)

Ubiquitin-Proteasome Pathway

in the Pathogenesis of Liver Disease

Samuel W. French, Fawzia Bardag-Gorce

32

32.1

Introduction

The discovery of the ubiquitin-proteasome pathway as a regulated system of protein digestion within all cells [32] has led to an appreciation of the impor- tance of protein turnover control. This control is a mechanism for regulation of cellular processes and quality control of intracellular proteins. Many liver cell functions are regulated by this mechanism of protein degradation. These include cell cycle check points and activation of transcription factors such as nuclear factor- κB (NF-κB) (Chapter 29), and hy- poxia inducible factor-1 α (HIF-1α) (Chapter 26) [28, 67, 71]. The loss of proteasomes or the inhibi- tion of the ubiquitin-proteasome pathway can lead to hepatocellular injury including proliferation and apoptosis [74, 107], and hepatic inclusions of ag- gregated cytokeratins [24]. Liver cell gene expres- sions, dependent on transcription factor activation 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 in liver injury is the focus of this review.

32.2

Ubiquitin-Proteasome Pathway

The enzymes that catalyze ubiquitin activation, conjugation and ligation are depicted as E1, E2, and E3, respectively in Fig. 32.1. Ubiquitin is indicated as a shaded circle. Note that the polyubiquitinated protein indicated by Cn+1 docks at the 19S protea- some. Alpha ( α) units of the 20S proteasome par- ticle are shown in gray, while the beta ( β) subunits are shown in white. Both types of subunits make up the 20S catalytic core of the proteasome. The arrows shown at the 19S proteasome indicate that the ubiq- uitinated protein is deubiquitinated by proteasomal deubiquitinase before the protein unfolds and enters

the chamber of the proteasome to undergo digestion.

Ubiquitin (shown as Ub) exists as a pool of free mol- ecules (upper left of Fig. 32.1), which can be attached either singly or in polyubiquitin chains to the sub- strate protein (bottom center). The polyubiquitinat- ed protein substrate is then degraded by the 26S pro- teasome to smaller peptides, which can then either be degraded to amino acids by other proteinases or be presented on the surface of the cell membrane in the immune cells. The beta subunits in the middle of the 20S proteasome include chymotrypsin-like and trypsin-like catalytic enzymes, which digest the proteins that enter the chamber. The entrance of the proteins into the digestion chamber is gated by alpha subunits. In this way the digestion of proteins is selectively regulated. Proteins that are digested by the 26S proteasomes must first be targeted to pro- teasomes by at least a 4 polyubiquitin chain where the C-terminal glycine is covalently bound to lysine residues in the protein destined to be digested by the proteasome.

32.3

Ubiquitination of Proteins Destined for Digestion by the Proteasome

Initially, in the conjugation phase of ubiquitin to the protein destined for proteolysis by the proteasome, ubiquitin is first activated through the ATP-depend- ent formation of thiol ester with a cysteine residue of the ubiquitination-activating enzyme (E

1

) (Fig. 32.1).

The carboxyl group of the last amino acid of ubiqui- tin, a glycine-glycine dipeptide, is first activated by adenylation. A thiol group in the activating enzyme (E

1

), which initiates the ubiquitination enzyme cas- cade, attaches the ubiquitin-carboxyl-adenosine 5’- monophosphate to form a E

1

-ubiquitin thiol ester.

Ubiquitin is then transferred to a cysteine residue on a ubiquitin-conjugating enzyme (E

2

) (Fig. 32.1).

Ubiquitin is then transferred to a lysine residue of

the protein substrate in a reaction catalyzed by a

ubiquitin-protein ligase (E

3

) (Fig. 32.1) [38, 40]. All

(2)

known E3s utilize one of two catalytic domains, HECT domains or a RING finger [81].

The ubiquitin-proteasome system of regulated protein digestion is highly selective where signal- dependent proteolysis directs the time and location of protein turnover in the cell [38, 40]. The selection of proteins to be degraded by the proteasome oc- curs at the point of conjugation, though the E

2

s and E

3

s have different substrate specificities. Each E

3

, in conjunction with its cognate E

2

, binds substrates that share a particular ubiquitination signal, usual- ly a small primary sequence motif [53, 62, 80]. Rec- ognition of the primary signal targets the substrate to be labeled with a second signal, i.e. the polyu- biquitin chain formation. Only the polyubiquitin chain signal is recognized by the proteasome. The phenomenon of polyubiquitinated secondary signal formation provides a large number of substrates for the proteasome, which are predestined for selective digestion.

The recognition of the secondary signal by the proteasome is true only for the proteasome and differs in this way from other ATP-dependent pro- teases in the cell [100]. The ubiquitin molecules are linked covalently to one another through K48-G76 isopeptide bonds. The first ubiquitin binds a lysine in the substrate. This supports residues in the sub- strate to generate an isopeptide bond [16]. The af- finity of binding of the polyubiquitin chain to the proteasome increases 100-fold as the chain length increases from two to four ubiquitins, but increases

only tenfold when eight more ubiquitin molecules are added [100]. Therefore, the length of the polyu- biquitin chain determines the rate of substrate deg- radation by the proteasome, after the chain binds to the 19S proteasome, possibly because of the nature of the deubiquitinating enzyme chain reaction. If the chain is too short before degradation is initiat- ed, the substrate will be deubiquitinated, but not de- graded. A longer ubiquitin chain would allow more time for unfolding of the substrate and opening the gate to allow entrance of the substrate into the cata- lytic chamber of the 20S proteasome for digestion.

The ubiquitin-proteasome system is regulated at the level of ubiquitination or at the level of the proteasome activity. The targeting motif can be the N-terminal residue, a sequence, a domain, or a post- translational change such as phosphorylation that results from externally generated signal transduc- tion. Phosphorylation of either the substrate or the ligase can activate binding of the ligase. Binding can be enhanced by ubiquitin-like proteins (UBL) by in- creasing the affinity of the ligases to the E

2

compo- nent of the conjugating apparatus, especially in the case of phosphorylated substrates. UBL proteins in- teract with the polyubiquitinin domain (UBA). The UBL domain probably acts to facilitate the interac- tion with UB-containing proteins, targeting them to the proteasome [17]. Linking elements on proteins such as UBA and UBL may increase the efficiency of the proteasome process.

ATP

Deubiquitination

Fig. 32.1. Scheme of the ubiquitin-proteasome pathway

(3)

32.4

Ubiquitin-Proteasome Mechanics

The processing of short-lived regulatory and struc- 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 intracellular proteins by heat shock proteins and chaperones [35, 37]; polyubiquitination by three enzymes in sequence (ubiquitin-activating enzyme, E1; ubiquitin-conjugating enzyme, E2s; and ubiqui- tin-protein ligase, E

3

s); assembly of the 20S protea- some containing the catalytic subunits; assembly of the 19S proteasome including the base and lid;

assembly of the 26S subunit; ATP, and deubiquiti- nating enzyme. There are thousands of proteasomes in hepatocytes that are mobile in the cytosol and localized to the nuclear pore, endoplasmic reticu- lum, and the nucleus [33]. In pathologic conditions, where proteins accumulate to form an aggresome, the proteasomes localize in the aggresomes [42].

When isolated from the liver they are found as 20S, 20S with one 19S, and 20S with two 19S attachments (26S) [28]. The 20S proteasome does not require ubiquitination of the proteins or ATP to digest pro- teins. This includes oxidized proteins [91, 97]. Thus, the digestion of proteins by the 26S proteasome is regulated by the polyubiquitinated protein and ATP-dependent mechanisms.

Other proteins involved in delivering the ubiq- uitinated proteins to the proteasome for digestion include CHIP, Rad 23, AG-1, and P62 (ZIP). These proteins dimerize and bind to polyubiquitin chains and the proteasome to facilitate proteolysis through a hypothetical UBA/UBL [18, 30, 35–37, 83, 102]. The protein substrates for proteasome digestion are nu- merous and the list is still growing [24, 101]. These include p53, Cdc 25, Cdc 18, IKB α, β-catenin, CD4, HIF- α, EGF-R, PDGF-R, Mdm

2

, and MAT α2, which are involved in liver diseases. Each has a different ubiquitin ligase.

32.5

Experimental Alcoholic Liver Disease and Proteasomal Catalytic Activity

Donohue [22] has recently reviewed the literature summarizing the evidence implicating the ubiq- uitin-proteasome system in ethanol-induced liver disease. He first introduced the concept [21] but, to date, it has largely been ignored in recent reviews of the mechanisms of alcohol-induced liver injury

[69]. The concept of liver injury through this mech- anism applies to both alcoholic and non-alcoholic liver disease in man [98, 99], as well as experimental ethanol-induced liver disease in rats and mice [4, 6, 21, 23, 28].

32.6

Functional Consequences of Ethanol Inhibition

of the Proteasome in the Liver

Donohue et al. [21] first showed that the activities of three proteolytic enzymes located in the 20S pro- teasome, i.e., chymotrypsin-like, trypsin-like, and peptidyl-glutamyl-peptide hydrolase, were inhibited in rats fed ethanol intragastrically for 2 months but not in rats fed ethanol ad lib. The inhibition of these proteasome enzyme activities correlated with an in- crease in liver weight, compared to pair-fed controls.

The liver total proteins also increased significantly compared to pair-fed controls, suggesting that the increase in liver weight and protein content was a result of the inhibition of protein proteolysis asso- ciated with ethanol intake. The relative amounts of proteasome subunits did not change with ethanol, suggesting that the inhibition of catalytic activity was not due to reduced enzyme protein levels. The proteasome inhibition was shown in the standard activities, i.e. in the SDS-stimulated 20S proteasome activities, and in the ATP-stimulated 26S protea- some activities, indicating that the defect involved the 26S proteasome as well as the 20S proteasome.

The inhibition of proteasome activity corre- lated with an increase in malondialdehyde in liver homogenates, suggesting that malondialdehyde ad- ducts resulting from increased lipid peroxidation caused by alcohol could be involved in proteasome inhibition. None of these changes was noted in the rats fed ethanol ad lib, suggesting that a dose-re- sponse relationship is responsible for proteasome 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-

tivity was significantly reduced, suggesting that free

radical damage may be the mechanism of inhibition

of the proteasome caused by ethanol feeding. Li-

pid peroxidation correlated inversely with chymot-

rypsin-like activity. Again, ad lib feeding of ethanol

did not cause these associations with proteasome

inhibition, nor was there an increase in formation of

malondialdehyde, protein thiols, or protein carbon-

yls. Also, ad lib feeding of ethanol did not increase

the total liver protein levels.

(4)

Rouach et al. [88] found that rats fed ethanol intragastrically had increased levels of malondial- dehyde, protein thiols, and protein carbonyls in the liver. The increase in oxidized proteins correlated positively with the severity of the liver pathology.

Thus, the inhibition of proteasome activity and the increase in protein oxidation may be related because the proteasome eliminates oxidized proteins [91].

32.7

Other Functional Consequences of Proteasome Inhibition

Ethanol stabilizes proteins normally degraded by the proteasome. Hypothetically, this would include CYP2E1, NF- κB, and HIF-1α. In the case of CYP2E1, it has been shown that ethanol withdrawal leads to an overnight reduction in the ethanol-induced CYP2E1 and that this rapid loss in CYP2E1 in the liver can be prevented by injecting a potent proteasome inhibi- tor, PS-341 [7]. This change in CYP2E1 levels cor- related with a return of the proteasome, inhibited by ethanol, to normal levels of activity 24 h after ethanol withdrawal. This is compelling evidence that CYP2E1 was stabilized by ethanol inhibition of the proteasome. Proteasome inhibitor treatment of HepG2 cells in vitro increases the level of CYP2E1 threefold by preventing degradation by the protea- some (stabilizes CYP2E1) [79]. In the case of NF- κB stabilization, it was shown that ethanol fed intragas- trically for 2 months was associated with the lack of NF- κB activation [34], which could be the result of inhibition of proteasome digestion of I κBα that pre- vents NF- κB activation. NF-κB would be expected to be activated by oxidative stress caused by ethanol in this model. In the case of HIF-1 α, the nuclear activa- tion of this transcription factor is increased at high blood alcohol levels and expression of genes regu- lated by HIF-1 α is also increased, including eryth- ropoietin, iNOS, and VEGF gene expression [7, 70], suggesting that the normal elimination of HIF-1 α by the proteasome is inhibited by ethanol feeding.

32.8

The Mechanism of Ethanol-Induced Inhibition of Proteasome Activity in the Liver

Ethanol feeding does not inhibit proteasome cata- lytic activity directly [10]. The inhibition does not occur after 2 weeks of ethanol feeding. Inhibition of the proteasome was not found until 1 month of feed- ing rats ethanol. This suggested that an oxidized product of CYP2E1 generated by ethanol CYP2E1

would be a likely mechanism of inhibition. Indeed, transgenic mice, in which the gene for CYP2E1 had been knocked out, failed to develop proteasome activity inhibition even after 1 month of ethanol feeding [4]. One candidate product is 4-hydrox- ynonenal (4-HNE), an end product of lipid peroxi- dation, which results from the hydroxyethyl radical 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 feeding, peaking when the proteasome catalytic ac- tivity had become inhibited [10]. 4-HNE adducts are formed in the liver in alcoholic liver disease. Their presence correlated with the morphologic severity of liver disease [89].

The possibility of adduct forming between a pro- teasome subunit and 4-HNE accounting for the loss of proteolytic activity was investigated. When the 20S proteasome was isolated from the livers of rats fed ethanol and controls, no loss of catalytic activity was found. Also, no 4-HNE adduct was found when the 20S proteasome was studied by two-dimension- al (2D)-electrophoresis and Western blot. However, when the 26S proteasome was isolated, and studied by 2D-electrophoresis and Western blot, there was loss of proteasome activity, and a 4-HNE adduct to a 44-kDa subunit was found in the alcohol-fed rats.

The 2D membrane, stripped of the 4-HNE antibody and stained with an antibody to Rpt4, an ATPase subunit of the regulatory complex 19S part of the 26S proteasome, showed that 4-HNE was adducted to this subunit in the ethanol-fed rat liver. Thus, proteasomal catalytic activity could be lost because the subunit that controls the opening of the catalytic channel was altered by 4-HNE adduction [10].

Another possible mechanism that could explain

the loss of proteasome catalytic activity after ethanol

feeding is phosphorylation of a regulatory subunit of

the proteasome. Previous in vitro studies had shown

that ethanol stimulated phosphorylation of proteins

in liver cells through a protein kinase C (PKC) route

[49]. Okadaic acid, which inhibits phosphatase ac-

tivity and increases phosphorylated proteins in the

liver, caused profound inhibition of proteasome

activity in vivo [8]. Proteasomes isolated from the

liver of rats fed ethanol and controls were studied

for subunit phosphorylation using an antibody to

phosphothreonine. Using 2D-electrophoresis and

Western blotting, a hyperphosphorylated subunit,

alpha 7, was found in the proteasomes isolated from

the liver. Inhibition of proteolytic activity of the

20S proteasome could result from phosphorylation

of this subunit because the α subunits are also in-

volved in opening the mouth of the catalytic cham-

ber of the 26S proteasome.

(5)

32.9

Some Consequences of Inhibition of the Proteasome: Apoptosis

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 of the proteasome is one mechanism involved in apoptosis. When rats were injected with the potent proteasome inhibitor PS-341 (IP 0.5 mg/kg), apop- tosis increased compared to controls [28] (Fig. 32.2).

The livers were examined for apoptosis 4 h post in- jection using TUNEL staining. Positive-stained nuclei were increased, as were cells that stained positive for activated caspase-3 (hepatocytes and sinusoidal cells). Alanine aminotransferase (ALT) was increased. Proteasome chymotrypsin-like ac- tivity was decreased in liver homogenates (~ 85%

decrease). The way in which proteasome inhibition leads to apoptosis is complex. Many pro- and anti- apoptosis proteins are regulated by the proteasome [28]. These include Bax, Bcl-2, p53, mdm 2, IKB α, caspase 3, caspase 8 c-1AP1, XiAP, NF- κB precursor, and Bid [74]. Pro-apoptosis proteins are p53, Bax, and Bid. Anti-apoptosis proteins are c-IAPI, XIAP,

Bd-2, and NF- κB. Thus, the ubiquitin-proteasome system is an essential player in the regulation of proteins involved in programmed cell death in the liver and other organs.

32.10

Role of the Ubiquitin-Proteasome Pathway in Liver Cell Aggresome (Mallory Body) Formation

Mallory bodies are formed by damaged liver cells in a great variety of different chronic liver diseases [41]. Thus, they are a sign of more severe liver injury [3, 90]. In alcoholic liver disease, ubiquitin conju- gates (polyubiquitinated proteins) are increased in the blood [99] and ubiquitin in the liver correlates with the presence of Mallory bodies in non-alco- holic steatohepatitis [3]. Thus, there is a general in- crease in polyubiquitinated proteins in an environ- ment in which Mallory bodies form. The association of Mallory bodies with ubiquitinated cytokeratins in liver cells has been known since 1988 [64, 75].

Only recently has the role of the ubiquitin-protea- some pathway in the pathogenesis of Mallory body

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)

(6)

formation been appreciated. The new concept is based on the aggresome phenomenon, which biolo- gists have described in a variety of tissues where protein aggregates accumulate around the centro- some [42, 104, 106]. The idea was first formulated as a consequence of events in the degradation pathway of liver cell cytokeratins. It started with a conforma- tional change in the cytokeratin proteins caused by phosphorylation, where α helices decreased and β sheets increased, causing the cytokeratins to stick to each other [109]. According to the concept of ag- gresome formation, β-sheet conformational change is an important mechanism for protein aggregation [54]. Conformational changes in Mallory bodies have been documented by infrared spectroscopy [44]. Hyperphosphorylation of cytokeratin 8 by hepatocytes from mouse livers forming Mallory bodies was documented by

32

P incorporation into 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.

Phosphothreonine antibody was used in Western blots of Mallory body protein [72]. Hyperphosphor- ylation of cytokeratins in Mallory-body-forming liv- ers has been characterized further by finding phos- phorylation of multiple sites on amino acid residues in the cytokeratin proteins [94]. The turnover of cy- tokeratins is signaled by phosphorylation, followed

by ubiquitination. Proteolysis of the cytokeratins by the proteasome then occurs [56].

To test the hypothesis that hyperphosphorylation induces Mallory body formation, drug-primed mice were given an acute dose of okadaic acid and the liv- ers were monitored for the appearance of Mallory bodies. Fifteen minutes after okadaic acid injection, phosphorylated protein aggregates formed in liver cells. The aggregates stained positive with cytokera- tin antibody [109]. True Mallory bodies formed by day 2 and progressed with daily injections of oka- daic acid. The Mallory bodies stained positive for phosphothreonine in tissue slices and by Western blot in the Mallory body band. Human liver cell Mallory bodies also stained positive with the phos- phothreonine antibody. Similar cytokeratin aggre- gates formed in hepatocytes in tissue culture when exposed to okadaic acid [13, 92].

32.11

Role of Heat Shock Proteins in Mallory Body Formation

Heat shock proteins act as chaperones for altered cytoplasmic proteins that have been targeted for digestion by the proteasome after polyubiquitina- tion (Fig. 32.3) [105]. Heat shock treatment of drug-

Fig. 32.3a, b. Theoretical model of the role of heat shock pro- teins (hsp) in normal protein degradation and MB formation. (a) Normal cytokeratin degradation. (b) Cytokeratin aggregation and formation of MBs in the presence of stress and/or UBB+1. Ub

ubiquitin, hsp 70 heat shock protein 70, hsp 90 heat shock protein 90, MB Mallory body, UBB+1 mutant ubiquitin, cytokeratin. (Re- printed from [86], with permission from Elsevier)

(7)

primed mice induces Mallory bodies after upregula- tion of heat shock protein expression [108]. When human or mice livers containing Mallory bodies were stained with antibodies to heat shock proteins 70 and 90, they stained positive, indicating that both heat shock proteins were pathogenetically linked to Mallory body formation [86, 95]. The expression of heat shock protein 70 and 90 as determined by West- ern blot correlated with the number of Mallory bod- ies expressed [9].

32.12 Role of p62

in Mallory Body Formation

P62 (Z1P) links polyubiquitinated proteins to the proteasome. P62 is a major component of hyaline bodies in hepatocellular carcinomas [93], Mallory bodies [96], as well as Alzheimer’s disease neurofi- brillary tangles and synucleipathies [59, 60] are in- volved in p62-ubiquitin binding in aggresome for- mation. ZIP/p62 is a scaffolding protein that binds to polyubiquitin at its UBA domain [30, 82]. Human Mallory bodies stain strongly with p62 antibody (Fig. 32.4).

32.13

Role of Microtubules in Mallory Body Formation

Aggresomes require microtubules to be present, in order for them to form. Colchicine and nocoda-

zole prevent aggresome formation [42, 104, 106].

Miniaggregates are transported on microtubules towards the microtubule organizing center at the centrosome, driven by the minus-end motor protein dynein [29, 43]. Using a new tissue culture model of Mallory body formation, microtubule inhibitors totally blocked Mallory body formation [87]. Only small cytokeratin-ubiquitin positive aggregates formed at the cell periphery, whereas the control liv- 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].

32.14

Role of Transglutaminase in Mallory Body Formation

Transglutaminase was found in the Mallory bodies and in Western blots and was induced when Mallory bodies were induced [9]. Northern blots showed that transglutaminase gene expression was upregulated when Mallory bodies were formed at the same time that cytokeratin 8 was overexpressed. Increased 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 bodies in the SDS cocktail used in Western blots [78].

Fig. 32.4a–c. Human liver cells with numerous Mallory bodies due to alcohol abuse. The liver section was double stained with antibodies to cytokeratin 18 (a) and to p62 (b) and viewed with

a confocal microscope. a, and b images were merged in c. Note the merged image shows the Mallory bodies, indicating colo- calization of p62 and cytokeratin 18 (×2,500)

(8)

32.15

Role of Proteasome Inhibition in the Formation of Mallory Bodies

Proteasome inhibitors induced aggresome forma- tion in vitro, presumably because the inhibition of proteolysis causes an accumulation of proteins in the cell [42, 106]. This means that inhibition of the proteasome is central to Mallory body formation [109]. In fact, injection of a potent proteasome in- hibitor in drug-primed mice induces Mallory body formation after 2 days [24]. The proteasome inhibi- tor PS-341 is selective in its action on the protea- some and is an effective anticancer drug because proteasome inhibition causes apoptosis of cancer cells [1]. PS-341 also induces aggresome formation in hepatocytes in vitro after going through a stage of collapse of cytokeratin from the plasma membrane followed by retraction of the cytokeratin around the nucleus [85] at 5 h. Later (24 h), aggresomes are formed in place of the contracted cytokeratin. When Mallory bodies form in mouse liver, the cytokeratin disappears in the cytoplasm (ghost cell formation).

Proteasomes are localized at the edge of the Mallory body and the proteasomes in the cytoplasm are di- minished [5].

32.16

Role of the Frameshift Mutant

of Ubiquitin in Mallory Body Formation

Recently, a frameshift mutant of ubiquitin, UBB

+1

, was shown to be present in Mallory bodies in hu-

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

+1

mutant was first reported to be present in neuronal cell ag- gresomes in Alzheimer’s disease [103]. This mutant is a result of molecular misreading of the ubiquitin B gene during transcription (a dinucleotide dele- tion), which results in a mutant ubiquitin that has lost the binding site in the C prime end of ubiquitin.

It therefore cannot function to bind misfolded pro- teins destined for proteolysis by the ubiquitin-pro- teasome pathway. This is not a functional ubiquitin.

Worse, UBB

+1

is polyubiquitinated by wild-type ubiquitin, but the resultant polyubiquitin chains are refractory to disassembly by deubiquitinating enzymes (Fig. 32.6). This potentially inhibits the degradation of a polyubiquitinated substrate by pu- rified 26S proteasomes [61]. In vitro studies using primary liver cultures showed that UBB

+1

transfec- tion induced cytokeratin-ubiquitin aggresomes in hepatocytes within 4 h of incubation [11]. In vitro studies used a ubiquitin-proteasome pathway mix- ture to form a reconstituted ubiquitin-proteasome, which includes a cytokeratin immunoprecipitate.

This mixture contained Mallory bodies, added ubiquitin, a ubiquitination system with the enzymes E

1

, E

2

, and E

3

, purified proteasomes, and an enzyme mixture for generating ATP. The mixture was in- 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- creased. Adding UBB

+1

to the mixture increased the Mallory body band further [11]. Adding PS-341 also increased the Mallory body band. The combination of UBB

+1

and PS-341 did not increase the Mallory body band further, suggesting that the two treat- ments were acting through the same mechanism.

Fig. 32.5a–c. Primary culture of mouse liver cells forming Mal- lory bodies. The liver cells were double stained for cytokeratin 18 (green, a) and for ubiquitin (red, b) and viewed with a confocal microscope. c a merged picture from a and b. Note the merged

image shows the Mallory bodies in yellow, indicating colocaliza- tion of ubiquitin and cytokeratin 18. Confocal fluorescent micro- scopy (×2,500)

(9)

Ubiquitination of UBB

+1

occurred. The proteasomes localized within the Mallory body, indicating that, as occurs in aggresomes in vivo, the proteasomes were bound to the aggresome. This might explain how UBB

+1

interferes with the ubiquitin-proteasome pathway of proteolysis.

The question arises: does UBB

+1

expression lead to cell death? In the case of neurons, UBB

+1

expres- sion does lead to cell death [20]. Aggresome forma- tion under many differing circumstances may cause death of the cell [12, 63].

32.17

Role of Cytokeratin 8 and 18 Overexpression in Mallory Body Formation

During primary tissue culture of liver cells from Mallory-body-forming livers, there was an increase in synthesis of cytokeratin 8 and cytokeratin 18 as measured by Western blot [15]. Cytokeratin 8 gene expression was also markedly upregulated in Mal- 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-

tin 18 to cause overexpression, led to the formation of cytokeratin aggresomes. But these aggresomes failed to form the characteristic filaments seen in Mallory bodies [14, 39, 73].

Mallory bodies incorporate newly synthesized cytokeratin directly in tissue culture as shown by [

35

S]methionine pulse labeling using Western blots and electron microscopy autoradiography of deter- gent-extracted hepatocytes in culture where only the cytoskeleton remained [45]. In Western blots, the 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- ments. Cytokeratin 18 cDNA was used to transfect Mallory-body-forming cells in mouse liver tissue culture using a green fluorescent protein tag. The cytokeratin localized in both the normally distrib- uted cytokeratin but was also mainly concentrated in the Mallory body. This suggested that the nascent human cytokeratin went both to the normal fila- ments as well as directly to the Mallory body after synthesis [84, 85].

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)

(10)

32.18

Functional Consequence

of the Loss of Cytokeratin Organization Due to Aggresome (Mallory Body) Formation

The function of liver cell cytokeratins and the vul- nerability of liver cells exposed to toxins and viral damage have been reviewed elsewhere [52, 57, 76, 77]. Mutations in cytokeratin 8 and cytokeratin 18 were found in patients with cryptogenic cirrhosis, hepatitis C, and other liver diseases. The question is: do changes in the cytokeratin organization due to mutations or other etiologies interfere with liver function and increase vulnerability of the liver to in- jury? For instance, hyperphosphorylation of the liv- er cell cytokeratins changes their organization, but does this lead to loss of function [55]? Cytokeratin reorganizes extensively during mitoses, apoptosis and polarization of the cell [66]. In in vitro studies using transfection of cells with mutant cytokeratins, the cells were more vulnerable to injury. Cytokerat- ins have viscoelastic properties, which make cells resistant to physical stress. Lack of cytokeratins in K18/8-deficient mice hepatocytes disturbs the pro- gression of the cell cycle. These cells are more likely to develop apoptosis in response to FAS [31, 58, 65].

The cytokeratins of the liver anchor and support the bile canaliculus, forming the pericanalicular sheath supporting bile flow [47, 48]. Likewise the cytokera- tin filaments of the liver cell are involved in transhe- patic transport of vesicles [50].

The cytokeratin cytoskeleton of liver cells that form Mallory bodies is profoundly altered so that only the Mallory body stains for keratin, leaving the rest of the cell unstained, the so-called “empty cell”

or “ghost cell” [46]. Mallory body cells lose their bile canaliculi and cytokeratin in the cytoplasm. This is associated with cell enlargement and rounding. The cytokeratin network supports the nucleus, the nu- clear pores and the centrioles. The Mallory-body- containing cells in primary tissue culture were un- able to form bile canaliculi or secrete fluorescent diacetate, compared with controls [25].

32.19

Concluding Remarks

It can be concluded that the inhibition of the ubiq- uitin-proteasome pathway of protein degradation has profound effects on the structure and function of liver cells, including loss of cell shape and inter- nal organization of organelles, polarity, and the bile secretory apparatus. Vulnerability of the liver cell

to FAS-induced apoptosis is increased. Cytokeratin aggresome formation (Mallory bodies) may further reduce proteolysis by the proteasome.

References

1. Adams J. Development of the proteasome inhibitor PS-341.

Oncologist 2002;7:9–16.

2. Albano E, Clot P, Morimoto M et al. Role of cytochrome P4502E1-dependent formation of hydroxyethyl free radical in the development of liver damage in rats intragastrically fed with ethanol. Hepatology 1996;23:155–163.

3. Banner BF, Savas L, Zivny J et al. Ubiquitin as a marker of cell injury in non alcoholic steatohepatitis. Am J Clin Pathol 2000;114:860–866.

4. Bardag-Gorce F, Yuan QX, Li J et al. The effect of ethanol- induced cytochrome P450 2E1 on the inhibition of protea- some activity by alcohol. Biochem Biophys Res Commun 2000;279:23–29.

5. Bardag-Gorce F, French BA, Lue YH et al. Mallory bodies formed in proteasome-depleted hepatocytes: an immuno- histochemical study. Exp Molec Pathol 2001;70:7–18.

6. Bardag-Gorce F, French BA, Li J et al. The importance of cy- cling of blood alcohol levels in the pathogenesis of experi- mental alcoholic liver disease in rats fed ethanol intragastri- cally. Gastroenterology 2002;123:325–335.

7. Bardag-Gorce F, Li J, French BA, French SW. Ethanol with- drawal induced CYP2E1 degradation in vivo, blocked by pro- teasomal inhibitor PS-341. Free Radical Biol Med 2002;32:17–

21.

8. Bardag-Gorce F, Li J, French BA et al. Chronic alcohol-in- duced phosphorylation of proteasome subunits proteolysis of the proteasome. FASEB J 2002;16:A245.

9. Bardag-Gorce F, van Leeuwen FW, Nguyen V et al. The role of the ubiquitin-proteasome pathway in the formation of Mal- lory bodies. Exp Molec Pathol 2002;73:75–83.

10. Bardag-Gorce F, Li J, French BA, French SW. The effect of ethanol-induced CYP2E1 lipid peroxidation by product 4- hydroxynonenal on the 26S proteasome. FASEB J 2003;17:

A325.

11. Bardag-Gorce F, Riley N, Nguyen V et al. The mechanism of 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 2001;292:1552–1555.

13. Blankson H, Holen I, Seglen PO. Disruption of the cytokera- tin cytoskeleton and inhibition of hepatocyte autophage by okadaic acid. Exp Cell Res 1995;218:522–530.

14. Blouin R, Kawahara H, French SW, Marceau N. Selective ac- cumulation of IF proteins at a focal juxtanuclear site in COS-1 cells transfected with mouse keratin 18 cDNA. Exp Cell Res 1990;187:234–242.

(11)

15. Cadrin M, Anderson M, Aasheim LH et al. Modifications in cytokeratin and actin in cultured liver cells derived from gri- seofulvin-fed mice. Lab Invest 1995;72:453–460.

16. Chan V, Tobias JW, Bachmair A et al. A multiubiquitin chain is confined to specific lysone in a targeted short-lived protein.

Science 1989;243:1576–1584.

17. Ciechanover A, Brundin P. The ubiquitin proteasome sys- tem in neurodegenerative diseases: sometimes the chicken, sometimes the egg. Neuron 2003;40:427–446.

18. Connell P, Ballinger CA, Jiang J et al. The co-chaperone CHIP regulates protein triage decisions mediated by heat-shock proteins. Nature Cell Biol 2001;3:93–96.

19. Denk H, Bernklau G, Krepler R. Effect of griseofulvin treat- ment and neoplastic transformation on transglutaminase activity in mouse liver. Liver 1984;4:208–213.

20. De Vrij FM, Sluijs JA, Gregori L et al. Mutant ubiquitin ex- pressed in Alzheimer’s disease causes neuronal death.

FASEB J 2001;15:2680—2688.

21. Donohue Jr TM, Zetterman RK, Zhang-Gouillon Z-Q, French SW. Peptidase activities of the multicatalytic protease in rat liver after voluntary and intragastric ethanol administration.

Hepatology 1998;28:486–491.

22. Donohue TM. The ubiquitin-proteasome system and its role in ethanol-induced disorders. Addict Biol 2002;7(1):15–28 23. Fataccioli V, Andraud E, Gentil M et al. Effects of chronic

ethanol administration on rat liver proteasome activities: re- lationship with oxidative stress. Hepatology 1999;29:14–20.

24. French, BA, van Leeuwen F, Riley NE et al. Aggresome for- mation in liver cells in response to different toxic mecha- nisms. Role of the ubiquitin-proteasome pathway and the frameshift mutant of ubiquitin. Exp Molec Pathol 2001;71:241–246.

25. French SW, Kawahara H, Katsuma Y et al. Interaction of in- termediate filaments with nuclear lamina and cell periphery.

Electron Microsc Rev 1989;2:17–51.

26. French SW, Wong KW, Jui L et al. Effect of ethanol on cyto- chrome P450 2E1 (CYP2E1) lipid peroxidation and serum protein adduct formation in relation to liver pathology pathogenesis. Exp Molec Pathol 1993;58:61–75.

27. French SW, Nash J, Shitabata P et al. and the VA Coop- erative Study Group. Pathology of Alcoholic Liver Disease 1993;13:154–169.

28. French SW, Bardag-Gorce F. The ubiquitin-proteasome 26S pathway in liver cell protein turnover: effect of ethanol and drugs. Alcoholism: Clin Exp Res 2001;25:225s–229s.

29. Garcia-Mata R, Gao Y-S, Sztul E. Hassles with taking out the garbage: aggravating aggresomes. Traffic 2002;3:388–396.

30. Geetha T, Wooten MW. Structure and functional properties of the ubiquitin binding protein p62. FEBS Lett 2002;512:19–

24.

31. Gilbert S, Loranger A, Daigle N, Marceau N. Simple epithe- lium keratins 8 and 18 provide resistance to FAS-mediated apoptosis. The protection occurs through a receptor-target- ing modulation. J Cell Biol 2001;154:763–773.

32. Glickman MH, Ciechanover A. The ubiquitin-proteasome proteolytic pathway: destruction for the sake of construc- tion. Physiol Rev 2002;82:373–428.

33. Gordon C. The intracellular localization of the proteasome.

Curr Topics Microbiol Immunol 2002;268:175–184.

34. Gouillon ZQ, Miyamoto K, Donohue TM et al. Role of CYP2E1 in the pathogenesis of alcoholic liver disease: modifications by cAMP and ubiquitin-proteasome pathway. Frontiers Bio- sci 1999;4:16–25.

35. Hartl FU, Hayer-Hartl M. Molecular chaperones in the cytosol: from nascent chain to folded protein. Science 2002;295:1852–1858.

36. Hartmann-Petersen R, Seeger M, Gordon C. Transferring substrates to the 26S proteasome. Trends Biochem Sci 2003;28:26–31.

37. Hartmann-Petersen R, Semple CA, Ponting CP et al. UBA do- main containing proteins in fission yeast. Intern J Biochem Cell Biol 2003;35:629–636.

38. Hershko A, Ciechanover A. The ubiquitin system. Annu Rev Biochem 1998;67s:425–480.

39. Hirano K, Roth J, Zuber C, Ziak M. Expression of a mutant ER- retained polytope membrane protein in cultured rat hepa- tocytes results in Mallory body formation. Histochem Cell Biol 2002;117:41–53.

40. Hochstrasser M. Ubiquitin-dependent protein degradation.

Annu Rev Genet 1996;30:405–439.

41. Jensen K, Gluud C. The Mallory body: morphological, clini- cal and experimental studies (Part 1 of a literature survey).

Hepatology 1994;20:1061–1077.

42. Johnston JA, Ward CL, Kopito RR. Aggresomes: a cellular re- sponse to misfolded protein. J Cell Biol 1998;143:1883–1889.

43. Johnston JA, Illing ME, Kopito RR. Cytoplasmic dynein/dyn- actin mediates the assembly of aggresomes. Cell Motil Cy- toskeleton 2002;53:26–38.

44. Kachi K, Wong TT, French SW. Molecular structural changes in Mallory body proteins in human and mouse livers: an in- frared spectroscopy study. Exp Molec Pathol 1993;59:197–

210.

45. Kachi K, Cadrin M, French SW. Synthesis of Mallory body, in- termediate filament, and microfilament proteins in liver cell primary cultures. Lab Invest 1993;68:71–81.

46. Katsuma Y, Swierenga SH, Khettry U et al. Changes in the cytokeratin intermediate filament cytoskeleton associated with Mallory body formation in mouse and human liver.

Hepatology 1987;7:1215–1223.

47. Katsuma Y, Marceau N, Ohta M, French SW. Cytokeratin inter- mediated filaments of rat hepatocytes: different cytoskele- tal domains and their three dimensional structure. Hepatol- ogy 1988;8:559–568.

48. Kawahara H, Marceau N, French SW. Effect of agents which rearrange the cytoskeleton in vitro on the structure and function of hepatocyte canaliculi. Lab Invest 1989;60:692–

704.

49. Kawahara H, Cadrin M, French SW. Ethanol-induced phos- phorylation of cytokeratin in cultured hepatocytes. Life Sci 1990;47:859–863.

(12)

50. Kawahara H, Cadrin M, Perry L et al. Role of the cytokeratin intermediate filaments in transhepatic transport and canal- icular secretion. Hepatology 1990;11:435–448.

51. Kenner L, el Shabrawi Y, Hutter H et al. Expression of three- and four-repeat tau isoforms in mouse liver. Hepatology 1994;20:1086–1089.

52. Kirfel J, Magin TM, Reichelt J. Keratins: a structural scaffold with emerging functions. Cell Mol Life Sci 2003;60:56–71.

53. Koepp DM, Harper JW, Elledge SJ. How the cyclin be- came a cyclin: regulated proteolysis in the cell cycle. Cell 1999;97:431–434.

54. Kopito RR, Ron D. Conformational disease. Nat Cell Biol 2000;2:E207–E209.

55. Ku NO, Michie SA, Soetikno RM et al. Susceptibility to hepato- toxicity in transgenic mice that express dominant-negative human keratin 18 mutant. J Clin Invest 1996;98:1034–1046.

56. Ku NO, Omary MB. Keratins turn over by ubiquitina- tion in a phosphorylation-modulated fashion. J Cell Biol 2000;149:547–552.

57. Ku NO, Gish R, Wright TL, Omary MB. Keratin 8 mutations in patients with cryptogenic liver disease. N Engl J Med 2001;344:1580–1587.

58. Ku NO, Soetikno RM, Omary ME. Keratin mutation in trans- genic mice predisposes Fas but not TNF-induced apoptosis and massive liver injury. Hepatology 2003;37:1006–1014.

59. Kuusisto E, Salminen A, Alafuzoff I. Ubiquitin-binding pro- tein p62 is present in neuronal and glial inclusions in hu- man tauopathies and synucleinopathies. Neuroreport 2001;12:2085–2090.

60. Kuusisto E, Salminen A, Alafuzoff I. Early accumulation of p62 in neurofibrillary tangles in Alzheimer’s disease: pos- sible role in tangle formation. Neuropathol Appl Neurobiol 2002;28:228–237.

61. Lam YA, Pickart CM, Alban A et al. Inhibition of the ubiquitin- proteasome system in Alzheimer’s disease. Proc Natl Acad Sci USA 2000;97:9902–9906.

62. Laney JD, Hochstrasser M. Substrate targeting in the ubiqui- tin system. Cell 1999;97:427–430.

63. Lang-Rollin I, Rideout H, Stefanis L. Ubiquitinated inclusions and neuronal cell death. Histol Histopathol 2003;18:509–

517.

64. Lowe J, Blanchard A, Morrell K et al. Ubiquitin is a common factor in intermediate filament inclusion bodies of diverse type in man, including those of Parkinson’s disease, Pick’s disease, and Alzheimer’s disease as well as Rosenthal fib- ers in cerebellar astrocytomas, cytoplasmic bodies in mus- cle, and Mallory bodies in alcoholic liver disease. J Pathol 1988;155:9–15.

65. Marceau N, Loranger A, Gilbert S et al. Keratin-medi- ated resistance to stress and apoptosis in simple epithe- lial cells in relation to health and disease. Biochem Cell Biol 2001;79:543–555.

66. Marceau N. Keratin 8 mutations in patients with cryptogenic liver disease. J Hepatol 2002;36:710–711.

67. Mayer JR. The meteoric rise of regulated intracellular prote- olysis. Nature Rev: Molec Cell Biol 2000;1:145–148.

68. McPhaul LW, Wang J, Hol EM et al. Molecular misreading of the ubiquitin B gene and hepatic Mallory body formation.

Gastroenterology 2002;122:1878–1885.

69. Molina PE, Hoek JB, Nelson S et al. Mechanisms of alcohol-in- duced tissue injury. Alcohol Clin Exp Res 2003;27:563–575.

70. Montgomery R, French SW. Alcohol-induced liver hypoxia in rats: role of HIF-1 induction. FASEB J 2003;17:A1283.

71. Muratani M, Tansey WP. How the ubiquitin-proteasome sys- tem controls transcription. Nat Rev Mol Cell Biol 2003;4:192–

201.

72. Nagao Y, Yuan Q-X, Wan Y-JY et al. Pathogenesis of Mal- lory body formation. Studies using the drug-primed mouse model. Hepatol Res 1998;13:15–24.

73. Nakamichi I, Hatakeyama S, Nakayama KI. Formation of Mal- lory body-like inclusions and cell death induced by deregu- lated expression of keratin 18. Mol Biol Cell 2002;13:3441–

3451.

74. Naujokat C, Hoffmann S. Role and function of the 26S proteasome in proliferation and apoptosis. Lab Invest 2002;82:965–980.

75. Ohta M, Marceau N, Perry G et al. Ubiquitin is present on the cytokeratin intermediate filaments and Mallory bodies of hepatocytes. Lab Invest 1988;58:848–856.

76. Omary MB, Ku NO. Intermediate filament proteins of the liv- er: emerging disease association and functions. Hepatology 1997;25:1043–1048.

77. Omary MB, Ku NO, Toivola DM. Keratins: guardians of the liver. Hepatology 2002;35:251–257.

78. Osborn M, Weber K. Keratins, transglutaminase, and Mallory bodies. The really insoluble stuff. Lab Invest 1989;61:585–

587.

79. Perez MJ, Cederbaum AI. Proteasome inhibition potenti- ates CYP2E1-mediated toxicity in HepG2 cells. Hepatology 2003;37(6):1395–1404.

80. Pickart CM. Ubiquitin in chains. Trends Biochem Sci 2000;25:

544–548.

81. Pickart CM. Mechanisms underlying ubiquitination. Annu Rev Biochem 2001;70:503–533.

82. Puls A, Schmidt S, Grawe F, Stabel S. Interaction of protein kinase C with ZIP, a novel protein kinase C-binding protein.

Proc Natl Acad Sci USA 1997;94:6191–6196.

83. Raasi S, Pickart CM. Rad23 ubiquitin-associated domains (UBA) inhibit 26S proteasome-catalyzed proteolysis by sequestering lysine 48-linked polyubiquitin chains. J Biol Chem 2003;278:8951–8959.

84. Riley NE, Li J, Worrall S et al. Mallory body as an aggresome:

in vitro studies. Exp Molec Pathol 2002;72:17–23.

85. Riley NE, Bardag-Gorce F, French SW. Proteasome inhibi- tor PS-341 causes aggresome formation, and loss of cell cohesion in a mouse hepatoma cell line. Molec Biol Cell 2002;13s:265A.

86. Riley NE, Li J, McPhaul LW et al. Heat shock proteins are present in Mallory bodies (cytokeratin aggresomes) in hu- man liver biopsy specimens. Exp Molec Pathol 2003;74:168–

172.

(13)

87. Riley NE, Bardag-Gorce F, Montgomery RO et al. Microtu- bules are required for cytokeratin aggresome formation in hepatocytes (Mallory bodies): an in vitro study. Exp Molec Pathol 2003;74:173–179.

88. Rouach H, Fataccioli V, Gentil M et al. Effect of chronic etha- nol feeding on lipid peroxidation and protein oxidation in relation to liver pathology. Hepatology 1997;25(2):351–355.

89. Seki S, Kitada T, Sakaguchi H et al. Pathological significance of oxidative cellular damage in human alcoholic liver dis- ease. Histopathology 2003;42:365–371.

90. Shimada M, Hashimoto E, Kaneda H et al. Nonalcoholic steatohepatitis: risk factors for liver fibrosis. Hepatol Res 2002;24(4):429–438.

91. Shringarpure R, Grune T, Mehlhase J, Davies KJ. Ubiquitin conjugation is not required for the degradation of oxidized proteins by proteasome. J Biol Chem 2003;278:311–318.

92. Strnad P, Windoffer R, Leube RE. In vivo detection of cytok- eratin filament network breakdown in cells treated with the phosphatase inhibitor okadaic acid. Cell Tissue Res 2001;306:277–293.

93. Stumptner C, Heid H, Fuchsbichler A et al. Analysis of intra- cytoplasmic hyaline bodies in a hepatocellular carcinoma.

Am J Pathol 1999;154:1701–1710.

94. Stumptner C, Omary MB, Fickert P et al. Hepatocyte cytok- eratins are hyperphosphorylated at multiple sites in human alcoholic hepatitis and in a Mallory body mouse model. Am J Pathol 2001;156:77–90.

95. Stumptner C, Fuchsbichler M, Lehner M et al. Sequence of events in the assembly of Mallory body components in mouse liver: clues to the pathogenesis and significance of Mallory body formation. J Hepatol 2001;34:665–675.

96. Stumptner C, Fuchsbichler A, Heid H et al. Mallory body:

a disease-associated type of sequestosome. Hepatology 2002;35:1035–1062.

97. Szweda PA, Friguet B, Szweda LI. Proteolysis, free radicals, and aging. Free Radical Biol Med 2002;33:29–36.

98. Takagi M, Yamauchi M, Toda G et al. Serum ubiquitin levels in patients with alcoholic liver disease. Alcohol Clin Exp Res 1999;23:76s–80s.

99. Takagi M, Yamauchi M, Takada K, Ohkawa K. Serum ubiqui- tin-protein conjugates in normal subjects and patients with

alcoholic liver diseases: immunoaffinity isolation and elec- trophoretic mobility. Alcohol Clin Exp Res 2002;26:1692–

1696.

100. Thrower JS, Hoffman L, Rechsteiner M, Pickart CM. Rec- ognition of the polyubiquitin proteolytic signal. EMBO J 2000;19:94–104.

101. Ulrich HD. Natural substrates of the proteasome and their recognition by the ubiquitin system. Curr Topics Microbiol Immunol 2002;268:137–174.

102. Vadlamudi RK, Joung I, Strominger JL, Shin J. P62, a phos- photyrosine-independent ligand of the SH2 domain of 56lck, belongs to a new class of ubiquitin-binding proteins.

J Biol Chem 1996;271:20235–20237.

103. van Leeuwen FW, de Kleijn DP, van den Hurk HH et al.

Frameshift mutants of β-amyloid precursor protein and ubiquitin-B in Alzheimer's down patients. Science 1998;279:242–247.

104. Wigley WC, Fabunmi RP, Lee MG et al. Dynamic association of proteasomal machinery with the centrosome. J Cell Biol 1999;145:481–490.

105. Wilkinson KD. Ubiquitination and deubiquitination: target- ing of proteins for degradation by the proteasome. Semin Cell Develop Biol 2000;11:141–148.

106. Wojcik C, Shroeter D, Wilk S et al. Ubiquitin-mediated prote- olysis centers in HeLa cells: indication from studies of an in- hibitor of the chymotrypsin-like activity of the proteasome.

Eur J Cell Biol 1996;7:311–318.

107. Wojcik C. Regulation of apoptosis by the ubiquitin and pro- teasome pathway. J Cell Mol Med 2002;6:25–48.

108. Yuan QX, Marceau N, French BA, French SW. Heat shock in vivo induces Mallory body formation in drug primed mouse liver. Exp Molec Pathol 1995;63:63–76.

109. Yuan QX, Nagao Y, Gaal K et al. Mechanisms of Mallory body formation induced by Okadaic acid in drug-primed mice.

Exp Molec Pathol 1998;65:87–103.

110. Zatloukal K, Denk H, Lackinger E, Rainer I. Hepatocellular cytokeratins as substrates of transglutaminases. Lab Invest 1989;61:603–608.

111. Zatloukal K, Fesus L, Denk H et al. High amount of Σ-(γ- glutamyl)lysine cross-links in Mallory bodies. Lab Invest 1992;66:774–777.

Riferimenti

Documenti correlati

The parent hull of the series, C1 model, was derived from a pre- existing model, C954, that had shown good performance, registered by an intensive experimental program in a towing

Our data show that primary fibroblasts har- vested from sensitized mice displayed an increased proliferation rate ( Fig. 5 A) as well as increased expression of ␣-SMA ( Fig. 5 B and

- the potential alternative responses (A i ) to population growth, assumed to moderate the effects of natural growth on international migration. We consider four such

Accordingly, platelet adhesion to liver sinusoids occurred less efficiently in CD44 / mice than wild-type (WT) controls ( Fig- ure S2 C; note that CD44 / mice have normal blood

Analogie e differenze tra Bulimia Nervosa e Binge Eating

In these three years, I started my research activities on topics related to Software Engineering, Cloud.. Computing,

Sublobar resection versus lobectomy for stage I non-small cell lung cancer: an appropriate choice in elderly patients..